1
|
Stratigi A, Soler-García M, Krout M, Shukla S, De Bono M, Richmond JE, Laurent P. Neuroendocrine Control of Synaptic Transmission by PHAC-1 in C. elegans. J Neurosci 2025; 45:e1767232024. [PMID: 39919830 PMCID: PMC11949478 DOI: 10.1523/jneurosci.1767-23.2024] [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: 09/15/2023] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 02/09/2025] Open
Abstract
A dynamic interplay between fast synaptic signals and slower neuromodulatory signals controls the excitatory/inhibitory (E/I) balance within neuronal circuits. The mechanisms by which neuropeptide signaling is regulated to maintain E/I balance remain uncertain. We designed a genetic screen to isolate genes involved in the peptidergic maintenance of the E/I balance in the C. elegans motor circuit. This screen identified the C. elegans orthologs of the presynaptic phosphoprotein synapsin (snn-1) and the protein phosphatase 1 (PP1) regulatory subunit PHACTR1 (phac-1). We demonstrate that both phac-1 and snn-1 alter the motor behavior of C. elegans, and genetic interactions suggest that SNN-1 contributes to PP1-PHAC-1 holoenzyme signaling. De novo variants of human PHACTR1, associated with early-onset epilepsies [developmental and epileptic encephalopathy 70 (DEE70)], when expressed in C. elegans resulted in constitutive PP1-PHAC-1 holoenzyme activity. Unregulated PP1-PHAC-1 signaling alters the synapsin and actin cytoskeleton and increases neuropeptide release by cholinergic motor neurons, which secondarily affects the presynaptic vesicle cycle. Together, these results clarify the dominant mechanisms of action of the DEE70 alleles and suggest that altered neuropeptide release may alter E/I balance in DEE70.
Collapse
Affiliation(s)
- Aikaterini Stratigi
- Laboratory of Neurophysiology, ULB Institute for Neuroscience, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Miguel Soler-García
- Laboratory of Neurophysiology, ULB Institute for Neuroscience, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Mia Krout
- Department of Biological Sciences, University of Illinois Chicago, Chicago, Illinois 60607
| | - Shikha Shukla
- Laboratory of Neurophysiology, ULB Institute for Neuroscience, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Mario De Bono
- Institute of Science and Technology, Klosterneuburg 3400, Austria
| | - Janet E Richmond
- Department of Biological Sciences, University of Illinois Chicago, Chicago, Illinois 60607
| | - Patrick Laurent
- Laboratory of Neurophysiology, ULB Institute for Neuroscience, Université Libre de Bruxelles, Brussels 1070, Belgium
| |
Collapse
|
2
|
Ihara D, Oishi R, Kasahara S, Yamamoto A, Kaito M, Tabuchi A. The BDNF-ERK/MAPK axis reduces phosphatase and actin regulator1, 2 and 3 (PHACTR1, 2 and 3) mRNA expressions in cortical neurons. Drug Discov Ther 2024; 18:255-259. [PMID: 39183043 DOI: 10.5582/ddt.2024.01048] [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] [Indexed: 08/27/2024]
Abstract
Actin rearrangement and phosphorylation-dephosphorylation in the nervous system contribute to plastic alteration of neuronal structure and function. Phosphatase and actin regulator (PHACTR) family members are actin- and protein phosphatase 1 (PP1)-binding proteins. Because some family members act as regulators of neuronal morphology, studying the regulatory mechanisms of PHACTR is valuable for understanding the basis of neuronal circuit formation. Although expression patterns of PHACTR family molecules (PHACTR1-4) vary across distinct brain areas, little is known about the extracellular ligands that influence their mRNA levels. In this study, we focused on an important neurotrophin, brain-derived neurotrophic factor (BDNF), and examined its effect on mRNA expression of PHACTR family member in cortical neurons. PHACTR1-3, but not PHACTR4, were affected by stimulation of primary cultured cortical neurons with BDNF; namely, sustained downregulation of their mRNA levels was observed. The observed downregulation was blocked by an inhibitor of the extracellular signal-regulated protein kinase/mitogen-activated protein kinase (ERK/MAPK) pathway, U0126, suggesting that ERK/MAPK plays an inhibitory role for gene induction of PHACTR1-3. These findings aid the elucidation of how BDNF regulates actin- and PP1-related neuronal functions.
Collapse
Affiliation(s)
- Daisuke Ihara
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Ryotaro Oishi
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Shiho Kasahara
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Aimi Yamamoto
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Maki Kaito
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Akiko Tabuchi
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| |
Collapse
|
3
|
Xu Z, Sadleir L, Goel H, Jiao X, Niu Y, Zhou Z, de Valles-Ibáñez G, Poke G, Hildebrand M, Lieffering N, Qin J, Yang Z. Genotype and phenotype correlation of PHACTR1-related neurological disorders. J Med Genet 2024; 61:536-542. [PMID: 38272663 DOI: 10.1136/jmg-2023-109638] [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: 09/14/2023] [Accepted: 01/16/2024] [Indexed: 01/27/2024]
Abstract
BACKGROUND PHACTR1 (phosphatase and actin regulators) plays a key role in cortical migration and synaptic activity by binding and regulating G-actin and PPP1CA. This study aimed to expand the genotype and phenotype of patients with de novo variants in PHACTR1 and analyse the impact of variants on protein-protein interaction. METHODS We identified seven patients with PHACTR1 variants by trio-based whole-exome sequencing. Additional two subjects were ascertained from two centres through GeneMatcher. The genotype-phenotype correlation was determined, and AlphaFold-Multimer was used to predict protein-protein interactions and interfaces. RESULTS Eight individuals carried missense variants and one had CNV in the PHACTR1. Infantile epileptic spasms syndrome (IESS) was the unifying phenotype in eight patients with missense variants of PHACTR1. They could present with other types of seizures and often exhibit drug-resistant epilepsy with a poor prognosis. One patient with CNV displayed a developmental encephalopathy phenotype. Using AlphaFold-Multimer, our findings indicate that PHACTR1 and G-actin-binding sequences overlap with PPP1CA at the RPEL3 domain, which suggests possible competition between PPP1CA and G-actin for binding to PHACTR1 through a similar polymerisation interface. In addition, patients carrying missense variants located at the PHACTR1-PPP1CA or PHACTR1-G-actin interfaces consistently exhibit the IESS phenotype. These missense variants are mostly concentrated in the overlapping sequence (RPEL3 domain). CONCLUSIONS Patients with variants in PHACTR1 can have a phenotype of developmental encephalopathy in addition to IESS. Moreover, our study confirmed that the variants affect the binding of PHACTR1 to G-actin or PPP1CA, resulting in neurological disorders in patients.
Collapse
Affiliation(s)
- Zhao Xu
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
- Epilepsy Center, Peking University People's Hospital, Beijing, China
| | - Lynette Sadleir
- Department of Paediatrics and Child Health, University of Otago Wellington, Wellington, New Zealand
| | - Himanshu Goel
- Hunter Genetics, Waratah, New South Wales, Australia
| | - Xianru Jiao
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
- Epilepsy Center, Peking University People's Hospital, Beijing, China
| | - Yue Niu
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
- Epilepsy Center, Peking University People's Hospital, Beijing, China
| | - Zongpu Zhou
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
- Epilepsy Center, Peking University People's Hospital, Beijing, China
| | - Guillem de Valles-Ibáñez
- Department of Paediatrics and Child Health, University of Otago Wellington, Wellington, New Zealand
| | - Gemma Poke
- Department of Paediatrics and Child Health, University of Otago Wellington, Wellington, New Zealand
| | - Michael Hildebrand
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Heidelberg, Victoria, Australia
- Neuroscience Research Group, Murdoch Children's Research Institute, Royal Children's Hospital, South Brisbane, Queensland, Australia
| | - Nico Lieffering
- Department of Paediatrics and Child Health, University of Otago Wellington, Wellington, New Zealand
| | - Jiong Qin
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
- Epilepsy Center, Peking University People's Hospital, Beijing, China
| | - Zhixian Yang
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
- Epilepsy Center, Peking University People's Hospital, Beijing, China
| |
Collapse
|
4
|
Chowdhury MM, Zimmerman S, Leeson H, Nefzger CM, Mar JC, Laslett A, Polo JM, Wolvetang E, Cooper-White JJ. Superior Induced Pluripotent Stem Cell Generation through Phactr3-Driven Mechanomodulation of Both Early and Late Phases of Cell Reprogramming. Biomater Res 2024; 28:0025. [PMID: 38774128 PMCID: PMC11106629 DOI: 10.34133/bmr.0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/25/2024] [Indexed: 05/24/2024] Open
Abstract
Human cell reprogramming traditionally involves time-intensive, multistage, costly tissue culture polystyrene-based cell culture practices that ultimately produce low numbers of reprogrammed cells of variable quality. Previous studies have shown that very soft 2- and 3-dimensional hydrogel substrates/matrices (of stiffnesses ≤ 1 kPa) can drive ~2× improvements in human cell reprogramming outcomes. Unfortunately, these similarly complex multistage protocols lack intrinsic scalability, and, furthermore, the associated underlying molecular mechanisms remain to be fully elucidated, limiting the potential to further maximize reprogramming outcomes. In screening the largest range of polyacrylamide (pAAm) hydrogels of varying stiffness to date (1 kPa to 1.3 MPa), we have found that a medium stiffness gel (~100 kPa) increased the overall number of reprogrammed cells by up to 10-fold (10×), accelerated reprogramming kinetics, improved both early and late phases of reprogramming, and produced induced pluripotent stem cells (iPSCs) having more naïve characteristics and lower remnant transgene expression, compared to the gold standard tissue culture polystyrene practice. Functionalization of these pAAm hydrogels with poly-l-dopamine enabled, for the first-time, continuous, single-step reprogramming of fibroblasts to iPSCs on hydrogel substrates (noting that even the tissue culture polystyrene practice is a 2-stage process). Comparative RNA sequencing analyses coupled with experimental validation revealed that a novel reprogramming regulator, protein phosphatase and actin regulator 3, up-regulated under the gel condition at a very early time point, was responsible for the observed enhanced reprogramming outcomes. This study provides a novel culture protocol and substrate for continuous hydrogel-based cell reprogramming and previously unattained clarity of the underlying mechanisms via which substrate stiffness modulates reprogramming kinetics and iPSC quality outcomes.
Collapse
Affiliation(s)
- Mohammad Mahfuz Chowdhury
- Australian Institute of Bioengineering and Nanotechnology (AIBN),
The University of Queensland, St. Lucia, QLD 4072, Australia
| | | | - Hannah Leeson
- Australian Institute of Bioengineering and Nanotechnology (AIBN),
The University of Queensland, St. Lucia, QLD 4072, Australia
| | | | - Jessica Cara Mar
- Australian Institute of Bioengineering and Nanotechnology (AIBN),
The University of Queensland, St. Lucia, QLD 4072, Australia
- Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Andrew Laslett
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Jose Maria Polo
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute and the Australian Regenerative Medicine Institute,
Monash University, Clayton, VIC 3800, Australia
- Adelaide Centre for Epigenetics and the South Australian Immunogenomics Cancer Institute, Faculty of Health and Medical Sciences,
The University of Adelaide, Adelaide, SA 5005, Australia
| | - Ernst Wolvetang
- Australian Institute of Bioengineering and Nanotechnology (AIBN),
The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Justin John Cooper-White
- Australian Institute of Bioengineering and Nanotechnology (AIBN),
The University of Queensland, St. Lucia, QLD 4072, Australia
- School of Chemical Engineering, Andrew N. Liveris Building,
The University of Queensland, St. Lucia, QLD 4072, Australia
| |
Collapse
|
5
|
Zang L, Song Y, Tian Y, Hu N. PHACTR1 promotes the mobility of papillary thyroid carcinoma cells by inducing F-actin formation. Heliyon 2023; 9:e20461. [PMID: 37876444 PMCID: PMC10590795 DOI: 10.1016/j.heliyon.2023.e20461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 09/13/2023] [Accepted: 09/26/2023] [Indexed: 10/26/2023] Open
Abstract
Papillary thyroid carcinoma (PTC) limits effective biomarkers for predicting prognosis and targeted therapy. Phosphatase and actin regulator 1 (PHACTR1) is a mobility-promoting molecule due to its regulation on F-actin formation, which is valuable for the investigation of PTC. Our study aimed to investigate the relationship between PHACTR1 and PTC carcinogenesis, especially mobility. Our results displayed that PHACTR1 expression was elevated in metastatic or larger PTC tissues. In addition, PTC cells K1 with more obvious mobility had higher PHACTR1 expression whereas weakly mobile cells TPC-1 was contrary. Moreover, PHACTR1 silencing inhibited the invasion, migration and tumorigenicity of K1 cells, while PHACTR1 overexpression promoted the invasion, migration and tumorigenicity in TPC-1 cells. Furthermore, PHACTR1 overexpression increased the fluorescent intensity of F-actin in TPC-1 cells. Importantly, the enhanced invasion and migration in TPC-1 cells caused by PHACTR1 overexpression were significantly reversed by the disruption of F-actin assembly with swinholide A. In conclusion, PHACTR1 can promote the mobility of PTC cells, which results in the carcinogenesis of PTC. PHACTR1-regulated F-actin formation determines the mobility of PTC cells. Therefore, PHACTR1 can function as a potential biomarker for predicting prognosis and targeting therapy in PTC.
Collapse
Affiliation(s)
- Leilei Zang
- Department 5 of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050005, Hebei, China
| | - Yanmei Song
- Department of Infection Management/Public Health, Hebei People's Hospital, Shijiazhuang, 050057, Hebei, China
| | - Yanhua Tian
- Department 2 of Oncology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050005, Hebei, China
| | - Ning Hu
- Department 4 of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050005, Hebei, China
| |
Collapse
|
6
|
Jiang D, Liu H, Zhu G, Li X, Fan L, Zhao F, Xu C, Wang S, Rose Y, Rhen J, Yu Z, Yin Y, Gu Y, Xu X, Fisher EA, Ge J, Xu Y, Pang J. Endothelial PHACTR1 Promotes Endothelial Activation and Atherosclerosis by Repressing PPARγ Activity Under Disturbed Flow in Mice. Arterioscler Thromb Vasc Biol 2023; 43:e303-e322. [PMID: 37199156 PMCID: PMC10524336 DOI: 10.1161/atvbaha.122.318173] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 05/02/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND Numerous genome-wide association studies revealed that SNPs (single nucleotide polymorphisms) at the PHACTR1 (phosphatase and actin regulator 1) locus strongly correlate with coronary artery disease. However, the biological function of PHACTR1 remains poorly understood. Here, we identified the proatherosclerotic effect of endothelial PHACTR1, contrary to macrophage PHACTR1. METHODS We generated global (Phactr1-/-) and endothelial cell (EC)-specific (Phactr1ECKO) Phactr1 KO (knockout) mice and crossed these mice with apolipoprotein E-deficient (Apoe-/-) mice. Atherosclerosis was induced by feeding the high-fat/high-cholesterol diet for 12 weeks or partially ligating carotid arteries combined with a 2-week high-fat/high-cholesterol diet. PHACTR1 localization was identified by immunostaining of overexpressed PHACTR1 in human umbilical vein ECs exposed to different types of flow. The molecular function of endothelial PHACTR1 was explored by RNA sequencing using EC-enriched mRNA from global or EC-specific Phactr1 KO mice. Endothelial activation was evaluated in human umbilical vein ECs transfected with siRNA targeting PHACTR1 and in Phactr1ECKO mice after partial carotid ligation. RESULTS Global or EC-specific Phactr1 deficiency significantly inhibited atherosclerosis in regions of disturbed flow. PHACTR1 was enriched in ECs and located in the nucleus of disturbed flow areas but shuttled to cytoplasm under laminar flow in vitro. RNA sequencing showed that endothelial Phactr1 depletion affected vascular function, and PPARγ (peroxisome proliferator-activated receptor gamma) was the top transcription factor regulating differentially expressed genes. PHACTR1 functioned as a PPARγ transcriptional corepressor by binding to PPARγ through the corepressor motifs. PPARγ activation protects against atherosclerosis by inhibiting endothelial activation. Consistently, PHACTR1 deficiency remarkably reduced endothelial activation induced by disturbed flow in vivo and in vitro. PPARγ antagonist GW9662 abolished the protective effects of Phactr1 KO on EC activation and atherosclerosis in vivo. CONCLUSIONS Our results identified endothelial PHACTR1 as a novel PPARγ corepressor to promote atherosclerosis in disturbed flow regions. Endothelial PHACTR1 is a potential therapeutic target for atherosclerosis treatment.
Collapse
Affiliation(s)
- Dongyang Jiang
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China (D. J., H. L., G. Z., X. L., L. F., F. Z., C. X., Z. Y., Y. Y., J. G., Y. X.)
| | - Hao Liu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China (D. J., H. L., G. Z., X. L., L. F., F. Z., C. X., Z. Y., Y. Y., J. G., Y. X.)
| | - Guofu Zhu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China (D. J., H. L., G. Z., X. L., L. F., F. Z., C. X., Z. Y., Y. Y., J. G., Y. X.)
| | - Xiankai Li
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China (D. J., H. L., G. Z., X. L., L. F., F. Z., C. X., Z. Y., Y. Y., J. G., Y. X.)
| | - Linlin Fan
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China (D. J., H. L., G. Z., X. L., L. F., F. Z., C. X., Z. Y., Y. Y., J. G., Y. X.)
| | - Faxue Zhao
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China (D. J., H. L., G. Z., X. L., L. F., F. Z., C. X., Z. Y., Y. Y., J. G., Y. X.)
| | - Chong Xu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China (D. J., H. L., G. Z., X. L., L. F., F. Z., C. X., Z. Y., Y. Y., J. G., Y. X.)
| | - Shumin Wang
- Aab Cardiovascular Research Institute, Department of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA (S. W., Y. R., J. R., X. X., J. P.)
| | - Yara Rose
- Aab Cardiovascular Research Institute, Department of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA (S. W., Y. R., J. R., X. X., J. P.)
| | - Jordan Rhen
- Aab Cardiovascular Research Institute, Department of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA (S. W., Y. R., J. R., X. X., J. P.)
| | - Ze Yu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China (D. J., H. L., G. Z., X. L., L. F., F. Z., C. X., Z. Y., Y. Y., J. G., Y. X.)
| | - Yiheng Yin
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China (D. J., H. L., G. Z., X. L., L. F., F. Z., C. X., Z. Y., Y. Y., J. G., Y. X.)
| | - Yuling Gu
- Shanghai Naturethink Life Science&Technology Co., Itd, Shanghai 201809, China (Y. G.)
| | - Xiangbin Xu
- Aab Cardiovascular Research Institute, Department of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA (S. W., Y. R., J. R., X. X., J. P.)
| | - Edward A. Fisher
- Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA (E. A. F.)
| | - Junbo Ge
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China (D. J., H. L., G. Z., X. L., L. F., F. Z., C. X., Z. Y., Y. Y., J. G., Y. X.)
| | - Yawei Xu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China (D. J., H. L., G. Z., X. L., L. F., F. Z., C. X., Z. Y., Y. Y., J. G., Y. X.)
| | - Jinjiang Pang
- Aab Cardiovascular Research Institute, Department of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA (S. W., Y. R., J. R., X. X., J. P.)
| |
Collapse
|
7
|
Rezvan A. PHACTR1 and Atherosclerosis: It's Complicated. Arterioscler Thromb Vasc Biol 2023; 43:1409-1411. [PMID: 37317846 PMCID: PMC10527601 DOI: 10.1161/atvbaha.123.319545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Affiliation(s)
- Amir Rezvan
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
8
|
Wood A, Antonopoulos A, Chuaiphichai S, Kyriakou T, Diaz R, Al Hussaini A, Marsh AM, Sian M, Meisuria M, McCann G, Rashbrook VS, Drydale E, Draycott S, Polkinghorne MD, Akoumianakis I, Antoniades C, Watkins H, Channon KM, Adlam D, Douglas G. PHACTR1 modulates vascular compliance but not endothelial function: a translational study. Cardiovasc Res 2023; 119:599-610. [PMID: 35653516 PMCID: PMC10064844 DOI: 10.1093/cvr/cvac092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/09/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS The non-coding locus at 6p24 located in Intron 3 of PHACTR1 has consistently been implicated as a risk allele in myocardial infarction and multiple other vascular diseases. Recent murine studies have identified a role for Phactr1 in the development of atherosclerosis. However, the role of PHACTR1 in vascular tone and in vivo vascular remodelling has yet to be established. The aim of this study was to investigate the role of PHACTR1 in vascular function. METHODS AND RESULTS Prospectively recruited coronary artery disease (CAD) patients undergoing bypass surgery and retrospectively recruited spontaneous coronary artery dissection (SCAD) patients and matched healthy volunteers were genotyped at the PHACTR1 rs9349379 locus. We observed a significant association between the PHACTR1 loci and changes in distensibility in both the ascending aorta (AA = 0.0053 ± 0.0004, AG = 0.0041 ± 0.003, GG = 0.0034 ± 0.0009, P < 0.05, n = 58, 54, and 7, respectively) and carotid artery (AA = 12.83 ± 0.51, AG = 11.14 ± 0.38, GG = 11.69 ± 0.66, P < 0.05, n = 70, 65, and 18, respectively). This association was not observed in the descending aorta or in SCAD patients. In contrast, the PHACTR1 locus was not associated with changes in endothelial cell function with no association between the rs9349379 locus and in vivo or ex vivo vascular function observed in CAD patients. This finding was confirmed in our murine model where the loss of Phactr1 on the pro-atherosclerosis ApoE-/- background did not alter ex vivo vascular function. CONCLUSION In conclusion, we have shown a role for PHACTR1 in arterial compliance across multiple vascular beds. Our study suggests that PHACTR1 has a key structural role within the vasculature.
Collapse
Affiliation(s)
- Alice Wood
- Department of Cardiovascular Sciences, Glenfield Hospital, Leicester, UK
- National Institute for Health Research (NIHR) Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Alexios Antonopoulos
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Surawee Chuaiphichai
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Theodosios Kyriakou
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Rebeca Diaz
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Abtehale Al Hussaini
- Department of Cardiovascular Sciences, Glenfield Hospital, Leicester, UK
- National Institute for Health Research (NIHR) Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Anna-Marie Marsh
- Department of Cardiovascular Sciences, Glenfield Hospital, Leicester, UK
- National Institute for Health Research (NIHR) Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Manjit Sian
- Department of Cardiovascular Sciences, Glenfield Hospital, Leicester, UK
- National Institute for Health Research (NIHR) Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Mitul Meisuria
- Department of Cardiovascular Sciences, Glenfield Hospital, Leicester, UK
- National Institute for Health Research (NIHR) Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Gerry McCann
- Department of Cardiovascular Sciences, Glenfield Hospital, Leicester, UK
- National Institute for Health Research (NIHR) Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Victoria S Rashbrook
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Edward Drydale
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Sally Draycott
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Murray David Polkinghorne
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Ioannis Akoumianakis
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Charalambos Antoniades
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Hugh Watkins
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Keith M Channon
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - David Adlam
- Department of Cardiovascular Sciences, Glenfield Hospital, Leicester, UK
- National Institute for Health Research (NIHR) Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Gillian Douglas
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| |
Collapse
|
9
|
Ma Y, Kemp SS, Yang X, Wu MH, Yuan SY. Cellular mechanisms underlying the impairment of macrophage efferocytosis. Immunol Lett 2023; 254:41-53. [PMID: 36740099 PMCID: PMC9992097 DOI: 10.1016/j.imlet.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/23/2023] [Accepted: 02/02/2023] [Indexed: 02/05/2023]
Abstract
The phagocytosis and clearance of dying cells by macrophages, a process termed efferocytosis, is essential for both maintaining homeostasis and promoting tissue repair after infection or sterile injury. If not removed in a timely manner, uncleared cells can undergo secondary necrosis, and necrotic cells lose membrane integrity, release toxic intracellular components, and potentially induce inflammation or autoimmune diseases. Efferocytosis also initiates the repair process by producing a wide range of pro-reparative factors. Accumulating evidence has revealed that macrophage efferocytosis defects are involved in the development and progression of a variety of inflammatory and autoimmune diseases. The underlying mechanisms of efferocytosis impairment are complex, disease-dependent, and incompletely understood. In this review, we will first summarize the current knowledge about the normal signaling and metabolic processes of macrophage efferocytosis and its importance in maintaining tissue homeostasis and repair. We then will focus on analyzing the molecular and cellular mechanisms underlying efferocytotic abnormality (impairment) in disease or injury conditions. Next, we will discuss the potential molecular targets for enhanced efferocytosis in animal models of disease. To provide a balanced view, we will also discuss some deleterious effects of efferocytosis.
Collapse
Affiliation(s)
- Yonggang Ma
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Scott S Kemp
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Xiaoyuan Yang
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Mack H Wu
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Sarah Y Yuan
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA.
| |
Collapse
|
10
|
Majdalani P, Levitas A, Krymko H, Slanovic L, Braiman A, Hadad U, Dabsan S, Horev A, Zarivach R, Parvari R. A Missense Variation in PHACTR2 Associates with Impaired Actin Dynamics, Dilated Cardiomyopathy, and Left Ventricular Non-Compaction in Humans. Int J Mol Sci 2023; 24:ijms24021388. [PMID: 36674904 PMCID: PMC9864900 DOI: 10.3390/ijms24021388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/24/2022] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
Dilated cardiomyopathy (DCM) with left ventricular non-compaction (LVNC) is a primary myocardial disease leading to contractile dysfunction, progressive heart failure, and excessive risk of sudden cardiac death. Using whole-exome sequencing to investigate a possible genetic cause of DCM with LVNC in a consanguineous child, a homozygous nucleotide change c.1532G>A causing p.Arg511His in PHACTR2 was found. The missense change can affect the binding of PHACTR2 to actin by eliminating the hydrogen bonds between them. The amino acid change does not change PHACTR2 localization to the cytoplasm. The patient’s fibroblasts showed a decreased globular to fibrillary actin ratio compared to the control fibroblasts. The re-polymerization of fibrillary actin after treatment with cytochalasin D, which disrupts the actin filaments, was slower in the patient’s fibroblasts. Finally, the patient’s fibroblasts bridged a scar gap slower than the control fibroblasts because of slower and indirect movement. This is the first report of a human variation in this PHACTR family member. The knock-out mouse model presented no significant phenotype. Our data underscore the importance of PHACTR2 in regulating the monomeric actin pool, the kinetics of actin polymerization, and cell movement, emphasizing the importance of actin regulation for the normal function of the human heart.
Collapse
Affiliation(s)
- Pierre Majdalani
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- The National Institute for Biotechnology in the Negev, Marcus Campus, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Aviva Levitas
- Department of Pediatric Cardiology, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| | - Hanna Krymko
- Department of Pediatric Cardiology, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| | - Leonel Slanovic
- Department of Pediatric Cardiology, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| | - Alex Braiman
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Uzi Hadad
- The Ilse Katz Institute for Nanoscale Science and Technology, Marcus Campus, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Salam Dabsan
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- The National Institute for Biotechnology in the Negev, Marcus Campus, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Amir Horev
- Pediatric Dermatology Service, Soroka University Medical Center, Beer-Sheva 84101, Israel
| | - Raz Zarivach
- The National Institute for Biotechnology in the Negev, Marcus Campus, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- Department of Life Sciences, Faculty of Natural Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Ruti Parvari
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- The National Institute for Biotechnology in the Negev, Marcus Campus, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- Correspondence: ; Tel.: +972-8-647-9967
| |
Collapse
|
11
|
Berrandou TE, Bouatia-Naji N. [Fibromuscular dysplasia as a polygenic disease]. Med Sci (Paris) 2022; 38:870-873. [PMID: 36448890 DOI: 10.1051/medsci/2022134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Affiliation(s)
- Takiy-Eddine Berrandou
- Université Paris Cité, Inserm, PARCC, F-75015 Paris, France - Quantitative genetics and genomics (QGG), Aarhus university, Aarhus, Danemark
| | | |
Collapse
|
12
|
The complex genetic basis of fibromuscular dysplasia, a systemic arteriopathy associated with multiple forms of cardiovascular disease. Clin Sci (Lond) 2022; 136:1241-1255. [PMID: 36043395 PMCID: PMC9434409 DOI: 10.1042/cs20210990] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/28/2022] [Accepted: 08/16/2022] [Indexed: 12/03/2022]
Abstract
Artery stenosis is a common cause of hypertension and stroke and can be due to atherosclerosis accumulation in the majority of cases and in a small fraction of patients to arterial fibromuscular dysplasia (FMD). Artery stenosis due to atherosclerosis is widely studied with known risk factors (e.g. increasing age, male gender, and dyslipidemia) to influence its etiology, including genetic factors. However, the causes of noninflammatory and nonatherosclerotic stenosis in FMD are less understood. FMD occurs predominantly in early middle-age women, a fraction of the population where cardiovascular risk is different and understudied. FMD arteriopathies are often diagnosed in the context of hypertension and stroke and co-occur mainly with spontaneous coronary artery dissection, an atypical cause of acute myocardial infarction. In this review, we provide a comprehensive overview of the recent advances in the understanding of molecular origins of FMD. Data were obtained from genetic studies using complementary methodological approaches applied to familial, syndromic, and sporadic forms of this intriguing arteriopathy. Rare variation analyses point toward mechanisms related to impaired prostacyclin signaling and defaults in fibrillar collagens. The study of common variation, mainly through a recent genome-wide association study, describes a shared genetic link with blood pressure, in addition to point at potential risk genes involved in actin cytoskeleton and intracellular calcium homeostasis supporting impaired vascular contraction as a key mechanism. We conclude this review with future strategies and approaches needed to fully understand the genetic and molecular mechanisms related to FMD.
Collapse
|
13
|
Rubin S, Bougaran P, Martin S, Abelanet A, Delobel V, Pernot M, Jeanningros S, Bats ML, Combe C, Dufourcq P, Debette S, Couffinhal T, Duplàa C. PHACTR-1 (Phosphatase and Actin Regulator 1) Deficiency in Either Endothelial or Smooth Muscle Cells Does Not Predispose Mice to Nonatherosclerotic Arteriopathies in 3 Transgenic Mice. Arterioscler Thromb Vasc Biol 2022; 42:597-609. [PMID: 35387477 DOI: 10.1161/atvbaha.122.317431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Genome-wide association studies have revealed robust associations of common genetic polymorphisms in an intron of the PHACTR-1 (phosphatase and actin regulator 1) gene (chr6p24), with cervical artery dissection, spontaneous coronary artery dissection, and fibromuscular dysplasia. The aim was to assess its role in the pathogenesis of cervical artery dissection or fibromuscular dysplasia. METHODS Using various tissue-specific Cre-driver mouse lines, Phactr1 was deleted either in endothelial cells using 2 tissue-specific Cre-driver (PDGFB [platelet-derived growth factor B]-CreERT2 mice and Tie2 [tyrosine kinase with immunoglobulin and EGF homology domains]-Cre) and smooth muscle cells (smooth muscle actin-CreERT2) with a third tissue-specific Cre-driver. RESULTS To test the efficacy of the Phactr1 deletion after cre-induction, we confirmed first, a decrease in Phactr1 transcription and Phactr1 expression in endothelial cell and smooth muscle cell isolated from Phactr1iPDGFB and Phactr1iSMA mice. Irrespective to the tissue or the duration of the deletion, mice did not spontaneously display pathological phenotype or vascular impairment: mouse survival, growth, blood pressure, large vessel morphology, or actin organization were not different in knockout mice than their comparatives littermates. Challenging vascular function and repair either by angiotensin II-induced hypertension or limb ischemia did not lead to vascular morphology or function impairment in Phactr1-deleted mice. Similarly, there were no more consequences of Phactr1 deletion during embryogenesis in endothelial cells. CONCLUSIONS Loss of PHACTR-1 function in the cells involved in vascular physiology does not appear to induce a pathological vascular phenotype. The in vivo effect of the intronic variation described in genome-wide association studies is unlikely to involve downregulation in PHACTR-1 expression.
Collapse
Affiliation(s)
- Sébastien Rubin
- University of Bordeaux, INSERM, Biologie des Maladies Cardiovasculaires, U1034, Pessac, France (S.R., P.B., S.M., A.A., V.D., M.P., S.J., M.-L.B., P.D., T.C., C.D.).,Service de Néphrologie, Transplantation, Dialyse et Aphérèses (S.R., C.C.), Hôpital Pellegrin, CHU de Bordeaux, France
| | - Pauline Bougaran
- University of Bordeaux, INSERM, Biologie des Maladies Cardiovasculaires, U1034, Pessac, France (S.R., P.B., S.M., A.A., V.D., M.P., S.J., M.-L.B., P.D., T.C., C.D.)
| | - Soizic Martin
- University of Bordeaux, INSERM, Biologie des Maladies Cardiovasculaires, U1034, Pessac, France (S.R., P.B., S.M., A.A., V.D., M.P., S.J., M.-L.B., P.D., T.C., C.D.)
| | - Alice Abelanet
- University of Bordeaux, INSERM, Biologie des Maladies Cardiovasculaires, U1034, Pessac, France (S.R., P.B., S.M., A.A., V.D., M.P., S.J., M.-L.B., P.D., T.C., C.D.)
| | - Valentin Delobel
- University of Bordeaux, INSERM, Biologie des Maladies Cardiovasculaires, U1034, Pessac, France (S.R., P.B., S.M., A.A., V.D., M.P., S.J., M.-L.B., P.D., T.C., C.D.)
| | - Mathieu Pernot
- University of Bordeaux, INSERM, Biologie des Maladies Cardiovasculaires, U1034, Pessac, France (S.R., P.B., S.M., A.A., V.D., M.P., S.J., M.-L.B., P.D., T.C., C.D.)
| | - Sylvie Jeanningros
- University of Bordeaux, INSERM, Biologie des Maladies Cardiovasculaires, U1034, Pessac, France (S.R., P.B., S.M., A.A., V.D., M.P., S.J., M.-L.B., P.D., T.C., C.D.)
| | - Marie-Lise Bats
- University of Bordeaux, INSERM, Biologie des Maladies Cardiovasculaires, U1034, Pessac, France (S.R., P.B., S.M., A.A., V.D., M.P., S.J., M.-L.B., P.D., T.C., C.D.).,Service de Biochimie (M.-L.B.), Hôpital Pellegrin, CHU de Bordeaux, France
| | - Christian Combe
- Service de Néphrologie, Transplantation, Dialyse et Aphérèses (S.R., C.C.), Hôpital Pellegrin, CHU de Bordeaux, France.,University of Bordeaux, Unité INSERM 1026, Université de Bordeaux, France (C.C.)
| | - Pascale Dufourcq
- University of Bordeaux, INSERM, Biologie des Maladies Cardiovasculaires, U1034, Pessac, France (S.R., P.B., S.M., A.A., V.D., M.P., S.J., M.-L.B., P.D., T.C., C.D.)
| | - Stéphanie Debette
- University of Bordeaux, INSERM, Bordeaux Population Health Center, UMR1219, France (S.D.).,Bordeaux University Hospital, Department of Neurology, Institute of Neurodegenerative Diseases, France (S.D.)
| | - Thierry Couffinhal
- University of Bordeaux, INSERM, Biologie des Maladies Cardiovasculaires, U1034, Pessac, France (S.R., P.B., S.M., A.A., V.D., M.P., S.J., M.-L.B., P.D., T.C., C.D.).,Service des Maladies Cardiaques et Vasculaires, Hôpital Haut-Léveque CHU de Bordeaux, Pessac, France (T.C.)
| | - Cécile Duplàa
- University of Bordeaux, INSERM, Biologie des Maladies Cardiovasculaires, U1034, Pessac, France (S.R., P.B., S.M., A.A., V.D., M.P., S.J., M.-L.B., P.D., T.C., C.D.)
| |
Collapse
|
14
|
Kikuchi N, Moreland E, Homma H, Semenova EA, Saito M, Larin AK, Kobatake N, Yusupov RA, Okamoto T, Nakazato K, Williams AG, Generozov EV, Ahmetov II. Genes and Weightlifting Performance. Genes (Basel) 2021; 13:25. [PMID: 35052366 PMCID: PMC8775245 DOI: 10.3390/genes13010025] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/16/2021] [Accepted: 12/22/2021] [Indexed: 11/17/2022] Open
Abstract
A recent case-control study identified 28 DNA polymorphisms associated with strength athlete status. However, studies of genotype-phenotype design are required to support those findings. The aim of the present study was to investigate both individually and in combination the association of 28 genetic markers with weightlifting performance in Russian athletes and to replicate the most significant findings in an independent cohort of Japanese athletes. Genomic DNA was collected from 53 elite Russian (31 men and 22 women, 23.3 ± 4.1 years) and 100 sub-elite Japanese (53 men and 47 women, 21.4 ± 4.2 years) weightlifters, and then genotyped using PCR or micro-array analysis. Out of 28 DNA polymorphisms, LRPPRC rs10186876 A, MMS22L rs9320823 T, MTHFR rs1801131 C, and PHACTR1 rs6905419 C alleles positively correlated (p < 0.05) with weightlifting performance (i.e., total lifts in snatch and clean and jerk in official competitions adjusted for sex and body mass) in Russian athletes. Next, using a polygenic approach, we found that carriers of a high (6-8) number of strength-related alleles had better competition results than carriers of a low (0-5) number of strength-related alleles (264.2 (14.7) vs. 239.1 (21.9) points; p = 0.009). These findings were replicated in the study of Japanese athletes. More specifically, Japanese carriers of a high number of strength-related alleles were stronger than carriers of a low number of strength-related alleles (212.9 (22.6) vs. 199.1 (17.2) points; p = 0.0016). In conclusion, we identified four common gene polymorphisms individually or in combination associated with weightlifting performance in athletes from East European and East Asian geographic ancestries.
Collapse
Affiliation(s)
- Naoki Kikuchi
- Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo 158-8508, Japan; (N.K.); (H.H.); (M.S.); (T.O.); (K.N.)
- Faculty of Sport Science, Nippon Sport Science University, Tokyo 158-8508, Japan;
| | - Ethan Moreland
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool L3 5AF, UK;
| | - Hiroki Homma
- Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo 158-8508, Japan; (N.K.); (H.H.); (M.S.); (T.O.); (K.N.)
| | - Ekaterina A. Semenova
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia; (E.A.S.); (A.K.L.); (E.V.G.)
- Research Institute of Physical Culture and Sport, Volga Region State University of Physical Culture, Sport and Tourism, 420010 Kazan, Russia
| | - Mika Saito
- Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo 158-8508, Japan; (N.K.); (H.H.); (M.S.); (T.O.); (K.N.)
| | - Andrey K. Larin
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia; (E.A.S.); (A.K.L.); (E.V.G.)
| | - Naoyuki Kobatake
- Faculty of Sport Science, Nippon Sport Science University, Tokyo 158-8508, Japan;
| | - Rinat A. Yusupov
- Department of Physical Culture and Sport, Kazan National Research Technical University Named after A.N. Tupolev-KAI, 420111 Kazan, Russia;
| | - Takanobu Okamoto
- Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo 158-8508, Japan; (N.K.); (H.H.); (M.S.); (T.O.); (K.N.)
- Faculty of Sport Science, Nippon Sport Science University, Tokyo 158-8508, Japan;
| | - Koichi Nakazato
- Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo 158-8508, Japan; (N.K.); (H.H.); (M.S.); (T.O.); (K.N.)
- Faculty of Medical Science, Nippon Sport Science University, Tokyo 158-8508, Japan
| | - Alun G. Williams
- Sports Genomics Laboratory, Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester M15 6BH, UK;
- Institute of Sport, Exercise and Health, University College London, London W1T 7HA, UK
| | - Edward V. Generozov
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia; (E.A.S.); (A.K.L.); (E.V.G.)
| | - Ildus I. Ahmetov
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool L3 5AF, UK;
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia; (E.A.S.); (A.K.L.); (E.V.G.)
- Department of Physical Education, Plekhanov Russian University of Economics, 115093 Moscow, Russia
- Laboratory of Molecular Genetics, Kazan State Medical University, 420012 Kazan, Russia
| |
Collapse
|
15
|
Georges A, Yang ML, Berrandou TE, Bakker MK, Dikilitas O, Kiando SR, Ma L, Satterfield BA, Sengupta S, Yu M, Deleuze JF, Dupré D, Hunker KL, Kyryachenko S, Liu L, Sayoud-Sadeg I, Amar L, Brummett CM, Coleman DM, d’Escamard V, de Leeuw P, Fendrikova-Mahlay N, Kadian-Dodov D, Li JZ, Lorthioir A, Pappaccogli M, Prejbisz A, Smigielski W, Stanley JC, Zawistowski M, Zhou X, Zöllner S, FEIRI investigators de LeeuwPeter1213, International Stroke Genetics Consortium (ISGC) Intracranial Aneurysm Working Group, MEGASTROKE, Amouyel P, De Buyzere ML, Debette S, Dobrowolski P, Drygas W, Gornik HL, Olin JW, Piwonski J, Rietzschel ER, Ruigrok YM, Vikkula M, Warchol Celinska E, Januszewicz A, Kullo IJ, Azizi M, ARCADIA Investigators, Jeunemaitre X, Persu A, Kovacic JC, Ganesh SK, Bouatia-Naji N. Genetic investigation of fibromuscular dysplasia identifies risk loci and shared genetics with common cardiovascular diseases. Nat Commun 2021; 12:6031. [PMID: 34654805 PMCID: PMC8521585 DOI: 10.1038/s41467-021-26174-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 09/17/2021] [Indexed: 12/23/2022] Open
Abstract
Fibromuscular dysplasia (FMD) is an arteriopathy associated with hypertension, stroke and myocardial infarction, affecting mostly women. We report results from the first genome-wide association meta-analysis of six studies including 1556 FMD cases and 7100 controls. We find an estimate of SNP-based heritability compatible with FMD having a polygenic basis, and report four robustly associated loci (PHACTR1, LRP1, ATP2B1, and LIMA1). Transcriptome-wide association analysis in arteries identifies one additional locus (SLC24A3). We characterize open chromatin in arterial primary cells and find that FMD associated variants are located in arterial-specific regulatory elements. Target genes are broadly involved in mechanisms related to actin cytoskeleton and intracellular calcium homeostasis, central to vascular contraction. We find significant genetic overlap between FMD and more common cardiovascular diseases and traits including blood pressure, migraine, intracranial aneurysm, and coronary artery disease.
Collapse
Affiliation(s)
- Adrien Georges
- grid.508487.60000 0004 7885 7602PARCC, INSERM, Université de Paris, F-750015 Paris, France
| | - Min-Lee Yang
- grid.214458.e0000000086837370Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI USA ,grid.214458.e0000000086837370Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI USA
| | - Takiy-Eddine Berrandou
- grid.508487.60000 0004 7885 7602PARCC, INSERM, Université de Paris, F-750015 Paris, France
| | - Mark K. Bakker
- grid.5477.10000000120346234Department of Neurology and Neurosurgery, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Ozan Dikilitas
- grid.66875.3a0000 0004 0459 167XDepartment of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55902 USA
| | - Soto Romuald Kiando
- grid.508487.60000 0004 7885 7602PARCC, INSERM, Université de Paris, F-750015 Paris, France
| | - Lijiang Ma
- grid.59734.3c0000 0001 0670 2351Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Benjamin A. Satterfield
- grid.66875.3a0000 0004 0459 167XDepartment of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55902 USA
| | - Sebanti Sengupta
- grid.214458.e0000000086837370Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI USA
| | - Mengyao Yu
- grid.508487.60000 0004 7885 7602PARCC, INSERM, Université de Paris, F-750015 Paris, France
| | - Jean-François Deleuze
- grid.418135.a0000 0004 0641 3404Centre National de Recherche en Génomique Humaine, Institut de Génomique, CEA and Fondation Jean Dausset-CEPH, Evry, France
| | - Delia Dupré
- grid.508487.60000 0004 7885 7602PARCC, INSERM, Université de Paris, F-750015 Paris, France
| | - Kristina L. Hunker
- grid.214458.e0000000086837370Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI USA ,grid.214458.e0000000086837370Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI USA
| | - Sergiy Kyryachenko
- grid.508487.60000 0004 7885 7602PARCC, INSERM, Université de Paris, F-750015 Paris, France
| | - Lu Liu
- grid.508487.60000 0004 7885 7602PARCC, INSERM, Université de Paris, F-750015 Paris, France
| | - Ines Sayoud-Sadeg
- grid.508487.60000 0004 7885 7602PARCC, INSERM, Université de Paris, F-750015 Paris, France
| | - Laurence Amar
- grid.508487.60000 0004 7885 7602PARCC, INSERM, Université de Paris, F-750015 Paris, France ,grid.414093.b0000 0001 2183 5849Hypertension Unit, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Chad M. Brummett
- grid.214458.e0000000086837370Department of Anesthesiology, Michigan Medicine, University of Michigan, Ann Arbor, MI USA
| | - Dawn M. Coleman
- grid.214458.e0000000086837370Vascular Surgery Section, Department of Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109 USA
| | - Valentina d’Escamard
- grid.59734.3c0000 0001 0670 2351Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Peter de Leeuw
- grid.412966.e0000 0004 0480 1382Department of Internal Medicine, Division of General Internal Medicine, Section Vascular Medicine, Maastricht University Medical Centre, Maastricht University, Maastricht, the Netherlands ,grid.5012.60000 0001 0481 6099CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht University, Maastricht, the Netherlands
| | - Natalia Fendrikova-Mahlay
- grid.239578.20000 0001 0675 4725Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH 44195 USA
| | - Daniella Kadian-Dodov
- grid.59734.3c0000 0001 0670 2351Zena and Michael A. Wiener Cardiovascular Institute and Marie-Josée and Henry R, Kravis Center for Cardiovascular Health Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Jun Z. Li
- grid.214458.e0000000086837370Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI USA
| | - Aurélien Lorthioir
- grid.508487.60000 0004 7885 7602PARCC, INSERM, Université de Paris, F-750015 Paris, France ,grid.414093.b0000 0001 2183 5849Hypertension Unit, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Marco Pappaccogli
- grid.7942.80000 0001 2294 713XDivision of Cardiology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 1200 Brussels, Belgium ,grid.7605.40000 0001 2336 6580Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Aleksander Prejbisz
- grid.418887.aDepartment of Hypertension, National Institute of Cardiology, Warsaw, Poland
| | - Witold Smigielski
- grid.10789.370000 0000 9730 2769Department of Demography, University of Lodz, Lodz, Poland
| | - James C. Stanley
- grid.214458.e0000000086837370Vascular Surgery Section, Department of Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109 USA
| | - Matthew Zawistowski
- grid.214458.e0000000086837370Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI USA
| | - Xiang Zhou
- grid.214458.e0000000086837370Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI USA
| | - Sebastian Zöllner
- grid.214458.e0000000086837370Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI USA
| | | | | | | | - Philippe Amouyel
- grid.503422.20000 0001 2242 6780Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Labex DISTALZ - Risk factors and molecular determinants of aging-related disease, F-59000 Lille, France
| | - Marc L. De Buyzere
- grid.5342.00000 0001 2069 7798Department of Cardiovascular Diseases, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Stéphanie Debette
- grid.42399.350000 0004 0593 7118Department of Neurology, Bordeaux University Hospital, Inserm U1219, Bordeaux, France
| | - Piotr Dobrowolski
- grid.418887.aDepartment of Hypertension, National Institute of Cardiology, Warsaw, Poland
| | - Wojciech Drygas
- grid.418887.aDepartment of Epidemiology, Cardiovascular Disease Prevention, and Health Promotion, National Institute of Cardiology, Warsaw, Poland
| | - Heather L. Gornik
- grid.239578.20000 0001 0675 4725Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH 44195 USA
| | - Jeffrey W. Olin
- grid.59734.3c0000 0001 0670 2351Zena and Michael A. Wiener Cardiovascular Institute and Marie-Josée and Henry R, Kravis Center for Cardiovascular Health Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Jerzy Piwonski
- grid.418887.aDepartment of Epidemiology, Cardiovascular Disease Prevention, and Health Promotion, National Institute of Cardiology, Warsaw, Poland
| | - Ernst R. Rietzschel
- grid.5342.00000 0001 2069 7798Department of Cardiovascular Diseases, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Ynte M. Ruigrok
- grid.66875.3a0000 0004 0459 167XDepartment of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55902 USA
| | - Miikka Vikkula
- grid.7942.80000 0001 2294 713XHuman Molecular Genetics, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Ewa Warchol Celinska
- grid.418887.aDepartment of Hypertension, National Institute of Cardiology, Warsaw, Poland
| | - Andrzej Januszewicz
- grid.418887.aDepartment of Hypertension, National Institute of Cardiology, Warsaw, Poland
| | - Iftikhar J. Kullo
- grid.66875.3a0000 0004 0459 167XDepartment of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55902 USA ,grid.66875.3a0000 0004 0459 167XGonda Vascular Center, Mayo Clinic, Rochester, MN 55902 USA
| | - Michel Azizi
- grid.414093.b0000 0001 2183 5849Hypertension Unit, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, F-75015 Paris, France ,grid.512950.aUniversité de Paris, Inserm, Centre d’Investigation Clinique 1418, F-75006 Paris, France
| | | | - Xavier Jeunemaitre
- grid.508487.60000 0004 7885 7602PARCC, INSERM, Université de Paris, F-750015 Paris, France ,grid.414093.b0000 0001 2183 5849Department of Genetics, Assistance-Publiques-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Alexandre Persu
- grid.7942.80000 0001 2294 713XDivision of Cardiology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 1200 Brussels, Belgium ,grid.7942.80000 0001 2294 713XPole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Jason C. Kovacic
- grid.59734.3c0000 0001 0670 2351Zena and Michael A. Wiener Cardiovascular Institute and Marie-Josée and Henry R, Kravis Center for Cardiovascular Health Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.1057.30000 0000 9472 3971Victor Chang Cardiac Research Institute, Darlinghurst, NSW Australia ,grid.1005.40000 0004 4902 0432St. Vincent’s Clinical School, University of New South Wales, Sydney, NSW Australia
| | - Santhi K. Ganesh
- grid.214458.e0000000086837370Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI USA ,grid.214458.e0000000086837370Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI USA
| | - Nabila Bouatia-Naji
- grid.508487.60000 0004 7885 7602PARCC, INSERM, Université de Paris, F-750015 Paris, France
| |
Collapse
|
16
|
Matos B, Howl J, Jerónimo C, Fardilha M. Modulation of serine/threonine-protein phosphatase 1 (PP1) complexes: A promising approach in cancer treatment. Drug Discov Today 2021; 26:2680-2698. [PMID: 34390863 DOI: 10.1016/j.drudis.2021.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/23/2021] [Accepted: 08/05/2021] [Indexed: 01/21/2023]
Abstract
Cancer is the second leading cause of death worldwide. Despite the availability of numerous therapeutic options, tumor heterogeneity and chemoresistance have limited the success of these treatments, and the development of effective anticancer therapies remains a major focus in oncology research. The serine/threonine-protein phosphatase 1 (PP1) and its complexes have been recognized as potential drug targets. Research on the modulation of PP1 complexes is currently at an early stage, but has immense potential. Chemically diverse compounds have been developed to disrupt or stabilize different PP1 complexes in various cancer types, with the objective of inhibiting disease progression. Beneficial results obtained in vitro now require further pre-clinical and clinical validation. In conclusion, the modulation of PP1 complexes seems to be a promising, albeit challenging, therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Bárbara Matos
- Laboratory of Signal Transduction, Department of Medical Sciences, Institute of Biomedicine-iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal; Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal
| | - John Howl
- Molecular Pharmacology Group, Research Institute in Healthcare Science, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal; Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), 4050-513 Porto, Portugal
| | - Margarida Fardilha
- Laboratory of Signal Transduction, Department of Medical Sciences, Institute of Biomedicine-iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
17
|
Kasikara C, Schilperoort M, Gerlach B, Xue C, Wang X, Zheng Z, Kuriakose G, Dorweiler B, Zhang H, Fredman G, Saleheen D, Reilly MP, Tabas I. Deficiency of macrophage PHACTR1 impairs efferocytosis and promotes atherosclerotic plaque necrosis. J Clin Invest 2021; 131:145275. [PMID: 33630758 DOI: 10.1172/jci145275] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Efferocytosis, the process through which apoptotic cells (ACs) are cleared through actin-mediated engulfment by macrophages, prevents secondary necrosis, suppresses inflammation, and promotes resolution. Impaired efferocytosis drives the formation of clinically dangerous necrotic atherosclerotic plaques, the underlying etiology of coronary artery disease (CAD). An intron of the gene encoding PHACTR1 contains rs9349379 (A>G), a common variant associated with CAD. As PHACTR1 is an actin-binding protein, we reasoned that if the rs9349379 risk allele G causes lower PHACTR1 expression in macrophages, it might link the risk allele to CAD via impaired efferocytosis. We show here that rs9349379-G/G was associated with lower levels of PHACTR1 and impaired efferocytosis in human monocyte-derived macrophages and human atherosclerotic lesional macrophages compared with rs9349379-A/A. Silencing PHACTR1 in human and mouse macrophages compromised AC engulfment, and Western diet-fed Ldlr-/- mice in which hematopoietic Phactr1 was genetically targeted showed impaired lesional efferocytosis, increased plaque necrosis, and thinner fibrous caps - all signs of vulnerable plaques in humans. Mechanistically, PHACTR1 prevented dephosphorylation of myosin light chain (MLC), which was necessary for AC engulfment. In summary, rs9349379-G lowered PHACTR1, which, by lowering phospho-MLC, compromised efferocytosis. Thus, rs9349379-G may contribute to CAD risk, at least in part, by impairing atherosclerotic lesional macrophage efferocytosis.
Collapse
Affiliation(s)
- Canan Kasikara
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Maaike Schilperoort
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Brennan Gerlach
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Chenyi Xue
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Ze Zheng
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - George Kuriakose
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | | | - Hanrui Zhang
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Gabrielle Fredman
- Department of Molecular and Cellular Physiology, Albany Medical Center, Albany, New York, USA
| | - Danish Saleheen
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Muredach P Reilly
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA.,Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, New York, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA.,Department of Physiology and Cellular Biophysics and.,Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
18
|
Herman L, Legois B, Todeschini AL, Veitia RA. Genomic exploration of the targets of FOXL2 and ESR2 unveils their implication in cell migration, invasion, and adhesion. FASEB J 2021; 35:e21355. [PMID: 33749886 DOI: 10.1096/fj.202002444r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 01/01/2023]
Abstract
FOXL2 and ESR2 are key transcriptional regulators in ovarian granulosa cells. To explore their transcriptional roles and their interplay, we have depleted Foxl2 and Esr2 in mouse primary granulosa cells to assess their ability to bind their targets and/or to modulate gene expression and cellular functions. We show that FOXL2 is involved in a large number of regulatory actions essential for the maintenance of granulosa cell fate. A parallel ChIP-seq analysis showed that FOXL2 mainly binds to sites located in intergenic regions quite far from its targets. A bioinformatic analysis demonstrated that FOXL2-activated genes were enriched in peaks associated with the H3K27ac mark, whereas FOXL2-repressed genes were not, suggesting that FOXL2 can activate transcription through binding to enhancer sites. We also identified about 500 deregulated genes upon Esr2 silencing, of which one third are also targets of FOXL2. We provide evidence showing that both factors modulate, through a coherent feed-forward loop, a number of common targets. Many of the FOXL2/ESR2 targets are involved in cell motility and, consistently, granulosa cells depleted for either Foxl2 or Esr2 exhibit decreased migration, invasion and adhesion. This effect is paralleled by the depletion of their target Phactr1, involved in actin cytoskeleton dynamics. Our analysis expands the number of direct and indirect transcriptional targets of both FOXL2 and ESR2, which deserve investigation in the context of adult-type granulosa cell tumors whose molecular diagnostic hallmark is the presence of the C134W FOXL2 pathogenic variant.
Collapse
Affiliation(s)
- Laetitia Herman
- Faculté des Sciences, Université de Paris, Paris, France.,Institut Jacques Monod, Université de Paris, CNRS, Paris, France
| | - Bérangère Legois
- Faculté des Sciences, Université de Paris, Paris, France.,Institut Jacques Monod, Université de Paris, CNRS, Paris, France
| | - Anne-Laure Todeschini
- Faculté des Sciences, Université de Paris, Paris, France.,Institut Jacques Monod, Université de Paris, CNRS, Paris, France
| | - Reiner A Veitia
- Faculté des Sciences, Université de Paris, Paris, France.,Institut Jacques Monod, Université de Paris, CNRS, Paris, France.,Institut de Biologie F. Jacob, Université Paris-Saclay, Commissariat à l'Energie Atomique, Fontenay aux Roses, France
| |
Collapse
|
19
|
Genetic deficiency of Phactr1 promotes atherosclerosis development via facilitating M1 macrophage polarization and foam cell formation. Clin Sci (Lond) 2021; 134:2353-2368. [PMID: 32857129 DOI: 10.1042/cs20191241] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 08/13/2020] [Accepted: 08/28/2020] [Indexed: 12/19/2022]
Abstract
Genetic variants in phosphatase and actin regulator-1 (Phactr1) are reported to be associated with arteriosclerotic cardiovascular disease (ASCVD). However, the function of Phactr1 in atherosclerosis remains unclear. Patients with acute coronary syndrome (ACS) who underwent coronary angiography and optical coherence tomography (OCT) were enrolled and divided into non-ST segment elevation (NST-ACS) group and ST-ACS group. The expression of Phactr1 on monocytes was higher in NST-ACS and ST-ACS groups as compared with control group. Furthermore, NST-ACS patients who have more vulnerable features including thin-cap fibroatheroma (TCFA) and large lipid area showed higher levels of Phactr1 on monocytes than those with stable plaques. Through mouse models of atherosclerosis, Phactr1-/-Apoe-/- mice (double knockout mice, DKO) developed more severe atherosclerotic plaques, recruiting more macrophages into subendothelium and having elevated levels of proinflammatory cytokines in plaques. Similarly, Apoe knockout mice (Apoe-/-) receiving DKO bone marrow (BM) exhibited elevated plaque burden compared with Apoe-/- mice receiving Apoe-/- BM, indicating the protective effect of Phactr1 in hematopoietic cells. We found that depletion of Phactr1 in BM-derived macrophages (BMDMs) tended to differentiate into M1 phenotype, produced more proatherogenic cytokines and eventually converted into foam cells driven by oxidized low-density lipoprotein (ox-LDL). Mechanistically, Phactr1 activated CREB signaling via directly binding to CREB, up-regulating CREB phosphorylation and inducing KLF4 expression. Finally, overexpression of KLF4 partly rescued the excessive inflammation response and foam cell formation induced by deficiency of Phactr1. In conclusion, our study demonstrates that elevated Phactr1 in monocytes is a promising biomarker for vulnerable plaques, while increased Phactr1 attenuates atherosclerotic development via activation of CREB and M2 macrophage differentiation.
Collapse
|
20
|
Marakhonov AV, Přechová M, Konovalov FA, Filatova AY, Zamkova MA, Kanivets IV, Solonichenko VG, Semenova NA, Zinchenko RA, Treisman R, Skoblov MY. Mutation in PHACTR1 associated with multifocal epilepsy with infantile spasms and hypsarrhythmia. Clin Genet 2021; 99:673-683. [PMID: 33463715 PMCID: PMC8629116 DOI: 10.1111/cge.13926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/07/2021] [Accepted: 01/13/2021] [Indexed: 11/28/2022]
Abstract
A young boy with multifocal epilepsy with infantile spasms and hypsarrhythmia with minimal organic lesions of brain structures underwent DNA diagnosis using whole‐exome sequencing. A heterozygous amino‐acid substitution p.L519R in a PHACTR1 gene was identified. PHACTR1 belongs to a protein family of G‐actin binding protein phosphatase 1 (PP1) cofactors and was not previously associated with a human disease. The missense single nucleotide variant in the proband was shown to occur de novo in the paternal allele. The mutation was shown in vitro to reduce the affinity of PHACTR1 for G‐actin, and to increase its propensity to form complexes with the catalytic subunit of PP1. These properties are associated with altered subcellular localization of PHACTR1 and increased ability to induce cytoskeletal rearrangements. Although the molecular role of the PHACTR1 in neuronal excitability and differentiation remains to be defined, PHACTR1 has been previously shown to be involved in Slack channelopathy pathogenesis, consistent with our findings. We conclude that this activating mutation in PHACTR1 causes a severe type of sporadic multifocal epilepsy in the patient.
Collapse
Affiliation(s)
- Andrey V Marakhonov
- Laboratory of Genetic Epidemiology, Laboratory of Functional Genomics, Department of Genetic Counseling, Research Centre for Medical Genetics, Moscow, Russia
| | - Magdalena Přechová
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.,Signalling and Transcription Laboratory, Francis Crick Institute, London, UK
| | | | - Alexandra Yu Filatova
- Laboratory of Genetic Epidemiology, Laboratory of Functional Genomics, Department of Genetic Counseling, Research Centre for Medical Genetics, Moscow, Russia
| | - Maria A Zamkova
- Laboratory of Regulatory Mechanisms in Immunity, Institute of Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia
| | - Ilya V Kanivets
- Laboratory of Molecular Pathology, Genomed Ltd., Moscow, Russia.,Medical Genetic Centre, Filatov Moscow Pediatric Clinical Hospital, Moscow, Russia
| | | | - Natalia A Semenova
- Laboratory of Genetic Epidemiology, Laboratory of Functional Genomics, Department of Genetic Counseling, Research Centre for Medical Genetics, Moscow, Russia
| | - Rena A Zinchenko
- Laboratory of Genetic Epidemiology, Laboratory of Functional Genomics, Department of Genetic Counseling, Research Centre for Medical Genetics, Moscow, Russia.,N.A. Semashko National Research Institute of Public Health, Moscow, Russia
| | - Richard Treisman
- Signalling and Transcription Laboratory, Francis Crick Institute, London, UK
| | - Mikhail Yu Skoblov
- Laboratory of Genetic Epidemiology, Laboratory of Functional Genomics, Department of Genetic Counseling, Research Centre for Medical Genetics, Moscow, Russia
| |
Collapse
|
21
|
Fedoryshchak RO, Přechová M, Butler AM, Lee R, O'Reilly N, Flynn HR, Snijders AP, Eder N, Ultanir S, Mouilleron S, Treisman R. Molecular basis for substrate specificity of the Phactr1/PP1 phosphatase holoenzyme. eLife 2020; 9:61509. [PMID: 32975518 PMCID: PMC7599070 DOI: 10.7554/elife.61509] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 09/24/2020] [Indexed: 01/01/2023] Open
Abstract
PPP-family phosphatases such as PP1 have little intrinsic specificity. Cofactors can target PP1 to substrates or subcellular locations, but it remains unclear how they might confer sequence-specificity on PP1. The cytoskeletal regulator Phactr1 is a neuronally enriched PP1 cofactor that is controlled by G-actin. Structural analysis showed that Phactr1 binding remodels PP1's hydrophobic groove, creating a new composite surface adjacent to the catalytic site. Using phosphoproteomics, we identified mouse fibroblast and neuronal Phactr1/PP1 substrates, which include cytoskeletal components and regulators. We determined high-resolution structures of Phactr1/PP1 bound to the dephosphorylated forms of its substrates IRSp53 and spectrin αII. Inversion of the phosphate in these holoenzyme-product complexes supports the proposed PPP-family catalytic mechanism. Substrate sequences C-terminal to the dephosphorylation site make intimate contacts with the composite Phactr1/PP1 surface, which are required for efficient dephosphorylation. Sequence specificity explains why Phactr1/PP1 exhibits orders-of-magnitude enhanced reactivity towards its substrates, compared to apo-PP1 or other PP1 holoenzymes.
Collapse
Affiliation(s)
- Roman O Fedoryshchak
- Signalling and Transcription Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Magdalena Přechová
- Signalling and Transcription Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Abbey M Butler
- Signalling and Transcription Laboratory, The Francis Crick Institute, London, United Kingdom.,Structural Biology Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Rebecca Lee
- Signalling and Transcription Laboratory, The Francis Crick Institute, London, United Kingdom.,Structural Biology Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Nicola O'Reilly
- Peptide Chemistry Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Helen R Flynn
- Proteomics Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Ambrosius P Snijders
- Proteomics Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Noreen Eder
- Proteomics Science Technology Platform, The Francis Crick Institute, London, United Kingdom.,Kinases and Brain Development Laboratory The Francis Crick Institute, London, United Kingdom
| | - Sila Ultanir
- Kinases and Brain Development Laboratory The Francis Crick Institute, London, United Kingdom
| | - Stephane Mouilleron
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Richard Treisman
- Signalling and Transcription Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
22
|
Associations between PHACTR1 gene polymorphisms and pulse pressure in Chinese Han population. Biosci Rep 2020; 40:224380. [PMID: 32420588 PMCID: PMC7276519 DOI: 10.1042/bsr20193779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 05/12/2020] [Accepted: 05/15/2020] [Indexed: 11/25/2022] Open
Abstract
A genome-wide association study (GWAS) in Chinese twins was performed to explore associations between genes and pulse pressure (PP) in 2012, and detected a suggestive association in the phosphatase and actin regulator 1 (PHACTR1) gene on chromosome 6p24.1 (rs1223397, P=1.04e−07). The purpose of the present study was to investigate associations of PHACTR1 gene polymorphisms with PP in a Chinese population. We recruited 347 subjects with PP ≥ 65 mmHg as cases and 359 subjects with 30 ≤ PP ≤ 45 mmHg as controls. Seven single nucleotide polymorphisms (SNPs) in the PHACTR1 gene were genotyped. Logistic regression was performed to explore associations between SNPs and PP in codominant, additive, dominant, recessive and overdominant models. The Pearson’s χ2 test was applied to assess the relationships of haplotypes and PP. The A allele of rs9349379 had a positive effect on high PP. Multivariate logistic regression analysis showed that rs9349379 was significantly related to high PP in codominant [AA vs GG, 2.255 (1.132–4.492)], additive [GG vs GA vs AA, 1.368 (1.049–1.783)] and recessive [AA vs GA + GG, 2.062 (1.051–4.045)] models. The positive association between rs499818 and high PP was significant in codominant [AA vs GG, 3.483 (1.044–11.613)] and recessive [AA vs GG + GA, 3.716 (1.119–12.339)] models. No significant association of haplotypes with PP was detected. There was no significant interaction between six SNPs without strong linkage. In conclusion, the present study presents that rs9349379 and rs499818 in the PHACTR1 gene were significantly associated with PP in Chinese population. Future research should be conducted to confirm them.
Collapse
|
23
|
Ali SR, Malone TJ, Zhang Y, Prechova M, Kaczmarek LK. Phactr1 regulates Slack (KCNT1) channels via protein phosphatase 1 (PP1). FASEB J 2020; 34:1591-1601. [PMID: 31914597 PMCID: PMC6956700 DOI: 10.1096/fj.201902366r] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 12/14/2022]
Abstract
The Slack (KCNT1) gene encodes sodium-activated potassium channels that are abundantly expressed in the central nervous system. Human mutations alter the function of Slack channels, resulting in epilepsy and intellectual disability. Most of the disease-causing mutations are located in the extended cytoplasmic C-terminus of Slack channels and result in increased Slack current. Previous experiments have shown that the C-terminus of Slack channels binds a number of cytoplasmic signaling proteins. One of these is Phactr1, an actin-binding protein that recruits protein phosphatase 1 (PP1) to certain phosphoprotein substrates. Using co-immunoprecipitation, we found that Phactr1 is required to link the channels to actin. Using patch clamp recordings, we found that co-expression of Phactr1 with wild-type Slack channels reduces the current amplitude but has no effect on Slack channels in which a conserved PKC phosphorylation site (S407) that regulates the current amplitude has been mutated. Furthermore, a Phactr1 mutant that disrupts the binding of PP1 but not that of actin fails to alter Slack currents. Our data suggest that Phactr1 regulates the Slack by linking PP1 to the channel. Targeting Slack-Phactr1 interactions may therefore be helpful in developing the novel therapies for brain disorders associated with the malfunction of Slack channels.
Collapse
Affiliation(s)
- Syed Rydwan Ali
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | | | - Yalan Zhang
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Magdalena Prechova
- Signalling and Transcription Group, The Francis Crick Institute, London, UK
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, CZ
| | - Leonard Konrad Kaczmarek
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
24
|
Hamada N, Ogaya S, Nakashima M, Nishijo T, Sugawara Y, Iwamoto I, Ito H, Maki Y, Shirai K, Baba S, Maruyama K, Saitsu H, Kato M, Matsumoto N, Momiyama T, Nagata KI. De novo PHACTR1 mutations in West syndrome and their pathophysiological effects. Brain 2019; 141:3098-3114. [PMID: 30256902 DOI: 10.1093/brain/awy246] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022] Open
Abstract
Trio-based whole exome sequencing identified two de novo heterozygous missense mutations [c.1449T > C/p.(Leu500Pro) and c.1436A > T/p.(Asn479Ile)] in PHACTR1, encoding a molecule critical for the regulation of protein phosphatase 1 (PP1) and the actin cytoskeleton, in unrelated Japanese individuals with West syndrome (infantile spasms with intellectual disability). We then examined the role of Phactr1 in the development of mouse cerebral cortex and the pathophysiological significance of these two mutations and others [c.1561C > T/p.(Arg521Cys) and c.1553T > A/p.(Ile518Asn)], which had been reported in undiagnosed patients with intellectual disability. Immunoprecipitation analyses revealed that actin-binding activity of PHACTR1 was impaired by the p.Leu500Pro, p.Asn479Ile and p.Ile518Asn mutations while the p.Arg521Cys mutation exhibited impaired binding to PP1. Acute knockdown of mouse Phactr1 using in utero electroporation caused defects in cortical neuron migration during corticogenesis, which were rescued by an RNAi-resistant PHACTR1 but not by the four mutants. Experiments using knockdown combined with expression mutants, aimed to mimic the effects of the heterozygous mutations under conditions of haploinsufficiency, suggested a dominant negative effect of the mutant allele. As for dendritic development in vivo, only the p.Arg521Cys mutant was determined to have dominant negative effects, because the three other mutants appeared to be degraded with these experimental conditions. Electrophysiological analyses revealed abnormal synaptic properties in Phactr1-deficient excitatory cortical neurons. Our data show that the PHACTR1 mutations may cause morphological and functional defects in cortical neurons during brain development, which is likely to be related to the pathophysiology of West syndrome and other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Nanako Hamada
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya, Kasugai, Aichi, Japan.,Research Fellow of Japan Society for the Promotion of Science, Japan
| | - Shunsuke Ogaya
- Department of Pediatric Neurology, Central Hospital, Aichi Human Service Center, 713-8 Kamiya, Kasugai, Aichi, Japan
| | - Mitsuko Nakashima
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Japan
| | - Takuma Nishijo
- Department of Pharmacology, Jikei University School of Medicine, 3-19-18 Nishishimbashi, Minato-ku, Tokyo, Japan
| | - Yuji Sugawara
- Department of Pediatrics, Soka Municipal Hospital, 2-21-1 Soka, Soka, Saitama, Japan
| | - Ikuko Iwamoto
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya, Kasugai, Aichi, Japan
| | - Hidenori Ito
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya, Kasugai, Aichi, Japan
| | - Yuki Maki
- Department of Pediatric Neurology, Central Hospital, Aichi Human Service Center, 713-8 Kamiya, Kasugai, Aichi, Japan
| | - Kentaro Shirai
- Department of Pediatrics, Tsuchiura Kyodo Hospital, 4-1-1 Ootsuno, Tsuchiura, Ibaraki, Japan
| | - Shimpei Baba
- Department of Child Neurology, Comprehensive Epilepsy Center, Seirei-Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Naka-ku, Hamamatsu, Shizuoka, Japan
| | - Koichi Maruyama
- Department of Pediatric Neurology, Central Hospital, Aichi Human Service Center, 713-8 Kamiya, Kasugai, Aichi, Japan
| | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Japan
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Japan
| | - Toshihiko Momiyama
- Department of Pharmacology, Jikei University School of Medicine, 3-19-18 Nishishimbashi, Minato-ku, Tokyo, Japan
| | - Koh-Ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya, Kasugai, Aichi, Japan.,Department of Neurochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
| |
Collapse
|
25
|
Diring J, Mouilleron S, McDonald NQ, Treisman R. RPEL-family rhoGAPs link Rac/Cdc42 GTP loading to G-actin availability. Nat Cell Biol 2019; 21:845-855. [PMID: 31209295 PMCID: PMC6960015 DOI: 10.1038/s41556-019-0337-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 04/29/2019] [Indexed: 12/29/2022]
Abstract
RPEL proteins, which contain the G-actin-binding RPEL motif, coordinate cytoskeletal processes with actin dynamics. We show that the ArhGAP12- and ArhGAP32-family GTPase-activating proteins (GAPs) are RPEL proteins. We determine the structure of the ArhGAP12/G-actin complex, and show that G-actin contacts the RPEL motif and GAP domain sequences. G-actin inhibits ArhGAP12 GAP activity, and this requires the G-actin contacts identified in the structure. In B16 melanoma cells, ArhGAP12 suppresses basal Rac and Cdc42 activity, F-actin assembly, invadopodia formation and experimental metastasis. In this setting, ArhGAP12 mutants defective for G-actin binding exhibit more effective downregulation of Rac GTP loading following HGF stimulation and enhanced inhibition of Rac-dependent processes, including invadopodia formation. Potentiation or disruption of the G-actin/ArhGAP12 interaction, by treatment with the actin-binding drugs latrunculin B or cytochalasin D, has corresponding effects on Rac GTP loading. The interaction of G-actin with RPEL-family rhoGAPs thus provides a negative feedback loop that couples Rac activity to actin dynamics.
Collapse
Affiliation(s)
- Jessica Diring
- Signalling and Transcription Group, The Francis Crick Institute, London, UK
| | - Stephane Mouilleron
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, UK
| | - Neil Q McDonald
- Signalling and Structural Biology Group, The Francis Crick Institute, London, UK
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, London, UK
| | - Richard Treisman
- Signalling and Transcription Group, The Francis Crick Institute, London, UK.
| |
Collapse
|
26
|
Viita T, Kyheröinen S, Prajapati B, Virtanen J, Frilander MJ, Varjosalo M, Vartiainen MK. Nuclear actin interactome analysis links actin to KAT14 histone acetyl transferase and mRNA splicing. J Cell Sci 2019; 132:jcs226852. [PMID: 30890647 PMCID: PMC6503952 DOI: 10.1242/jcs.226852] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 03/05/2019] [Indexed: 12/25/2022] Open
Abstract
In addition to its essential functions within the cytoskeleton, actin also localizes to the cell nucleus, where it is linked to many important nuclear processes from gene expression to maintenance of genomic integrity. However, the molecular mechanisms by which actin operates in the nucleus remain poorly understood. Here, we have used two complementary mass spectrometry (MS) techniques, AP-MS and BioID, to identify binding partners for nuclear actin. Common high-confidence interactions highlight the role of actin in chromatin-remodeling complexes and identify the histone-modifying complex human Ada-Two-A-containing (hATAC) as a novel actin-containing nuclear complex. Actin binds directly to the hATAC subunit KAT14, and modulates its histone acetyl transferase activity in vitro and in cells. Transient interactions detected through BioID link actin to several steps of transcription as well as to RNA processing. Alterations in nuclear actin levels disturb alternative splicing in minigene assays, likely by affecting the transcription elongation rate. This interactome analysis thus identifies both novel direct binding partners and functional roles for nuclear actin, as well as forms a platform for further mechanistic studies on how actin operates during essential nuclear processes.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Tiina Viita
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland
- Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Salla Kyheröinen
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland
- Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Bina Prajapati
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland
- Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Jori Virtanen
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland
- Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Mikko J Frilander
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland
- Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland
- Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
- Proteomics Unit, University of Helsinki, Helsinki 00014, Finland
| | - Maria K Vartiainen
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland
- Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| |
Collapse
|
27
|
Gengenbach BB, Müschen CR, Buyel JF. Expression and purification of human phosphatase and actin regulator 1 (PHACTR1) in plant-based systems. Protein Expr Purif 2018; 151:46-55. [PMID: 29894805 DOI: 10.1016/j.pep.2018.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 05/31/2018] [Accepted: 06/07/2018] [Indexed: 12/21/2022]
Abstract
Cardiovascular diseases are a prevalent cause of morbidity and mortality especially in industrialized countries. The human phosphatase and actin regulator 1 (PHACTR1) may be involved in such diseases, but its precise regulatory function remains unclear due to the large number of potential interaction partners. The same phenomenon makes this protein difficult to express in mammalian cells, but it is also an intrinsically disordered protein that likely aggregates when expressed in bacteria due to the absence of chaperones. We therefore used a design of experiments approach to test the suitability of three plant-based systems for the expression of satisfactory quantities of recombinant PHACTR1, namely transient expression in tobacco (Nicotiana tabacum) BY-2 plant cell packs (PCPs), whole N. benthamiana leaves and BY-2 cell lysate (BYL). The highest yield was achieved using the BYL: up to 120 mg product kg-1 biomass equivalent within 48 h of translation. This was 1.3-fold higher than transient expression in N. benthamiana together with the silencing inhibitor p19, and 6-fold higher than the PCP system. The presence of Triton X-100 in the extraction buffer increased the recovery of PHACTR1 by 2-200-fold depending on the conditions. PHACTR1 was incompatible with biomass blanching and was stable for less than 16 h in raw plant extracts. Purification using a DDK-tag proved inefficient whereas 15% purity was achieved by immobilized metal affinity chromatography.
Collapse
Affiliation(s)
- B B Gengenbach
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstraße 6, 52074, Aachen, Germany.
| | - C R Müschen
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstraße 6, 52074, Aachen, Germany.
| | - J F Buyel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstraße 6, 52074, Aachen, Germany; Institute for Molecular Biotechnology, Worringerweg 1, RWTH Aachen University, 52074, Aachen, Germany.
| |
Collapse
|
28
|
Zhang Z, Jiang F, Zeng L, Wang X, Tu S. PHACTR1 regulates oxidative stress and inflammation to coronary artery endothelial cells via interaction with NF-κB/p65. Atherosclerosis 2018; 278:180-189. [PMID: 30293016 DOI: 10.1016/j.atherosclerosis.2018.08.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 08/06/2018] [Accepted: 08/29/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Genome-wide association studies have showed that genetic variants in phosphatase and actin regulator 1 (PHACTR1) are associated with coronary artery disease and myocardial infarction. However, the underlying mechanism of PHACTR1 in atherosclerosis remains unknown. METHODS Immunoblots were performed to evaluate the expression of PHACTR1 and phosphorylation of NF-κB signaling. Reactive oxygen species (ROS) labeled with DCFH-DA were assessed by flow cytometry. Fluorescence microscope was used to detect the translocation of p65 in human coronary artery endothelial cells (HACECs). Co-immunoprecipitation was performed to determine the interaction of PHACTR1 with MRTF-A. RESULTS The mRNA and protein levels of PHACTR1 were markedly increased in carotid plaquescompared with normal carotid arteries. Immunofluorescence staining indicated that PHACTR1 was constitutively expressed in endothelial cells in carotid plaques. Knockdown of PHACTR1 reduced excessive ICAM-1, VCAM-1 and VE-cadherin expression induced by oxidized low density lipoprotein (ox-LDL) in HCAECs. Additionally, silencing PHACTR1 alleviated p47phox phosphorylation and intracellular oxidative stress reflected by the reduction of ROS. Molecular experiments revealed that knockdown of PHACTR1 attenuated NF-κB activity without affecting IκBα and IKKα/β phosphorylation. In contrast, nuclear translation of p65 was blocked by depletion of PHACTR1. Furthermore, co-immunoprecipitation showed that PHACTR1 interacted with MRTF-A and p65 in HCAECs. Knockdown of MRTF-A suppressed the interaction of PHACTR1 with p65, subsequently blocking the nuclear translocation of p65. CONCLUSIONS Our finding suggest that silencing PHACTR1 alleviates the nuclear accumulation of p65 and NF-κB via interaction with MRTF-A, ensuing attenuating oxidative stress and inflammation in HCAECs.
Collapse
Affiliation(s)
- Zhihui Zhang
- Department of Cardiology, The Third Xiangya Hospital of Central South University, China
| | - Fenglin Jiang
- Department of Cardiology, The Third Xiangya Hospital of Central South University, China
| | - Lixiong Zeng
- Department of Cardiology, The Third Xiangya Hospital of Central South University, China
| | - Xiaoyan Wang
- Department of Cardiology, The Third Xiangya Hospital of Central South University, China
| | - Shan Tu
- Department of Cardiology, The Third Xiangya Hospital of Central South University, China.
| |
Collapse
|
29
|
Codina-Fauteux VA, Beaudoin M, Lalonde S, Lo KS, Lettre G. PHACTR1 splicing isoforms and eQTLs in atherosclerosis-relevant human cells. BMC MEDICAL GENETICS 2018; 19:97. [PMID: 29884117 PMCID: PMC5994109 DOI: 10.1186/s12881-018-0616-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/23/2018] [Indexed: 11/20/2022]
Abstract
Background Genome-wide association studies (GWAS) have identified a variant (rs9349379) at the phosphatase and actin regulator 1 (PHACTR1) locus that is associated with coronary artery disease (CAD). The same variant is also an expression quantitative trait locus (eQTL) for PHACTR1 in human coronary arteries (hCA). Here, we sought to characterize PHACTR1 splicing pattern in atherosclerosis-relevant human cells. We also explored how rs9349379 modulates the expression of the different PHACTR1 splicing isoforms. Methods We combined rapid amplification of cDNA ends (RACE) with next-generation long-read DNA sequencing to discover all PHACTR1 transcripts in many human tissues and cell types. We measured PHACTR1 transcripts by qPCR to identify transcript-specific eQTLs. Results We confirmed a brain-specific long transcript, a short transcript expressed in monocytes and four intermediate transcripts that are different due to alternative splicing of two in-frame exons. In contrast to a previous report, we confirmed that the PHACTR1 protein is present in vascular smooth muscle cells. In 158 hCA from our collection and the GTEx dataset, rs9349379 was only associated with the expression levels of the intermediate PHACTR1 transcripts. Conclusions Our comprehensive transcriptomic profiling of PHACTR1 indicates that this gene encodes six main transcripts. Five of them are expressed in hCA, where atherosclerotic plaques develop. In this tissue, genotypes at rs9349379 are associated with the expression of the intermediate transcripts, but not the immune-specific short transcript. This result suggests that rs9349379 may in part influence CAD by modulating the expression of intermediate PHACTR1 transcripts in endothelial or vascular smooth muscle cells found in hCA. Electronic supplementary material The online version of this article (10.1186/s12881-018-0616-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Valérie-Anne Codina-Fauteux
- Montreal Heart Institute, 5000 Bélanger Street, Montréal, Québec, H1T 1C8, Canada.,Université de Montréal, 2900 Boul. Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| | - Mélissa Beaudoin
- Montreal Heart Institute, 5000 Bélanger Street, Montréal, Québec, H1T 1C8, Canada
| | - Simon Lalonde
- Montreal Heart Institute, 5000 Bélanger Street, Montréal, Québec, H1T 1C8, Canada
| | - Ken Sin Lo
- Montreal Heart Institute, 5000 Bélanger Street, Montréal, Québec, H1T 1C8, Canada
| | - Guillaume Lettre
- Montreal Heart Institute, 5000 Bélanger Street, Montréal, Québec, H1T 1C8, Canada. .,Université de Montréal, 2900 Boul. Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada.
| |
Collapse
|
30
|
Actin Cross-Linking Toxin Is a Universal Inhibitor of Tandem-Organized and Oligomeric G-Actin Binding Proteins. Curr Biol 2018; 28:1536-1547.e9. [PMID: 29731300 DOI: 10.1016/j.cub.2018.03.065] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/06/2018] [Accepted: 03/28/2018] [Indexed: 11/20/2022]
Abstract
Delivery of bacterial toxins to host cells is hindered by host protective barriers. This obstruction dictates a remarkable efficiency of toxins, a single copy of which may kill a host cell. Efficiency of actin-targeting toxins is further hampered by an overwhelming abundance of their target. The actin cross-linking domain (ACD) toxins of Vibrio species and related bacterial genera catalyze the formation of covalently cross-linked actin oligomers. Recently, we reported that the ACD toxicity can be amplified via a multivalent inhibitory association of actin oligomers with actin assembly factors formins, suggesting that the oligomers may act as secondary toxins. Importantly, many proteins involved in nucleation, elongation, severing, branching, and bundling of actin filaments contain G-actin-binding Wiskott-Aldrich syndrome protein (WASP)-homology motifs 2 (WH2) organized in tandem and therefore may act as a multivalent platform for high-affinity interaction with the ACD-cross-linked actin oligomers. Using live-cell single-molecule speckle (SiMS) microscopy, total internal reflection fluorescence (TIRF) microscopy, and actin polymerization assays, we show that, in addition to formins, the oligomers bind with high affinity and potently inhibit several families of actin assembly factors: Ena/vasodilator-stimulated phosphorprotein (VASP); Spire; and the Arp2/3 complex, both in vitro and in live cells. As a result, ACD blocks the actin retrograde flow and membrane dynamics and disrupts association of Ena/VASP with adhesion complexes. This study defines ACD as a universal inhibitor of tandem-organized G-actin binding proteins that overcomes the abundance of actin by redirecting the toxicity cascade toward less abundant targets and thus leading to profound disorganization of the actin cytoskeleton and disruption of actin-dependent cellular functions.
Collapse
|
31
|
Ito H, Mizuno M, Noguchi K, Morishita R, Iwamoto I, Hara A, Nagata KI. Expression analyses of Phactr1 (phosphatase and actin regulator 1) during mouse brain development. Neurosci Res 2018; 128:50-57. [DOI: 10.1016/j.neures.2017.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 08/03/2017] [Accepted: 08/07/2017] [Indexed: 12/18/2022]
|
32
|
Aherrahrou R, Aherrahrou Z, Schunkert H, Erdmann J. Coronary artery disease associated gene Phactr1 modulates severity of vascular calcification in vitro. Biochem Biophys Res Commun 2017; 491:396-402. [DOI: 10.1016/j.bbrc.2017.07.090] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 12/20/2022]
|
33
|
Ben-Avraham D, Karasik D, Verghese J, Lunetta KL, Smith JA, Eicher JD, Vered R, Deelen J, Arnold AM, Buchman AS, Tanaka T, Faul JD, Nethander M, Fornage M, Adams HH, Matteini AM, Callisaya ML, Smith AV, Yu L, De Jager PL, Evans DA, Gudnason V, Hofman A, Pattie A, Corley J, Launer LJ, Knopman DS, Parimi N, Turner ST, Bandinelli S, Beekman M, Gutman D, Sharvit L, Mooijaart SP, Liewald DC, Houwing-Duistermaat JJ, Ohlsson C, Moed M, Verlinden VJ, Mellström D, van der Geest JN, Karlsson M, Hernandez D, McWhirter R, Liu Y, Thomson R, Tranah GJ, Uitterlinden AG, Weir DR, Zhao W, Starr JM, Johnson AD, Ikram MA, Bennett DA, Cummings SR, Deary IJ, Harris TB, Kardia SLR, Mosley TH, Srikanth VK, Windham BG, Newman AB, Walston JD, Davies G, Evans DS, Slagboom EP, Ferrucci L, Kiel DP, Murabito JM, Atzmon G. The complex genetics of gait speed: genome-wide meta-analysis approach. Aging (Albany NY) 2017; 9:209-246. [PMID: 28077804 PMCID: PMC5310665 DOI: 10.18632/aging.101151] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 12/26/2016] [Indexed: 01/08/2023]
Abstract
Emerging evidence suggests that the basis for variation in late-life mobility is attributable, in part, to genetic factors, which may become increasingly important with age. Our objective was to systematically assess the contribution of genetic variation to gait speed in older individuals. We conducted a meta-analysis of gait speed GWASs in 31,478 older adults from 17 cohorts of the CHARGE consortium, and validated our results in 2,588 older adults from 4 independent studies. We followed our initial discoveries with network and eQTL analysis of candidate signals in tissues. The meta-analysis resulted in a list of 536 suggestive genome wide significant SNPs in or near 69 genes. Further interrogation with Pathway Analysis placed gait speed as a polygenic complex trait in five major networks. Subsequent eQTL analysis revealed several SNPs significantly associated with the expression of PRSS16, WDSUB1 and PTPRT, which in addition to the meta-analysis and pathway suggested that genetic effects on gait speed may occur through synaptic function and neuronal development pathways. No genome-wide significant signals for gait speed were identified from this moderately large sample of older adults, suggesting that more refined physical function phenotypes will be needed to identify the genetic basis of gait speed in aging.
Collapse
Affiliation(s)
- Dan Ben-Avraham
- Department of Medicine and Genetics Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - David Karasik
- Institute for Aging Research, Hebrew SeniorLife, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02131, USA
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Joe Verghese
- Integrated Divisions of Cognitive & Motor Aging (Neurology) and Geriatrics (Medicine), Montefiore-Einstein Center for the Aging Brain, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kathryn L. Lunetta
- The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, MA 01702, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - John D. Eicher
- The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, MA 01702, USA
- Population Sciences Branch, National Heart Lung and Blood Institute, Framingham, MA 01702, USA
| | - Rotem Vered
- Psychology Department, University of Haifa, Haifa, Israel
| | - Joris Deelen
- Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
- Max Planck Institute for Biology of Ageing, Köln, Germany
| | - Alice M. Arnold
- Department of Biostatistics, University of Washington, Seattle, WA 98115, USA
| | - Aron S. Buchman
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60614, USA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, Baltimore MD 21224, USA
| | - Jessica D. Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| | - Maria Nethander
- Bioinformatics Core Facility, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Myriam Fornage
- The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Hieab H. Adams
- Department of Epidemiology, Erasmus MC, Rotterdam, Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Amy M. Matteini
- Division of Geriatric Medicine, Johns Hopkins Medical Institutes, Baltimore, MD 21224, USA
| | - Michele L. Callisaya
- Medicine, Peninsula Health, Peninsula Clinical School, Central Clinical School, Frankston, Melbourne, Victoria, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Albert V. Smith
- Icelandic Heart Association, Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60614, USA
| | - Philip L. De Jager
- Broad Institute of Harvard and MIT, Cambridge, Harvard Medical School, Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Denis A. Evans
- Rush Institute for Healthy Aging and Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Albert Hofman
- Department of Epidemiology, Erasmus MC, Rotterdam, Netherlands
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Alison Pattie
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Janie Corley
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Lenore J. Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Neeta Parimi
- California Pacific Medical Center Research Institute, San Francisco, CA 94107, USA
| | - Stephen T. Turner
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Marian Beekman
- Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Danielle Gutman
- Department of Human Biology, Faculty of Natural Science, University of Haifa, Haifa, Israel
| | - Lital Sharvit
- Department of Human Biology, Faculty of Natural Science, University of Haifa, Haifa, Israel
| | - Simon P. Mooijaart
- Gerontology and Geriatrics, Leiden University Medical Center, Leiden, Netherland
| | - David C. Liewald
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Jeanine J. Houwing-Duistermaat
- Genetical Statistics, Leiden University Medical Center, Leiden, Netherland. Department of Statistics, University of Leeds, Leeds, UK
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska, Academy, University of Gothenburg, Gothenburg, Sweden
| | - Matthijs Moed
- Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Dan Mellström
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska, Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Magnus Karlsson
- Clinical and Molecular Osteoporosis Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Dena Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - Rebekah McWhirter
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Russell Thomson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
- School of Computing, Engineering and Mathematics, University of Western Sydney, Sydney, Australia
| | - Gregory J. Tranah
- California Pacific Medical Center Research Institute, San Francisco, CA 94107, USA
| | - Andre G. Uitterlinden
- Department of Internal Medicine, Erasmus MC, and Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, The Netherlands
| | - David R. Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - John M. Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - Andrew D. Johnson
- The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, MA 01702, USA
- Population Sciences Branch, National Heart Lung and Blood Institute, Framingham, MA 01702, USA
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus MC, Rotterdam, Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, Netherlands
| | - David A. Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60614, USA
| | - Steven R. Cummings
- California Pacific Medical Center Research Institute, San Francisco, CA 94107, USA
| | - Ian J. Deary
- Department of Psychology, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Tamara B. Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sharon L. R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Thomas H. Mosley
- University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Velandai K. Srikanth
- Medicine, Peninsula Health, Peninsula Clinical School, Central Clinical School, Frankston, Melbourne, Victoria, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | | | - Ann B. Newman
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jeremy D. Walston
- Division of Geriatric Medicine, Johns Hopkins Medical Institutes, Baltimore, MD 21224, USA
| | - Gail Davies
- Department of Psychology, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Daniel S. Evans
- California Pacific Medical Center Research Institute, San Francisco, CA 94107, USA
| | - Eline P. Slagboom
- Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore MD 21224, USA
| | - Douglas P. Kiel
- Institute for Aging Research, Hebrew SeniorLife, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02131, USA
- Broad Institute of Harvard and MIT, Boston, MA 02131, USA
| | - Joanne M. Murabito
- The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, MA 01702, USA
- Section of General Internal Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Gil Atzmon
- Department of Medicine and Genetics Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Human Biology, Faculty of Natural Science, University of Haifa, Haifa, Israel
| |
Collapse
|
34
|
Abstract
Thoracic aortic aneurysm is a potentially life-threatening condition in that it places patients at risk for aortic dissection or rupture. However, our modern understanding of the pathogenesis of thoracic aortic aneurysm is quite limited. A genetic predisposition to thoracic aortic aneurysm has been established, and gene discovery in affected families has identified several major categories of gene alterations. The first involves mutations in genes encoding various components of the transforming growth factor beta (TGF-β) signaling cascade (FBN1, TGFBR1, TGFBR2, TGFB2, TGFB3, SMAD2, SMAD3 and SKI), and these conditions are known collectively as the TGF-β vasculopathies. The second set of genes encode components of the smooth muscle contractile apparatus (ACTA2, MYH11, MYLK, and PRKG1), a group called the smooth muscle contraction vasculopathies. Mechanistic hypotheses based on these discoveries have shaped rational therapies, some of which are under clinical evaluation. This review discusses published data on genes involved in thoracic aortic aneurysm and attempts to explain divergent hypotheses of aneurysm origin.
Collapse
Affiliation(s)
- Eric M Isselbacher
- From Thoracic Aortic Center (E.M.I., C.L.L.C., M.E.L.), Cardiovascular Genetics Program (M.E.L.), Cardiovascular Research Center (C.L.L.C., M.E.L.), and Cardiology Division (E.M.I., C.L.L.C., M.E.L.), Department of Medicine, and Pediatric Cardiology Division, Department of Pediatrics (M.E.L.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Christian Lacks Lino Cardenas
- From Thoracic Aortic Center (E.M.I., C.L.L.C., M.E.L.), Cardiovascular Genetics Program (M.E.L.), Cardiovascular Research Center (C.L.L.C., M.E.L.), and Cardiology Division (E.M.I., C.L.L.C., M.E.L.), Department of Medicine, and Pediatric Cardiology Division, Department of Pediatrics (M.E.L.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Mark E Lindsay
- From Thoracic Aortic Center (E.M.I., C.L.L.C., M.E.L.), Cardiovascular Genetics Program (M.E.L.), Cardiovascular Research Center (C.L.L.C., M.E.L.), and Cardiology Division (E.M.I., C.L.L.C., M.E.L.), Department of Medicine, and Pediatric Cardiology Division, Department of Pediatrics (M.E.L.), Massachusetts General Hospital, Harvard Medical School, Boston.
| |
Collapse
|
35
|
Rodríguez-Pérez JM, Blachman-Braun R, Pomerantz A, Vargas-Alarcón G, Posadas-Sánchez R, Pérez-Hernández N. Possible role of intronic polymorphisms in the PHACTR1 gene on the development of cardiovascular disease. Med Hypotheses 2016; 97:64-70. [PMID: 27876132 DOI: 10.1016/j.mehy.2016.10.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 10/19/2016] [Indexed: 11/24/2022]
Abstract
Cardiovascular disease (CVD) is a complex multifactorial and polygenetic disease in which the interaction of numerous genes, genetic variants, and environmental factors plays a major role in its development. In an attempt to demonstrate the association between certain genetic variants and CVD, researchers have run large genomic wild association studies (GWAS) in recent decades. These studies have correlated several genomic variants with the presence of CVD. Recently, certain polymorphisms in the phosphatase and actin regulator 1 (PHACTR1) gene have been shown to be associated with CVD (i.e., coronary artery disease, coronary artery calcification, early onset myocardial infarction, cervical artery dissection and hypertension) in different ethnic groups. It is important to state that all of the described PHACTR1 genetic variants associated with CVD are located in non-translating gene regions known as introns. Thus, the purpose of this article is to hypothesize the effect of certain intronic polymorphisms in the PHACTR1 gene on pathological processes in the cardiovascular system. In addition, we present compelling evidence that supports this hypothesis as well as a methodology that could be used to assess the allelic effect using in vitro and in vivo models, which will ultimately demonstrate the pathophysiological contribution of PHACTR1 intronic polymorphisms to the development of CVD.
Collapse
Affiliation(s)
- José Manuel Rodríguez-Pérez
- Department of Molecular Biology, National Institute of Cardiology "Ignacio Chávez", Mexico City 14080, Mexico
| | - Ruben Blachman-Braun
- Department of Molecular Biology, National Institute of Cardiology "Ignacio Chávez", Mexico City 14080, Mexico
| | - Alan Pomerantz
- Department of Oncology and Hematology, National Institute of Medical Sciences and Nutrition "Salvador Zubirán", Mexico City 14080, Mexico
| | - Gilberto Vargas-Alarcón
- Department of Molecular Biology, National Institute of Cardiology "Ignacio Chávez", Mexico City 14080, Mexico
| | - Rosalinda Posadas-Sánchez
- Department of Endocrinology, National Institute of Cardiology "Ignacio Chávez", Mexico City 14080, Mexico
| | - Nonanzit Pérez-Hernández
- Department of Molecular Biology, National Institute of Cardiology "Ignacio Chávez", Mexico City 14080, Mexico.
| |
Collapse
|
36
|
Stimulation of Slack K(+) Channels Alters Mass at the Plasma Membrane by Triggering Dissociation of a Phosphatase-Regulatory Complex. Cell Rep 2016; 16:2281-8. [PMID: 27545877 DOI: 10.1016/j.celrep.2016.07.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 09/27/2015] [Accepted: 07/13/2016] [Indexed: 11/23/2022] Open
Abstract
Human mutations in the cytoplasmic C-terminal domain of Slack sodium-activated potassium (KNa) channels result in childhood epilepsy with severe intellectual disability. Slack currents can be increased by pharmacological activators or by phosphorylation of a Slack C-terminal residue by protein kinase C. Using an optical biosensor assay, we find that Slack channel stimulation in neurons or transfected cells produces loss of mass near the plasma membrane. Slack mutants associated with intellectual disability fail to trigger any change in mass. The loss of mass results from the dissociation of the protein phosphatase 1 (PP1) targeting protein, Phactr-1, from the channel. Phactr1 dissociation is specific to wild-type Slack channels and is not observed when related potassium channels are stimulated. Our findings suggest that Slack channels are coupled to cytoplasmic signaling pathways and that dysregulation of this coupling may trigger the aberrant intellectual development associated with specific childhood epilepsies.
Collapse
|
37
|
Panayiotou R, Miralles F, Pawlowski R, Diring J, Flynn HR, Skehel M, Treisman R. Phosphorylation acts positively and negatively to regulate MRTF-A subcellular localisation and activity. eLife 2016; 5:e15460. [PMID: 27304076 PMCID: PMC4963197 DOI: 10.7554/elife.15460] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 06/14/2016] [Indexed: 11/29/2022] Open
Abstract
The myocardin-related transcription factors (MRTF-A and MRTF-B) regulate cytoskeletal genes through their partner transcription factor SRF. The MRTFs bind G-actin, and signal-regulated changes in cellular G-actin concentration control their nuclear accumulation. The MRTFs also undergo Rho- and ERK-dependent phosphorylation, but the function of MRTF phosphorylation, and the elements and signals involved in MRTF-A nuclear export are largely unexplored. We show that Rho-dependent MRTF-A phosphorylation reflects relief from an inhibitory function of nuclear actin. We map multiple sites of serum-induced phosphorylation, most of which are S/T-P motifs and show that S/T-P phosphorylation is required for transcriptional activation. ERK-mediated S98 phosphorylation inhibits assembly of G-actin complexes on the MRTF-A regulatory RPEL domain, promoting nuclear import. In contrast, S33 phosphorylation potentiates the activity of an autonomous Crm1-dependent N-terminal NES, which cooperates with five other NES elements to exclude MRTF-A from the nucleus. Phosphorylation thus plays positive and negative roles in the regulation of MRTF-A.
Collapse
Affiliation(s)
- Richard Panayiotou
- Signaling and Transcription Group, Francis Crick Institute, London, United Kingdom
| | - Francesc Miralles
- Signaling and Transcription Group, Francis Crick Institute, London, United Kingdom
| | - Rafal Pawlowski
- Signaling and Transcription Group, Francis Crick Institute, London, United Kingdom
| | - Jessica Diring
- Signaling and Transcription Group, Francis Crick Institute, London, United Kingdom
| | - Helen R Flynn
- Mass Spectrometry Science Technology Platform, Francis Crick Institute, London, United Kingdom
| | - Mark Skehel
- Mass Spectrometry Science Technology Platform, Francis Crick Institute, London, United Kingdom
| | - Richard Treisman
- Signaling and Transcription Group, Francis Crick Institute, London, United Kingdom
| |
Collapse
|
38
|
Reschen ME, Lin D, Chalisey A, Soilleux EJ, O'Callaghan CA. Genetic and environmental risk factors for atherosclerosis regulate transcription of phosphatase and actin regulating gene PHACTR1. Atherosclerosis 2016; 250:95-105. [PMID: 27187934 PMCID: PMC4917897 DOI: 10.1016/j.atherosclerosis.2016.04.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 03/20/2016] [Accepted: 04/26/2016] [Indexed: 12/20/2022]
Abstract
Background and aims Coronary artery disease (CAD) risk is associated with non-coding genetic variants at the phosphatase and actin regulating protein 1(PHACTR1) gene locus. The PHACTR1 gene encodes an actin-binding protein with phosphatase regulating activity. The mechanism whereby PHACTR1 influences CAD risk is unknown. We hypothesized that PHACTR1 would be expressed in human cell types relevant to CAD and regulated by atherogenic or genetic factors. Methods and results Using immunohistochemistry, we demonstrate that PHACTR1 protein is expressed strongly in human atherosclerotic plaque macrophages, lipid-laden foam cells, adventitial lymphocytes and endothelial cells. Using a combination of genomic analysis and molecular techniques, we demonstrate that PHACTR1 is expressed as multiple previously uncharacterized transcripts in macrophages, foam cells, lymphocytes and endothelial cells. Immunoblotting confirmed a total absence of PHACTR1 in vascular smooth muscle cells. Real-time quantitative PCR showed that PHACTR1 is regulated by atherogenic and inflammatory stimuli. In aortic endothelial cells, oxLDL and TNF-alpha both upregulated an intermediate length transcript. A short transcript expressed only in immune cells was upregulated in macrophages by oxidized low-density lipoprotein, and oxidized phospholipids but suppressed by lipopolysaccharide or TNF-alpha. In primary human macrophages, we identified a novel expression quantitative trait locus (eQTL) specific for this short transcript, whereby the risk allele at CAD risk SNP rs9349379 is associated with reduced PHACTR1 expression, similar to the effect of an inflammatory stimulus. Conclusions Our data demonstrate that PHACTR1 is a key atherosclerosis candidate gene since it is regulated by atherogenic stimuli in macrophages and endothelial cells and we identify an effect of the genetic risk variant on PHACTR1 expression in macrophages that is similar to that of an inflammatory stimulus. PHACTR1 is expressed as two transcripts in both immune and endothelial cells in human atherosclerotic plaque. Oxidized-LDL upregulates a short PHACTR1 transcript, but suppresses an intermediate length transcript in macrophages. Lipopolysaccharide and TNF-alpha cause the opposite effect with strong suppression of the short transcript in macrophages. The coronary artery disease risk SNP, rs9349379, is associated with expression of the short transcript in macrophages. The effect of the coronary artery disease risk allele on PHACTR1 mirrors that of inflammatory stimuli.
Collapse
Affiliation(s)
- Michael E Reschen
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, United Kingdom
| | - Da Lin
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, United Kingdom
| | - Anil Chalisey
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, United Kingdom
| | - Elizabeth J Soilleux
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford and Department of Cellular Pathology, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Christopher A O'Callaghan
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, United Kingdom.
| |
Collapse
|
39
|
Schumann T, Adhikary T, Wortmann A, Finkernagel F, Lieber S, Schnitzer E, Legrand N, Schober Y, Nockher WA, Toth PM, Diederich WE, Nist A, Stiewe T, Wagner U, Reinartz S, Müller-Brüsselbach S, Müller R. Deregulation of PPARβ/δ target genes in tumor-associated macrophages by fatty acid ligands in the ovarian cancer microenvironment. Oncotarget 2016; 6:13416-33. [PMID: 25968567 PMCID: PMC4537024 DOI: 10.18632/oncotarget.3826] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/29/2015] [Indexed: 01/04/2023] Open
Abstract
The nuclear receptor peroxisome proliferator-activated receptor β/δ (PPARβ/δ) is a lipid ligand-inducible transcription factor associated with macrophage polarization. However, its function in tumor-associated macrophages (TAMs) has not been investigated to date. Here, we report the PPARβ/δ-regulated transcriptome and cistrome for TAMs from ovarian carcinoma patients. Comparison with monocyte-derived macrophages shows that the vast majority of direct PPARβ/δ target genes are upregulated in TAMs and largely refractory to synthetic agonists, but repressible by inverse agonists. Besides genes with metabolic functions, these include cell type-selective genes associated with immune regulation and tumor progression, e.g., LRP5, CD300A, MAP3K8 and ANGPTL4. This deregulation is not due to increased expression of PPARβ/δ or its enhanced recruitment to target genes. Instead, lipidomic analysis of malignancy-associated ascites revealed high concentrations of polyunsaturated fatty acids, in particular linoleic acid, acting as potent PPARβ/δ agonists in macrophages. These fatty acid ligands accumulate in lipid droplets in TAMs, thereby providing a reservoir of PPARβ/δ ligands. These observations suggest that the deregulation of PPARβ/δ target genes by ligands of the tumor microenvironment contributes to the pro-tumorigenic polarization of ovarian carcinoma TAMs. This conclusion is supported by the association of high ANGPTL4 expression with a shorter relapse-free survival in serous ovarian carcinoma.
Collapse
Affiliation(s)
- Tim Schumann
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Till Adhikary
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Annika Wortmann
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Florian Finkernagel
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Sonja Lieber
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Evelyn Schnitzer
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Nathalie Legrand
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Yvonne Schober
- Metabolomics Core Facility and Institute of Laboratory Medicine and Pathobiochemistry, Philipps University, Marburg, Germany
| | - W Andreas Nockher
- Metabolomics Core Facility and Institute of Laboratory Medicine and Pathobiochemistry, Philipps University, Marburg, Germany
| | - Philipp M Toth
- Medicinal Chemistry Core Facility and Institute of Pharmaceutical Chemistry, Philipps University, Marburg, Germany
| | - Wibke E Diederich
- Medicinal Chemistry Core Facility and Institute of Pharmaceutical Chemistry, Philipps University, Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Silke Reinartz
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | | | - Rolf Müller
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| |
Collapse
|
40
|
Navarro AP, Collins MA, Folker ES. The nucleus is a conserved mechanosensation and mechanoresponse organelle. Cytoskeleton (Hoboken) 2016; 73:59-67. [PMID: 26849407 DOI: 10.1002/cm.21277] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 01/22/2016] [Accepted: 01/22/2016] [Indexed: 11/05/2022]
Abstract
Cells in vivo exist in a dynamic environment where they experience variable mechanical influences. The precise mechanical environment influences cell-cell interactions, cell-extracellular matrix interactions, and in-turn, cell morphology and cell function. Therefore, the ability of each cell to constantly and rapidly alter their behavior in response to variations in their mechanical environment is essential for cell viability, development, and function. Mechanotransduction, the process by which mechanical force is translated into a biochemical signal to activate downstream cellular responses, is thus crucial to cell function during development and homeostasis. Although much research has focused on how protein complexes at the cell cortex respond to mechanical stress to initiate mechanotransduction, the nucleus has emerged as crucial to the ability of the cell to perceive and respond to changes in its mechanical environment. This additional method for mechanosensing allows for direct transmission of force through the cytoskeleton to the nucleus, which can increase the speed at which a cell changes its transcriptional profile. This review discusses recent work demonstrating the importance of the nucleus in mediating the cellular response to internal and external force, establishing the nucleus as an important mechanosensing organelle.
Collapse
Affiliation(s)
- Alexandra P Navarro
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02142
| | - Mary Ann Collins
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, 02467
| | - Eric S Folker
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, 02467
| |
Collapse
|
41
|
Turner AW, McPherson R. PHACTR1: Functional Clues Linking a Genome-Wide Association Study Locus to Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2015; 35:1293-5. [PMID: 25995042 DOI: 10.1161/atvbaha.115.305680] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Adam W Turner
- From the Departments of Medicine and Biochemistry, Atherogenomics Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Ruth McPherson
- From the Departments of Medicine and Biochemistry, Atherogenomics Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
42
|
Kelloniemi A, Szabo Z, Serpi R, Näpänkangas J, Ohukainen P, Tenhunen O, Kaikkonen L, Koivisto E, Bagyura Z, Kerkelä R, Leosdottir M, Hedner T, Melander O, Ruskoaho H, Rysä J. The Early-Onset Myocardial Infarction Associated PHACTR1 Gene Regulates Skeletal and Cardiac Alpha-Actin Gene Expression. PLoS One 2015; 10:e0130502. [PMID: 26098115 PMCID: PMC4476650 DOI: 10.1371/journal.pone.0130502] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/19/2015] [Indexed: 11/19/2022] Open
Abstract
The phosphatase and actin regulator 1 (PHACTR1) locus is a very commonly identified hit in genome-wide association studies investigating coronary artery disease and myocardial infarction (MI). However, the function of PHACTR1 in the heart is still unknown. We characterized the mechanisms regulating Phactr1 expression in the heart, used adenoviral gene delivery to investigate the effects of Phactr1 on cardiac function, and analyzed the relationship between MI associated PHACTR1 allele and cardiac function in human subjects. Phactr1 mRNA and protein levels were markedly reduced (60%, P<0.01 and 90%, P<0.001, respectively) at 1 day after MI in rats. When the direct myocardial effects of Phactr1 were studied, the skeletal α-actin to cardiac α-actin isoform ratio was significantly higher (1.5-fold, P<0.05) at 3 days but 40% lower (P<0.05) at 2 weeks after adenovirus-mediated Phactr1 gene delivery into the anterior wall of the left ventricle. Similarly, the skeletal α-actin to cardiac α-actin ratio was lower at 2 weeks in infarcted hearts overexpressing Phactr1. In cultured neonatal cardiac myocytes, adenovirus-mediated Phactr1 overexpression for 48 hours markedly increased the skeletal α-actin to cardiac α-actin ratio, this being associated with an enhanced DNA binding activity of serum response factor. Phactr1 overexpression exerted no major effects on the expression of other cardiac genes or LV structure and function in normal and infarcted hearts during 2 weeks’ follow-up period. In human subjects, MI associated PHACTR1 allele was not associated significantly with cardiac function (n = 1550). Phactr1 seems to regulate the skeletal to cardiac α-actin isoform ratio.
Collapse
Affiliation(s)
- Annina Kelloniemi
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Zoltan Szabo
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Raisa Serpi
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Juha Näpänkangas
- Department of Pathology, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Pauli Ohukainen
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Olli Tenhunen
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Leena Kaikkonen
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Elina Koivisto
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Zsolt Bagyura
- Heart Center, Semmelweis University, Budapest, Hungary
| | - Risto Kerkelä
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | | | - Thomas Hedner
- Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Olle Melander
- Lund University, Department of Clinical Sciences, Malmö, Sweden
| | - Heikki Ruskoaho
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- * E-mail: (JR); (HR)
| | - Jaana Rysä
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- * E-mail: (JR); (HR)
| |
Collapse
|
43
|
Zhang Y, Wang L, Zhang Z, Zhang Z, Zhou S, Cao L, Cai B, Liu K, Bai W, Xie X, Fan W, Liu X, Lu G, Xu G. Shared and discrepant susceptibility for carotid artery and aortic arch calcification: A genetic association study. Atherosclerosis 2015; 241:371-5. [PMID: 26071660 DOI: 10.1016/j.atherosclerosis.2015.05.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/19/2015] [Accepted: 05/22/2015] [Indexed: 11/16/2022]
Abstract
Genome-wide association studies (GWASs) have identified several risk loci for coronary artery calcification. Four single-nucleotide polymorphisms (SNPs, rs1537370, rs1333049, rs2026458 and rs9349379) were associated with coronary artery calcification with P values less than 5 × 10(-8) in GWASs. It is unclear if these associations exist in other vascular beds. Thus, we evaluated the impacts of these four SNPs on carotid artery and aortic arch calcification in this study. Computed tomography was applied to quantify the calcification of carotid artery and aortic arch. 860 patients with stroke completed calcification quantification and genotype testing were included in data analysis. Each SNP was evaluated for the association with carotid artery calcification, and with aortic arch calcification using generalized linear model. Among the four tested SNPs, rs2026458 was associated with calcification in both carotid artery (β = 0.31, 95% confidence interval [CI] 0.10-0.52, P = 0.003) and aortic arch (β = 0.32, 95% CI 0.10-0.54, P = 0.004), while rs1333049 was only associated with carotid artery calcification (β = 0.28, 95% CI 0.06-0.50, P = 0.011). In gender-stratified analyses, rs2026458 had significant impacts on carotid artery (P = 0.003) and aortic arch calcification (P = 0.008) in male, but not in female patients; while rs1537370 was significantly associated with carotid artery calcification in female (P = 0.013), but not in male patients. In conclusion, SNPs associated with coronary artery calcification may also increase the risk of calcification in other arteries such as carotid artery and aortic arch.
Collapse
Affiliation(s)
- Yumeng Zhang
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Li Wang
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Zhizhong Zhang
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Zongjun Zhang
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Shuyu Zhou
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Liping Cao
- Department of Neurology, Third Affiliated Hospital, Soochow University, Changzhou, Jiangsu, China
| | - Biyang Cai
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Keting Liu
- Department of Neurology, Jinling Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wen Bai
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Xia Xie
- Department of Neurology, Jinling Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenping Fan
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Xinfeng Liu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Guangming Lu
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China.
| | - Gelin Xu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China.
| |
Collapse
|
44
|
Adhikary T, Wortmann A, Schumann T, Finkernagel F, Lieber S, Roth K, Toth PM, Diederich WE, Nist A, Stiewe T, Kleinesudeik L, Reinartz S, Müller-Brüsselbach S, Müller R. The transcriptional PPARβ/δ network in human macrophages defines a unique agonist-induced activation state. Nucleic Acids Res 2015; 43:5033-51. [PMID: 25934804 PMCID: PMC4446423 DOI: 10.1093/nar/gkv331] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/01/2015] [Indexed: 02/06/2023] Open
Abstract
Peroxisome proliferator-activated receptor β/δ (PPARβ/δ) is a lipid ligand-inducible transcription factor with established metabolic functions, whereas its anti-inflammatory function is poorly understood. To address this issue, we determined the global PPARβ/δ-regulated signaling network in human monocyte-derived macrophages. Besides cell type-independent, canonical target genes with metabolic and immune regulatory functions we identified a large number of inflammation-associated NFκB and STAT1 target genes that are repressed by agonists. Accordingly, PPARβ/δ agonists inhibited the expression of multiple pro-inflammatory mediators and induced an anti-inflammatory, IL-4-like morphological phenotype. Surprisingly, bioinformatic analyses also identified immune stimulatory effects. Consistent with this prediction, PPARβ/δ agonists enhanced macrophage survival under hypoxic stress and stimulated CD8+ T cell activation, concomitantly with the repression of immune suppressive target genes and their encoded products CD274 (PD-1 ligand), CD32B (inhibitory Fcγ receptor IIB) and indoleamine 2,3-dioxygenase 1 (IDO-1), as well as a diminished release of the immune suppressive IDO-1 metabolite kynurenine. Comparison with published data revealed a significant overlap of the PPARβ/δ transcriptome with coexpression modules characteristic of both anti-inflammatory and pro-inflammatory cytokines. Our findings indicate that PPARβ/δ agonists induce a unique macrophage activation state with strong anti-inflammatory but also specific immune stimulatory components, pointing to a context-dependent function of PPARβ/δ in immune regulation.
Collapse
Affiliation(s)
- Till Adhikary
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Annika Wortmann
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Tim Schumann
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Florian Finkernagel
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Sonja Lieber
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Katrin Roth
- Cellular Imaging Core Facility, Philipps University, Center for Tumor Biology and Immunology (ZTI), 35043 Marburg, Germany
| | - Philipp M Toth
- Medicinal Chemistry Core Facility and Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Wibke E Diederich
- Medicinal Chemistry Core Facility and Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Lara Kleinesudeik
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Silke Reinartz
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Sabine Müller-Brüsselbach
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| |
Collapse
|
45
|
Beaudoin M, Gupta RM, Won HH, Lo KS, Do R, Henderson CA, Lavoie-St-Amour C, Langlois S, Rivas D, Lehoux S, Kathiresan S, Tardif JC, Musunuru K, Lettre G. Myocardial Infarction-Associated SNP at 6p24 Interferes With MEF2 Binding and Associates With PHACTR1 Expression Levels in Human Coronary Arteries. Arterioscler Thromb Vasc Biol 2015; 35:1472-1479. [PMID: 25838425 DOI: 10.1161/atvbaha.115.305534] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/18/2015] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Coronary artery disease (CAD), including myocardial infarction (MI), is the main cause of death in the world. Genome-wide association studies have identified dozens of single nucleotide polymorphisms (SNPs) associated with CAD/MI. One of the most robust CAD/MI genetic associations is with intronic SNPs in the gene PHACTR1 on chromosome 6p24. How these PHACTR1 SNPs influence CAD/MI risk, and whether PHACTR1 itself is the causal gene at the locus, is currently unknown. APPROACH AND RESULTS Using genetic fine-mapping and DNA resequencing experiments, we prioritized an intronic SNP (rs9349379) in PHACTR1 as causal variant. We showed that this variant is an expression quantitative trait locus for PHACTR1 expression in human coronary arteries. Experiments in endothelial cell extracts confirmed that alleles at rs9349379 are differentially bound by the transcription factors myocyte enhancer factor-2. We engineered a deletion of this myocyte enhancer factor-2-binding site using CRISPR/Cas9 genome-editing methodology. Heterozygous endothelial cells carrying this deletion express 35% less PHACTR1. Finally, we found no evidence that PHACTR1 expression levels are induced when stimulating human endothelial cells with vascular endothelial growth factor, tumor necrosis factor-α, or shear stress. CONCLUSIONS Our results establish a link between intronic SNPs in PHACTR1, myocyte enhancer factor-2 binding, and transcriptional functions at the locus, PHACTR1 expression levels in coronary arteries and CAD/MI risk. Because PHACTR1 SNPs are not associated with the traditional risk factors for CAD/MI (eg, blood lipids or pressure, diabetes mellitus), our results suggest that PHACTR1 may influence CAD/MI risk through as yet unknown mechanisms in the vascular endothelium.
Collapse
Affiliation(s)
- Mélissa Beaudoin
- Montreal Heart Institute, 5000 Bélanger Street, Montréal, Québec, H1T 1C8, Canada
| | - Rajat M Gupta
- Department of Stem Cell and Regenerative Biology, Harvard University, and Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Hong-Hee Won
- Center of Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Ken Sin Lo
- Montreal Heart Institute, 5000 Bélanger Street, Montréal, Québec, H1T 1C8, Canada
| | - Ron Do
- Center of Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Christopher A Henderson
- Department of Stem Cell and Regenerative Biology, Harvard University, and Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
| | | | - Simon Langlois
- Montreal Heart Institute, 5000 Bélanger Street, Montréal, Québec, H1T 1C8, Canada
| | - Daniel Rivas
- Lady Davis Institute for Medical Research, McGill University, 3755 Côte Sainte-Catherine, Montreal, Quebec, H3T 1E2, Canada
| | - Stephanie Lehoux
- Lady Davis Institute for Medical Research, McGill University, 3755 Côte Sainte-Catherine, Montreal, Quebec, H3T 1E2, Canada
| | - Sekar Kathiresan
- Center of Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Jean-Claude Tardif
- Montreal Heart Institute, 5000 Bélanger Street, Montréal, Québec, H1T 1C8, Canada
- Department of Medicine, Université de Montréal, 2900 Boul. Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| | - Kiran Musunuru
- Department of Stem Cell and Regenerative Biology, Harvard University, and Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Guillaume Lettre
- Montreal Heart Institute, 5000 Bélanger Street, Montréal, Québec, H1T 1C8, Canada
- Department of Medicine, Université de Montréal, 2900 Boul. Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| |
Collapse
|
46
|
Chambers JE, Dalton LE, Clarke HJ, Malzer E, Dominicus CS, Patel V, Moorhead G, Ron D, Marciniak SJ. Actin dynamics tune the integrated stress response by regulating eukaryotic initiation factor 2α dephosphorylation. eLife 2015; 4:e04872. [PMID: 25774599 PMCID: PMC4394351 DOI: 10.7554/elife.04872] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 03/12/2015] [Indexed: 12/23/2022] Open
Abstract
Four stress-sensing kinases phosphorylate the alpha subunit of eukaryotic translation initiation factor 2 (eIF2α) to activate the integrated stress response (ISR). In animals, the ISR is antagonised by selective eIF2α phosphatases comprising a catalytic protein phosphatase 1 (PP1) subunit in complex with a PPP1R15-type regulatory subunit. An unbiased search for additional conserved components of the PPP1R15-PP1 phosphatase identified monomeric G-actin. Like PP1, G-actin associated with the functional core of PPP1R15 family members and G-actin depletion, by the marine toxin jasplakinolide, destabilised the endogenous PPP1R15A-PP1 complex. The abundance of the ternary PPP1R15-PP1-G-actin complex was responsive to global changes in the polymeric status of actin, as was its eIF2α-directed phosphatase activity, while localised G-actin depletion at sites enriched for PPP1R15 enhanced eIF2α phosphorylation and the downstream ISR. G-actin's role as a stabilizer of the PPP1R15-containing holophosphatase provides a mechanism for integrating signals regulating actin dynamics with stresses that trigger the ISR.
Collapse
Affiliation(s)
- Joseph E Chambers
- Cambridge Institute for Medical Research, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Wellcome Trust MRC Building, University of Cambridge, Cambridge, United Kingdom
| | - Lucy E Dalton
- Cambridge Institute for Medical Research, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Wellcome Trust MRC Building, University of Cambridge, Cambridge, United Kingdom
| | - Hanna J Clarke
- Wellcome Trust MRC Building, University of Cambridge, Cambridge, United Kingdom
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Elke Malzer
- Cambridge Institute for Medical Research, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Wellcome Trust MRC Building, University of Cambridge, Cambridge, United Kingdom
| | - Caia S Dominicus
- Cambridge Institute for Medical Research, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Wellcome Trust MRC Building, University of Cambridge, Cambridge, United Kingdom
| | - Vruti Patel
- Cambridge Institute for Medical Research, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Wellcome Trust MRC Building, University of Cambridge, Cambridge, United Kingdom
| | - Greg Moorhead
- Department of Biological Sciences, University of Calgary, Calgary, Canada
| | - David Ron
- Wellcome Trust MRC Building, University of Cambridge, Cambridge, United Kingdom
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Stefan J Marciniak
- Wellcome Trust MRC Building, University of Cambridge, Cambridge, United Kingdom
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
47
|
Itoh A, Uchiyama A, Taniguchi S, Sagara J. Phactr3/scapinin, a member of protein phosphatase 1 and actin regulator (phactr) family, interacts with the plasma membrane via basic and hydrophobic residues in the N-terminus. PLoS One 2014; 9:e113289. [PMID: 25405772 PMCID: PMC4236165 DOI: 10.1371/journal.pone.0113289] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 10/21/2014] [Indexed: 11/22/2022] Open
Abstract
Proteins that belong to the protein phosphatase 1 and actin regulator (phactr) family are involved in cell motility and morphogenesis. However, the mechanisms that regulate the actin cytoskeleton are poorly understood. We have previously shown that phactr3, also known as scapinin, localizes to the plasma membrane, including lamellipodia and membrane ruffles. In the present study, experiments using deletion and point mutants showed that the basic and hydrophobic residues in the N-terminus play crucial roles in the localization to the plasma membrane. A BH analysis (http://helixweb.nih.gov/bhsearch) is a program developed to identify membrane-binding domains that comprise basic and hydrophobic residues in membrane proteins. We applied this program to phactr3. The results of the BH plot analysis agreed with the experimentally determined region that is responsible for the localization of phactr3 to the plasma membrane. In vitro experiments showed that the N-terminal itself binds to liposomes and acidic phospholipids. In addition, we showed that the interaction with the plasma membrane via the N-terminal membrane-binding sequence is required for phactr3-induced morphological changes in Cos7 cells. The membrane-binding sequence in the N-terminus is highly conserved in all members of the phactr family. Our findings may provide a molecular basis for understanding the mechanisms that allow phactr proteins to regulate cell morphogenesis.
Collapse
Affiliation(s)
- Akihiro Itoh
- Department of Biomedical Laboratory Sciences, Health Sciences, Shinshu University, Matsumoto, Japan
| | - Atsushi Uchiyama
- Department of Biomedical Laboratory Sciences, Health Sciences, Shinshu University, Matsumoto, Japan
| | - Shunichiro Taniguchi
- Department of Molecular Oncology, Medical Sciences, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Junji Sagara
- Department of Biomedical Laboratory Sciences, Health Sciences, Shinshu University, Matsumoto, Japan
- * E-mail:
| |
Collapse
|
48
|
RPEL proteins are the molecular targets for CCG-1423, an inhibitor of Rho signaling. PLoS One 2014; 9:e89016. [PMID: 24558465 PMCID: PMC3928398 DOI: 10.1371/journal.pone.0089016] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 01/13/2014] [Indexed: 01/25/2023] Open
Abstract
Epithelial–msenchymal transition (EMT) is closely associated with cancer and tissue fibrosis. The nuclear accumulation of myocardin-related transcription factor A (MRTF-A/MAL/MKL1) plays a vital role in EMT. In various cells treated with CCG-1423, a novel inhibitor of Rho signaling, the nuclear accumulation of MRTF-A is inhibited. However, the molecular target of this inhibitor has not yet been identified. In this study, we investigated the mechanism of this effect of CCG-1423. The interaction between MRTF-A and importin α/β1 was inhibited by CCG-1423, but monomeric G-actin binding to MRTF-A was not inhibited. We coupled Sepharose with CCG-1423 (CCG-1423 Sepharose) to investigate this mechanism. A pull-down assay using CCG-1423 Sepharose revealed the direct binding of CCG-1423 to MRTF-A. Furthermore, we found that the N-terminal basic domain (NB) of MRTF-A, which acts as a functional nuclear localization signal (NLS) of MRTF-A, was the binding site for CCG-1423. G-actin did not bind to CCG-1423 Sepharose, but the interaction between MRTF-A and CCG-1423 Sepharose was reduced in the presence of G-actin. We attribute this result to the high binding affinity of MRTF-A for G-actin and the proximity of NB to G-actin-binding sites (RPEL motifs). Therefore, when MRTF-A forms a complex with G-actin, the binding of CCG-1423 to NB is expected to be blocked. NF-E2 related factor 2, which contains three distinct basic amino acid-rich NLSs, did not bind to CCG-1423 Sepharose, but other RPEL-containing proteins such as MRTF-B, myocardin, and Phactr1 bound to CCG-1423 Sepharose. These results suggest that the specific binding of CCG-1423 to the NLSs of RPEL-containing proteins. Our proposal to explain the inhibitory action of CCG-1423 is as follows: When the G-actin pool is depleted, CCG-1423 binds specifically to the NLS of MRTF-A/B and prevents the interaction between MRTF-A/B and importin α/β1, resulting in inhibition of the nuclear import of MRTF-A/B.
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Since 2007, genome-wide association studies (GWAS) have led to the identification of numerous loci of atherosclerotic cardiovascular disease. The majority of these loci harbor genes previously not known to be involved in atherogenesis. In this review, we summarize the recent progress in understanding the pathophysiology of genetic variants in atherosclerosis. RECENT FINDINGS Fifty-eight loci with P < 10⁻⁷ have been identified in GWAS for coronary heart disease and myocardial infarction. Of these, 23 loci (40%) overlap with GWAS loci of classical risk factors such as lipids, blood pressure, and diabetes mellitus, suggesting a potential causal relation. The vast majority of the remaining 35 loci (60%) are at genomic regions where the mechanism in atherogenesis is unclear. Loci most frequently found in independent GWAS were at Chr9p21.3 (ANRIL/CDKN2B-AS1), Chr6p24.1 (PHACTR1), and Chr1p13.3 (CELSR2, PSRC1, MYBPHL, SORT1). Recent work suggests that Chr9p21.3 exerts its effects through epigenetic regulation of target genes, whereas mechanisms at Chr6p24.1 remain obscure, and Chr1p13.3 affects plasma LDL cholesterol. SUMMARY Novel GWAS loci indicate that our understanding of atherosclerosis is limited and implicate a role of hitherto unknown mechanisms, such as epigenetic gene regulation in atherogenesis.
Collapse
Affiliation(s)
- Lesca M Holdt
- Institute of Laboratory Medicine, University Hospital Munich-LMU and Ludwig-Maximilians-University Munich, Munich, Germany
| | | |
Collapse
|
50
|
Mouilleron S, Wiezlak M, O'Reilly N, Treisman R, McDonald NQ. Structures of the Phactr1 RPEL domain and RPEL motif complexes with G-actin reveal the molecular basis for actin binding cooperativity. Structure 2012; 20:1960-70. [PMID: 23041370 DOI: 10.1016/j.str.2012.08.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 08/10/2012] [Accepted: 08/14/2012] [Indexed: 12/11/2022]
Abstract
The Phactr family of PP1-binding proteins and the myocardin-related transcription factor family of transcriptional coactivators contain regulatory domains comprising three copies of the RPEL motif, a G-actin binding element. We report the structure of a Phactr1 G-actin⋅RPEL domain complex. Three G-actins surround the crank-shaped RPEL domain forming a closed helical assembly. Their spatial relationship is identical to the RPEL-actins within the pentavalent MRTF G-actin⋅RPEL domain complex, suggesting that conserved cooperative interactions between actin⋅RPEL units organize the assembly. In the trivalent Phactr1 complex, each G-actin⋅RPEL unit makes secondary contacts with its downstream actin involving distinct RPEL residues. Similar secondary contacts are seen in G-actin⋅RPEL peptide crystals. Loss-of-secondary-contact mutations destabilize the Phactr1 G-actin⋅RPEL assembly. Furthermore, actin-mediated inhibition of Phactr1 nuclear import requires secondary contact residues in the Phactr1 N-terminal RPEL-N motif, suggesting that it involves interaction of RPEL-N with the C-terminal assembly. Secondary actin contacts by actin-bound RPEL motifs thus govern formation of multivalent actin⋅RPEL assemblies.
Collapse
Affiliation(s)
- Stephane Mouilleron
- Structural Biology, CRUK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | | | | | | | | |
Collapse
|