1
|
Acosta-Alvear D, Harnoss JM, Walter P, Ashkenazi A. Homeostasis control in health and disease by the unfolded protein response. Nat Rev Mol Cell Biol 2025; 26:193-212. [PMID: 39501044 DOI: 10.1038/s41580-024-00794-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 10/07/2024] [Indexed: 02/27/2025]
Abstract
Cells rely on the endoplasmic reticulum (ER) to fold and assemble newly synthesized transmembrane and secretory proteins - essential for cellular structure-function and for both intracellular and intercellular communication. To ensure the operative fidelity of the ER, eukaryotic cells leverage the unfolded protein response (UPR) - a stress-sensing and signalling network that maintains homeostasis by rebalancing the biosynthetic capacity of the ER according to need. The metazoan UPR can also redirect signalling from cytoprotective adaptation to programmed cell death if homeostasis restoration fails. As such, the UPR benefits multicellular organisms by preserving optimally functioning cells while removing damaged ones. Nevertheless, dysregulation of the UPR can be harmful. In this Review, we discuss the UPR and its regulatory processes as a paradigm in health and disease. We highlight important recent advances in molecular and mechanistic understanding of the UPR that enable greater precision in designing and developing innovative strategies to harness its potential for therapeutic gain. We underscore the rheostatic character of the UPR, its contextual nature and critical open questions for its further elucidation.
Collapse
Affiliation(s)
| | - Jonathan M Harnoss
- Department of General, Visceral, Thoracic and Transplant Surgery, University Hospital Giessen, Giessen, Germany
| | - Peter Walter
- Altos Labs, Inc., Bay Area Institute of Science, Redwood City, CA, USA.
| | - Avi Ashkenazi
- Research Oncology, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
2
|
Lee GE, Bang G, Byun J, Lee CJ, Chen W, Jeung D, An HJ, Kang HC, Lee JY, Lee HS, Hong YS, Kim DJ, Keniry M, Kim JY, Choi JS, Fanto M, Cho SJ, Kim KD, Cho YY. Dysregulated CREB3 cleavage at the nuclear membrane induces karyoptosis-mediated cell death. Exp Mol Med 2024; 56:686-699. [PMID: 38480902 PMCID: PMC10985101 DOI: 10.1038/s12276-024-01195-1] [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] [Academic Contribution Register] [Received: 09/01/2023] [Revised: 12/17/2023] [Accepted: 01/02/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer cells often exhibit resistance to apoptotic cell death, but they may be vulnerable to other types of cell death. Elucidating additional mechanisms that govern cancer cell death is crucial for developing new therapies. Our research identified cyclic AMP-responsive element-binding protein 3 (CREB3) as a crucial regulator and initiator of a unique cell death mechanism known as karyoptosis. This process is characterized by nuclear shrinkage, deformation, and the loss of nuclear components following nuclear membrane rupture. We found that the N-terminal domain (aa 1-230) of full-length CREB3 (CREB3-FL), which is anchored to the nuclear inner membrane (INM), interacts with lamins and chromatin DNA. This interaction maintains a balance between the outward force exerted by tightly packed DNA and the inward constraining force, thereby preserving INM integrity. Under endoplasmic reticulum (ER) stress, aberrant cleavage of CREB3-FL at the INM leads to abnormal accumulation of the cleaved form of CREB3 (CREB3-CF). This accumulation disrupts the attachment of CREB3-FL to the INM, resulting in sudden rupture of the nuclear membrane and the onset of karyoptosis. Proteomic studies revealed that CREB3-CF overexpression induces a DNA damage response akin to that caused by UVB irradiation, which is associated with cellular senescence in cancer cells. These findings demonstrated that the dysregulation of CREB3-FL cleavage is a key factor in karyoptotic cell death. Consequently, these findings suggest new therapeutic strategies in cancer treatment that exploit the process of karyoptosis.
Collapse
Affiliation(s)
- Ga-Eun Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Geul Bang
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, Cheongju-si, Chungbuk, 28119, Republic of Korea
| | - Jiin Byun
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Cheol-Jung Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- Research Center for Materials Analysis, Korea Basic Science Institute, Daejeon, 34133, Republic of Korea
| | - Weidong Chen
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Dohyun Jeung
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Hyun-Jung An
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Han Chang Kang
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Joo Young Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Hye Suk Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Young-Soo Hong
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Chungbuk, 28116, Republic of Korea
| | - Dae Joon Kim
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, 78504, USA
| | - Megan Keniry
- Department of Biology, University of Texas Rio Grande Valley, Edinburg, TX, 78539, USA
| | - Jin Young Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, Cheongju-si, Chungbuk, 28119, Republic of Korea
| | - Jin-Sung Choi
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Manolis Fanto
- Department of Basic and Clinical Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Sung-Jun Cho
- University of Minnesota, Department of Medicine, 420 Delaware St SE, MMC 284, Minneapolis, MN, 55455, USA
| | - Kwang-Dong Kim
- Division of Applied Life Science (BK21 four), PMBBRC, Gyeongsang National University, Jinju, 52828, Korea
| | - Yong-Yeon Cho
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea.
- BK21-4th, and RCD Control∙Material Research Institute, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea.
| |
Collapse
|
3
|
Pascal LE, Frahm KA, Skalitzky KO, DeFranco DB, Rigatti LH, Lu R, Liu TT. Genetic alterations in CREBRF influence prostate cancer survival and impact prostate tissue homeostasis in mice. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2023; 11:27-39. [PMID: 36923723 PMCID: PMC10009309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Academic Contribution Register] [Received: 11/06/2022] [Accepted: 02/16/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND Risk factors for prostate cancer include age, environment, race and ethnicity. Genetic variants in cyclic-adenosine-monophosphate-response-element-binding protein 3 regulatory factor (CREBRF) gene are frequently observed in Pacific Islanders, a population with elevated prostate cancer incidence. CREBRF has been shown to play a role in other cancers, however its function in prostate homeostasis and tumorigenesis has not been previously explored. We determined the incidence of CREBRF alterations in publicly available databases and examined the impact of CREBRF deletion on the murine prostate in order to determine whether CREBRF impacts prostate physiology or pathophysiology. METHODS Alterations in CREBRF were identified in prostate cancer patients via in silico analysis of several publicly available datasets through cBioPortal. Male Crebrf knockout and wild-type littermate mice were generated and examined for prostate defects at 4 months of age. Immunohistochemical staining of murine prostate sections was used to determine the impact of Crebrf knockout on proliferation, apoptosis, inflammation and blood vessel density in the prostate. Serum adipokine levels were measured using a Luminex Multiplex Assay. RESULTS CREBRF alterations were identified in up to 4.05% of prostate tumors and the mutations identified were categorized as likely damaging. Median survival of prostate cancer patients with genetic alterations in CREBRF was 41.23 months, compared to 131 months for patients without these changes. In the murine model, the prostates of Crebrf knockout mice had reduced epithelial proliferation and increased TUNEL+ apoptotic cells. Circulating adipokines PAI-1 and MCP-1 were also altered in Crebrf knockout mice compared to age-matched controls. CONCLUSIONS Prostate cancer patients with genetic alterations in CREBRF had a significantly decreased overall survival suggesting that wild type CREBRF may play a role in limiting prostate tumorigenesis and progression. The murine knockout model demonstrated that CREBRF could modulate proliferation and apoptosis and macrophage density in the prostate. Serum levels of adipokines PAI-1 and MCP-1 were also altered and may contribute to the phenotypic changes observed in the prostates of Crebrf knockout mice. Future studies focused on populations susceptible to CREBRF mutations and mechanistic studies will be required to fully elucidate the potential role of CREBRF in prostate tumorigenesis.
Collapse
Affiliation(s)
- Laura E Pascal
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine Pittsburgh, PA, USA.,UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| | - Krystle A Frahm
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| | | | - Donald B DeFranco
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine Pittsburgh, PA, USA.,Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| | - Lora H Rigatti
- Division of Laboratory Animal Resources, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| | - Ray Lu
- Department of Molecular and Cellular Biology, University of Guelph Guelph, ON, Canada
| | - Teresa T Liu
- Department of Urology, University of Wisconsin Madison, WI, USA
| |
Collapse
|
4
|
Glat M, Gundacker A, Cuenca Rico L, Czuczu B, Ben‐Simon Y, Harkany T, Pollak DD. An accessory prefrontal cortex-thalamus circuit sculpts maternal behavior in virgin female mice. EMBO J 2022; 41:e111648. [PMID: 36341708 PMCID: PMC9753463 DOI: 10.15252/embj.2022111648] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/12/2022] [Revised: 10/08/2022] [Accepted: 10/14/2022] [Indexed: 11/09/2022] Open
Abstract
The ability to care for the young is innate and readily displayed by postpartum females after delivery to ensure offspring survival. Upon pup exposure, rodent virgin (nulliparous) females also develop parental behavior that over time becomes displayed at levels equivalent to parenting mothers. Although maternal behavior in postpartum females and the associated neurocircuits are well characterized, the neural mechanisms underlying the acquisition of maternal behavior without prior experience remain poorly understood. Here, we show that the development of maternal care behavior in response to first-time pup exposure in virgin females is initiated by the activation of the anterior cingulate cortex (ACC). ACC activity is dependent on feedback excitation by Vglut2+ /Galanin+ neurons of the centrolateral nucleus of the thalamus (CL), with their activity sufficient to display parenting behaviors. Accordingly, acute bidirectional chemogenetic manipulation of neuronal activity in the ACC facilitates or impairs the attainment of maternal behavior, exclusively in virgin females. These results reveal an ACC-CL neurocircuit as an accessory loop in virgin females for the initiation of maternal care upon first-time exposure to pups.
Collapse
Affiliation(s)
- Micaela Glat
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - Anna Gundacker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - Laura Cuenca Rico
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - Barbara Czuczu
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - Yoav Ben‐Simon
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain ResearchMedical University of ViennaViennaAustria
- Department of Neuroscience, Biomedicum 7DKarolinska InstitutetSolnaSweden
| | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and PharmacologyMedical University of ViennaViennaAustria
| |
Collapse
|
5
|
Li Y, Wang H, Chen H, Liao Y, Gou S, Yan Q, Zhuang Z, Li H, Wang J, Suo Y, Lan T, Liu Y, Zhao Y, Zou Q, Nie T, Hui X, Lai L, Wu D, Fan N. Generation of a genetically modified pig model with CREBRF R457Q variant. FASEB J 2022; 36:e22611. [PMID: 36250915 DOI: 10.1096/fj.202201117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/13/2022] [Accepted: 10/03/2022] [Indexed: 11/11/2022]
Abstract
Obesity is among the strongest risk factors for type 2 diabetes (T2D). The CREBRF missense allele rs373863828 (p. Arg457Gln, p. R457Q) is associated with increased body mass index but reduced risk of T2D in people of Pacific ancestry. To investigate the functional consequences of the CREBRF variant, we introduced the corresponding human mutation R457Q into the porcine genome. The CREBRFR457Q pigs displayed dramatically increased fat deposition, which was mainly distributed in subcutaneous adipose tissue other than visceral adipose tissue. The CREBRFR457Q variant promoted preadipocyte differentiation. The increased differentiation capacity of precursor adipocytes conferred pigs the unique histological phenotype that adipocytes had a smaller size but a greater number in subcutaneous adipose tissue (SAT) of CREBRFR457Q variant pigs. In addition, in SAT of CREBRFR457Q pigs, the contents of the peroxidative metabolites 4-hydroxy-nonenal and malondialdehyde were significantly decreased, while the activity of antioxidant enzymes, such as glutathione peroxidase, superoxide dismutase, and catalase, was increased, which was in accordance with the declined level of the reactive oxygen species (ROS) in CREBRFR457Q pigs. Together, these data supported a causal role of the CREBRFR457Q variant in the pathogenesis of obesity, partly via adipocyte hyperplasia, and further suggested that reduced oxidative stress in adipose tissue may mediate the relative metabolic protection afforded by this variant despite the related obesity.
Collapse
Affiliation(s)
- Yingying Li
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Hai Wang
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Huangyao Chen
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Yuan Liao
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Institute of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Shixue Gou
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Quanmei Yan
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhenpeng Zhuang
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Hao Li
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Jiaowei Wang
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Yangyang Suo
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Ting Lan
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Yang Liu
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Yu Zhao
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Qingjian Zou
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, China
| | - Tao Nie
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaoyan Hui
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong SAR
| | - Liangxue Lai
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.,Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, China
| | - Donghai Wu
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Nana Fan
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| |
Collapse
|
6
|
Qu X, Tao J, Xie J. Circ_0009035 regulates the progression of cervical cancer by targeting miR-1305/CREBRF axis. Anticancer Drugs 2022; 33:539-552. [PMID: 35389936 DOI: 10.1097/cad.0000000000001278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/26/2022]
Abstract
Circular RNAs (circRNAs) have a crucial role in the occurrence of many diseases, such as tumors. Yet the roles of circ_0009035 (circRACGAP1) in cervical cancer are not fully characterized. The expression levels of circRACGAP1, miR-1305 and cAMP-responsive element-binding protein 3 regulatory factor (CREBRF) were detected by using real-time quantitative PCR or western blot. Cell counting kit-8 assay, 5-ethynyl-2'-deoxyuridine, colony formation assay, transwell assay and tube formation assay were used to detect cell proliferation, migration and invasion and angiogenesis, respectively. Flow cytometry assay was used to analyze the cell apoptosis. Dual-luciferase reporter assay and RNA immunoprecipitation assay were performed to analyze the targeting about miR-1305 and circ_0009035 or CREBRF. Xenograft model was built to study the role of circ_0009035 in vivo. Immunohistochemistry was used to detect the expression of Ki67, epithelial cadherin and vimentin. First, we found that circ_0009035 expression was significantly upregulated in tumor cells and tissues; second, knockdown of circ_0009035 could inhibit cell proliferation, migration and invasion and promote cell apoptosis. Subsequently, circ_0009035 was found to be able to target miR-1305, and the expression of miR-1305 in tumor tissues and cells was significantly lower. MiR-1305 inhibitor could restore cell-related progression of cervical cancer inhibited by si-circ_0009035. Finally, miR-1305 could target CREBRF, and circ_0009035 could regulate CREBRF expression by targeting miR-1305, thereby affecting cervical cancer tumorigenesis. In summary, our study confirmed that circ_0009035 could influence the development of cervical cancer through the targeted regulation of miR-1305/CREBRF.
Collapse
Affiliation(s)
- Xiangdong Qu
- Department of Obstetrics and Gynecology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou City, Zhejiang Province, China
| | | | | |
Collapse
|
7
|
Lee K, Vakili S, Burden HJ, Adams S, Smith GC, Kulatea B, Wright-McNaughton M, Sword D, Watene-O'Sullivan C, Atiola RD, Paul RG, Plank LD, Kallingappa P, King F, Wilcox P, Merriman TR, Krebs JD, Hall RM, Murphy R, Merry TL, Shepherd PR. The minor allele of the CREBRF rs373863828 p.R457Q coding variant is associated with reduced levels of myostatin in males: Implications for body composition. Mol Metab 2022; 59:101464. [PMID: 35218947 PMCID: PMC8927835 DOI: 10.1016/j.molmet.2022.101464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 07/22/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 12/04/2022] Open
Abstract
OBJECTIVE The minor allele (A) of the rs373863828 variant (p.Arg457Gln) in CREBRF is restricted to indigenous peoples of the Pacific islands (including New Zealand Māori and peoples of Polynesia), with a frequency of up to 25% in these populations. This allele associates with a large increase in body mass index (BMI) but with significantly lower risk of type-2 diabetes (T2D). It remains unclear whether the increased BMI is driven by increased adiposity or by increased lean mass. METHODS We undertook body composition analysis using DXA in 189 young men of Māori and Pacific descent living in Aotearoa New Zealand. Further investigation was carried out in two orthologous Arg458Gln knockin mouse models on FVB/NJ and C57BL/6j backgrounds. RESULTS The rs373863828 A allele was associated with lower fat mass when adjusted for BMI (p < 0.05) and was associated with significantly lower circulating levels of the muscle inhibitory hormone myostatin (p < 0.05). Supporting the human data, significant reductions in adipose tissue mass were observed in the knockin mice. This was more significant in older mice in both backgrounds and appeared to be the result of reduced age-associated increases in fat mass. The older male knockin mice on C57BL/6j background also had increased grip strength (p < 0.01) and lower levels of myostatin (p < 0.05). CONCLUSION Overall, these results prove that the rs373863828 A-allele is associated with a reduction of myostatin levels which likely contribute to an age-dependent lowering of fat mass, at least in males.
Collapse
Affiliation(s)
- Kate Lee
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Sanaz Vakili
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Hannah J Burden
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Shannon Adams
- Discipline of Nutrition, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Greg C Smith
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Kensington, Australia
| | - Braydon Kulatea
- Discipline of Nutrition, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | | | - Danielle Sword
- Department of Medicine, University of Otago Wellington, Wellington, New Zealand
| | | | - Robert D Atiola
- Discipline of Nutrition, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Ryan G Paul
- Waikato Medical Research Centre, University of Waikato, Hamilton, New Zealand
| | - Lindsay D Plank
- Department of Surgery, School of Medicine, The University of Auckland, Auckland, New Zealand
| | - Prasanna Kallingappa
- Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Frances King
- Ngati Porou Hauora, Te Puia Springs, New Zealand
| | - Phillip Wilcox
- Department of Mathematics and Statistics, University of Otago, New Zealand
| | - Tony R Merriman
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Biochemistry, School of Biomedical Sciences, University of Otago, New Zealand; Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Alabama, United States
| | - Jeremy D Krebs
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Medicine, University of Otago Wellington, Wellington, New Zealand
| | - Rosemary M Hall
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Medicine, University of Otago Wellington, Wellington, New Zealand
| | - Rinki Murphy
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Medicine, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Troy L Merry
- Discipline of Nutrition, School of Medical Sciences, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Peter R Shepherd
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
8
|
Limited Metabolic Effect of the CREBRF R457Q Obesity Variant in Mice. Cells 2022; 11:cells11030497. [PMID: 35159305 PMCID: PMC8833978 DOI: 10.3390/cells11030497] [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] [Academic Contribution Register] [Received: 11/22/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 11/17/2022] Open
Abstract
The Arg457Gln missense variant in the CREBRF gene has previously been identified as driving excess body weight in Pacific/Oceanic populations. Intriguingly, Arg457Gln variant carriers also demonstrate paradoxical reductions in diabetes risk, indicating that the gene has a critical role in whole-body metabolism. To study the function of this variant in more detail, we generated mice on an FVB/N background with the Crebrf Arg458Gln variant knocked in to replace the endogenous Crebrf. The whole-body metabolic phenotype was characterized for male and female mice on a regular chow diet or an 8-week high-fat challenge. Regular assessment of body composition found that the Crebrf variant had no influence on total body weight or fat mass at any time point. Glucose tolerance tests demonstrated no obvious genotype effect on glucose homeostasis, with indirect calorimetry measures of whole-body energy expenditure likewise unaffected. Male chow-fed variant carriers displayed a trend towards increased lean mass and significantly reduced sensitivity to insulin administration. Overall, this novel mouse model showed only limited phenotypic effects associated with the Crebrf missense variant. The inability to recapitulate results of human association studies may invite reconsideration of the precise mechanistic link between CREBRF function and the risks of obesity and diabetes in variant allele carriers.
Collapse
|
9
|
Larson S, Arrazola A, Parra R, Morrissey K, Faulkner T, Jafarikia M, Mandell I, Bergeron R, Lu R. Genetic variation in LUMAN/CREB3 and association with stress and meat quality traits in Yorkshire pigs. CANADIAN JOURNAL OF ANIMAL SCIENCE 2021. [DOI: 10.1139/cjas-2020-0156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/22/2022]
Abstract
LUMAN/CREB3 is a stress regulatory gene that affects the activity of the hypothalamic–pituitary–adrenal axis in mice and presents a promising avenue for exploring variable stress-responsiveness in pigs. Pigs with similar characteristics to LUMAN-deficient mice, including greater resilience to stress and receptivity to human handling, would be valuable in the pork industry from animal welfare and production efficiency perspectives. We previously identified eight genetic variations and five haplotypes throughout the LUMAN locus in Yorkshire pigs. In this study, we analysed associations between LUMAN variations with behavioural stress response during three tests (open field test, novel object test, and human approach test), physiological stress responsiveness (cortisol), and carcass/meat quality measurements from purebred Yorkshire pigs. Haplotypes A1 and A2 were associated with decreased activity levels in novel environments and greater plasma cortisol concentrations at slaughter. Haplotype A1 was associated with lower carcass scratch scores and meat with lower cooking losses and greater tenderness. Haplotypes B1 and B2 were associated with the opposite traits including increased activity levels in novel environments and characteristics for lower meat quality including greater cooking losses, lower marbling, and paler coloured meat. We conclude that DNA variations in the LUMAN locus could potentially be used as genetic markers for stress resistance and meat quality in pig breeding.
Collapse
Affiliation(s)
- Shayla Larson
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Aitor Arrazola
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Rebecca Parra
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Krysta Morrissey
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Tess Faulkner
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Mohsen Jafarikia
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
- Canadian Centre for Swine Improvement Inc., Central Experimental Farm, Building #75, 960 Carling Avenue, Ottawa, ON K1A 0C6, Canada
| | - Ira Mandell
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Renée Bergeron
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Ray Lu
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
10
|
Kanshana JS, Mattila PE, Ewing MC, Wood AN, Schoiswohl G, Meyer AC, Kowalski A, Rosenthal SL, Gingras S, Kaufman BA, Lu R, Weeks DE, McGarvey ST, Minster RL, Hawley NL, Kershaw EE. A murine model of the human CREBRFR457Q obesity-risk variant does not influence energy or glucose homeostasis in response to nutritional stress. PLoS One 2021; 16:e0251895. [PMID: 34520472 PMCID: PMC8439463 DOI: 10.1371/journal.pone.0251895] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/27/2021] [Accepted: 08/09/2021] [Indexed: 01/02/2023] Open
Abstract
Obesity and diabetes have strong heritable components, yet the genetic contributions to these diseases remain largely unexplained. In humans, a missense variant in Creb3 regulatory factor (CREBRF) [rs373863828 (p.Arg457Gln); CREBRFR457Q] is strongly associated with increased odds of obesity but decreased odds of diabetes. Although virtually nothing is known about CREBRF's mechanism of action, emerging evidence implicates it in the adaptive transcriptional response to nutritional stress downstream of TORC1. The objectives of this study were to generate a murine model with knockin of the orthologous variant in mice (CREBRFR458Q) and to test the hypothesis that this CREBRF variant promotes obesity and protects against diabetes by regulating energy and glucose homeostasis downstream of TORC1. To test this hypothesis, we performed extensive phenotypic analysis of CREBRFR458Q knockin mice at baseline and in response to acute (fasting/refeeding), chronic (low- and high-fat diet feeding), and extreme (prolonged fasting) nutritional stress as well as with pharmacological TORC1 inhibition, and aging to 52 weeks. The results demonstrate that the murine CREBRFR458Q model of the human CREBRFR457Q variant does not influence energy/glucose homeostasis in response to these interventions, with the exception of possible greater loss of fat relative to lean mass with age. Alternative preclinical models and/or studies in humans will be required to decipher the mechanisms linking this variant to human health and disease.
Collapse
Affiliation(s)
- Jitendra S. Kanshana
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Polly E. Mattila
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Michael C. Ewing
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Ashlee N. Wood
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Gabriele Schoiswohl
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
| | - Anna C. Meyer
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Aneta Kowalski
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Samantha L. Rosenthal
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Sebastien Gingras
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Brett A. Kaufman
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Ray Lu
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Daniel E. Weeks
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Stephen T. McGarvey
- International Health Institute, Department of Epidemiology, Brown University School of Public Health, Providence, Rhode Island, United States of America
- Department of Anthropology, Brown University, Providence, Rhode Island, United States of America
| | - Ryan L. Minster
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Nicola L. Hawley
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Erin E. Kershaw
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|
11
|
Alberca CD, Papale LA, Madrid A, Gianatiempo O, Cánepa ET, Alisch RS, Chertoff M. Perinatal protein malnutrition results in genome-wide disruptions of 5-hydroxymethylcytosine at regions that can be restored to control levels by an enriched environment. Epigenetics 2020; 16:1085-1101. [PMID: 33172347 DOI: 10.1080/15592294.2020.1841871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022] Open
Abstract
Maternal malnutrition remains one of the major adversities affecting brain development and long-term mental health outcomes, increasing the risk to develop anxiety and depressive disorders. We have previously shown that malnutrition-induced anxiety-like behaviours can be rescued by a social and sensory stimulation (enriched environment) in male mice. Here, we expand these findings to adult female mice and profiled genome-wide ventral hippocampal 5hmC levels related to malnutrition-induced anxiety-like behaviours and their rescue by an enriched environment. This approach revealed 508 differentially hydroxymethylated genes associated with protein malnutrition and that several genes (N = 34) exhibited a restored 5hmC abundance to control levels following exposure to an enriched environment, including genes involved in neuronal functions like dendrite outgrowth, axon guidance, and maintenance of neuronal circuits (e.g. Fltr3, Itsn1, Lman1, Lsamp, Nav, and Ror1) and epigenetic mechanisms (e.g. Hdac9 and Dicer1). Sequence motif predictions indicated that 5hmC may be modulating the binding of transcription factors for several of these transcripts, suggesting a regulatory role for 5hmC in response to perinatal malnutrition and exposure to an enriched environment. Together, these findings establish a role for 5hmC in early-life malnutrition and reveal genes linked to malnutrition-induced anxious behaviours that are mitigated by an enriched environment.
Collapse
Affiliation(s)
- Carolina D Alberca
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Neuroepigenetica, Buenos Aires, Argentina
| | - Ligia A Papale
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Andy Madrid
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.,Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
| | - Octavio Gianatiempo
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Neuroepigenetica, Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales - CONICET (IQUIBICEN), Buenos Aires, Argentina
| | - Eduardo T Cánepa
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Neuroepigenetica, Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales - CONICET (IQUIBICEN), Buenos Aires, Argentina
| | - Reid S Alisch
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Mariela Chertoff
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Neuroepigenetica, Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales - CONICET (IQUIBICEN), Buenos Aires, Argentina
| |
Collapse
|
12
|
Chen YC, Kuo HC, Lo WS, Hung CM. Avian phenotypic convergence is subject to low genetic constraints based on genomic evidence. BMC Evol Biol 2020; 20:147. [PMID: 33160317 PMCID: PMC7648321 DOI: 10.1186/s12862-020-01711-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/26/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Phenotypic convergence between distinct species provides an opportunity to examine the predictability of genetic evolution. Unrelated species sharing genetic underpinnings for phenotypic convergence suggests strong genetic constraints, and thus high predictability of evolution. However, there is no clear big picture of the genomic constraints on convergent evolution. Genome-based phylogenies have confirmed many cases of phenotypic convergence in birds, making them a good system for examining genetic constraints in phenotypic convergence. In this study, we used hierarchical genomic approaches to estimate genetic constraints in three convergent avian traits: nocturnality, raptorial behavior and foot-propelled diving. RESULTS Phylogeny-based hypothesis tests and positive selection tests were applied to compare 16 avian genomes, representing 14 orders, and identify genes with strong convergence signals. We found 43 adaptively convergent genes (ACGs) associated with the three phenotypic convergence cases and assessed genetic constraints in all three cases, from (amino acid) site mutations to genetic pathways. We found that the avian orders shared few site mutations in the ACGs that contributed to the convergent phenotypes, and that these ACGs were not enriched in any genetic pathways. In addition, different pairs of orders with convergent foot-propelled diving or raptorial behaviors shared few ACGs. We also found that closely related orders that shared foot-propelled diving behavior did not share more ACGs than did distinct orders, suggesting that convergence among these orders could not be explained by their initial genomic backgrounds. CONCLUSIONS Our analyses of three avian convergence events suggest low constraints for phenotypic convergence across multiple genetic levels, implying that genetic evolution is unpredictable at the phylogenetic level of avian order. Ours is one of first studies to apply hierarchical genomic examination to multiple avian convergent cases to assess the genetic constraints in life history trait evolution.
Collapse
Affiliation(s)
- Yu-Chi Chen
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Hao-Chih Kuo
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Wen-Sui Lo
- Department of Evolutionary Biology, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Chih-Ming Hung
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
13
|
Frahm KA, Williams AA, Wood AN, Ewing MC, Mattila PE, Chuan BW, Guo L, Shah FA, O’Donnell CP, Lu R, DeFranco DB. Loss of CREBRF Reduces Anxiety-like Behaviors and Circulating Glucocorticoids in Male and Female Mice. Endocrinology 2020; 161:bqaa163. [PMID: 32901804 PMCID: PMC7567405 DOI: 10.1210/endocr/bqaa163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/05/2020] [Accepted: 09/04/2020] [Indexed: 12/24/2022]
Abstract
Glucocorticoid signaling controls many key biological functions ranging from stress responses to affective states. The putative transcriptional coregulator CREB3 regulatory factor (CREBRF) reduces glucocorticoid receptor levels in vitro, suggesting that CREBRF may impact behavioral and physiological outputs. In the present study, we examined adult male and female mice with global loss of CREBRF (CrebrfKO) for anxiety-like behaviors and circulating glucocorticoids in response to various acute stress conditions. Results demonstrate that both male and female CrebrfKO mice have preserved locomotor activity but reduced anxiety-like behaviors during the light-dark box and elevated plus maze. These behavioral phenotypes were associated with lower plasma corticosterone after restraint stress. Further studies using unhandled female mice also demonstrated a loss of the diurnal circulating corticosterone rhythm in CrebrfKO mice. These results suggest that CREBRF impacts anxiety-like behavior and circulating glucocorticoids in response to acute stressors and serves as a basis for future mechanistic studies to define the impact of CREBRF in glucocorticoid-associated behavioral and physiological responses.
Collapse
Affiliation(s)
- Krystle A Frahm
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Akeem A Williams
- Division of Endocrinology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ashlee N Wood
- Division of Endocrinology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Michael C Ewing
- Division of Endocrinology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Polly E Mattila
- Division of Endocrinology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Byron W Chuan
- Division of Pulmonology, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Lanping Guo
- Division of Pulmonology, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Faraaz A Shah
- Division of Pulmonology, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Christopher P O’Donnell
- Division of Pulmonology, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ray Lu
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Donald B DeFranco
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
14
|
Krishnan M, Murphy R, Okesene-Gafa KAM, Ji M, Thompson JMD, Taylor RS, Merriman TR, McCowan LME, McKinlay CJD. The Pacific-specific CREBRF rs373863828 allele protects against gestational diabetes mellitus in Māori and Pacific women with obesity. Diabetologia 2020; 63:2169-2176. [PMID: 32654027 DOI: 10.1007/s00125-020-05202-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 10/29/2019] [Accepted: 05/06/2020] [Indexed: 01/13/2023]
Abstract
AIMS/HYPOTHESIS The CREBRF rs373863828 minor (A) allele is associated with increased BMI but reduced prevalence of type 2 diabetes in Māori and Pacific people. Given the shared aetiology of type 2 diabetes and gestational diabetes mellitus (GDM), we tested for an association between the CREBRF rs373863828 variant and GDM. METHODS We conducted a prospective cohort study of Māori and Pacific women nested within a nutritional intervention study for pregnant women with obesity. Women were enrolled at 12-17 weeks' gestation and underwent anthropometry and collection of buffy coats for later genetic testing. GDM was diagnosed by 75 g OGTT at 24-28 weeks' gestation using the International Association of Diabetes and Pregnancy Study Groups criteria. Genotyping was performed by real-time PCR with a custom CREBRF rs373863828 probe-set. The association between CREBRF rs373863828 and GDM was analysed separately by ethnic group using logistic regression, with effect estimates combined in a meta-analysis. RESULTS Of 112 Māori and Pacific pregnant women with obesity, 31 (28%) carried the CREBRF rs373863828 A allele (A/G or A/A) and 35 (31%) developed GDM. Women who carried the CREBRF rs373863828 A allele did not differ in BMI when compared with non-carriers (G/G). There was a fivefold reduction in the likelihood of GDM per CREBRF rs373863828 A allele (OR 0.19 [95% CI 0.05, 0.69], p = 0.01), independent of age, BMI and family history of diabetes (adjusted OR 0.13 [95% CI 0.03, 0.53], p = 0.004). GDM was diagnosed in 10% and 40% of women with and without the CREBRF rs373863828 A allele, respectively (no woman with the A/A genotype developed GDM). CONCLUSIONS/INTERPRETATION The CREBRF rs373863828 (A) allele is associated with reduced likelihood of GDM in Māori and Pacific women with obesity and may improve GDM risk prediction. Graphical abstract.
Collapse
Affiliation(s)
- Mohanraj Krishnan
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
- Department of Medicine, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Rinki Murphy
- Department of Medicine, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
- Counties Manukau Health, Auckland, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.
| | - Karaponi A M Okesene-Gafa
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
- Counties Manukau Health, Auckland, New Zealand
| | - Maria Ji
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - John M D Thompson
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
- Department of Paediatrics, University of Auckland, Auckland, New Zealand
| | - Rennae S Taylor
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Tony R Merriman
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Lesley M E McCowan
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
- Counties Manukau Health, Auckland, New Zealand
| | - Christopher J D McKinlay
- Counties Manukau Health, Auckland, New Zealand.
- Liggins Institute, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| |
Collapse
|
15
|
Hawley NL, Pomer A, Rivara AC, Rosenthal SL, Duckham RL, Carlson JC, Naseri T, Reupena MS, Selu M, Lupematisila V, Unasa F, Vesi L, Fatu T, Unasa S, Faasalele-Savusa K, Wetzel AI, Soti-Ulberg C, Prescott AT, Siufaga G, Penaia C, To SB, LaMonica LC, Lameko V, Choy CC, Crouter SE, Redline S, Deka R, Kershaw EE, Urban Z, Minster RL, Weeks DE, McGarvey ST. Exploring the Paradoxical Relationship of a Creb 3 Regulatory Factor Missense Variant With Body Mass Index and Diabetes Among Samoans: Protocol for the Soifua Manuia (Good Health) Observational Cohort Study. JMIR Res Protoc 2020; 9:e17329. [PMID: 32706746 PMCID: PMC7413272 DOI: 10.2196/17329] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/06/2019] [Revised: 02/24/2020] [Accepted: 03/02/2020] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The prevalence of obesity and diabetes in Samoa, like many other Pacific Island nations, has reached epidemic proportions. Although the etiology of these conditions can be largely attributed to the rapidly changing economic and nutritional environment, a recently identified genetic variant, rs373863828 (CREB 3 regulatory factor, CREBRF: c.1370G>A p.[R457Q]) is associated with increased odds of obesity, but paradoxically, decreased odds of diabetes. OBJECTIVE The overarching goal of the Soifua Manuia (Good Health) study was to precisely characterize the association of the CREBRF variant with metabolic (body composition and glucose homeostasis) and behavioral traits (dietary intake, physical activity, sleep, and weight control behaviors) that influence energy homeostasis in 500 adults. METHODS A cohort of adult Samoans who participated in a genome-wide association study of adiposity in Samoa in 2010 was followed up, based on the presence or absence of the CREBRF variant, between August 2017 and March 2019. Over a period of 7-10 days, each participant completed the main study protocol, which consisted of anthropometric measurements (weight, height, circumferences, and skinfolds), body composition assessment (bioelectrical impedance and dual-energy x-ray absorptiometry), point-of-care glycated hemoglobin measurement, a fasting blood draw and oral glucose tolerance test, urine collection, blood pressure measurement, hand grip strength measurement, objective physical activity and sleep apnea monitoring, and questionnaire measures (eg, health interview, cigarette and alcohol use, food frequency questionnaire, socioeconomic position, stress, social support, food and water insecurity, sleep, body image, and dietary preferences). In January 2019, a subsample of the study participants (n=118) completed a buttock fat biopsy procedure to collect subcutaneous adipose tissue samples. RESULTS Enrollment of 519 participants was completed in March 2019. Data analyses are ongoing, with results expected in 2020 and 2021. CONCLUSIONS While the genetic variant rs373863828, in CREBRF, has the largest known effect size of any identified common obesity gene, very little is currently understood about the mechanisms by which it confers increased odds of obesity but paradoxically lowered odds of type 2 diabetes. The results of this study will provide insights into how the gene functions on a whole-body level, which could provide novel targets to prevent or treat obesity, diabetes, and associated metabolic disorders. This study represents the human arm of a comprehensive and integrated approach involving humans as well as preclinical models that will provide novel insights into metabolic disease. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) RR1-10.2196/17329.
Collapse
Affiliation(s)
- Nicola L Hawley
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, United States
| | - Alysa Pomer
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, United States
| | - Anna C Rivara
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, United States
| | - Samantha L Rosenthal
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rachel L Duckham
- Institute of Physical Activity and Nutrition, Deakin University, Geelong, Australia
- Australian Institute for Musculoskeletal Sciences, The University of Melbourne and Western Health, St Albans, Australia
| | - Jenna C Carlson
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | | | | | - Melania Selu
- Obesity, Lifestyle and Genetic Adaptations Study Group, Apia, Samoa
| | | | - Folla Unasa
- Obesity, Lifestyle and Genetic Adaptations Study Group, Apia, Samoa
| | - Lupesina Vesi
- Obesity, Lifestyle and Genetic Adaptations Study Group, Apia, Samoa
| | - Tracy Fatu
- Obesity, Lifestyle and Genetic Adaptations Study Group, Apia, Samoa
| | - Seipepa Unasa
- Obesity, Lifestyle and Genetic Adaptations Study Group, Apia, Samoa
| | | | - Abigail I Wetzel
- International Health Institute, Department of Epidemiology, School of Public Health, Brown University, Providence, RI, United States
| | | | - Angela T Prescott
- Department of Plastic and Reconstructive Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Gloria Siufaga
- Obesity, Lifestyle and Genetic Adaptations Study Group, Apia, Samoa
| | - Corina Penaia
- Asian Pacific Islander Forward Movement, Los Angeles, CA, United States
| | - Sophie B To
- Department of Social and Behavioral Sciences, Yale School of Public Health, New Haven, CT, United States
| | - Lauren C LaMonica
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, United States
| | | | - Courtney C Choy
- International Health Institute, Department of Epidemiology, School of Public Health, Brown University, Providence, RI, United States
| | - Scott E Crouter
- Department of Kinesiology, Recreation, and Sport Studies, The University of Tennessee Knoxville, Knoxville, TN, United States
| | - Susan Redline
- Departments of Medicine, Brigham and Women's Hospital and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Ranjan Deka
- Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Erin E Kershaw
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Zsolt Urban
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ryan L Minster
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Daniel E Weeks
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Stephen T McGarvey
- International Health Institute, Department of Epidemiology, School of Public Health, Brown University, Providence, RI, United States
- Department of Anthropology, Brown University, Providence, RI, United States
| |
Collapse
|
16
|
Carlson JC, Rosenthal SL, Russell EM, Hawley NL, Sun G, Cheng H, Naseri T, Reupena MS, Tuitele J, Deka R, McGarvey ST, Weeks DE, Minster RL. A missense variant in CREBRF is associated with taller stature in Samoans. Am J Hum Biol 2020; 32:e23414. [PMID: 32190945 DOI: 10.1002/ajhb.23414] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/09/2019] [Revised: 02/25/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Studies have demonstrated that rs373863828, a missense variant in CREBRF, is associated with a number of anthropometric traits including body mass index (BMI), obesity, percent body fat, hip circumference, and abdominal circumference. Given the biological relationship between height and adiposity, we hypothesized that the effect of this variant on BMI might be due in part to an association of this variant with height. METHODS We tested the hypothesis that minor allele of rs373863828 is associated with height in a Samoan population in two adult cohorts and in a separate cohort of children (age 5-18 years old) using linear mixed modeling. RESULTS We found evidence of a strong relationship between rs373863828 and greater mean height in Samoan adults (0.77 cm greater average height for each copy of the minor allele) with the same direction of effect in Samoan children. CONCLUSIONS These results suggest that the missense variant rs373863828 in CREBRF, first identified through an association with larger BMI, may be related to an underlying biological mechanism affecting overall body size including stature.
Collapse
Affiliation(s)
- Jenna C Carlson
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Samantha L Rosenthal
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Emily M Russell
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nicola L Hawley
- Department of Chronic Disease Epidemiology, School of Public Health, Yale University, New Haven, Connecticut, USA
| | - Guangyun Sun
- Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Hong Cheng
- Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Take Naseri
- Ministry of Health, Government of Samoa, Apia, Samoa
| | | | - John Tuitele
- Department of Public Health, Government of American Samoa, Pago Pago, American Samoa
| | - Ranjan Deka
- Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Stephen T McGarvey
- International Health Institute and Department of Epidemiology, School of Public Health, Brown University, Providence, Rhode Island, USA.,Department of Anthropology, Brown University, Providence, Rhode Island, USA
| | - Daniel E Weeks
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ryan L Minster
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
17
|
Abstract
We previously identified Drosophila REPTOR and REPTOR-BP as transcription factors downstream of mTORC1 that play an important role in regulating organismal metabolism. We study here the mammalian ortholog of REPTOR-BP, Crebl2. We find that Crebl2 mediates part of the transcriptional induction caused by mTORC1 inhibition. In C2C12 myoblasts, Crebl2 knockdown leads to elevated glucose uptake, elevated glycolysis as observed by lactate secretion, and elevated triglyceride biosynthesis. In Hepa1-6 hepatoma cells, Crebl2 knockdown also leads to elevated triglyceride levels. In sum, this works identifies Crebl2 as a regulator of cellular metabolism that can link nutrient sensing via mTORC1 to the metabolic response of cells.
Collapse
|
18
|
Yang D, Jiang T, Liu J, Zhang B, Lin P, Chen H, Zhou D, Tang K, Wang A, Jin Y. CREB3 regulatory factor -mTOR-autophagy regulates goat endometrial function during early pregnancy. Biol Reprod 2019; 98:713-721. [PMID: 29447354 DOI: 10.1093/biolre/ioy044] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/29/2017] [Accepted: 02/12/2018] [Indexed: 02/06/2023] Open
Abstract
In domestic ruminants, a receptive endometrium is crucial for successful pregnancy. Although many essential molecular modulators and pathways have been identified during early pregnancy, the precise mechanisms regulating goat endometrial function remains largely unknown. Here, we describe a novel regulator during early pregnancy, whereby hormones increased CREB3 regulatory factor (CREBRF) expression and act as a potential activator of autophagy in endometrial epithelial cells (EECs) via the mTOR pathway. Our results showed that knockdown of CREBRF via shCREBRF hampered EECs proliferation by S-phase cell cycle arrest and significantly inhibited endometrial function. We also reported that CREBRF-mTOR-autophagy pathway plays a vital role in regulating endometrial function, with a blockade of the mTOR by rapamycin demonstrating the regulatory function on prostaglandin (PGs) secretion and cell attachment in EECs. Moreover, chloroquine pretreatment also proved the above conclusion. Collectively, our findings provide new insight into the molecular mechanisms of goat endometrial function and indicate that the CREBRF-mTOR-autophagy pathway plays a central role in PGs secretion and cell attachment.
Collapse
Affiliation(s)
- Diqi Yang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Tingting Jiang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jianguo Liu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Beibei Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Pengfei Lin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Huatao Chen
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Dong Zhou
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Keqiong Tang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Aihua Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yaping Jin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
19
|
Khan HA, Margulies CE. The Role of Mammalian Creb3-Like Transcription Factors in Response to Nutrients. Front Genet 2019; 10:591. [PMID: 31293620 PMCID: PMC6598459 DOI: 10.3389/fgene.2019.00591] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/03/2018] [Accepted: 06/04/2019] [Indexed: 12/18/2022] Open
Abstract
Our ability to overcome the challenges behind metabolic disorders will require a detailed understanding of the regulation of responses to nutrition. The Creb3 transcription factor family appears to have a unique regulatory role that links cellular secretory capacity with development, nutritional state, infection, and other stresses. This role in regulating individual secretory capacity genes could place this family of transcription factors at an important regulatory intersection mediating an animal’s responses to nutrients and other environmental challenges. Interestingly, in both humans and mice, individuals with mutations in Creb3L3/CrebH, one of the Creb3 family members, exhibit hypertriglyceridemia (HTG) thus linking this transcription factor to lipid metabolism. We are beginning to understand how Creb3L3 and related family members are regulated and to dissect the potential redundancy and cross talk between distinct family members, thereby mediating both healthy and pathological responses to the environment. Here, we review the current knowledge on the regulation of Creb3 family transcription factor activity, their target genes, and their role in metabolic disease.
Collapse
Affiliation(s)
- Haris A Khan
- Physiological Chemistry, Biomedical Center, Ludwig-Maximilians-Universität, Munich, Germany
| | - Carla E Margulies
- Physiological Chemistry, Biomedical Center, Ludwig-Maximilians-Universität, Munich, Germany
| |
Collapse
|
20
|
St. John ME, McGirr JA, Martin CH. The behavioral origins of novelty: did increased aggression lead to scale-eating in pupfishes? Behav Ecol 2019; 30:557-569. [PMID: 30971862 PMCID: PMC6450202 DOI: 10.1093/beheco/ary196] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/02/2018] [Revised: 11/08/2018] [Accepted: 12/14/2018] [Indexed: 11/13/2022] Open
Abstract
Behavioral changes in a new environment are often assumed to precede the origins of evolutionary novelties. Here, we examined whether an increase in aggression is associated with a novel scale-eating trophic niche within a recent radiation of Cyprinodon pupfishes endemic to San Salvador Island, Bahamas. We measured aggression using multiple behavioral assays and used transcriptomic analyses to identify differentially expressed genes in aggression and other behavioral pathways across 3 sympatric species in the San Salvador radiation (generalist, snail-eating specialist, and scale-eating specialist) and 2 generalist outgroups. Surprisingly, we found increased behavioral aggression and differential expression of aggression-related pathways in both the scale-eating and snail-eating specialists, despite their independent evolutionary origins. Increased behavioral aggression varied across both sex and stimulus context in both species. Our results indicate that aggression is not unique to scale-eating specialists. Instead, selection may increase aggression in other contexts such as niche specialization in general or mate competition. Alternatively, increased aggression may result from indirect selection on craniofacial traits, pigmentation, or metabolism-all traits which are highly divergent, exhibit signs of selective sweeps, and are affected by aggression-related genetic pathways which are differentially expressed in this system. In conclusion, the evolution of a novel predatory trophic niche within a recent adaptive radiation does not have clear-cut behavioral origins as previously assumed, highlighting the multivariate nature of adaptation and the complex integration of behavior with other phenotypic traits.
Collapse
Affiliation(s)
| | - Joseph A McGirr
- Department of Biology, University of North Carolina at Chapel Hill, NC, USA
| | - Christopher H Martin
- Department of Biology, University of North Carolina at Chapel Hill, NC, USA
- Department of Integrative Biology and Museum of Vertebrate Zoology, University of California, Berkeley, CA, USA
| |
Collapse
|
21
|
Garfinkel BP, Arad S, Neuner SM, Netser S, Wagner S, Kaczorowski CC, Rosen CJ, Gal M, Soreq H, Orly J. HP1BP3 expression determines maternal behavior and offspring survival. GENES BRAIN AND BEHAVIOR 2017; 15:678-88. [PMID: 27470444 DOI: 10.1111/gbb.12312] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 03/31/2016] [Revised: 07/03/2016] [Accepted: 07/26/2016] [Indexed: 12/17/2022]
Abstract
Maternal care is an indispensable behavioral component necessary for survival and reproductive success in mammals, and postpartum maternal behavior is mediated by an incompletely understood complex interplay of signals including effects of epigenetic regulation. We approached this issue using our recently established mice with targeted deletion of heterochromatin protein 1 binding protein 3 (HP1BP3), which we found to be a novel epigenetic repressor with critical roles in postnatal growth. Here, we report a dramatic reduction in the survival of pups born to Hp1bp3(-/-) deficient mouse dams, which could be rescued by co-fostering with wild-type dams. Hp1bp3(-/-) females failed to retrieve both their own pups and foster pups in a pup retrieval test, and showed reduced anxiety-like behavior in the open-field and elevated-plus-maze tests. In contrast, Hp1bp3(-/-) females showed no deficits in behaviors often associated with impaired maternal care, including social behavior, depression, motor coordination and olfactory capability; and maintained unchanged anxiety-associated hallmarks such as cholinergic status and brain miRNA profiles. Collectively, our results suggest a novel role for HP1BP3 in regulating maternal and anxiety-related behavior in mice and call for exploring ways to manipulate this epigenetic process.
Collapse
Affiliation(s)
- B P Garfinkel
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel. .,Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - S Arad
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.,Biomedical Sciences, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - S M Neuner
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - S Netser
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - S Wagner
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - C C Kaczorowski
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - C J Rosen
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - M Gal
- Biomedical Sciences, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.,The IVF Unit - Obstetrics and Gynecology Department, Shaare Zedek Medical Center, Jerusalem, Israel
| | - H Soreq
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.,Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - J Orly
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
22
|
Penney J, Mendell A, Zeng M, Tran K, Lymer J, Turner PV, Choleris E, MacLusky N, Lu R. LUMAN/CREB3 is a key regulator of glucocorticoid-mediated stress responses. Mol Cell Endocrinol 2017; 439:95-104. [PMID: 27789393 DOI: 10.1016/j.mce.2016.10.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 05/06/2016] [Revised: 10/21/2016] [Accepted: 10/21/2016] [Indexed: 01/28/2023]
Abstract
Altered glucocorticoid sensitivity is believed to contribute to a number of human diseases, including inflammatory and autoimmune conditions as well as disorders characterized by abnormal hypothalamic-pituitary-adrenal axis (HPA) function. LUMAN (or CREB3), originally identified through its interaction with a cell cycle regulator HCFC1, is an endoplasmic reticulum membrane-bound transcription factor that is involved in the unfolded protein response. Here we demonstrate that LUMAN changes the glucocorticoid response by modulating the expression of the glucocorticoid receptor leading to an overall increase in GR activity. Luman-deficient mice exhibited a blunted stress response characterized by low levels of both anxiety and depressive-like behaviour in addition to low circulating corticosterone levels. These mice also have reduced dendritic branching in the CA3 region of the hippocampus, consistent with increased GR responses. These findings are consistent with the notion that elevated GR activities are the primary cause of the observed phenotype in these LUMAN-deficient mice. We thus postulate that LUMAN is a key regulator of GR-mediated signaling and modulates HPA axis reactivity.
Collapse
Affiliation(s)
- Jenna Penney
- Department of Molecular and Cellular Biology, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Ari Mendell
- Department of Biomedical Sciences, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Minghua Zeng
- Department of Molecular and Cellular Biology, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Khoa Tran
- Department of Biomedical Sciences, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Jennifer Lymer
- Department of Psychology, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Patricia V Turner
- Department of Pathobiology, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Elena Choleris
- Department of Psychology, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Neil MacLusky
- Department of Biomedical Sciences, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Ray Lu
- Department of Molecular and Cellular Biology, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada.
| |
Collapse
|
23
|
Audas TE, Hardy-Smith PW, Penney J, Taylor T, Lu R. Characterization of nuclear foci-targeting of Luman/CREB3 recruitment factor (LRF/CREBRF) and its potential role in inhibition of herpes simplex virus-1 replication. Eur J Cell Biol 2016; 95:611-622. [PMID: 28029379 DOI: 10.1016/j.ejcb.2016.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/11/2016] [Revised: 10/07/2016] [Accepted: 10/24/2016] [Indexed: 01/18/2023] Open
Abstract
The recently identified Luman/CREB3-binding partner LRF (Luman/CREB3 recruitment factor) was shown to localize to discrete sub-nuclear foci. Luman is implicated in herpes simplex virus-1 (HSV-1) latency/reactivation and the unfolded protein response (UPR) pathway; therefore, we sought to characterize the formation of the LRF nuclear foci in the context of cellular signaling and HSV-1 replication. Here, we mapped the nuclear foci-targeting sequence to the central region containing the first leucine zipper (a.a.415-519), and found that the integrity of the whole region appears essential for LRF foci formation. LRF foci integrity was unaffected by inhibition of cellular DNA replication and translation, however, disruption of transcription resulted in altered LRF localization. When compared to other cellular and viral foci LRF co-localized with the nuclear receptor co-activator GRIP1, while the HSV-1 gene products ICP4, ICP27 and VP13/14 disrupted foci formation to varying degrees. Interestingly, cells over-expressing LRF were resistant to productive HSV-1 infection and this resistance was dependent upon protein targeting and an N-terminal transactivation domain. When LRF knockdown cells were subjected to primary infection, HSV-1 gene expression and progeny virus yield were enhanced by ∼3 fold compared to wildtype cells. Taken together, these results indicate that LRF is a key regulator that may act direct or indirectly as a repressor of essential genes required for productive viral infection.
Collapse
Affiliation(s)
- Timothy E Audas
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada; Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, B.C., V5A 1S6, Canada
| | - Philip W Hardy-Smith
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Jenna Penney
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Tiegh Taylor
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Ray Lu
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
24
|
A thrifty variant in CREBRF strongly influences body mass index in Samoans. Nat Genet 2016; 48:1049-1054. [PMID: 27455349 PMCID: PMC5069069 DOI: 10.1038/ng.3620] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/07/2015] [Accepted: 06/15/2016] [Indexed: 12/14/2022]
Abstract
Samoans are a unique founder population with a high prevalence of obesity, making them well suited for identifying new genetic contributors to obesity. We conducted a genome-wide association study (GWAS) in 3,072 Samoans, discovered a variant, rs12513649, strongly associated with body mass index (BMI) (P = 5.3 × 10(-14)), and replicated the association in 2,102 additional Samoans (P = 1.2 × 10(-9)). Targeted sequencing identified a strongly associated missense variant, rs373863828 (p.Arg457Gln), in CREBRF (meta P = 1.4 × 10(-20)). Although this variant is extremely rare in other populations, it is common in Samoans (frequency of 0.259), with an effect size much larger than that of any other known common BMI risk variant (1.36-1.45 kg/m(2) per copy of the risk-associated allele). In comparison to wild-type CREBRF, the Arg457Gln variant when overexpressed selectively decreased energy use and increased fat storage in an adipocyte cell model. These data, in combination with evidence of positive selection of the allele encoding p.Arg457Gln, support a 'thrifty' variant hypothesis as a factor in human obesity.
Collapse
|
25
|
Li X, Lin P, Chen F, Wang N, Zhao F, Wang A, Jin Y. Luman recruiting factor is involved in stromal cell proliferation during decidualization in mice. Cell Tissue Res 2016; 365:437-47. [DOI: 10.1007/s00441-016-2392-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/26/2015] [Accepted: 03/03/2016] [Indexed: 11/29/2022]
|
26
|
Arensdorf AM, Diedrichs D, Rutkowski DT. Regulation of the transcriptome by ER stress: non-canonical mechanisms and physiological consequences. Front Genet 2013; 4:256. [PMID: 24348511 PMCID: PMC3844873 DOI: 10.3389/fgene.2013.00256] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/24/2013] [Accepted: 11/08/2013] [Indexed: 12/29/2022] Open
Abstract
The mammalian unfolded protein response (UPR) is propagated by three ER-resident transmembrane proteins, each of which initiates a signaling cascade that ultimately culminates in production of a transcriptional activator. The UPR was originally characterized as a pathway for upregulating ER chaperones, and a comprehensive body of subsequent work has shown that protein synthesis, folding, oxidation, trafficking, and degradation are all transcriptionally enhanced by the UPR. However, the global reach of the UPR extends to genes involved in diverse physiological processes having seemingly little to do with ER protein folding, and this includes a substantial number of mRNAs that are suppressed by stress rather than stimulated. Through multiple non-canonical mechanisms emanating from each of the UPR pathways, the cell dynamically regulates transcription and mRNA degradation. Here we highlight these mechanisms and their increasingly appreciated impact on physiological processes.
Collapse
Affiliation(s)
- Angela M Arensdorf
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine Iowa City, IA, USA
| | - Danilo Diedrichs
- Department of Mathematics and Computer Science, Wheaton College Wheaton, IL, USA
| | - D Thomas Rutkowski
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine Iowa City, IA, USA ; Department of Internal Medicine, University of Iowa Carver College of Medicine Iowa City, IA, USA
| |
Collapse
|
27
|
Yang Y, Jin Y, Lin P, Hu L, Cui C, Li X, Li Q, Wang A. THE EXPRESSION AND LOCALIZATION OF LRF IN THE FEMALE REPRODUCTIVE TRACT OF CYCLING MICE THROUGHOUT THE ESTROUS CYCLE. J Immunoassay Immunochem 2013; 34:313-22. [DOI: 10.1080/15321819.2012.732169] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/11/2023]
|
28
|
Zalachoras I, Houtman R, Meijer OC. Understanding stress-effects in the brain via transcriptional signal transduction pathways. Neuroscience 2013; 242:97-109. [PMID: 23545270 DOI: 10.1016/j.neuroscience.2013.03.038] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/22/2012] [Revised: 03/22/2013] [Accepted: 03/23/2013] [Indexed: 12/22/2022]
Abstract
Glucocorticoid hormones exert crucial effects on the brain in relation to physiology, endocrine regulation, mood and cognition. Their two receptor types, glucocorticoid and mineralocorticoid receptors (GR and MR), are members of the nuclear receptor superfamily and act in large measure as transcription factors. The outcome of MR/GR action on the genome depends on interaction with members from different protein families, which are of crucial importance for cross-talk with other neuronal and hormonal signals that impinge on the glucocorticoid sensitive circuitry. Relevant interacting proteins include other transcription factors that may either tether the receptor to the DNA, or that bind in the vicinity of GR and MR to tune the transcriptional response. In addition, transcriptional coregulator proteins constitute the actual signal transduction pathway to the transcription machinery. We review the current evidence for involvement of individual coregulators in GR-dependent effects on stress responses, and learning and memory. We discuss the use of in vitro and in silico tools to predict those coregulators that are of importance for particular brain processes. Finally, we discuss the potential of selective receptor modulators that may only allow a subset of all interactions, thus allowing more selective targeting of glucocorticoid-dependent processes in the brain.
Collapse
Affiliation(s)
- I Zalachoras
- Department of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | |
Collapse
|
29
|
Yang Y, Jin Y, Martyn AC, Lin P, Song Y, Chen F, Hu L, Cui C, Li X, Li Q, Lu R, Wang A. Expression pattern implicates a potential role for luman recruitment factor in the process of implantation in uteri and development of preimplantation embryos in mice. J Reprod Dev 2013; 59:245-51. [PMID: 23400243 PMCID: PMC3934142 DOI: 10.1262/jrd.2012-137] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/20/2022] Open
Abstract
Luman/CREB3 recruitment factor (LRF or CREBRF) was identified as a regulator of Luman
(or CREB3) that is involved in the unfolded protein response during endoplasmic reticulum
stress. Luman is implicated in a multitude of functions ranging from viral infection and
immunity to cancer. The biological function of LRF, however, is unknown. In this paper, we
report that uteri of pregnant mice and embryos displayed enhanced LRF expression at all
stages, and the expressed LRF was found to be localized specifically at implantation
sites. On the other hand, uteri of mice induced for delayed implantation or pseudopregnant
mice showed low levels of LRF expression, suggesting that LRF mediates uterine receptivity
during implantation. Further, expression of LRF was found to be modulated by steroid
hormones such as progesterone and estradiol. This study thereby identifies a potential
role for LRF in the process of implantation in uteri and development of preimplantation
embryos in mice.
Collapse
Affiliation(s)
- Yanzhou Yang
- Key Open Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling Shaanxi 712100, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|