1
|
Brinkmeier ML, Cheung LYM, O'Connell SP, Gutierrez DK, Rhoads EC, Camper SA, Davis SW. Nucleoredoxin regulates WNT signaling during pituitary stem cell differentiation. Hum Mol Genet 2025:ddaf032. [PMID: 40044116 DOI: 10.1093/hmg/ddaf032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 02/23/2025] [Indexed: 03/12/2025] Open
Abstract
Nucleoredoxin (Nxn) encodes a multi-functional enzyme with oxidoreductase activity that regulates many different signaling pathways and cellular processes in a redox-dependent manner. Rare NXN mutations are reported in individuals with recessive Robinow syndrome, which involves mesomelic skeletal dysplasia, short stature, craniofacial dysmorphisms, and incompletely penetrant heart and palate defects. Here we report that Nxn is expressed in the ventral diencephalon and developing pituitary gland, and that Nxn deficient mice have pituitary dysmorphology and craniofacial abnormalities that include defects in the skull base and cleft palate. Nxn mutant mice exhibit reduced WNT signaling and reduced differentiation of pituitary stem cells into hormone-producing cells. These results suggest patients with Robinow syndrome could benefit from evaluation by endocrinologists for pituitary structural imaging and hormone insufficiency.
Collapse
Affiliation(s)
- Michelle L Brinkmeier
- Department of Human Genetics, 5805 Medical Science II, 1241 Catherine St, University of Michigan, Ann Arbor, MI 48109-5618, United States
| | - Leonard Y M Cheung
- Department of Human Genetics, 5805 Medical Science II, 1241 Catherine St, University of Michigan, Ann Arbor, MI 48109-5618, United States
| | - Sean P O'Connell
- Department of Biological Sciences, 715 Sumter St, CLS room 401, University of South Carolina, Columbia, SC 29208, United States
| | - Diana K Gutierrez
- Department of Biological Sciences, 715 Sumter St, CLS room 401, University of South Carolina, Columbia, SC 29208, United States
| | - Eve C Rhoads
- Department of Human Genetics, 5805 Medical Science II, 1241 Catherine St, University of Michigan, Ann Arbor, MI 48109-5618, United States
| | - Sally A Camper
- Department of Human Genetics, 5805 Medical Science II, 1241 Catherine St, University of Michigan, Ann Arbor, MI 48109-5618, United States
| | - Shannon W Davis
- Department of Biological Sciences, 715 Sumter St, CLS room 401, University of South Carolina, Columbia, SC 29208, United States
| |
Collapse
|
2
|
Brinkmeier ML, Cheung LYM, O’Connell SP, Gutierrez DK, Rhoads EC, Camper SA, Davis SW. Nucleoredoxin regulates WNT signaling during pituitary stem cell differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635771. [PMID: 39975280 PMCID: PMC11838423 DOI: 10.1101/2025.01.30.635771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Nucleoredoxin (Nxn) encodes a multi-functional enzyme with oxidoreductase activity that regulates many different signaling pathways and cellular processes in a redox-dependent manner. Rare NXN mutations are reported in individuals with recessive Robinow syndrome, which involves mesomelic skeletal dysplasia, short stature, craniofacial dysmorphisms, and incompletely penetrant heart and palate defects. Here we report that Nxn is expressed in the ventral diencephalon and developing pituitary gland, and that Nxn deficient mice have pituitary dysmorphology and craniofacial abnormalities that include defects in the skull base and cleft palate. Nxn mutant mice exhibit reduced WNT signaling and reduced differentiation of pituitary stem cells into hormone-producing cells. These results suggest patients with Robinow syndrome could benefit from evaluation by endocrinologists for pituitary structural imaging and hormone insufficiency.
Collapse
Affiliation(s)
| | - Leonard Y. M. Cheung
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, 48109-5618, USA
- Current address: Department of Physiology and Biophysics, Renaissance School of Medicine, State University of New York, Stonybrook, NY 11794, USA
| | - Sean P. O’Connell
- Department of Biological Sciences, University of South Carolina, Columbia, SC, 29208, USA
| | - Diana K. Gutierrez
- Department of Biological Sciences, University of South Carolina, Columbia, SC, 29208, USA
| | - Eve C. Rhoads
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, 48109-5618, USA
| | - Sally A. Camper
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, 48109-5618, USA
| | - Shannon W. Davis
- Department of Biological Sciences, University of South Carolina, Columbia, SC, 29208, USA
| |
Collapse
|
3
|
Yang S, Bussing A, Marra G, Brinkmeier ML, Camper SA, Davis SW, Ho YY. TIME-CoExpress: Temporal Trajectory Modeling of Dynamic Gene Co-expression Patterns Using Single-Cell Transcriptomics Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634392. [PMID: 39896591 PMCID: PMC11785143 DOI: 10.1101/2025.01.23.634392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The rapid advancements of single-cell RNA sequencing (scRNAseq) technology provide high-resolution views of transcriptomic activity within a single cell. Most routine analyses of scRNAseq data focus on individual genes; however, the one-gene-at-a-time analysis is likely to miss meaningful genetic interactions. Gene co-expression analysis addresses this issue by identifying coordinated gene expression changes in response to cellular conditions, such as developmental or temporal trajectory. Identifying differential co-expression gene combinations along the cell temporal trajectory using scRNAseq data can provide deeper insight into the biological processes. Existing approaches for gene co-expression analysis assume a restrictive linear change of gene co-expression. In this paper, we propose a copula-based approach with proper data-driven smoothing functions to model non-linear gene co-expression changes along cellular temporal trajectories. Our proposed approach provides flexibility to incorporate characteristics such as over-dispersion and zero-inflation rate observed in scRNAseq data into the modeling framework. We conducted a series of simulation analyses to evaluate the performance of the proposed algorithm. We demonstrate the implementation of the proposed algorithm using a scRNAseq dataset and identify differential co-expression gene pairs along cell temporal trajectory in pituitary embryonic development comparing Nxn - / - mutated versus wild-type mice.
Collapse
|
4
|
Fei S, Rule BD, Godwin JS, Mobley CB, Roberts MD, von Walden F, Vechetti IJ. miRNA-1 regulation is necessary for mechanical overload-induced muscle hypertrophy in male mice. Physiol Rep 2025; 13:e70166. [PMID: 39761956 PMCID: PMC11705529 DOI: 10.14814/phy2.70166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
MicroRNAs (miRNAs) are small, noncoding RNAs that play a critical role in regulating gene expression post-transcriptionally. They are involved in various developmental and physiological processes, and their dysregulation is linked to various diseases. Skeletal muscle-specific miRNAs, including miR-1, play a crucial role in the development and maintenance of skeletal muscle. It has been demonstrated that the expression of miR-1 decreases by approximately 50% in response to hypertrophic stimuli, suggesting its potential involvement in muscle hypertrophy. In our study, we hypothesize that reduction of miR-1 levels is necessary for skeletal muscle growth due to its interaction to essential pro-growth genes. Promoting a smaller reduction of miR-1 levels, we observed a blunted hypertrophic response in mice undergoing a murine model of muscle hypertrophy. In addition, our results suggest that miR-1 inhibits the expression of Itm2a, a membrane-related protein, as potential miR-1-related candidate for skeletal muscle hypertrophy. While the exact mechanism in muscle hypertrophy has not been identified, our results suggest that miR-1-regulated membrane proteins are important for skeletal muscle hypertrophy.
Collapse
Affiliation(s)
- Shengyi Fei
- Department of Nutrition and Health SciencesUniversity of Nebraska‐LincolnLincolnNebraskaUSA
| | - Blake D. Rule
- Department of Nutrition and Health SciencesUniversity of Nebraska‐LincolnLincolnNebraskaUSA
| | | | | | | | | | - Ivan J. Vechetti
- Department of Nutrition and Health SciencesUniversity of Nebraska‐LincolnLincolnNebraskaUSA
| |
Collapse
|
5
|
Bellanti F, Mangieri D, Vendemiale G. Redox Biology and Liver Fibrosis. Int J Mol Sci 2023; 25:410. [PMID: 38203581 PMCID: PMC10778611 DOI: 10.3390/ijms25010410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/21/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Hepatic fibrosis is a complex process that develops in chronic liver diseases. Even though the initiation and progression of fibrosis rely on the underlying etiology, mutual mechanisms can be recognized and targeted for therapeutic purposes. Irrespective of the primary cause of liver disease, persistent damage to parenchymal cells triggers the overproduction of reactive species, with the consequent disruption of redox balance. Reactive species are important mediators for the homeostasis of both hepatocytes and non-parenchymal liver cells. Indeed, other than acting as cytotoxic agents, reactive species are able to modulate specific signaling pathways that may be relevant to hepatic fibrogenesis. After a brief introduction to redox biology and the mechanisms of fibrogenesis, this review aims to summarize the current evidence of the involvement of redox-dependent pathways in liver fibrosis and focuses on possible therapeutic targets.
Collapse
Affiliation(s)
- Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
| | - Domenica Mangieri
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Gianluigi Vendemiale
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
| |
Collapse
|
6
|
Zhang L, Li Z, Zhang M, Zou H, Bai Y, Liu Y, Lv J, Lv L, Liu P, Deng Z, Liu C. Advances in the molecular mechanism and targeted therapy of radioactive-iodine refractory differentiated thyroid cancer. Med Oncol 2023; 40:258. [PMID: 37524925 DOI: 10.1007/s12032-023-02098-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/21/2023] [Indexed: 08/02/2023]
Abstract
Most patients with differentiated thyroid cancer have a good prognosis after radioactive iodine-131 treatment, but there are still a small number of patients who are not sensitive to radioiodine treatment and may subsequently show disease progression. Therefore, radioactive-iodine refractory differentiated thyroid cancer treated with radioiodine usually shows reduced radioiodine uptake. Thus, when sodium iodine symporter expression, basolateral membrane localization and recycling degradation are abnormal, radioactive-iodine refractory differentiated thyroid cancer may occur. In recent years, with the deepening of research into the pathogenesis of this disease, an increasing number of molecules have become or are expected to become therapeutic targets. The application of corresponding inhibitors or combined treatment regimens for different molecular targets may be effective for patients with advanced radioactive-iodine refractory differentiated thyroid cancer. Currently, some targeted drugs that can improve the progression-free survival of patients with radioactive-iodine refractory differentiated thyroid cancer, such as sorafenib and lenvatinib, have been approved by the FDA for the treatment of radioactive-iodine refractory differentiated thyroid cancer. However, due to the adverse reactions and drug resistance caused by some targeted drugs, their application is limited. In response to targeted drug resistance and high rates of adverse reactions, research into new treatment combinations is being carried out; in addition to kinase inhibitor therapy, gene therapy and rutin-assisted iodine-131 therapy for radioactive-iodine refractory thyroid cancer have also made some progress. Thus, this article mainly focuses on sodium iodide symporter changes leading to the main molecular mechanisms in radioactive-iodine refractory differentiated thyroid cancer, some targeted drug resistance mechanisms and promising new treatments.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Zhi Li
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Meng Zhang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Huangren Zou
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Yuke Bai
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Yanlin Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Juan Lv
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Ling Lv
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Pengjie Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Zhiyong Deng
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China.
| | - Chao Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| |
Collapse
|
7
|
Craig SEL, Michalski MN, Williams BO. Got WNTS? Insight into bone health from a WNT perspective. Curr Top Dev Biol 2023; 153:327-346. [PMID: 36967199 DOI: 10.1016/bs.ctdb.2023.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
WNT signaling, essential for many aspects of development, is among the most commonly altered pathways associated with human disease. While initially studied in cancer, dysregulation of WNT signaling has been determined to be essential for skeletal development and the maintenance of bone health throughout life. In this review, we discuss the role of Wnt signaling in bone development and disease with a particular focus on two areas. First, we discuss the roles of WNT signaling pathways in skeletal development, with an emphasis on congenital and idiopathic skeletal syndromes and diseases that are associated with genetic variations in WNT signaling components. Next, we cover a topic that has long been an interest of our laboratory, how high and low levels of WNT signaling affects the establishment and maintenance of healthy bone mass. We conclude with a discussion of the status of WNT-based therapeutics in the treatment of skeletal disease.
Collapse
Affiliation(s)
- Sonya E L Craig
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI, United States
| | - Megan N Michalski
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI, United States
| | - Bart O Williams
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI, United States.
| |
Collapse
|
8
|
Zhang Y, Zuo D, Qiu J, Li K, Niu Y, Yuan Y, Qiu Y, Qiao L, He W, Wang C, Yuan Y, Li B. NXN suppresses metastasis of hepatocellular carcinoma by promoting degradation of Snail through binding to DUB3. Cell Death Dis 2022; 13:676. [PMID: 35927236 PMCID: PMC9352874 DOI: 10.1038/s41419-022-05135-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 07/04/2022] [Accepted: 07/25/2022] [Indexed: 01/21/2023]
Abstract
The poor prognosis of hepatocellular carcinoma (HCC) could be attributed to its high metastasis rate. Here, we report the role of nucleoredoxin (NXN), a multifunctional redox-active protein, in HCC metastasis. The expression of NXN in HCC tissues was measured by immunohistochemistry. The role of NXN on HCC proliferation was determined by CCK-8, EdU and colony formation assays in vitro and subcutaneous tumor formation model in vivo. Transwell and wound healing assays and tail vein injection model were performed to assess the function of NXN on HCC metastasis. Co-immunoprecipitation assay was performed to examine the interaction among NXN, Snail and DUB3. Our results showed that NXN was downregulated in HCC tissues compared to adjacent liver tissues. Patients with low NXN expression had shorter overall survival (OS) time (P < 0.001) than those with high NXN expression. Biologically, ectopic expression of NXN significantly inhibited the proliferation and metastasis of HCC cells both in vitro and in vivo by suppressing epithelial-mesenchymal transition (EMT). Mechanistically, NXN promoted the ubiquitin-proteasome-mediated degradation of Snail through interaction with DUB3. Further, depletion of Snail abolished NXN-inhibited cell proliferation and metastasis. In summary, NXN suppressed the proliferation and metastasis of HCC by inhibiting DUB3-mediated deubiquitylation of Snail protein. Our study demonstrates that NXN, DUB3 and Snail complex functioned as an important regulatory mechanism of HCC progression and indicates a potential therapeutic approach for the treatment of HCC metastasis.
Collapse
Affiliation(s)
- Yuanping Zhang
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Dinglan Zuo
- grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jiliang Qiu
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Kai Li
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yi Niu
- grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yichuan Yuan
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yuxiong Qiu
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Liang Qiao
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wei He
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Chenwei Wang
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yunfei Yuan
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Binkui Li
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
9
|
Guo CL. Self-Sustained Regulation or Self-Perpetuating Dysregulation: ROS-dependent HIF-YAP-Notch Signaling as a Double-Edged Sword on Stem Cell Physiology and Tumorigenesis. Front Cell Dev Biol 2022; 10:862791. [PMID: 35774228 PMCID: PMC9237464 DOI: 10.3389/fcell.2022.862791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/29/2022] [Indexed: 12/19/2022] Open
Abstract
Organ development, homeostasis, and repair often rely on bidirectional, self-organized cell-niche interactions, through which cells select cell fate, such as stem cell self-renewal and differentiation. The niche contains multiplexed chemical and mechanical factors. How cells interpret niche structural information such as the 3D topology of organs and integrate with multiplexed mechano-chemical signals is an open and active research field. Among all the niche factors, reactive oxygen species (ROS) have recently gained growing interest. Once considered harmful, ROS are now recognized as an important niche factor in the regulation of tissue mechanics and topology through, for example, the HIF-YAP-Notch signaling pathways. These pathways are not only involved in the regulation of stem cell physiology but also associated with inflammation, neurological disorder, aging, tumorigenesis, and the regulation of the immune checkpoint molecule PD-L1. Positive feedback circuits have been identified in the interplay of ROS and HIF-YAP-Notch signaling, leading to the possibility that under aberrant conditions, self-organized, ROS-dependent physiological regulations can be switched to self-perpetuating dysregulation, making ROS a double-edged sword at the interface of stem cell physiology and tumorigenesis. In this review, we discuss the recent findings on how ROS and tissue mechanics affect YAP-HIF-Notch-PD-L1 signaling, hoping that the knowledge can be used to design strategies for stem cell-based and ROS-targeting therapy and tissue engineering.
Collapse
Affiliation(s)
- Chin-Lin Guo
- Institute of Physics, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
10
|
Idelfonso-García OG, Alarcón-Sánchez BR, Vásquez-Garzón VR, Baltiérrez-Hoyos R, Villa-Treviño S, Muriel P, Serrano H, Pérez-Carreón JI, Arellanes-Robledo J. Is Nucleoredoxin a Master Regulator of Cellular Redox Homeostasis? Its Implication in Different Pathologies. Antioxidants (Basel) 2022; 11:antiox11040670. [PMID: 35453355 PMCID: PMC9030443 DOI: 10.3390/antiox11040670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
Nucleoredoxin (NXN), an oxidoreductase enzyme, contributes to cellular redox homeostasis by regulating different signaling pathways in a redox-dependent manner. By interacting with seven proteins so far, namely disheveled (DVL), protein phosphatase 2A (PP2A), phosphofructokinase-1 (PFK1), translocation protein SEC63 homolog (SEC63), myeloid differentiation primary response gene-88 (MYD88), flightless-I (FLII), and calcium/calmodulin-dependent protein kinase II type alpha (CAMK2A), NXN is involved in the regulation of several key cellular processes, including proliferation, organogenesis, cell cycle progression, glycolysis, innate immunity and inflammation, motility, contraction, protein transport into the endoplasmic reticulum, neuronal plasticity, among others; as a result, NXN has been implicated in different pathologies, such as cancer, alcoholic and polycystic liver disease, liver fibrogenesis, obesity, Robinow syndrome, diabetes mellitus, Alzheimer’s disease, and retinitis pigmentosa. Together, this evidence places NXN as a strong candidate to be a master redox regulator of cell physiology and as the hub of different redox-sensitive signaling pathways and associated pathologies. This review summarizes and discusses the current insights on NXN-dependent redox regulation and its implication in different pathologies.
Collapse
Affiliation(s)
- Osiris Germán Idelfonso-García
- Laboratory of Liver Diseases, National Institute of Genomic Medicine–INMEGEN, Mexico City 14610, Mexico; (O.G.I.-G.); (B.R.A.-S.); (J.I.P.-C.)
- Department of Health Sciences, Metropolitan Autonomous University-Iztapalapa Campus, Mexico City 09340, Mexico;
| | - Brisa Rodope Alarcón-Sánchez
- Laboratory of Liver Diseases, National Institute of Genomic Medicine–INMEGEN, Mexico City 14610, Mexico; (O.G.I.-G.); (B.R.A.-S.); (J.I.P.-C.)
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute–CINVESTAV-IPN, Mexico City 07360, Mexico;
| | - Verónica Rocío Vásquez-Garzón
- Laboratory of Fibrosis and Cancer, Faculty of Medicine and Surgery, ‘Benito Juárez’ Autonomous University of Oaxaca–UABJO, Oaxaca 68020, Mexico; (V.R.V.-G.); (R.B.-H.)
- Directorate of Cátedras, National Council of Science and Technology–CONACYT, Mexico City 03940, Mexico
| | - Rafael Baltiérrez-Hoyos
- Laboratory of Fibrosis and Cancer, Faculty of Medicine and Surgery, ‘Benito Juárez’ Autonomous University of Oaxaca–UABJO, Oaxaca 68020, Mexico; (V.R.V.-G.); (R.B.-H.)
- Directorate of Cátedras, National Council of Science and Technology–CONACYT, Mexico City 03940, Mexico
| | - Saúl Villa-Treviño
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute–CINVESTAV-IPN, Mexico City 07360, Mexico;
| | - Pablo Muriel
- Laboratory of Experimental Hepatology, Department of Pharmacology, Center for Research and Advanced Studies of the National Polytechnic Institute–CINVESTAV-IPN, Mexico City 07360, Mexico;
| | - Héctor Serrano
- Department of Health Sciences, Metropolitan Autonomous University-Iztapalapa Campus, Mexico City 09340, Mexico;
| | - Julio Isael Pérez-Carreón
- Laboratory of Liver Diseases, National Institute of Genomic Medicine–INMEGEN, Mexico City 14610, Mexico; (O.G.I.-G.); (B.R.A.-S.); (J.I.P.-C.)
| | - Jaime Arellanes-Robledo
- Laboratory of Liver Diseases, National Institute of Genomic Medicine–INMEGEN, Mexico City 14610, Mexico; (O.G.I.-G.); (B.R.A.-S.); (J.I.P.-C.)
- Directorate of Cátedras, National Council of Science and Technology–CONACYT, Mexico City 03940, Mexico
- Correspondence: ; Tel.: +52-55-5350-1900 (ext. 1218)
| |
Collapse
|
11
|
Ascorbic acid regulates mouse spermatogonial stem cell proliferation in a Wnt/β-catenin/ROS signaling dependent manner. Theriogenology 2022; 184:61-72. [DOI: 10.1016/j.theriogenology.2022.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 11/17/2022]
|
12
|
Cowan AD, Ciulli A. Driving E3 Ligase Substrate Specificity for Targeted Protein Degradation: Lessons from Nature and the Laboratory. Annu Rev Biochem 2022; 91:295-319. [PMID: 35320687 DOI: 10.1146/annurev-biochem-032620-104421] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Methods to direct the degradation of protein targets with proximity-inducing molecules that coopt the cellular degradation machinery are advancing in leaps and bounds, and diverse modalities are emerging. The most used and well-studied approach is to hijack E3 ligases of the ubiquitin-proteasome system. E3 ligases use specific molecular recognition to determine which proteins in the cell are ubiquitinated and degraded. This review focuses on the structural determinants of E3 ligase recruitment of natural substrates and neo-substrates obtained through monovalent molecular glues and bivalent proteolysis-targeting chimeras. We use structures to illustrate the different types of substrate recognition and assess the basis for neo-protein-protein interactions in ternary complex structures. The emerging structural and mechanistic complexity is reflective of the diverse physiological roles of protein ubiquitination. This molecular insight is also guiding the application of structure-based design approaches to the development of new and existing degraders as chemical tools and therapeutics. Expected final online publication date for the Annual Review of Biochemistry, Volume 91 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Angus D Cowan
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, United Kingdom;
| | - Alessio Ciulli
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, United Kingdom;
| |
Collapse
|
13
|
Impact of Oxidative Stress on Embryogenesis and Fetal Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1391:221-241. [PMID: 36472825 DOI: 10.1007/978-3-031-12966-7_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple cellular processes are regulated by oxygen radicals or reactive oxygen species (ROS) where they play crucial roles as primary or secondary messengers, particularly during cell proliferation, differentiation, and apoptosis. Embryogenesis and organogenesis encompass all these processes; therefore, their role during these crucial life events cannot be ignored, more so when there is an imbalance in redox homeostasis. Perturbed redox homeostasis is responsible for damaging the biomolecules such as lipids, proteins, and nucleic acids resulting in leaky membrane, altered protein, enzyme function, and DNA damage which have adverse impact on the embryo and fetal development. In this article, we attempt to summarize the available data in literature for an in-depth understanding of redox regulation during development that may help in optimizing the pregnancy outcome both under natural and assisted conditions.
Collapse
|
14
|
Funato Y, Yamazaki D, Okuzaki D, Yamamoto N, Miki H. Importance of the renal ion channel TRPM6 in the circadian secretion of renin to raise blood pressure. Nat Commun 2021; 12:3683. [PMID: 34140503 PMCID: PMC8211686 DOI: 10.1038/s41467-021-24063-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 06/01/2021] [Indexed: 11/30/2022] Open
Abstract
Blood pressure has a daily pattern, with higher values in the active period. Its elevation at the onset of the active period substantially increases the risk of fatal cardiovascular events. Renin secretion stimulated by renal sympathetic neurons is considered essential to this process; however, its regulatory mechanism remains largely unknown. Here, we show the importance of transient receptor potential melastatin-related 6 (TRPM6), a Mg2+-permeable cation channel, in augmenting renin secretion in the active period. TRPM6 expression is significantly reduced in the distal convoluted tubule of hypotensive Cnnm2-deficient mice. We generate kidney-specific Trpm6-deficient mice and observe a decrease in blood pressure and a disappearance of its circadian variation. Consistently, renin secretion is not augmented in the active period. Furthermore, renin secretion after pharmacological activation of β-adrenoreceptor, the target of neuronal stimulation, is abrogated, and the receptor expression is decreased in renin-secreting cells. These results indicate crucial roles of TRPM6 in the circadian regulation of blood pressure. Circadian variation of blood pressure, with higher values in the active period, is associated with the risk of fatal cardiovascular events. Here, we show the importance of renal TRPM6, a Magnesium-permeable cation channel, in raising blood pressure by stimulating renin secretion.
Collapse
Affiliation(s)
- Yosuke Funato
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Daisuke Yamazaki
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Nobuhiko Yamamoto
- Neuroscience Laboratories, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Hiroaki Miki
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
| |
Collapse
|
15
|
Zhang C, Mazzeu JF, Eisfeldt J, Grochowski CM, White J, Akdemir ZC, Jhangiani SN, Muzny DM, Gibbs RA, Lindstrand A, Lupski JR, Sutton VR, Carvalho CMB. Novel pathogenic genomic variants leading to autosomal dominant and recessive Robinow syndrome. Am J Med Genet A 2020; 185:3593-3600. [PMID: 33048444 DOI: 10.1002/ajmg.a.61908] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/11/2020] [Accepted: 09/19/2020] [Indexed: 12/29/2022]
Abstract
Robinow syndrome (RS) is a genetically heterogeneous disorder characterized by skeletal dysplasia and a distinctive facial appearance. Previous studies have revealed locus heterogeneity with rare variants in DVL1, DVL3, FZD2, NXN, ROR2, and WNT5A underlying the etiology of RS. The aforementioned "Robinow-associated genes" and their gene products all play a role in the WNT/planar cell polarity signaling pathway. We performed gene-targeted Sanger sequencing, exome sequencing, genome sequencing, and array comparative genomic hybridization on four subjects with a clinical diagnosis of RS who had not had prior DNA testing. Individuals in our cohort were found to carry pathogenic or likely pathogenic variants in three RS related genes: DVL1, ROR2, and NXN. One subject was found to have a nonsense variant (c.817C > T [p.Gln273*]) in NXN in trans with an ~1 Mb telomeric deletion on chromosome 17p containing NXN, which supports our contention that biallelic NXN variant alleles are responsible for a novel autosomal recessive RS locus. These findings provide increased understanding of the role of WNT signaling in skeletal development and maintenance. These data further support the hypothesis that dysregulation of the noncanonical WNT pathway in humans gives rise to RS.
Collapse
Affiliation(s)
- Chaofan Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Juliana F Mazzeu
- University of Brasilia, Brasilia, Brazil.,Robinow Syndrome Foundation, Anoka, Minnesota, USA
| | - Jesper Eisfeldt
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | | | - Janson White
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Zeynep C Akdemir
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Shalini N Jhangiani
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Donna M Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Richard A Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Anna Lindstrand
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Hospital, Houston, Texas, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - V Reid Sutton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Hospital, Houston, Texas, USA
| | - Claudia M B Carvalho
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Pacific Northwest Research Institute (PNRI), Seattle, Washington, USA
| |
Collapse
|
16
|
Abstract
Significance: The selenium-containing Glutathione peroxidases (GPxs)1-4 protect against oxidative challenge, inhibit inflammation and oxidant-induced regulated cell death. Recent Advances: GPx1 and GPx4 dampen phosphorylation cascades predominantly via prevention of inactivation of phosphatases by H2O2 or lipid hydroperoxides. GPx2 regulates the balance between regeneration and apoptotic cell shedding in the intestine. It inhibits inflammation-induced carcinogenesis in the gut but promotes growth of established cancers. GPx3 deficiency facilitates platelet aggregation likely via disinhibition of thromboxane biosynthesis. It is also considered a tumor suppressor. GPx4 is expressed in three different forms. The cytosolic form proved to inhibit interleukin-1-driven nuclear factor κB activation and leukotriene biosynthesis. Moreover, it is a key regulator of ferroptosis, because it reduces hydroperoxy groups of complex lipids and silences lipoxygenases. By alternate substrate use, the nuclear form contributes to chromatin compaction. Mitochondrial GPx4 forms the mitochondrial sheath of spermatozoa and, thus, guarantees male fertility. Out of the less characterized GPxs, the cysteine-containing GPx7 and GPx8 are unique in contributing to oxidative protein folding in the endoplasmic reticulum by reacting with protein isomerase as an alternate substrate. A yeast 2-Cysteine glutathione peroxidase equipped with CP and CR was reported to sense H2O2 for inducing an adaptive response. Critical Issues: Most of the findings compiled are derived from tissue culture and/or animal studies only. Their impact on human physiology is sometimes questionable. Future Directions: The expression of individual GPxs and GPx-dependent regulatory phenomena are to be further investigated, in particular in respect to human health.
Collapse
Affiliation(s)
- Regina Brigelius-Flohé
- Department of Biochemistry of Micronutrients, German Institute of Human Nutrition-Potsdam-Rehbrücke (DIfE), Nuthetal, Germany
| | - Leopold Flohé
- Depatamento de Biochímica, Universidad de la República, Montevideo, Uruguay.,Dipartimento di Medicina Moleculare, Università degli Studi di Padova, Padova, Italy
| |
Collapse
|
17
|
Chen Z, Wasney GA, Picaud S, Filippakopoulos P, Vedadi M, D'Angiolella V, Bullock AN. Identification of a PGXPP degron motif in dishevelled and structural basis for its binding to the E3 ligase KLHL12. Open Biol 2020; 10:200041. [PMID: 32574548 PMCID: PMC7333892 DOI: 10.1098/rsob.200041] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Wnt signalling is dependent on dishevelled proteins (DVL1-3), which assemble an intracellular Wnt signalosome at the plasma membrane. The levels of DVL1-3 are regulated by multiple Cullin-RING E3 ligases that mediate their ubiquitination and degradation. The BTB-Kelch protein KLHL12 was the first E3 ubiquitin ligase to be identified for DVL1-3, but the molecular mechanisms determining its substrate interactions have remained unknown. Here, we mapped the interaction of DVL1-3 to a ‘PGXPP' motif that is conserved in other known partners and substrates of KLHL12, including PLEKHA4, PEF1, SEC31 and DRD4. To determine the binding mechanism, we solved a 2.4 Å crystal structure of the Kelch domain of KLHL12 in complex with a DVL1 peptide that bound with low micromolar affinity. The DVL1 substrate adopted a U-shaped turn conformation that enabled hydrophobic interactions with all six blades of the Kelch domain β-propeller. In cells, the mutation or deletion of this motif reduced the binding and ubiquitination of DVL1 and increased its stability confirming this sequence as a degron motif for KLHL12 recruitment. These results define the molecular mechanisms determining DVL regulation by KLHL12 and establish the KLHL12 Kelch domain as a new protein interaction module for a novel proline-rich motif.
Collapse
Affiliation(s)
- Zhuoyao Chen
- Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Gregory A Wasney
- Structural Genomics Consortium, University of Toronto, MaRS Centre, South Tower, 101 College Street, Toronto, M5G 1L7, Canada
| | - Sarah Picaud
- Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Panagis Filippakopoulos
- Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Masoud Vedadi
- Structural Genomics Consortium, University of Toronto, MaRS Centre, South Tower, 101 College Street, Toronto, M5G 1L7, Canada
| | - Vincenzo D'Angiolella
- Department of Oncology, Cancer Research UK and Medical Research Council Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Alex N Bullock
- Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| |
Collapse
|
18
|
Huybrechts Y, Mortier G, Boudin E, Van Hul W. WNT Signaling and Bone: Lessons From Skeletal Dysplasias and Disorders. Front Endocrinol (Lausanne) 2020; 11:165. [PMID: 32328030 PMCID: PMC7160326 DOI: 10.3389/fendo.2020.00165] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/09/2020] [Indexed: 12/15/2022] Open
Abstract
Skeletal dysplasias are a diverse group of heritable diseases affecting bone and cartilage growth. Throughout the years, the molecular defect underlying many of the diseases has been identified. These identifications led to novel insights in the mechanisms regulating bone and cartilage growth and homeostasis. One of the pathways that is clearly important during skeletal development and bone homeostasis is the Wingless and int-1 (WNT) signaling pathway. So far, three different WNT signaling pathways have been described, which are all activated by binding of the WNT ligands to the Frizzled (FZD) receptors. In this review, we discuss the skeletal disorders that are included in the latest nosology of skeletal disorders and that are caused by genetic defects involving the WNT signaling pathway. The number of skeletal disorders caused by defects in WNT signaling genes and the clinical phenotype associated with these disorders illustrate the importance of the WNT signaling pathway during skeletal development as well as later on in life to maintain bone mass. The knowledge gained through the identification of the genes underlying these monogenic conditions is used for the identification of novel therapeutic targets. For example, the genes underlying disorders with altered bone mass are all involved in the canonical WNT signaling pathway. Consequently, targeting this pathway is one of the major strategies to increase bone mass in patients with osteoporosis. In addition to increasing the insights in the pathways regulating skeletal development and bone homeostasis, knowledge of rare skeletal dysplasias can also be used to predict possible adverse effects of these novel drug targets. Therefore, this review gives an overview of the skeletal and extra-skeletal phenotype of the different skeletal disorders linked to the WNT signaling pathway.
Collapse
|
19
|
George S, Hamblin MR, Abrahamse H. Differentiation of Mesenchymal Stem Cells to Neuroglia: in the Context of Cell Signalling. Stem Cell Rev Rep 2019; 15:814-826. [PMID: 31515658 PMCID: PMC6925073 DOI: 10.1007/s12015-019-09917-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The promise of engineering specific cell types from stem cells and rebuilding damaged or diseased tissues has fascinated stem cell researchers and clinicians over last few decades. Mesenchymal Stem Cells (MSCs) have the potential to differentiate into non-mesodermal cells, particularly neural-lineage, consisting of neurons and glia. These multipotent adult stem cells can be used for implementing clinical trials in neural repair. Ongoing research identifies several molecular mechanisms involved in the speciation of neuroglia, which are tightly regulated and interconnected by various components of cell signalling machinery. Growing MSCs with multiple inducers in culture media will initiate changes on intricately interlinked cell signalling pathways and processes. Net result of these signal flow on cellular architecture is also dependent on the type of ligands and stem cells investigated in vitro. However, our understanding about this dynamic signalling machinery is limited and confounding, especially with spheroid structures, neurospheres and organoids. Therefore, the results for differentiating neurons and glia in vitro have been inconclusive, so far. Added to this complication, we have no convincing evidence about the electrical conductivity and functionality status generated in differentiating neurons and glia. This review has taken a step forward to tailor the information on differentiating neuroglia with the common methodologies, in practice.
Collapse
Affiliation(s)
- Sajan George
- Laser Research Centre, University of Johannesburg, P.O. Box 17011, Doornfontein, 2028, South Africa
| | - Michael R Hamblin
- Laser Research Centre, University of Johannesburg, P.O. Box 17011, Doornfontein, 2028, South Africa
- Wellman Centre for Photomedicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
| | - Heidi Abrahamse
- Laser Research Centre, University of Johannesburg, P.O. Box 17011, Doornfontein, 2028, South Africa.
| |
Collapse
|
20
|
Harnoš J, Cañizal MCA, Jurásek M, Kumar J, Holler C, Schambony A, Hanáková K, Bernatík O, Zdráhal Z, Gömöryová K, Gybeľ T, Radaszkiewicz TW, Kravec M, Trantírek L, Ryneš J, Dave Z, Fernández-Llamazares AI, Vácha R, Tripsianes K, Hoffmann C, Bryja V. Dishevelled-3 conformation dynamics analyzed by FRET-based biosensors reveals a key role of casein kinase 1. Nat Commun 2019; 10:1804. [PMID: 31000703 PMCID: PMC6472409 DOI: 10.1038/s41467-019-09651-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 03/20/2019] [Indexed: 01/17/2023] Open
Abstract
Dishevelled (DVL) is the key component of the Wnt signaling pathway. Currently, DVL conformational dynamics under native conditions is unknown. To overcome this limitation, we develop the Fluorescein Arsenical Hairpin Binder- (FlAsH-) based FRET in vivo approach to study DVL conformation in living cells. Using this single-cell FRET approach, we demonstrate that (i) Wnt ligands induce open DVL conformation, (ii) DVL variants that are predominantly open, show more even subcellular localization and more efficient membrane recruitment by Frizzled (FZD) and (iii) Casein kinase 1 ɛ (CK1ɛ) has a key regulatory function in DVL conformational dynamics. In silico modeling and in vitro biophysical methods explain how CK1ɛ-specific phosphorylation events control DVL conformations via modulation of the PDZ domain and its interaction with DVL C-terminus. In summary, our study describes an experimental tool for DVL conformational sampling in living cells and elucidates the essential regulatory role of CK1ɛ in DVL conformational dynamics.
Collapse
Affiliation(s)
- Jakub Harnoš
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic.,Department of Cell, Developmental & Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Maria Consuelo Alonso Cañizal
- Department of Pharmacology and Toxicology, University of Würzburg, Würzburg, 97078, Germany.,Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, 97078, Germany.,Institute for Molecular Cell Biology, CMB-Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, 07745, Germany
| | - Miroslav Jurásek
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, 62500, Czech Republic.,National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
| | - Jitender Kumar
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, 62500, Czech Republic
| | - Cornelia Holler
- Max Planck Institute for the Science of Light, Erlangen, 91058, Germany.,Biology Department, Developmental Biology, Friedrich-Alexander University Erlangen-Nüremberg, Erlangen, 91058, Germany
| | - Alexandra Schambony
- Max Planck Institute for the Science of Light, Erlangen, 91058, Germany.,Biology Department, Developmental Biology, Friedrich-Alexander University Erlangen-Nüremberg, Erlangen, 91058, Germany
| | - Kateřina Hanáková
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, 62500, Czech Republic.,National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
| | - Ondřej Bernatík
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
| | - Zbyněk Zdráhal
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, 62500, Czech Republic.,National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
| | - Kristína Gömöryová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
| | - Tomáš Gybeľ
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
| | | | - Marek Kravec
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
| | - Lukáš Trantírek
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, 62500, Czech Republic.,Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Brno, 612 65, Czech Republic
| | - Jan Ryneš
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, 62500, Czech Republic
| | - Zankruti Dave
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
| | | | - Robert Vácha
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, 62500, Czech Republic.,National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
| | - Konstantinos Tripsianes
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, 62500, Czech Republic
| | - Carsten Hoffmann
- Department of Pharmacology and Toxicology, University of Würzburg, Würzburg, 97078, Germany.,Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, 97078, Germany.,Institute for Molecular Cell Biology, CMB-Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, 07745, Germany
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic. .,Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Brno, 612 65, Czech Republic.
| |
Collapse
|
21
|
Nucleoredoxin-Dependent Targets and Processes in Neuronal Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4829872. [PMID: 30584462 PMCID: PMC6280245 DOI: 10.1155/2018/4829872] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/24/2018] [Accepted: 09/12/2018] [Indexed: 12/15/2022]
Abstract
Nucleoredoxin (Nrx) is an oxidoreductase of the thioredoxin family of proteins. It was shown to act as a signal transducer in some pathways; however, so far, no comprehensive analysis of its regulated substrates and functions was available. Here, we used a combination of two different strategies to fill this gap. First, we analyzed the thiol-redox state of the proteome of SH-SY5Y neuroblastoma cells depleted of Nrx compared to control cells using a differential thiol-labeling technique and quantitative mass spectrometry. 171 proteins were identified with an altered redox state; 161 of these were more reduced in the absence of Nrx. This suggests functions of Nrx in the oxidation of protein thiols. Second, we utilized the active site mutant Cys208Ser of Nrx, which stabilizes a mixed disulfide intermediate with its substrates and therefore trapped interacting proteins from the mouse brain (identifying 1710 proteins) and neuronal cell culture extracts (identifying 609 proteins). Profiling of the affected biological processes and molecular functions in cells of neuronal origin suggests numerous functions of Nrx in the redox regulation of metabolic pathways, cellular morphology, and signal transduction. These results characterize Nrx as a cellular oxidase that itself may be oxidized by the formation of disulfide relays with peroxiredoxins.
Collapse
|
22
|
Abstract
SIGNIFICANCE Numerous studies have demonstrated the actions of reactive oxygen species (ROS) as regulators of several physiological processes. In this study, we discuss how redox signaling mechanisms operate to control different processes such as neuronal differentiation, oligodendrocyte differentiation, dendritic growth, and axonal growth. Recent Advances: Redox homeostasis regulates the physiology of neural stem cells (NSCs). Notably, the neuronal differentiation process of NSCs is determined by a change toward oxidative metabolism, increased levels of mitochondrial ROS, increased activity of NADPH oxidase (NOX) enzymes, decreased levels of Nrf2, and differential regulation of different redoxins. Furthermore, during the neuronal maturation processes, NOX and MICAL produce ROS to regulate cytoskeletal dynamics, which control the dendritic and axonal growth, as well as the axonal guidance. CRITICAL ISSUES The redox homeostasis changes are, in part, attributed to cell metabolism and compartmentalized production of ROS, which is regulated, sensed, and transduced by different molecules such as thioredoxins, glutaredoxins, peroxiredoxins, and nucleoredoxin to control different signaling pathways in different subcellular regions. The study of how these elements cooperatively act is essential for the understanding of nervous system development, as well as the application of regenerative therapies that recapitulate these processes. FUTURE DIRECTIONS The information about these topics in the last two decades leads us to the conclusion that the role of ROS signaling in development of the nervous system is more important than it was previously believed and makes clear the importance of exploring in more detail the mechanisms of redox signaling. Antioxid. Redox Signal. 28, 1603-1625.
Collapse
Affiliation(s)
- Mauricio Olguín-Albuerne
- División de Neurociencias, Instituto de Fisiología Celular , Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Julio Morán
- División de Neurociencias, Instituto de Fisiología Celular , Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
23
|
Yang X, Chung JY, Rai U, Esumi N. Cadherins in the retinal pigment epithelium (RPE) revisited: P-cadherin is the highly dominant cadherin expressed in human and mouse RPE in vivo. PLoS One 2018; 13:e0191279. [PMID: 29338041 PMCID: PMC5770047 DOI: 10.1371/journal.pone.0191279] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 01/02/2018] [Indexed: 01/26/2023] Open
Abstract
The retinal pigment epithelium (RPE) supports the health and function of retinal photoreceptors and is essential for normal vision. RPE cells are post-mitotic, terminally differentiated, and polarized epithelial cells. In pathological conditions, however, they lose their epithelial integrity, become dysfunctional, even dedifferentiate, and ultimately die. The integrity of epithelial cells is maintained, in part, by adherens junctions, which are composed of cadherin homodimers and p120-, β-, and α-catenins linking to actin filaments. While E-cadherin is the major cadherin for forming the epithelial phenotype in most epithelial cell types, it has been reported that cadherin expression in RPE cells is different from other epithelial cells based on results with cultured RPE cells. In this study, we revisited the expression of cadherins in the RPE to clarify their relative contribution by measuring the absolute quantity of cDNAs produced from mRNAs of three classical cadherins (E-, N-, and P-cadherins) in the RPE in vivo. We found that P-cadherin (CDH3) is highly dominant in both mouse and human RPE in situ. The degree of dominance of P-cadherin is surprisingly large, with mouse Cdh3 and human CDH3 accounting for 82-85% and 92-93% of the total of the three cadherin mRNAs, respectively. We confirmed the expression of P-cadherin protein at the cell-cell border of mouse RPE in situ by immunofluorescence. Furthermore, we found that oxidative stress induces dissociation of P-cadherin and β-catenin from the cell membrane and subsequent translocation of β-catenin into the nucleus, resulting in activation of the canonical Wnt/β-catenin pathway. This is the first report of absolute comparison of the expression of three cadherins in the RPE, and the results suggest that the physiological role of P-cadherin in the RPE needs to be reevaluated.
Collapse
Affiliation(s)
- Xue Yang
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jin-Yong Chung
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Usha Rai
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Noriko Esumi
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
24
|
White JJ, Mazzeu JF, Coban-Akdemir Z, Bayram Y, Bahrambeigi V, Hoischen A, van Bon BWM, Gezdirici A, Gulec EY, Ramond F, Touraine R, Thevenon J, Shinawi M, Beaver E, Heeley J, Hoover-Fong J, Durmaz CD, Karabulut HG, Marzioglu-Ozdemir E, Cayir A, Duz MB, Seven M, Price S, Ferreira BM, Vianna-Morgante AM, Ellard S, Parrish A, Stals K, Flores-Daboub J, Jhangiani SN, Gibbs RA, Brunner HG, Sutton VR, Lupski JR, Carvalho CMB. WNT Signaling Perturbations Underlie the Genetic Heterogeneity of Robinow Syndrome. Am J Hum Genet 2018; 102:27-43. [PMID: 29276006 DOI: 10.1016/j.ajhg.2017.10.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 10/06/2017] [Indexed: 12/12/2022] Open
Abstract
Locus heterogeneity characterizes a variety of skeletal dysplasias often due to interacting or overlapping signaling pathways. Robinow syndrome is a skeletal disorder historically refractory to molecular diagnosis, potentially stemming from substantial genetic heterogeneity. All current known pathogenic variants reside in genes within the noncanonical Wnt signaling pathway including ROR2, WNT5A, and more recently, DVL1 and DVL3. However, ∼70% of autosomal-dominant Robinow syndrome cases remain molecularly unsolved. To investigate this missing heritability, we recruited 21 families with at least one family member clinically diagnosed with Robinow or Robinow-like phenotypes and performed genetic and genomic studies. In total, four families with variants in FZD2 were identified as well as three individuals from two families with biallelic variants in NXN that co-segregate with the phenotype. Importantly, both FZD2 and NXN are relevant protein partners in the WNT5A interactome, supporting their role in skeletal development. In addition to confirming that clustered -1 frameshifting variants in DVL1 and DVL3 are the main contributors to dominant Robinow syndrome, we also found likely pathogenic variants in candidate genes GPC4 and RAC3, both linked to the Wnt signaling pathway. These data support an initial hypothesis that Robinow syndrome results from perturbation of the Wnt/PCP pathway, suggest specific relevant domains of the proteins involved, and reveal key contributors in this signaling cascade during human embryonic development. Contrary to the view that non-allelic genetic heterogeneity hampers gene discovery, this study demonstrates the utility of rare disease genomic studies to parse gene function in human developmental pathways.
Collapse
Affiliation(s)
- Janson J White
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX 77030, USA
| | - Juliana F Mazzeu
- University of Brasilia, Brasilia 70910, Brazil; Robinow Syndrome Foundation, Anoka, MN 55303, USA
| | - Zeynep Coban-Akdemir
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX 77030, USA
| | - Yavuz Bayram
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX 77030, USA
| | - Vahid Bahrambeigi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX 77030, USA; Graduate Program in Diagnostic Genetics, School of Health Professions, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alexander Hoischen
- Department of Human Genetics, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands; Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Bregje W M van Bon
- Department of Human Genetics, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Alper Gezdirici
- Department of Medical Genetics, Kanuni Sultan Suleyman Training and Research Hospital, Istanbul 34303, Turkey
| | - Elif Yilmaz Gulec
- Department of Medical Genetics, Kanuni Sultan Suleyman Training and Research Hospital, Istanbul 34303, Turkey
| | - Francis Ramond
- Service de Génétique, CHU-Hôpital Nord, 42000 Saint-Etienne, France
| | - Renaud Touraine
- Service de Génétique, CHU-Hôpital Nord, 42000 Saint-Etienne, France
| | - Julien Thevenon
- Inserm UMR 1231 GAD team, Genetics of Developmental Anomalies, Université de Bourgogne-Franche Comté, 21000 Dijon, France; FHU-TRANSLAD, Université de Bourgogne, 21000 CHU Dijon, France; Centre de génétique, Hôpital Couple-Enfant, CHU de Grenoble-Alpes, 38700 La Tronche, France
| | - Marwan Shinawi
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Erin Beaver
- Mercy Clinic-Kids Genetics, Mercy Children's Hospital St. Louis, St. Louis, MO 63141, USA
| | - Jennifer Heeley
- Mercy Clinic-Kids Genetics, Mercy Children's Hospital St. Louis, St. Louis, MO 63141, USA
| | - Julie Hoover-Fong
- Greenberg Center for Skeletal Dysplasias, McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Ceren D Durmaz
- Department of Medical Genetics, Ankara University School of Medicine, 06100 Ankara, Turkey
| | - Halil Gurhan Karabulut
- Department of Medical Genetics, Ankara University School of Medicine, 06100 Ankara, Turkey
| | - Ebru Marzioglu-Ozdemir
- Department of Medical Genetics, Erzurum Regional and Training Hospital, 25070 Erzurum, Turkey
| | - Atilla Cayir
- Erzurum Training and Research Hospital, Department of Pediatric Endocrinology, 25070 Erzurum, Turkey
| | - Mehmet B Duz
- Department of Medical Genetics, Cerrahpasa Medical School, Istanbul University, 34452 Istanbul, Turkey
| | - Mehmet Seven
- Department of Medical Genetics, Cerrahpasa Medical School, Istanbul University, 34452 Istanbul, Turkey
| | - Susan Price
- Oxford Centre for Genomic Medicine, Nuffield Orthopaedic Centre, Oxford OX3 7LD, UK
| | | | - Angela M Vianna-Morgante
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, Sao Paulo - SP 05508-090, Brazil
| | - Sian Ellard
- Department of Molecular Genetics, Royal Devon and Exeter NHS Foundation Trust, Exeter EX2 5DW, UK; Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter EX1 2LU, UK
| | - Andrew Parrish
- Department of Molecular Genetics, Royal Devon and Exeter NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Karen Stals
- Department of Molecular Genetics, Royal Devon and Exeter NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Josue Flores-Daboub
- Department of Pediatric Genetics, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Shalini N Jhangiani
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Richard A Gibbs
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX 77030, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Han G Brunner
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands; Department of Clinical Genetics, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, 6202 AZ Maastricht, the Netherlands
| | - V Reid Sutton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX 77030, USA; Texas Children's Hospital, Houston, TX 77030, USA
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX 77030, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Hospital, Houston, TX 77030, USA
| | - Claudia M B Carvalho
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX 77030, USA.
| |
Collapse
|
25
|
Wald JH, Hatakeyama J, Printsev I, Cuevas A, Fry WH, Saldana MJ, Vorst KV, Rowson-Hodel A, Angelastro JM, Sweeney C, Carraway KL. Suppression of planar cell polarity signaling and migration in glioblastoma by Nrdp1-mediated Dvl polyubiquitination. Oncogene 2017; 36:5158-5167. [PMID: 28481871 PMCID: PMC5589482 DOI: 10.1038/onc.2017.126] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 02/15/2017] [Accepted: 03/23/2017] [Indexed: 12/16/2022]
Abstract
The lethality of the aggressive brain tumor glioblastoma multiforme (GBM) results in part from its strong propensity to invade surrounding normal brain tissue. Although oncogenic drivers such as epidermal growth factor receptor activation and Phosphatase and Tensin homolog inactivation are thought to promote the motility and invasiveness of GBM cells via phosphatidylinostitol 3-kinase activation, other unexplored mechanisms may also contribute to malignancy. Here we demonstrate that several components of the planar cell polarity (PCP) arm of non-canonical Wnt signaling including VANGL1, VANGL2 and FZD7 are transcriptionally upregulated in glioma and correlate with poorer patient outcome. Knockdown of the core PCP pathway component VANGL1 suppresses the motility of GBM cell lines, pointing to an important mechanistic role for this pathway in glioblastoma malignancy. We further observe that restoration of Nrdp1, a RING finger type E3 ubiquitin ligase whose suppression in GBM also correlates with poor prognosis, reduces GBM cell migration and invasiveness by suppressing PCP signaling. Our observations indicate that Nrdp1 physically interacts with the Vangl1 and Vangl2 proteins to mediate the K63-linked polyubiquitination of the Dishevelled, Egl-10 and Pleckstrin (DEP) domain of the Wnt pathway protein Dishevelled (Dvl). Ubiquitination hinders Dvl binding to phosphatidic acid, an interaction necessary for efficient Dvl recruitment to the plasma membrane upon Wnt stimulation of Fzd receptor and for the propagation of downstream signals. We conclude that the PCP pathway contributes significantly to the motility and hence the invasiveness of GBM cells, and that Nrdp1 acts as a negative regulator of PCP signaling by inhibiting Dvl through a novel polyubiquitination mechanism. We propose that the upregulation of core PCP components, together with the loss of the key negative regulator Nrdp1, act coordinately to promote GBM invasiveness and malignancy.
Collapse
Affiliation(s)
- Jessica H. Wald
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Jason Hatakeyama
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Ignat Printsev
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Antonio Cuevas
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - William H.D. Fry
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Matthew J. Saldana
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Kacey Vander Vorst
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Ashley Rowson-Hodel
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - James M. Angelastro
- Department of Molecular Biosciences, University of California Davis School of Veterinary Medicine, Davis, CA, USA
| | - Colleen Sweeney
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Kermit L. Carraway
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| |
Collapse
|
26
|
Kan F, Ye L, Yan T, Cao J, Zheng J, Li W. Proteomic and transcriptomic studies of HBV-associated liver fibrosis of an AAV-HBV-infected mouse model. BMC Genomics 2017; 18:641. [PMID: 28830339 PMCID: PMC5568174 DOI: 10.1186/s12864-017-3984-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 08/01/2017] [Indexed: 01/05/2023] Open
Abstract
Background Human hepatitis B virus (HBV) infection is an important public health issue in the Asia-Pacific region and is associated with chronic hepatitis, liver fibrosis, cirrhosis and even liver cancer. However, the underlying mechanisms of HBV-associated liver fibrosis remain incompletely understood. Results In the present study, proteomic and transcriptomic approaches as well as biological network analyses were performed to investigate the differentially expressed molecular signature and key regulatory networks that were associated with HBV-mediated liver fibrosis. RNA sequencing and 2DE-MALDI-TOF/TOF were performed on liver tissue samples obtained from HBV-infected C57BL/6 mouse generated via AAV8-HBV virus. The results showed that 322 genes and 173 proteins were differentially expressed, and 28 HBV-specific proteins were identified by comprehensive proteomic and transcriptomic analysis. GO analysis indicated that the differentially expressed proteins were predominantly involved in oxidative stress, which plays a key role in HBV-related liver fibrosis. Importantly, CAT, PRDX1, GSTP1, NXN and BLVRB were shown to be associated with oxidative stress among the differentially expressed proteins. The most striking results were validated by Western blot and RT-qPCR. The RIG-I like receptor signaling pathway was found to be the major signal pathway that changed during HBV-related fibrosis. Conclusions This study provides novel insights into HBV-associated liver fibrosis and reveals the significant role of oxidative stress in liver fibrosis. Furthermore, CAT, BLVRB, NXN, PRDX1, and IDH1 may be candidates for detection of liver fibrosis or therapeutic targets for the treatment of liver fibrosis. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3984-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fangming Kan
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lei Ye
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tao Yan
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiaqi Cao
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianhua Zheng
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Wuping Li
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
27
|
Wills ES, te Morsche RHM, van Reeuwijk J, Horn N, Geomini I, van de Laarschot LFM, Mans DA, Ueffing M, Boldt K, Drenth JPH, Roepman R. Liver cyst gene knockout in cholangiocytes inhibits cilium formation and Wnt signaling. Hum Mol Genet 2017; 26:4190-4202. [DOI: 10.1093/hmg/ddx308] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/26/2017] [Indexed: 01/07/2023] Open
|
28
|
Rong X, Zhou Y, Liu Y, Zhao B, Wang B, Wang C, Gong X, Tang P, Lu L, Li Y, Zhao C, Zhou J. Glutathione peroxidase 4 inhibits Wnt/β-catenin signaling and regulates dorsal organizer formation in zebrafish embryos. Development 2017; 144:1687-1697. [PMID: 28302747 DOI: 10.1242/dev.144261] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 03/07/2017] [Indexed: 12/20/2022]
Abstract
The Wnt/β-catenin signaling pathway plays pivotal roles in axis formation during embryogenesis and in adult tissue homeostasis. Glutathione peroxidase 4 (GPX4) is a selenoenzyme and participates in the reduction of peroxides. Its synthesis depends on the availability of the element selenium. However, the roles of GPX4 in vertebrate embryonic development and underlying mechanisms are largely unknown. Here, we show that maternal loss of zebrafish gpx4b promotes embryonic dorsal organizer formation, whereas overexpression of gpx4b inhibits the development of the dorsal organizer. Depletion of human GPX4 and zebrafish gpx4b (GPX4/gpx4b) increases, while GPX4/gpx4b overexpression decreases, Wnt/β-catenin signaling in vivo and in vitro Functional and epistatic studies showed that GPX4 functions at the Tcf/Lef level, independently of selenocysteine activation. Mechanistically, GPX4 interacts with Tcf/Lefs and inhibits Wnt activity by preventing the binding of Tcf/Lefs to the promoters of Wnt target genes, resulting in inhibitory action in the presence of Wnt/β-catenin signaling. Our findings unravel GPX4 as a suppressor of Wnt/β-catenin signals, suggesting a possible relationship between the Wnt/β-catenin pathway and selenium via the association of Tcf/Lef family proteins with GPX4.
Collapse
Affiliation(s)
- Xiaozhi Rong
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.,Institute of Evolution and Marine Biodiversity and College of Marine Biology, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China.,Laboratory for Marine Drugs and Biological Products, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Yumei Zhou
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Yunzhang Liu
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Beibei Zhao
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Bo Wang
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Caixia Wang
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Xiaoxia Gong
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Peipei Tang
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Ling Lu
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Yun Li
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Chengtian Zhao
- Institute of Evolution and Marine Biodiversity and College of Marine Biology, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Jianfeng Zhou
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China .,Laboratory for Marine Drugs and Biological Products, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
| |
Collapse
|
29
|
Dickson BJ, Gatie MI, Spice DM, Kelly GM. NOX1 and NOX4 are required for the differentiation of mouse F9 cells into extraembryonic endoderm. PLoS One 2017; 12:e0170812. [PMID: 28152080 PMCID: PMC5289483 DOI: 10.1371/journal.pone.0170812] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 01/11/2017] [Indexed: 01/23/2023] Open
Abstract
Mouse F9 cells differentiate to primitive endoderm (PrE) when treated with retinoic acid (RA). Differentiation is accompanied by increased reactive oxygen species (ROS) levels, and while treating F9 cells with antioxidants attenuates differentiation, H2O2 treatment alone is sufficient to induce PrE. We identified the NADPH oxidase (NOX) complexes as candidates for the source of this endogenous ROS, and within this gene family, and over the course of differentiation, Nox1 and Nox 4 show the greatest upregulation induced by RA. Gata6, encoding a master regulator of extraembryonic endoderm is also up-regulated by RA and we provide evidence that NOX1 and NOX4 protein levels increase in F9 cells overexpressing Gata6. Pan-NOX and NOX1-specific inhibitors significantly reduced the ability of RA to induce PrE, and this was recapitulated using a genetic approach to knockdown Nox1 and/or Nox4 transcripts. Interestingly, overexpressing either gene in untreated F9 cells did not induce differentiation, even though each elevated ROS levels. Thus, the data suggests that ROS produced during PrE differentiation is dependent in part on increased NOX1 and NOX4 levels, which is under the control of GATA6. Furthermore, these results suggest that the combined activity of multiple NOX proteins is necessary for the differentiation of F9 cells to primitive endoderm.
Collapse
Affiliation(s)
- Benjamin J. Dickson
- Department of Biology, Molecular Genetics Unit, Western University, London, Ontario, Canada
| | - Mohamed I. Gatie
- Department of Biology, Molecular Genetics Unit, Western University, London, Ontario, Canada
| | - Danielle M. Spice
- Department of Biology, Molecular Genetics Unit, Western University, London, Ontario, Canada
| | - Gregory M. Kelly
- Department of Biology, Molecular Genetics Unit, Western University, London, Ontario, Canada
- Child Health Research Institute, London, Ontario, Canada
- Ontario Institute for Regenerative Medicine, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
30
|
Mak CCY, Chow PC, Liu APY, Chan KYK, Chu YWY, Mok GTK, Leung GKC, Yeung KS, Chau AKT, Lowther C, Scherer SW, Marshall CR, Bassett AS, Chung BHY. De novo large rare copy-number variations contribute to conotruncal heart disease in Chinese patients. NPJ Genom Med 2016; 1:16033. [PMID: 29263819 PMCID: PMC5685312 DOI: 10.1038/npjgenmed.2016.33] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/02/2016] [Accepted: 08/05/2016] [Indexed: 01/05/2023] Open
Abstract
Conotruncal heart anomalies (CTDs) are particularly prevalent congenital heart diseases (CHD) in Hong Kong. We surveyed large (>500 kb), rare (<1% frequency in controls) copy-number variations (CNVs) in Chinese patients with CTDs to identify potentially disease-causing variations. Adults who tested negative for 22q11.2 deletions were recruited from the adult CHD clinic in Hong Kong. Using a stringent calling criteria, high-confidence CNV calls were obtained, and a large control set comprising 3,987 Caucasian and 1,945 Singapore Chinese subjects was used to identify rare CNVs. Ten large rare CNVs were identified, and 3 in 108 individuals were confirmed to harbour de novo CNVs. All three patients were syndromic with a more complex phenotype, and each of these CNVs overlapped regions likely to be important in CHD. One was a 611 kb deletion at 17p13.3, telomeric to the Miller-Dieker syndrome (MDS) critical region, overlapping the NXN gene. Another was a 5 Mb deletion at 13q33.3, within a previously described critical region for CHD. A third CNV, previously unreported, was a large duplication at 2q22.3 overlapping the ZEB2 gene. The commonly reported 1q21.1 recurrent duplication was not observed in this Chinese cohort. We provide detailed phenotypic and genotypic descriptions of large rare genic CNVs that may represent CHD loci in the East Asian population. Larger samples of Chinese origin will be required to determine whether the genome-wide distribution differs from that found in predominantly European CHD cohorts.
Collapse
Affiliation(s)
- Christopher C Y Mak
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Pak Cheong Chow
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Anthony P Y Liu
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kelvin Y K Chan
- Department of Obstetrics and Gynecology, Queen Mary Hospital, Hong Kong, China
| | - Yoyo W Y Chu
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Gary T K Mok
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Gordon K C Leung
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kit San Yeung
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Adolphus K T Chau
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chelsea Lowther
- The Clinical Genetics Research Program at The Centre for Addiction and Mental Health, The Dalglish Family 22q Clinic at The University Health Network, and The Department of Psychiatry at The University of Toronto, Toronto, ON, Canada
| | - Stephen W Scherer
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Christian R Marshall
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Anne S Bassett
- The Clinical Genetics Research Program at The Centre for Addiction and Mental Health, The Dalglish Family 22q Clinic at The University Health Network, and The Department of Psychiatry at The University of Toronto, Toronto, ON, Canada
| | - Brian H Y Chung
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
31
|
Torigata K, Daisuke O, Mukai S, Hatanaka A, Ohka F, Motooka D, Nakamura S, Ohkawa Y, Yabuta N, Kondo Y, Nojima H. LATS2 Positively Regulates Polycomb Repressive Complex 2. PLoS One 2016; 11:e0158562. [PMID: 27434182 PMCID: PMC4951031 DOI: 10.1371/journal.pone.0158562] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/19/2016] [Indexed: 11/19/2022] Open
Abstract
LATS2, a pivotal Ser/Thr kinase of the Hippo pathway, plays important roles in many biological processes. LATS2 also function in Hippo-independent pathway, including mitosis, DNA damage response and epithelial to mesenchymal transition. However, the physiological relevance and molecular basis of these LATS2 functions remain obscure. To understand novel functions of LATS2, we constructed a LATS2 knockout HeLa-S3 cell line using TAL-effector nuclease (TALEN). Integrated omics profiling of this cell line revealed that LATS2 knockout caused genome-wide downregulation of Polycomb repressive complex 2 (PRC2) and H3K27me3. Cell-cycle analysis revealed that downregulation of PRC2 was not due to cell cycle aberrations caused by LATS2 knockout. Not LATS1, a homolog of LATS2, but LATS2 bound PRC2 on chromatin and phosphorylated it. LATS2 positively regulates histone methyltransferase activity of PRC2 and their expression at both the mRNA and protein levels. Our findings reveal a novel signal upstream of PRC2, and provide insight into the crucial role of LATS2 in coordinating the epigenome through regulation of PRC2.
Collapse
Affiliation(s)
- Kosuke Torigata
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Suita City, Osaka, Japan
| | - Okuzaki Daisuke
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Suita City, Osaka, Japan
- DNA-chip Development Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita City, Osaka, Japan
| | - Satomi Mukai
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Suita City, Osaka, Japan
| | - Akira Hatanaka
- Department of Epigenomics, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, Japan
| | - Fumiharu Ohka
- Department of Epigenomics, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, Japan
| | - Daisuke Motooka
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita City, Osaka, Japan
| | - Shota Nakamura
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita City, Osaka, Japan
| | - Yasuyuki Ohkawa
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan
| | - Norikazu Yabuta
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Suita City, Osaka, Japan
| | - Yutaka Kondo
- Department of Epigenomics, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, Japan
| | - Hiroshi Nojima
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Suita City, Osaka, Japan
- DNA-chip Development Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita City, Osaka, Japan
- * E-mail:
| |
Collapse
|
32
|
Requirement of CXCL12-CXCR7 signaling for CD20(-) CD138(-) double-negative population in lymphoplasmacytic lymphoma. J Transl Med 2016; 96:517-25. [PMID: 26878134 DOI: 10.1038/labinvest.2016.28] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/13/2015] [Accepted: 12/25/2015] [Indexed: 12/12/2022] Open
Abstract
Cancer cells with tumorigenic potential are limited to a small subpopulation known as cancer-initiating cells (CICs). Recently we investigated a candidate of CICs of lymphoplasmacytic lymphoma (LPL), which is positive for both B-cell marker CD20 and plasma-cell marker CD138. We reported that the subpopulation of CD20(-) CD138(-) phenotype, in which both markers were negative was a candidate of CICs in LPL using LPL cell line, MWCL-1. CICs are known to be plastic under stressed condition, in which non-CICs are changed to CICs. In the present study, we investigated the plasticity of CICs of LPL, and found that hypoxia induced the conversion of CD20(+) CD138(-) to CD20(-) CD138(-) phenotype. We then searched for markers preferentially expressed in CD20(-) CD138(-) subpopulation, and the chemokine receptor CXCR7 was isolated. When cultured with CXCL12, a ligand of CXCR7, the number of CD20(-) CD138(-) cells increased in a time- and dose-dependent manner. In addition, hypoxia enhanced the expression level of CXCL12 in MWCL-1. In clinical samples of LPL, a few tumor cells expressed CXCR7, in which CD20 expression was not detected. These results indicated that hypoxia and CXCL12-CXCR7 axis appeared to be advantageous microenvironments to CD20(-) CD138(-) cells.
Collapse
|
33
|
Visweswaran M, Pohl S, Arfuso F, Newsholme P, Dilley R, Pervaiz S, Dharmarajan A. Multi-lineage differentiation of mesenchymal stem cells - To Wnt, or not Wnt. Int J Biochem Cell Biol 2015; 68:139-47. [PMID: 26410622 DOI: 10.1016/j.biocel.2015.09.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/21/2015] [Accepted: 09/22/2015] [Indexed: 01/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotent precursor cells originating from several adult connective tissues. MSCs possess the ability to self-renew and differentiate into several lineages, and are recognized by the expression of unique cell surface markers. Several lines of evidence suggest that various signal transduction pathways and their interplay regulate MSC differentiation. To that end, a critical player in regulating MSC differentiation is a group of proteins encoded by the Wnt gene family, which was previously known for influencing various stages of embryonic development and cell fate determination. As MSCs have gained significant clinical attention for their potential applications in regenerative medicine, it is imperative to unravel the mechanisms by which molecular regulators control differentiation of MSCs for designing cell-based therapeutics. It is rather coincidental that the functional outcome(s) of Wnt-induced signals share similarities with cellular redox-mediated networks from the standpoint of MSC biology. Furthermore, there is evidence for a crosstalk between Wnt and redox signalling, which begs the question whether Wnt-mediated differentiation signals involve the intermediary role of reactive oxygen species. In this review, we summarize the impact of Wnt signalling on multi-lineage differentiation of MSCs, and attempt to unravel the intricate interplay between Wnt and redox signals.
Collapse
Affiliation(s)
- Malini Visweswaran
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Sebastian Pohl
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Philip Newsholme
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Rodney Dilley
- Ear Sciences Centre, University of Western Australia and Ear Science Institute Australia, Perth, Western Australia 6008, Australia
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; National University Cancer Institute, National University Health System, Singapore; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Arun Dharmarajan
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia.
| |
Collapse
|
34
|
|
35
|
Naito A, Yamamoto H, Kagawa Y, Naito Y, Okuzaki D, Otani K, Iwamoto Y, Maeda S, Kikuta J, Nishikawa K, Uemura M, Nishimura J, Hata T, Takemasa I, Mizushima T, Ishii H, Doki Y, Mori M, Ishii M. RFPL4A increases the G1 population and decreases sensitivity to chemotherapy in human colorectal cancer cells. J Biol Chem 2015; 290:6326-6337. [PMID: 25605732 PMCID: PMC4358269 DOI: 10.1074/jbc.m114.614859] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/09/2015] [Indexed: 01/06/2023] Open
Abstract
Cell cycle-arrested cancer cells are resistant to conventional chemotherapy that acts on the mitotic phases of the cell cycle, although the molecular mechanisms involved in halting cell cycle progression remain unclear. Here, we demonstrated that RFPL4A, an uncharacterized ubiquitin ligase, induced G1 retention and thus conferred decreased sensitivity to chemotherapy in the human colorectal cancer cell line, HCT116. Long term time lapse observations in HCT116 cells bearing a "fluorescence ubiquitin-based cell cycle indicator" identified a characteristic population that is viable but remains in the G1 phase for an extended period of time (up to 56 h). Microarray analyses showed that expression of RFPL4A was significantly up-regulated in these G1-arrested cells, not only in HCT116 cells but also in other cancer cell lines, and overexpression of RFPL4A increased the G1 population and decreased sensitivity to chemotherapy. However, knockdown of RFPL4A expression caused the cells to resume mitosis and induced their susceptibility to anti-cancer drugs in vitro and in vivo. These results indicate that RFPL4A is a novel factor that increases the G1 population and decreases sensitivity to chemotherapy and thus may be a promising therapeutic target for refractory tumor conditions.
Collapse
Affiliation(s)
- Atsushi Naito
- From the Departments of Immunology and Cell Biology, Gastroenterological Surgery, and
| | | | - Yoshinori Kagawa
- From the Departments of Immunology and Cell Biology, Gastroenterological Surgery, and the WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Yoko Naito
- From the Departments of Immunology and Cell Biology, the WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- the DNA-chip Developmental Center for Infectious Diseases, Research Institute for Microbial Diseases, 3-1 Yamada-oka, Osaka University, Suita, Osaka 565-0871, Japan, and
| | | | | | - Sakae Maeda
- From the Departments of Immunology and Cell Biology, Gastroenterological Surgery, and the WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Junichi Kikuta
- From the Departments of Immunology and Cell Biology, the WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan, the Japan Science and Technology Agency (JST), CREST, 5 Sanban-cho, Chiyoda-ku, Tokyo 102-0075, Japan
| | - Keizo Nishikawa
- From the Departments of Immunology and Cell Biology, the WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan, the Japan Science and Technology Agency (JST), CREST, 5 Sanban-cho, Chiyoda-ku, Tokyo 102-0075, Japan
| | | | | | | | | | | | - Hideshi Ishii
- Cancer Profiling Discovery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | | - Masaru Ishii
- From the Departments of Immunology and Cell Biology, the WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan, the Japan Science and Technology Agency (JST), CREST, 5 Sanban-cho, Chiyoda-ku, Tokyo 102-0075, Japan
| |
Collapse
|
36
|
Prozorovski T, Schneider R, Berndt C, Hartung HP, Aktas O. Redox-regulated fate of neural stem progenitor cells. Biochim Biophys Acta Gen Subj 2015; 1850:1543-54. [PMID: 25662818 DOI: 10.1016/j.bbagen.2015.01.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 01/29/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND Accumulated data indicate that self-renewal, multipotency, and differentiation of neural stem cells are under an intrinsic control mediated by alterations in the redox homeostasis. These dynamic redox changes not only reflect and support the ongoing metabolic and energetic processes, but also serve to coordinate redox-signaling cascades. Controlling particular redox couples seems to have a relevant impact on cell fate decision during development, adult neurogenesis and regeneration. SCOPE OF REVIEW Our own research provided initial evidence for the importance of NAD+-dependent enzymes in neural stem cell fate decision. In this review, we summarize recent knowledge on the active role of reactive oxygen species, redox couples and redox-signaling mechanisms on plasticity and function of neural stem and progenitor cells focusing on NAD(P)+/NAD(P)H-mediated processes. MAJOR CONCLUSIONS The compartmentalized subcellular sources and availability of oxidizing/reducing molecules in particular microenvironment define the specificity of redox regulation in modulating the delicate balance between stemness and differentiation of neural progenitors. The generalization of "reactive oxygen species" as well as the ambiguity of their origin might explain the diametrically-opposed findings in the field of redox-dependent cell fate reflected by the literature. GENERAL SIGNIFICANCE Increasing knowledge of temporary and spatially defined redox regulation is of high relevance for the development of novel approaches in the field of cell-based regeneration of nervous tissue in various pathological states. This article is part of a special issue entitled Redox regulation of differentiation and de-differentiation.
Collapse
Affiliation(s)
- Tim Prozorovski
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
| | - Reiner Schneider
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
37
|
Norman GL, Yang CY, Ostendorff HP, Shums Z, Lim MJ, Wang J, Awad A, Hirschfield GM, Milkiewicz P, Bloch DB, Rothschild KJ, Bowlus CL, Adamopoulos IE, Leung PS, Janssen HJ, Cheung AC, Coltescu C, Gershwin ME. Anti-kelch-like 12 and anti-hexokinase 1: novel autoantibodies in primary biliary cirrhosis. Liver Int 2015; 35:642-51. [PMID: 25243383 PMCID: PMC4305042 DOI: 10.1111/liv.12690] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 09/15/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Using high-density human recombinant protein microarrays, we identified two potential biomarkers, kelch-like 12 (KLHL12) and hexokinase-1 (HK1), in primary biliary cirrhosis (PBC). The objective of this study was to determine the diagnostic value of anti-KLHL12/HK1 autoantibodies in PBC. Initial discovery used sera from 22 patients with PBC and 62 non-PBC controls. KLHL12 and HK1 proteins were then analysed for immunoglobulin reactivity by immunoblot and enzyme-linked immunosorbent assay (ELISA) in two independent cohorts of PBC and disease/healthy control patients. METHODS Serum samples from 100 patients with PBC and 165 non-PBC disease controls were analysed by immunoblot and samples from 366 patients with PBC, 174 disease controls, and 80 healthy donors were tested by ELISA. RESULTS Anti-KLHL12 and anti-HK1 antibodies were each detected more frequently in PBC compared with non-PBC disease controls (P < 0.001). Not only are both markers highly specific for PBC (≥95%) but they also yielded higher sensitivity than anti-gp210 and anti-sp100 antibodies. Combining anti-HK1 and anti-KLHL12 with available markers (MIT3, gp210 and sp100), increased the diagnostic sensitivity for PBC. Most importantly, anti-KLHL12 and anti-HK1 antibodies were present in 10-35% of anti-mitochondrial antibody (AMA)-negative PBC patients and adding these two biomarkers to conventional PBC assays dramatically improved the serological sensitivity in AMA-negative PBC from 55% to 75% in immunoblot and 48.3% to 68.5% in ELISA. CONCLUSIONS The addition of tests for highly specific anti-KLHL12 and anti-HK1 antibodies to AMA and ANA serological assays significantly improves efficacy in the clinical detection and diagnosis of PBC, especially for AMA-negative subjects.
Collapse
Affiliation(s)
| | - Chen-Yen Yang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, USA
| | | | | | | | - Jinjun Wang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, USA
| | | | - Gideon M. Hirschfield
- Centre for Liver Research, Institute of Biomedical Research, University of Birmingham, UK
| | - Piotr Milkiewicz
- Department of General, Transplant and Liver Surgery, Warsaw Medical University, Poland
| | - Donald B. Bloch
- The Center for Immunology and Inflammatory Diseases and the Division of Rheumatology, Allergy and Immunology of the General Medical Services and the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Harvard, MA, USA
| | | | - Christopher L. Bowlus
- Division of Gastroenterology and Hepatology, University of California, Davis, CA, USA
| | - Iannis E. Adamopoulos
- Institute of Pediatric and Regenerative Medicine, Shriners Hospital for Northern California, Sacramento, CA, USA
| | - Patrick S.C. Leung
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, USA
| | - Harry J. Janssen
- Division of Gastroenterology, University of Toronto, Ontario, Canada,Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center Rotterdam, Netherlands
| | - Angela C. Cheung
- Division of Gastroenterology, University of Toronto, Ontario, Canada
| | - Catalina Coltescu
- Toronto Center for Liver Diseases, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - M. Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, USA
| |
Collapse
|
38
|
Bahn YJ, Lee KP, Lee SM, Choi JY, Seo YS, Kwon KS. Nucleoredoxin promotes adipogenic differentiation through regulation of Wnt/β-catenin signaling. J Lipid Res 2014; 56:294-303. [PMID: 25548260 DOI: 10.1194/jlr.m054056] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Nucleoredoxin (NRX) is a member of the thioredoxin family of proteins that controls redox homeostasis in cell. Redox homeostasis is a well-known regulator of cell differentiation into various tissue types. We found that NRX expression levels were higher in white adipose tissue of obese ob/ob mice and increased in the early adipogenic stage of 3T3-L1 preadipocyte differentiation. Knockdown of NRX decreased differentiation of 3T3-L1 cells, whereas overexpression increased differentiation. Adipose tissue-specific NRX transgenic mice showed increases in adipocyte size as well as number compared with WT mice. We further confirmed that the Wingless/int-1 class (Wnt)/β-catenin pathway was also involved in NRX-promoted adipogenesis, consistent with a previous report showing NRX regulation of this pathway. Genes involved in lipid metabolism were downregulated, whereas inflammatory genes, including those encoding macrophage markers, were significantly upregulated, likely contributing to the obesity in Adipo-NRX mice. Our results therefore suggest that NRX acts as a novel proadipogenic factor and controls obesity in vivo.
Collapse
Affiliation(s)
- Young Jae Bahn
- Department of Biological Science, Korea Advanced Institute Science and Technology (KAIST), Daejeon 305-701, Republic of Korea Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 305-806, Republic of Korea
| | - Kwang-Pyo Lee
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 305-806, Republic of Korea
| | - Seung-Min Lee
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 305-806, Republic of Korea
| | - Jeong Yi Choi
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 305-806, Republic of Korea
| | - Yeon-Soo Seo
- Department of Biological Science, Korea Advanced Institute Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Ki-Sun Kwon
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 305-806, Republic of Korea
| |
Collapse
|
39
|
Rharass T, Lemcke H, Lantow M, Kuznetsov SA, Weiss DG, Panáková D. Ca2+-mediated mitochondrial reactive oxygen species metabolism augments Wnt/β-catenin pathway activation to facilitate cell differentiation. J Biol Chem 2014; 289:27937-51. [PMID: 25124032 PMCID: PMC4183826 DOI: 10.1074/jbc.m114.573519] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Emerging evidence suggests that reactive oxygen species (ROS) can stimulate the Wnt/β-catenin pathway in a number of cellular processes. However, potential sources of endogenous ROS have not been thoroughly explored. Here, we show that growth factor depletion in human neural progenitor cells induces ROS production in mitochondria. Elevated ROS levels augment activation of Wnt/β-catenin signaling that regulates neural differentiation. We find that growth factor depletion stimulates the release of Ca(2+) from the endoplasmic reticulum stores. Ca(2+) subsequently accumulates in the mitochondria and triggers ROS production. The inhibition of mitochondrial Ca(2+) uptake with simultaneous growth factor depletion prevents the rise in ROS metabolism. Moreover, low ROS levels block the dissociation of the Wnt effector Dishevelled from nucleoredoxin. Attenuation of the response amplitudes of pathway effectors delays the onset of the Wnt/β-catenin pathway activation and results in markedly impaired neuronal differentiation. Our findings reveal Ca(2+)-mediated ROS metabolic cues that fine-tune the efficiency of cell differentiation by modulating the extent of the Wnt/β-catenin signaling output.
Collapse
Affiliation(s)
- Tareck Rharass
- From Electrochemical Signaling in Development and Disease, Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Strasse 10, D-13125 Berlin-Buch and Cell Biology and Biosystems Technology, Institute of Biological Sciences, and Live Cell Imaging Center, University of Rostock, Albert-Einstein-Strasse 3, D-18059 Rostock, Germany
| | - Heiko Lemcke
- Cell Biology and Biosystems Technology, Institute of Biological Sciences, and Live Cell Imaging Center, University of Rostock, Albert-Einstein-Strasse 3, D-18059 Rostock, Germany
| | - Margareta Lantow
- Cell Biology and Biosystems Technology, Institute of Biological Sciences, and
| | - Sergei A Kuznetsov
- Live Cell Imaging Center, University of Rostock, Albert-Einstein-Strasse 3, D-18059 Rostock, Germany
| | - Dieter G Weiss
- Cell Biology and Biosystems Technology, Institute of Biological Sciences, and Live Cell Imaging Center, University of Rostock, Albert-Einstein-Strasse 3, D-18059 Rostock, Germany
| | - Daniela Panáková
- From Electrochemical Signaling in Development and Disease, Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Strasse 10, D-13125 Berlin-Buch and
| |
Collapse
|
40
|
Sandieson L, Hwang JTK, Kelly GM. Redox regulation of canonical Wnt signaling affects extraembryonic endoderm formation. Stem Cells Dev 2014; 23:1037-49. [PMID: 24471440 DOI: 10.1089/scd.2014.0010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Retinoic acid (RA) induces mouse F9 cells to form primitive endoderm (PrE) and increased levels of reactive oxygen species (ROS) accompany differentiation. ROS are obligatory for differentiation and while H2O2 alone induces PrE, antioxidants attenuate the response to RA. Evidence shows that ROS can modulate the Wnt/β-catenin pathway and in this study, we show that extraembryonic endoderm formation is dependent on the redox state of nucleoredoxin (NRX). In undifferentiated F9 cells, NRX interacted with dishevelled 2 (Dvl2) and while this association was enhanced under reduced conditions, it decreased following H2O2 treatment. Depleting NRX levels caused morphological changes like those induced by RA, while increasing protein kinase A activity further induced these PrE cells to parietal endoderm. Reduced NRX levels also correlated to an increase in T-cell-factors-lymphoid enhancer factors-mediated transcription, indicative of canonical Wnt signaling. Together these results indicate that a mechanism exists whereby NRX maintains canonical Wnt signaling in the off state in F9 cells, while increased ROS levels lift these constraints. Dvl2 no longer bound to NRX is now positioned to prime the Wnt pathway(s) required for PrE formation.
Collapse
Affiliation(s)
- Leanne Sandieson
- Molecular Genetics Unit, Department of Biology, Child Health Research Institute, Western University , London, Canada
| | | | | |
Collapse
|
41
|
ROS, Notch, and Wnt signaling pathways: crosstalk between three major regulators of cardiovascular biology. BIOMED RESEARCH INTERNATIONAL 2014; 2014:318714. [PMID: 24689035 PMCID: PMC3932294 DOI: 10.1155/2014/318714] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 12/28/2013] [Indexed: 12/20/2022]
Abstract
Reactive oxygen species (ROS), traditionally viewed as toxic by-products that cause damage to biomolecules, now are clearly recognized as key modulators in a variety of biological processes and pathological states. The development and regulation of the cardiovascular system require orchestrated activities; Notch and Wnt/β-catenin signaling pathways are implicated in many aspects of them, including cardiomyocytes and smooth muscle cells survival, angiogenesis, progenitor cells recruitment and differentiation, arteriovenous specification, vascular cell migration, and cardiac remodelling. Several novel findings regarding the role of ROS in Notch and Wnt/β-catenin modulation prompted us to review their emerging function in the cardiovascular system during embryogenesis and postnatally.
Collapse
|
42
|
Marchal C, Delorme-Hinoux V, Bariat L, Siala W, Belin C, Saez-Vasquez J, Riondet C, Reichheld JP. NTR/NRX define a new thioredoxin system in the nucleus of Arabidopsis thaliana cells. MOLECULAR PLANT 2014; 7:30-44. [PMID: 24253198 DOI: 10.1093/mp/sst162] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Thioredoxins (TRX) are key components of cellular redox balance, regulating many target proteins through thiol/disulfide exchange reactions. In higher plants, TRX constitute a complex multigenic family whose members have been found in almost all cellular compartments. Although chloroplastic and cytosolic TRX systems have been largely studied, the presence of a nuclear TRX system has been elusive for a long time. Nucleoredoxins (NRX) are potential nuclear TRX found in most eukaryotic organisms. In contrast to mammals, which harbor a unique NRX, angiosperms generally possess multiple NRX organized in three subfamilies. Here, we show that Arabidopsis thaliana has two NRX genes (AtNRX1 and AtNRX2), respectively, belonging to subgroups I and III. While NRX1 harbors typical TRX active sites (WCG/PPC), NRX2 has atypical active sites (WCRPC and WCPPF). Nevertheless, both NRX1 and NRX2 have disulfide reduction capacities, although NRX1 alone can be reduced by the thioredoxin reductase NTRA. We also show that both NRX1 and NRX2 have a dual nuclear/cytosolic localization. Interestingly, we found that NTRA, previously identified as a cytosolic protein, is also partially localized in the nucleus, suggesting that a complete TRX system is functional in the nucleus. We show that NRX1 is mainly found as a dimer in vivo. nrx1 and nrx2 knockout mutant plants exhibit no phenotypic perturbations under standard growth conditions. However, the nrx1 mutant shows a reduced pollen fertility phenotype, suggesting a specific role of NRX1 at the haploid phase.
Collapse
Affiliation(s)
- Corinne Marchal
- Université Perpignan Via Domitia, CNRS, Laboratoire Génome et Développement des Plantes, UMR 5096, 58 Avenue Paul Alduy-Bat T, F-66860, Perpignan, France
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Kagawa Y, Matsumoto S, Kamioka Y, Mimori K, Naito Y, Ishii T, Okuzaki D, Nishida N, Maeda S, Naito A, Kikuta J, Nishikawa K, Nishimura J, Haraguchi N, Takemasa I, Mizushima T, Ikeda M, Yamamoto H, Sekimoto M, Ishii H, Doki Y, Matsuda M, Kikuchi A, Mori M, Ishii M. Cell cycle-dependent Rho GTPase activity dynamically regulates cancer cell motility and invasion in vivo. PLoS One 2013; 8:e83629. [PMID: 24386239 PMCID: PMC3875446 DOI: 10.1371/journal.pone.0083629] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 11/05/2013] [Indexed: 01/09/2023] Open
Abstract
The mechanism behind the spatiotemporal control of cancer cell dynamics and its possible association with cell proliferation has not been well established. By exploiting the intravital imaging technique, we found that cancer cell motility and invasive properties were closely associated with the cell cycle. In vivo inoculation of human colon cancer cells bearing fluorescence ubiquitination-based cell cycle indicator (Fucci) demonstrated an unexpected phenomenon: S/G2/M cells were more motile and invasive than G1 cells. Microarray analyses showed that Arhgap11a, an uncharacterized Rho GTPase-activating protein (RhoGAP), was expressed in a cell-cycle-dependent fashion. Expression of ARHGAP11A in cancer cells suppressed RhoA-dependent mechanisms, such as stress fiber formation and focal adhesion, which made the cells more prone to migrate. We also demonstrated that RhoA suppression by ARHGAP11A induced augmentation of relative Rac1 activity, leading to an increase in the invasive properties. RNAi-based inhibition of Arhgap11a reduced the invasion and in vivo expansion of cancers. Additionally, analysis of human specimens showed the significant up-regulation of Arhgap11a in colon cancers, which was correlated with clinical invasion status. The present study suggests that ARHGAP11A, a cell cycle-dependent RhoGAP, is a critical regulator of cancer cell mobility and is thus a promising therapeutic target in invasive cancers.
Collapse
Affiliation(s)
- Yoshinori Kagawa
- Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Laboratory of Cellular Dynamics, WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- JST, CREST, Chiyoda-ku, Tokyo, Japan
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shinji Matsumoto
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yuji Kamioka
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koshi Mimori
- Department of Surgery, Medical Institute of Bioregulation, Kyushu University, Beppu, Oita, Japan
| | - Yoko Naito
- Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Laboratory of Cellular Dynamics, WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Taeko Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Laboratory of Cellular Dynamics, WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Daisuke Okuzaki
- DNA-chip Developmental Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Naohiro Nishida
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Surgery, Medical Institute of Bioregulation, Kyushu University, Beppu, Oita, Japan
| | - Sakae Maeda
- Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Laboratory of Cellular Dynamics, WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Atsushi Naito
- Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Laboratory of Cellular Dynamics, WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Laboratory of Cellular Dynamics, WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- JST, CREST, Chiyoda-ku, Tokyo, Japan
| | - Keizo Nishikawa
- Laboratory of Cellular Dynamics, WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- JST, CREST, Chiyoda-ku, Tokyo, Japan
| | - Junichi Nishimura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Naotsugu Haraguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Ichiro Takemasa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tsunekazu Mizushima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Masataka Ikeda
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hirofumi Yamamoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Mitsugu Sekimoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hideshi Ishii
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Michiyuki Matsuda
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Kikuchi
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Laboratory of Cellular Dynamics, WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- JST, CREST, Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
44
|
Hanschmann EM, Godoy JR, Berndt C, Hudemann C, Lillig CH. Thioredoxins, glutaredoxins, and peroxiredoxins--molecular mechanisms and health significance: from cofactors to antioxidants to redox signaling. Antioxid Redox Signal 2013; 19:1539-605. [PMID: 23397885 PMCID: PMC3797455 DOI: 10.1089/ars.2012.4599] [Citation(s) in RCA: 507] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 02/01/2013] [Accepted: 02/07/2013] [Indexed: 12/19/2022]
Abstract
Thioredoxins (Trxs), glutaredoxins (Grxs), and peroxiredoxins (Prxs) have been characterized as electron donors, guards of the intracellular redox state, and "antioxidants". Today, these redox catalysts are increasingly recognized for their specific role in redox signaling. The number of publications published on the functions of these proteins continues to increase exponentially. The field is experiencing an exciting transformation, from looking at a general redox homeostasis and the pathological oxidative stress model to realizing redox changes as a part of localized, rapid, specific, and reversible redox-regulated signaling events. This review summarizes the almost 50 years of research on these proteins, focusing primarily on data from vertebrates and mammals. The role of Trx fold proteins in redox signaling is discussed by looking at reaction mechanisms, reversible oxidative post-translational modifications of proteins, and characterized interaction partners. On the basis of this analysis, the specific regulatory functions are exemplified for the cellular processes of apoptosis, proliferation, and iron metabolism. The importance of Trxs, Grxs, and Prxs for human health is addressed in the second part of this review, that is, their potential impact and functions in different cell types, tissues, and various pathological conditions.
Collapse
Affiliation(s)
- Eva-Maria Hanschmann
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, Ernst-Moritz Arndt University, Greifswald, Germany
| | - José Rodrigo Godoy
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Duesseldorf, Germany
| | - Christoph Hudemann
- Institute of Laboratory Medicine, Molecular Diagnostics, Philipps University, Marburg, Germany
| | - Christopher Horst Lillig
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, Ernst-Moritz Arndt University, Greifswald, Germany
| |
Collapse
|
45
|
Cai X, Seal S, McGinnis JF. Sustained inhibition of neovascularization in vldlr-/- mice following intravitreal injection of cerium oxide nanoparticles and the role of the ASK1-P38/JNK-NF-κB pathway. Biomaterials 2013; 35:249-58. [PMID: 24140045 DOI: 10.1016/j.biomaterials.2013.10.022] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 10/04/2013] [Indexed: 02/06/2023]
Abstract
Cerium oxide nanoparticles (nanoceria) are direct antioxidants; they inhibit pathological neovascularization following a single intravitreal injection into new born very low density lipoprotein receptor knockout (vldlr(-/-)) mice. However, the long-term therapeutic effects and mechanisms of nanoceria action on regression of the existing pathologic neovascularization in the eyes are unknown. We intravitreally injected P28 vldlr(-/-) mice and extended the endpoint for analysis until P70. The data demonstrate that nanoceria sustained their therapeutic function up to 6 weeks. Multiple parameters for nanoceria effects were examined including: regression of existing abnormal blood vessels, reduction of vascular leakage, down-regulation of the expression of vascular endothelial growth factor (VEGF), acrolein, glial fibrillary acidic protein (GFAP) and caspase 3 as well as up-regulation of the expression of rod- and cone-opsin genes. Regulation of ASK1-P38/JNK-NF-κB signaling pathway by nanoceria was investigated. Our data demonstrated that a single intravitreal injection of nanoceria in P28 vldlr(-/-) mice produced sustained regression of existing oxidative stress-induced neovascularizations, prevented blood vessel leakage and inhibited apoptosis via down-regulation of the ASK1-P38/JNK-NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xue Cai
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | | | | |
Collapse
|
46
|
Funato Y, Hayashi T, Irino Y, Takenawa T, Miki H. Nucleoredoxin regulates glucose metabolism via phosphofructokinase 1. Biochem Biophys Res Commun 2013; 440:737-42. [PMID: 24120946 DOI: 10.1016/j.bbrc.2013.09.138] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 09/30/2013] [Indexed: 10/26/2022]
Abstract
Phosphofructokinase (PFK) 1 is a glycolytic enzyme, and its abnormality contributes to the development of multiple human diseases, such as cancer. Here, we report that nucleoredoxin (NRX), a thioredoxin-related oxidoreductase, is a novel interacting partner of PFK1. NRX binds directly to PFK1, and endogenous NRX and PFK1 interact in vivo. In NRX(-/-) mouse embryonic fibroblasts (MEFs), the oligomerization status of PFK1 is altered and the catalytic activity of PFK1 is decreased. NRX deficiency augmented levels of NADPH and reduced glutathione, two major cellular antioxidants generated through the pentose phosphate pathway. Indeed, NRX(-/-) MEFs are significantly more resistant to oxidative stress than NRX(+/+) MEFs. These results reveal a novel role of NRX in the regulation of PFK1 activity and in the balance between glycolysis and the pentose phosphate pathway.
Collapse
Affiliation(s)
- Yosuke Funato
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | | | | | | | | |
Collapse
|
47
|
Identification of cilia genes that affect cell-cycle progression using whole-genome transcriptome analysis in Chlamydomonas reinhardtti. G3-GENES GENOMES GENETICS 2013; 3:979-91. [PMID: 23604077 PMCID: PMC3689809 DOI: 10.1534/g3.113.006338] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cilia are microtubule based organelles that project from cells. Cilia are found on almost every cell type of the human body and numerous diseases, collectively termed ciliopathies, are associated with defects in cilia, including respiratory infections, male infertility, situs inversus, polycystic kidney disease, retinal degeneration, and Bardet-Biedl Syndrome. Here we show that Illumina-based whole-genome transcriptome analysis in the biflagellate green alga Chlamydomonas reinhardtii identifies 1850 genes up-regulated during ciliogenesis, 4392 genes down-regulated, and 4548 genes with no change in expression during ciliogenesis. We examined four genes up-regulated and not previously known to be involved with cilia (ZMYND10, NXN, GLOD4, SPATA4) by knockdown of the human orthologs in human retinal pigment epithelial cells (hTERT-RPE1) cells to ask whether they are involved in cilia-related processes that include cilia assembly, cilia length control, basal body/centriole numbers, and the distance between basal bodies/centrioles. All of the genes have cilia-related phenotypes and, surprisingly, our data show that knockdown of GLOD4 and SPATA4 also affects the cell cycle. These results demonstrate that whole-genome transcriptome analysis during ciliogenesis is a powerful tool to gain insight into the molecular mechanism by which centrosomes and cilia are assembled.
Collapse
|
48
|
Abstract
KLHL3 is a BTB-BACK-Kelch family protein that serves as a substrate adapter in Cullin3 (Cul3) E3 ubiquitin ligase complexes. KLHL3 is highly expressed in distal nephron tubules where it is involved in the regulation of electrolyte homeostasis and blood pressure. Mutations in KLHL3 have been identified in patients with inherited hypertension disorders, and several of the disease-associated mutations are located in the presumed Cul3 binding region. Here, we report the crystal structure of a complex between the KLHL3 BTB-BACK domain dimer and two copies of an N terminal fragment of Cul3. We use isothermal titration calorimetry to directly demonstrate that several of the disease mutations in the KLHL3 BTB-BACK domains disrupt the association with Cul3. Both the BTB and BACK domains contribute to the Cul3 interaction surface, and an extended model of the dimeric CRL3 complex places the two E2 binding sites in a suprafacial arrangement with respect to the presumed substrate-binding sites.
Collapse
Affiliation(s)
- Alan X. Ji
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Gilbert G. Privé
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Ontario Cancer Institute, Campbell Family Institute for Cancer Research, University Health Network, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
49
|
Monk KR, Voas MG, Franzini-Armstrong C, Hakkinen IS, Talbot WS. Mutation of sec63 in zebrafish causes defects in myelinated axons and liver pathology. Dis Model Mech 2013; 6:135-45. [PMID: 22864019 PMCID: PMC3529346 DOI: 10.1242/dmm.009217] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 07/19/2012] [Indexed: 12/17/2022] Open
Abstract
Mutations in SEC63 cause polycystic liver disease in humans. Sec63 is a member of the endoplasmic reticulum (ER) translocon machinery, although it is unclear how mutations in SEC63 lead to liver cyst formation in humans. Here, we report the identification and characterization of a zebrafish sec63 mutant, which was discovered in a screen for mutations that affect the development of myelinated axons. Accordingly, we show that disruption of sec63 in zebrafish leads to abnormalities in myelinating glia in both the central and peripheral nervous systems. In the vertebrate nervous system, segments of myelin are separated by the nodes of Ranvier, which are unmyelinated regions of axonal membrane containing a high density of voltage-gated sodium channels. We show that sec63 mutants have morphologically abnormal and reduced numbers of clusters of voltage-gated sodium channels in the spinal cord and along peripheral nerves. Additionally, we observed reduced myelination in both the central and peripheral nervous systems, as well as swollen ER in myelinating glia. Markers of ER stress are upregulated in sec63 mutants. Finally, we show that sec63 mutants develop liver pathology. As in glia, the primary defect, detectable at 5 dpf, is fragmentation and swelling of the ER, indicative of accumulation of proteins in the lumen. At 8 dpf, ER swelling is severe; other pathological features include disrupted bile canaliculi, altered cytoplasmic matrix and accumulation of large lysosomes. Together, our analyses of sec63 mutant zebrafish highlight the possible role of ER stress in polycystic liver disease and suggest that these mutants will serve as a model for understanding the pathophysiology of this disease and other abnormalities involving ER stress.
Collapse
Affiliation(s)
- Kelly R. Monk
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Matthew G. Voas
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Clara Franzini-Armstrong
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6058, USA
| | - Ian S. Hakkinen
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - William S. Talbot
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
50
|
Yabuta N, Mukai S, Okamoto A, Okuzaki D, Suzuki H, Torigata K, Yoshida K, Okada N, Miura D, Ito A, Ikawa M, Okabe M, Nojima H. N-terminal truncation of Lats1 causes abnormal cell growth control and chromosomal instability. J Cell Sci 2012; 126:508-20. [PMID: 23230145 DOI: 10.1242/jcs.113431] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The tumor suppressors Lats1 and Lats2 are mediators of the Hippo pathway that regulates tissue growth and proliferation. Their N-terminal non-kinase regions are distinct except for Lats conserved domains 1 and 2 (LCD1 and LCD2), which may be important for Lats1/2-specific functions. Lats1 knockout mice were generated by disrupting the N-terminal region containing LCD1 (Lats1(ΔN/ΔN)). Some Lats1(ΔN/ΔN) mice were born safely and grew normally. However, mouse embryonic fibroblasts (MEFs) from Lats1(ΔN/ΔN) mice displayed mitotic defects, centrosomal overduplication, chromosomal misalignment, multipolar spindle formation, chromosomal bridging and cytokinesis failure. They also showed anchorage-independent growth and continued cell cycles and cell growth, bypassing cell-cell contact inhibition similar to tumor cells. Lats1(ΔN/ΔN) MEFs produced tumors in nude mice after subcutaneous injection, although the tumor growth rate was much slower than that of ordinary cancer cells. Yap, a key transcriptional coactivator of the Hippo pathway, was overexpressed and stably retained in Lats1(ΔN/ΔN) MEFs in a cell density independent manner, and Lats2 mRNA expression was downregulated. In conclusion, N-terminally truncated Lats1 induced Lats2 downregulation and Yap protein accumulation, leading to chromosomal instability and tumorigenesis.
Collapse
Affiliation(s)
- Norikazu Yabuta
- Department of Molecular Genetics, Osaka University, 3-1 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|