1
|
Ishikawa N, Watanabe Y, Maeda Y, Yoshida T, Kimura N, Abe H, Sakamaki A, Kamimura H, Yokoo T, Kamimura K, Tsuchiya A, Terai S. Human placental extract improves liver cirrhosis in mice with regulation of macrophages and senescent cells. Regen Ther 2025; 28:509-516. [PMID: 39991509 PMCID: PMC11846928 DOI: 10.1016/j.reth.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/06/2025] [Accepted: 01/19/2025] [Indexed: 02/25/2025] Open
Abstract
INTRODUCTION Cirrhosis is a disease with poor prognosis that requires the development of a novel therapeutic approach alternative to liver transplantation. In this study, we focused on the placenta and aimed to clarify the effects of human placental extract (HPE) on cirrhosis. METHODS A mouse model of carbon tetrachloride-induced cirrhosis was used to evaluate the effect of HPE administration subcutaneously and compared with the control group (n = 8 for each group). In vitro and in vivo, real time-PCR and immunostaining were performed for HPE mechanistic analysis. Spatial transcriptomics was also performed for detailed analysis of the effect of HPE on cirrhosis. RESULTS HPE administration improved serum ALT levels compared to control mice. Furthermore, there was a decrease in the number of senescent cells in the liver and the mRNA levels of secrete senescence-associated secretory phenotype factors and Cdkn2a (p16). In vitro, HPE induced macrophage polarization to the anti-inflammatory M2 phenotype. Spatial transcriptomics was also performed to analyze the underlying anti-inflammatory mechanism. The results showed that HPE strongly polarized macrophages to the M2 phenotype, especially in macrophage-rich regions in the liver. Gene expression pathway analysis using spatial transcriptomics also revealed the possibility of improving senescent cell-derived inflammation via mitochondrial function. CONCLUSIONS HPE improves serum ALT levels via anti-inflammatory mechanisms in macrophages and senescent cells. HPE serves as a novel agent for cirrhosis treatment.
Collapse
Affiliation(s)
- Natsuki Ishikawa
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yusuke Watanabe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Division of Preemptive Medicine for Digestive Disease and Healthy Active Life, School of Medicine, Niigata University, Niigata, Japan
| | - Yuichirou Maeda
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Tomoaki Yoshida
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Division of Preemptive Medicine for Digestive Disease and Healthy Active Life, School of Medicine, Niigata University, Niigata, Japan
| | - Naruhiro Kimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hiroyuki Abe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Akira Sakamaki
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hiroteru Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Takeshi Yokoo
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Division of Preemptive Medicine for Digestive Disease and Healthy Active Life, School of Medicine, Niigata University, Niigata, Japan
| | - Kenya Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Atsunori Tsuchiya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
2
|
Lee HH, Chuang HY, Lin K, Yeh CT, Wang YM, Chi HC, Lin KH. RNASE4 promotes malignant progression and chemoresistance in hypoxic glioblastoma via activation of AXL/AKT and NF-κB/cIAPs signaling pathways. Am J Cancer Res 2024; 14:4320-4336. [PMID: 39417186 PMCID: PMC11477813 DOI: 10.62347/udbj5986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/15/2024] [Indexed: 10/19/2024] Open
Abstract
Glioblastoma (GBM) is the most malignant brain tumor frequently characterized by a hypoxic microenvironment. In this investigation, we unveiled unprecedented role of Ribonuclease 4 (RNASE4) in GBM pathogenesis through integrative methodologies. Leveraging The Cancer Genome Atlas (TCGA) dataset and clinical specimens from normal brain tissues, low- and high-grade gliomas, alongside rigorous in vitro and in vivo functional analyses, we identified a consistent upregulation of RNASE4 correlating with advanced GBM pathological stages and poor clinical survival outcomes. Functional assays corroborated the pivotal influences of RNASE4 on key tumorigenic processes such as cell proliferation, migration, invasion, stemness properties and temozolomide (TMZ) resistance. Further, Gene Set Enrichment Analysis (GSEA) illuminated the involvement of RNASE4 in modulating epithelial-mesenchymal transition (EMT) via activation of AXL, AKT and NF-κB signaling pathways. Furthermore, recombinant human RNASE4 (hRNASE4)-mediated NF-κB activation through IκBα phosphorylation and degradation could result in the upregulation of inhibitors of apoptosis proteins (IAPs), such as cIAP1, cIAP2, and SURVIVIN. Notably, treating RNASE4-induced TMZ-resistant cells with the SURVIVIN inhibitor YM-155 significantly restored cellular sensitivity to TMZ therapy. Herein, this study positions RNASE4 as a potent prognostic biomarker and therapeutic target, offering new insights into molecular pathogenesis of GBM and new avenues for future therapeutic interventions.
Collapse
Affiliation(s)
- Hsun-Hua Lee
- Department of Neurology, Taipei Medical University Hospital, Taipei Medical UniversityTaipei 110, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical UniversityTaipei 110, Taiwan
- Dizziness and Balance Disorder Center, Taipei Medical University Hospital, Taipei Medical UniversityTaipei 110, Taiwan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical UniversityNew Taipei 23561, Taiwan
- Dizziness and Balance Disorder Center, Shuang Ho Hospital, Taipei Medical UniversityNew Taipei 23561, Taiwan
| | - Hao-Yu Chuang
- School of Medicine, China Medical UniversityTaichung 40447, Taiwan
- Translational Cell Therapy Center, Tainan Municipal An-Nan Hospital-China Medical UniversityTainan 709204, Taiwan
- Division of Neurosurgery, Tainan Municipal An-Nan Hospital-China Medical UniversityTainan 709204, Taiwan
- Division of Neurosurgery, China Medical University Beigang HospitalBeigang Township, Yunlin 65152, Taiwan
| | - Kent Lin
- Northern Clinical School, Faculty of Medicine and Health, The University of SydneyNSW 2006, Australia
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial HospitalLinkou, Taoyuan 330, Taiwan
| | - Yi-Min Wang
- Department of Neurosurgery, An Nan Hospital, China Medical UniversityTainan 709204, Taiwan
| | - Hsiang-Cheng Chi
- Institute of Biochemistry and Molecular Biology, China Medical UniversityTaichung 404333, Taiwan
- Chinese Medicine Research Center, China Medical UniversityTaichung 40447, Taiwan
| | - Kwang-Huei Lin
- Liver Research Center, Chang Gung Memorial HospitalLinkou, Taoyuan 330, Taiwan
- Department of Biochemistry, College of Medicine, Chang-Gung UniversityTaoyuan 330, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang-Gung UniversityTaoyuan 330, Taiwan
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and TechnologyTaoyuan 330, Taiwan
| |
Collapse
|
3
|
Yao Z, Bai R, Liu W, Liu Y, Zhou W, Xu Z, Sheng J. Activation of angiogenin expression in macrophages by lipopolysaccharide via the TLR4/NF-κB pathway in colitis. Acta Biochim Biophys Sin (Shanghai) 2024; 56:857-865. [PMID: 38567413 PMCID: PMC11214953 DOI: 10.3724/abbs.2024013] [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: 11/29/2023] [Accepted: 01/10/2024] [Indexed: 04/04/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a debilitating condition that can lead to life-threatening complications. Macrophages are crucial in IBD management because they secrete various cytokines and regulate tissue repair. Macrophage-derived angiogenin (ANG) has been shown to be essential for limiting colonic inflammation, but its upstream regulatory pathway and role in macrophages remain unclear. Here we show that ANG expression is up-regulated in macrophages during colitis treatment or upon lipopolysaccharides (LPS) treatment. Mechanistically, LPS activates Toll-like receptor 4 (TLR4) to initiate NF-κB translocation from the cytoplasm to the nucleus, where it binds to the ANG promoter and enhances its transcriptional activity, leading to increased ANG expression. Interestingly, our data also reveal that the deletion of ANG in macrophages has no adverse effect on key macrophage functions, such as phagocytosis, chemotaxis, and cell survival. Our findings establish a "LPS-TLR4-NF-κB-ANG" regulatory axis in inflammatory disorders and confirm that ANG controls inflammation in a paracrine manner, highlighting the importance of ANG as a key mediator in the complex network of inflammatory processes.
Collapse
Affiliation(s)
- Zhengrong Yao
- Institute of Environmental Medicine and Department of General SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Liangzhu LaboratoryZhejiang UniversityHangzhou311121China
| | - Rongpan Bai
- Institute of Environmental Medicine and Department of General SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Liangzhu LaboratoryZhejiang UniversityHangzhou311121China
| | - Wei Liu
- Department of General SurgerySir Run Run Shaw Hospital.Zhejiang University School of MedicineHangzhou310016China
| | - Yaxing Liu
- Institute of Environmental Medicine and Department of General SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Liangzhu LaboratoryZhejiang UniversityHangzhou311121China
| | - Wei Zhou
- Department of General SurgerySir Run Run Shaw Hospital.Zhejiang University School of MedicineHangzhou310016China
| | - Zhengping Xu
- Institute of Environmental Medicine and Department of General SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Liangzhu LaboratoryZhejiang UniversityHangzhou311121China
- Cancer CenterZhejiang UniversityHangzhou310012China
- Zhejiang Provincial Key Laboratory of BioelectromagneticsHangzhou310058China
| | - Jinghao Sheng
- Institute of Environmental Medicine and Department of General SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Liangzhu LaboratoryZhejiang UniversityHangzhou311121China
- Cancer CenterZhejiang UniversityHangzhou310012China
- Zhejiang Provincial Key Laboratory of BioelectromagneticsHangzhou310058China
| |
Collapse
|
4
|
Willemin G, Mange F, Praz V, Lorrain S, Cousin P, Roger C, Willis IM, Hernandez N. Contrasting effects of whole-body and hepatocyte-specific deletion of the RNA polymerase III repressor Maf1 in the mouse. Front Mol Biosci 2023; 10:1297800. [PMID: 38143800 PMCID: PMC10746880 DOI: 10.3389/fmolb.2023.1297800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/15/2023] [Indexed: 12/26/2023] Open
Abstract
MAF1 is a nutrient-sensitive, TORC1-regulated repressor of RNA polymerase III (Pol III). MAF1 downregulation leads to increased lipogenesis in Drosophila melanogaster, Caenorhabditis elegans, and mice. However, Maf1 -/- mice are lean as increased lipogenesis is counterbalanced by futile pre-tRNA synthesis and degradation, resulting in increased energy expenditure. We compared Chow-fed Maf1 -/- mice with Chow- or High Fat (HF)-fed Maf1 hep-/- mice that lack MAF1 specifically in hepatocytes. Unlike Maf1 -/- mice, Maf1 hep-/- mice become heavier and fattier than control mice with old age and much earlier under a HF diet. Liver ChIPseq, RNAseq and proteomics analyses indicate increased Pol III occupancy at Pol III genes, very few differences in mRNA accumulation, and protein accumulation changes consistent with increased lipogenesis. Futile pre-tRNA synthesis and degradation in the liver, as likely occurs in Maf1 hep-/- mice, thus seems insufficient to counteract increased lipogenesis. Indeed, RNAseq and metabolite profiling indicate that liver phenotypes of Maf1 -/- mice are strongly influenced by systemic inter-organ communication. Among common changes in the three phenotypically distinct cohorts, Angiogenin downregulation is likely linked to increased Pol III occupancy of tRNA genes in the Angiogenin promoter.
Collapse
Affiliation(s)
- Gilles Willemin
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - François Mange
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Viviane Praz
- Lausanne Genomic Technologies Facility, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Séverine Lorrain
- Protein Analysis Facility, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Pascal Cousin
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Catherine Roger
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Ian M. Willis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Nouria Hernandez
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
5
|
Wolf EJ, Grünberg S, Dai N, Chen TH, Roy B, Yigit E, Corrêa I. Human RNase 4 improves mRNA sequence characterization by LC–MS/MS. Nucleic Acids Res 2022; 50:e106. [PMID: 35871301 PMCID: PMC9561288 DOI: 10.1093/nar/gkac632] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/22/2022] [Accepted: 07/20/2022] [Indexed: 11/17/2022] Open
Abstract
With the rapid growth of synthetic messenger RNA (mRNA)-based therapeutics and vaccines, the development of analytical tools for characterization of long, complex RNAs has become essential. Tandem liquid chromatography–mass spectrometry (LC–MS/MS) permits direct assessment of the mRNA primary sequence and modifications thereof without conversion to cDNA or amplification. It relies upon digestion of mRNA with site-specific endoribonucleases to generate pools of short oligonucleotides that are then amenable to MS-based sequence analysis. Here, we showed that the uridine-specific human endoribonuclease hRNase 4 improves mRNA sequence coverage, in comparison with the benchmark enzyme RNase T1, by producing a larger population of uniquely mappable cleavage products. We deployed hRNase 4 to characterize mRNAs fully substituted with 1-methylpseudouridine (m1Ψ) or 5-methoxyuridine (mo5U), as well as mRNAs selectively depleted of uridine–two key strategies to reduce synthetic mRNA immunogenicity. Lastly, we demonstrated that hRNase 4 enables direct assessment of the 5′ cap incorporation into in vitro transcribed mRNA. Collectively, this study highlights the power of hRNase 4 to interrogate mRNA sequence, identity, and modifications by LC–MS/MS.
Collapse
Affiliation(s)
- Eric J Wolf
- New England Biolabs, Inc, 43/44 Dunham Ridge, Beverly, MA 01915, USA
| | | | - Nan Dai
- New England Biolabs, Inc, 43/44 Dunham Ridge, Beverly, MA 01915, USA
| | - Tien-Hao Chen
- New England Biolabs, Inc, 43/44 Dunham Ridge, Beverly, MA 01915, USA
| | - Bijoyita Roy
- New England Biolabs, Inc, 43/44 Dunham Ridge, Beverly, MA 01915, USA
| | - Erbay Yigit
- New England Biolabs, Inc, 43/44 Dunham Ridge, Beverly, MA 01915, USA
| | - Ivan R Corrêa
- To whom correspondence should be addressed. Tel: +1 978 380 7504;
| |
Collapse
|
6
|
Vanli N, Sheng J, Li S, Xu Z, Hu GF. Ribonuclease 4 is associated with aggressiveness and progression of prostate cancer. Commun Biol 2022; 5:625. [PMID: 35752711 PMCID: PMC9233706 DOI: 10.1038/s42003-022-03597-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/15/2022] [Indexed: 11/09/2022] Open
Abstract
Prostate specific antigen screening has resulted in a decrease in prostate cancer-related deaths. However, it also has led to over-treatment affecting the quality of life of many patients. New biomarkers are needed to distinguish prostate cancer from benign prostate hyperplasia (BPH) and to predict aggressiveness of the disease. Here, we report that ribonuclease 4 (RNASE4) serves as such a biomarker as well as a therapeutic target. RNASE4 protein level in the plasma is elevated in prostate cancer patients and is positively correlated with disease stage, grade, and Gleason score. Plasma RNASE4 level can be used to predict biopsy outcome and to enhance diagnosis accuracy. RNASE4 protein in prostate cancer tissues is enhanced and can differentiate prostate cancer and BPH. RNASE4 stimulates prostate cancer cell proliferation, induces tumor angiogenesis, and activates receptor tyrosine kinase AXL as well as AKT and S6K. An RNASE4-specific monoclonal antibody inhibits the growth of xenograft human prostate cancer cell tumors in athymic mice.
Collapse
Affiliation(s)
- Nil Vanli
- Divison of Hematology and Oncology, Department of Medicine, Tufts Medical Center, Boston, MA, USA.,Graduate Program in Biochemistry, Graduate School of Biomedical Sciences, Tufts University, Boston, MA, USA
| | - Jinghao Sheng
- Divison of Hematology and Oncology, Department of Medicine, Tufts Medical Center, Boston, MA, USA.,Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuping Li
- Divison of Hematology and Oncology, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Zhengping Xu
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Guo-Fu Hu
- Divison of Hematology and Oncology, Department of Medicine, Tufts Medical Center, Boston, MA, USA. .,Graduate Program in Biochemistry, Graduate School of Biomedical Sciences, Tufts University, Boston, MA, USA.
| |
Collapse
|
7
|
Yu D, Sun J, Weng Y, Luo L, Sheng J, Xu Z. Serum angiogenin as a potential biomarker for early detection of colorectal adenomas and colorectal cancer. Anticancer Drugs 2021; 32:703-708. [PMID: 33661188 DOI: 10.1097/cad.0000000000001047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Although colorectal cancer (CRC) is one of the most common causes of cancer mortality, early-stage detection dramatically improves survival rate. To explore the feasibility of serum angiogenin (ANG) as a biomarker for early detection of colorectal neoplasia, we collected serum samples from 781 participants, including 369 patients with CRC, 133 with colorectal adenoma and 279 healthy controls. We examined the levels of serum ANG by ELISA, calculated the diagnostic accuracy of ANG by plotted receiver operating characteristic curves (ROCs), and compared it with those obtained by carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9). We also analyzed the relationship between serum ANG level and TNM stage in CRC patients. The results showed that ANG serum levels were significantly elevated in patients with colorectal adenomas and CRC (P < 0.01). The area under the ROC curve (AUC) for ANG in distinguishing CRC patients from healthy controls was 0.740 [95% confidence interval (CI): 0.705-0.744], comparable to that of CEA (0.770; 95% CI: 0.735-0.802; P = 0.26) but significantly higher than that of CA19-9 (0.636; 95% CI: 0.598-0.674, P < 0.01), with much higher sensitivity (67.75%) than CEA (36.86%) or CA19-9 (12.20%). We observed no significant differences in ANG serum levels among CRCs at different TNM stages. Furthermore, sensitivity and specificity of ANG for distinguishing colorectal adenomas patients from healthy controls were 66.20% and 64.90%, respectively. ANG has the potential to serve as a serum biomarker for early detection of colorectal neoplasia.
Collapse
Affiliation(s)
| | - Jun Sun
- Institute of Environmental Medicine
| | - Yu Weng
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital
| | - Liang Luo
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | | | | |
Collapse
|
8
|
Angiogenin (ANG)-Ribonuclease Inhibitor (RNH1) System in Protein Synthesis and Disease. Int J Mol Sci 2021; 22:ijms22031287. [PMID: 33525475 PMCID: PMC7866052 DOI: 10.3390/ijms22031287] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/25/2021] [Accepted: 01/25/2021] [Indexed: 12/16/2022] Open
Abstract
Protein synthesis is a highly complex process executed by well-organized translation machinery. Ribosomes, tRNAs and mRNAs are the principal components of this machinery whereas RNA binding proteins and ribosome interacting partners act as accessory factors. Angiogenin (ANG)-Ribonuclease inhibitor (RNH1) system is one such accessory part of the translation machinery that came into focus afresh due to its unconventional role in the translation. ANG is conventionally known for its ability to induce blood vessel formation and RNH1 as a "sentry" to protect RNAs from extracellular RNases. However, recent studies suggest them to be important in translation regulation. During cell homeostasis, ANG in the nucleus promotes rRNA transcription. While under stress, ANG translocates to the cytosol and cleaves tRNA into fragments which inhibit ribosome biogenesis and protein synthesis. RNH1, which intimately interacts with ANG to inhibit its ribonucleolytic activity, can also bind to the 40S ribosomes and control translation by yet to be known mechanisms. Here, we review recent advancement in the knowledge of translation regulation by the ANG-RNH1 system. We also gather information about this system in cell homeostasis as well as in pathological conditions such as cancer and ribosomopathies. Additionally, we discuss the future research directions and therapeutic potential of this system.
Collapse
|
9
|
Luo C, Balsa E, Perry EA, Liang J, Tavares CD, Vazquez F, Widlund HR, Puigserver P. H3K27me3-mediated PGC1α gene silencing promotes melanoma invasion through WNT5A and YAP. J Clin Invest 2020; 130:853-862. [PMID: 31929186 DOI: 10.1172/jci130038] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 10/30/2019] [Indexed: 12/14/2022] Open
Abstract
Oncogene-targeted and immune checkpoint therapies have revolutionized the clinical management of malignant melanoma and now offer hope to patients with advanced disease. Intimately connected to patients' overall clinical risk is whether the initial primary melanoma lesion will metastasize and cause advanced disease, but underlying mechanisms are not entirely understood. A subset of melanomas display heightened peroxisome proliferator-activated receptor γ coactivator 1-α (PGC1α) expression that maintains cell survival cues by promoting mitochondrial function, but also suppresses metastatic spread. Here, we show that PGC1α expression in melanoma cells was silenced by chromatin modifications that involve promoter H3K27 trimethylation. Pharmacological EZH2 inhibition diminished H3K27me3 histone markers, increased PGC1α expression, and functionally suppressed invasion within PGC1α-silenced melanoma cells. Mechanistically, PGC1α silencing activated transcription factor 12 (TCF12), to increase expression of WNT5A, which in turn stabilized YAP protein levels to promote melanoma migration and metastasis. Accordingly, inhibition of components of this transcription-signaling axis, including TCF12, WNT5A, or YAP, blocked melanoma migration in vitro and metastasis in vivo. These results indicate that epigenetic control of melanoma metastasis involved altered expression of PGC1α and an association with the inherent metabolic state of the tumor.
Collapse
Affiliation(s)
- Chi Luo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Eduardo Balsa
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Elizabeth A Perry
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jiaxin Liang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Clint D Tavares
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Hans R Widlund
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Henry BA, Kanarek JP, Kotter A, Helm M, Lee N. 5-methylcytosine modification of an Epstein-Barr virus noncoding RNA decreases its stability. RNA (NEW YORK, N.Y.) 2020; 26:1038-1048. [PMID: 32354721 PMCID: PMC7373997 DOI: 10.1261/rna.075275.120] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 04/27/2020] [Indexed: 05/06/2023]
Abstract
Many cellular noncoding RNAs contain chemically modified nucleotides that are essential for their function. The Epstein-Barr virus expresses two highly abundant noncoding RNAs called EBV-encoded RNA 1 (EBER1) and EBER2. To examine whether these viral RNAs contain modified nucleotides, we purified native EBERs from EBV-infected cells and performed mass spectrometry analysis. While EBER2 contains no modified nucleotides at stoichiometric amounts, EBER1 was found to carry 5-methylcytosine (m5C) modification. Bisulfite sequencing indicated that a single cytosine of EBER1 is methylated in ∼95% of molecules, and the RNA methyltransferase NSUN2 was identified as the EBER1-specific writer. Intriguingly, ablation of NSUN2 and thus loss of m5C modification resulted in an increase in EBER1 levels. We further found that EBER1 is a substrate for the RNase Angiogenin and cleavage in vivo is dependent on the presence of m5C, providing an explanation as to why loss of m5C increases EBER1 levels. Taken together, our observations indicate that m5C, a modification previously shown for tRNAs to oppose Angiogenin-mediated degradation, can also adversely affect RNA stability.
Collapse
Affiliation(s)
- Belle A Henry
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, USA
| | - Jack P Kanarek
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, USA
| | - Annika Kotter
- Johannes Gutenberg University Mainz, Institute of Pharmaceutical and Biomedical Sciences, 55128 Mainz, Germany
| | - Mark Helm
- Johannes Gutenberg University Mainz, Institute of Pharmaceutical and Biomedical Sciences, 55128 Mainz, Germany
| | - Nara Lee
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, USA
| |
Collapse
|
11
|
Lee HH, Wang YN, Hung MC. Functional roles of the human ribonuclease A superfamily in RNA metabolism and membrane receptor biology. Mol Aspects Med 2019; 70:106-116. [PMID: 30902663 DOI: 10.1016/j.mam.2019.03.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/17/2019] [Indexed: 02/08/2023]
Abstract
The human ribonuclease A (hRNase A) superfamily is comprised of 13 members of secretory RNases, most of which are recognized as catabolic enzymes for their ribonucleolytic activity to degrade ribonucleic acids (RNAs) in the extracellular space, where they play a role in innate host defense and physiological homeostasis. Interestingly, human RNases 9-13, which belong to a non-canonical subgroup of the hRNase A superfamily, are ribonucleolytic activity-deficient proteins with unclear biological functions. Moreover, accumulating evidence indicates that secretory RNases, such as human RNase 5, can be internalized into cells facilitated by membrane receptors like the epidermal growth factor receptor to regulate intracellular RNA species, in particular non-coding RNAs, and signaling pathways by either a ribonucleolytic activity-dependent or -independent manner. In this review, we summarize the classical role of hRNase A superfamily in the metabolism of extracellular and intracellular RNAs and update its non-classical function as a cognate ligand of membrane receptors. We further discuss the biological significance and translational potential of using secretory RNases as predictive biomarkers or therapeutic agents in certain human diseases and the pathological settings for future investigations.
Collapse
Affiliation(s)
- Heng-Huan Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Unit 108, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Ying-Nai Wang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Unit 108, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Unit 108, 1515 Holcombe Boulevard, Houston, TX, 77030, USA; Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung, 404, Taiwan; Department of Biotechnology, Asia University, Taichung 413, Taiwan.
| |
Collapse
|
12
|
Farkas C, Fuentes-Villalobos F, Rebolledo-Jaramillo B, Benavides F, Castro AF, Pincheira R. Streamlined computational pipeline for genetic background characterization of genetically engineered mice based on next generation sequencing data. BMC Genomics 2019; 20:131. [PMID: 30755158 PMCID: PMC6373082 DOI: 10.1186/s12864-019-5504-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 01/31/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Genetically engineered mice (GEM) are essential tools for understanding gene function and disease modeling. Historically, gene targeting was first done in embryonic stem cells (ESCs) derived from the 129 family of inbred strains, leading to a mixed background or congenic mice when crossed with C57BL/6 mice. Depending on the number of backcrosses and breeding strategies, genomic segments from 129-derived ESCs can be introgressed into the C57BL/6 genome, establishing a unique genetic makeup that needs characterization in order to obtain valid conclusions from experiments using GEM lines. Currently, SNP genotyping is used to detect the extent of 129-derived ESC genome introgression into C57BL/6 recipients; however, it fails to detect novel/rare variants. RESULTS Here, we present a computational pipeline implemented in the Galaxy platform and in BASH/R script to determine genetic introgression of GEM using next generation sequencing data (NGS), such as whole genome sequencing (WGS), whole exome sequencing (WES) and RNA-Seq. The pipeline includes strategies to uncover variants linked to a targeted locus, genome-wide variant visualization, and the identification of potential modifier genes. Although these methods apply to congenic mice, they can also be used to describe variants fixed by genetic drift. As a proof of principle, we analyzed publicly available RNA-Seq data from five congenic knockout (KO) lines and our own RNA-Seq data from the Sall2 KO line. Additionally, we performed target validation using several genetics approaches. CONCLUSIONS We revealed the impact of the 129-derived ESC genome introgression on gene expression, predicted potential modifier genes, and identified potential phenotypic interference in KO lines. Our results demonstrate that our new approach is an effective method to determine genetic introgression of GEM.
Collapse
Affiliation(s)
- C Farkas
- Laboratorio de Transducción de Señales y Cáncer. Departamento de Bioquímica y Biología Molecular. Facultad Cs. Biológicas, Universidad de Concepción, Concepción, Chile
| | - F Fuentes-Villalobos
- Laboratorio de Transducción de Señales y Cáncer. Departamento de Bioquímica y Biología Molecular. Facultad Cs. Biológicas, Universidad de Concepción, Concepción, Chile
| | | | - F Benavides
- Department of Epigenetics and Molecular Carcinogenesis, M.D. Anderson Cancer Center, Smithville, TX, USA
| | - A F Castro
- Laboratorio de Transducción de Señales y Cáncer. Departamento de Bioquímica y Biología Molecular. Facultad Cs. Biológicas, Universidad de Concepción, Concepción, Chile
| | - R Pincheira
- Laboratorio de Transducción de Señales y Cáncer. Departamento de Bioquímica y Biología Molecular. Facultad Cs. Biológicas, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
13
|
Li HY, Yang HG, Li P, Wang YZ, Huang GX, Xing L, Wang JQ, Zheng N. Effect of Heat Treatment on the Antitumor Activity of Lactoferrin in Human Colon Tumor (HT29) Model. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:140-147. [PMID: 30418775 DOI: 10.1021/acs.jafc.8b05131] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
To investigate the effect of heat treatment on the antitumor activity of lactoferrin in colon cancer cells and colon tumors, the HT-29 (human intestinal epithelial tumor cell) cell line was exposed to lactoferrin and various heat treatments. The impacts on cell proliferation, invasion, and migration were observed in vitro, and nude mice bearing HT29 tumors were administered lactoferrin and underwent various heat treatments in vivo. In the HT29 cell proliferation test using transwell and scratch analyses, lactoferrin (20 mg/mL) without or with heat treatment (50 and 70 °C) significantly inhibited cell proliferation, migration, and invasion (compared with the control, p < 0.05), while lactoferrin with heat treatment (100 °C) did not affect these parameters. In vivo, HT29 tumor weight was significantly reduced in the lactoferrin (without heat treatment and with 50 and 70 °C treatment) groups (1.59 ± 0.20, 1.67 ± 0.25, and 2.41 ± 0.42 g, compared with the control, p < 0.05), and there was no significant difference between the control (3.73 ± 0.33 g) and the 100 °C treatment group (3.58 ± 0.29 g). Moreover, 100 °C heat treatment reduced inhibition of the VEGFR2/VEGFA/PI3K/Akt/Erk1/2 angiogenesis pathway by lactoferrin. In summary, HT29 tumors were effectively suppressed by lactoferrin via inhibition of VEGFR2/VEGFA/PI3K/Akt/Erk1/2 pathway, and heat treatment affected the antitumor activity of lactoferrin in a temperature-dependent manner.
Collapse
Affiliation(s)
- Hui-Ying Li
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
| | - Huai-Gu Yang
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
| | - Peng Li
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
| | - Yi-Zhen Wang
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
| | - Guo-Xin Huang
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
| | - Lei Xing
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
| | - Jia-Qi Wang
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
| | - Nan Zheng
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
| |
Collapse
|
14
|
The Immunomodulatory and Antimicrobial Properties of the Vertebrate Ribonuclease A Superfamily. Vaccines (Basel) 2018; 6:vaccines6040076. [PMID: 30463297 PMCID: PMC6313885 DOI: 10.3390/vaccines6040076] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/31/2018] [Accepted: 11/16/2018] [Indexed: 02/08/2023] Open
Abstract
The Ribonuclease A Superfamily is composed of cationic peptides that are secreted by immune cells and epithelial tissues. Although their physiological roles are unclear, several members of the vertebrate Ribonuclease A Superfamily demonstrate antimicrobial and immune modulation activities. The objective of this review is to provide an overview of the published literature on the Ribonuclease A Superfamily with an emphasis on each peptide’s regulation, antimicrobial properties, and immunomodulatory functions. As additional insights emerge regarding the mechanisms in which these ribonucleases eradicate invading pathogens and modulate immune function, these ribonucleases may have the potential to be developed as a novel class of therapeutics for some human diseases.
Collapse
|
15
|
DNA methylation as a mediator of HLA-DRB1*15:01 and a protective variant in multiple sclerosis. Nat Commun 2018; 9:2397. [PMID: 29921915 PMCID: PMC6008330 DOI: 10.1038/s41467-018-04732-5] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/17/2018] [Indexed: 01/28/2023] Open
Abstract
The human leukocyte antigen (HLA) haplotype DRB1*15:01 is the major risk factor for multiple sclerosis (MS). Here, we find that DRB1*15:01 is hypomethylated and predominantly expressed in monocytes among carriers of DRB1*15:01. A differentially methylated region (DMR) encompassing HLA-DRB1 exon 2 is particularly affected and displays methylation-sensitive regulatory properties in vitro. Causal inference and Mendelian randomization provide evidence that HLA variants mediate risk for MS via changes in the HLA-DRB1 DMR that modify HLA-DRB1 expression. Meta-analysis of 14,259 cases and 171,347 controls confirms that these variants confer risk from DRB1*15:01 and also identifies a protective variant (rs9267649, p < 3.32 × 10-8, odds ratio = 0.86) after conditioning for all MS-associated variants in the region. rs9267649 is associated with increased DNA methylation at the HLA-DRB1 DMR and reduced expression of HLA-DRB1, suggesting a modulation of the DRB1*15:01 effect. Our integrative approach provides insights into the molecular mechanisms of MS susceptibility and suggests putative therapeutic strategies targeting a methylation-mediated regulation of the major risk gene.
Collapse
|
16
|
Cătană CS, Pichler M, Giannelli G, Mader RM, Berindan-Neagoe I. Non-coding RNAs, the Trojan horse in two-way communication between tumor and stroma in colorectal and hepatocellular carcinoma. Oncotarget 2018; 8:29519-29534. [PMID: 28392501 PMCID: PMC5438748 DOI: 10.18632/oncotarget.15706] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/24/2017] [Indexed: 12/29/2022] Open
Abstract
In a continuous and mutual exchange of information, cancer cells are invariably exposed to microenvironment transformation. This continuous alteration of the genetic, molecular and cellular peritumoral stroma background has become as critical as the management of primary tumor progression events in cancer cells. The communication between stroma and tumor cells within the extracellular matrix is one of the triggers in colon and liver carcinogenesis. All non- codingRNAs including long non-coding RNAs, microRNAs and ultraconserved genes play a critical role in almost all cancers and are responsible for the modulation of the tumor microenvironment in several malignant processes such as initiation, progression and dissemination. This review details the involvement of non codingRNAs in the evolution of human colorectal carcinoma and hepatocellular carcinoma in relationship with the microenvironment. Recent research has shown that a considerable number of dysregulated non- codingRNAs could be valuable diagnostic and prognostic biomarkers in cancer. Therefore, more in-depth knowledge of the role non- codingRNAs play in stroma-tumor communication and of the complex regulatory mechanisms between ultraconserved genes and microRNAs supports the validation of future effective therapeutic targets in patients suffering from hepatocellular and colorectal carcinoma, two distinctive entities which share quite a lot common non-coding RNAs.
Collapse
Affiliation(s)
- Cristina- Sorina Cătană
- Department of Medical Biochemistry, ""Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Martin Pichler
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, Graz, Austria
| | - Gianluigi Giannelli
- Department of Internal Medicine, Immunology and Infectious Diseases, Section of Internal Medicine, University of Bari Medical School, Bari, Italy
| | - Robert M Mader
- Department of Medicine I, Comprehensive Cancer Center of the Medical University of Vienna, Austria
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Institute of Doctoral Studies, ""Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Experimental Pathology, "Ion Chiricuta" Institute of Oncology, Cluj-Napoca, Romania.,Medfuture Research Center for Advanced Medicine, Cluj-Napoca, Romania
| |
Collapse
|
17
|
Dong X, Cong S. Identification of differentially expressed genes and regulatory relationships in Huntington's disease by bioinformatics analysis. Mol Med Rep 2018; 17:4317-4326. [PMID: 29328442 PMCID: PMC5802203 DOI: 10.3892/mmr.2018.8410] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 12/20/2017] [Indexed: 12/14/2022] Open
Abstract
Huntington's disease (HD) is an inherited, progressive neurodegenerative disease caused by a CAG expansion in the huntingtin (HTT) gene; various dysfunctions of biological processes in HD have been proposed. However, at present the exact pathogenesis of HD is not fully understood. The present study aimed to explore the pathogenesis of HD using a computational bioinformatics analysis of gene expression. GSE11358 was downloaded from the Gene Expression Omnibus andthe differentially expressed genes (DEGs) in the mutant HTT knock-in cell model STHdhQ111/Q111 were predicted. DEGs between the HD and control samples were screened using the limma package in R. Functional and pathway enrichment analyses were conducted using the database for annotation, visualization and integrated discovery software. A protein-protein interaction (PPI) network was established by the search tool for the retrieval of interacting genes and visualized by Cytoscape. Module analysis of the PPI network was performed utilizing MCODE. A total of 471 DEGs were identified, including ribonuclease A family member 4 (RNASE4). In addition, 41 significantly enriched Kyoto Encyclopedia of Genes and Genomes pathways, as well as several significant Gene Ontology terms (including cytokine-cytokine receptor interaction and cytosolic DNA-sensing) were identified. A total of 18 significant modules were identified from the PPI network. Furthermore, a novel transcriptional regulatory relationship was identified, namely signal transducer and activator of transcription 3 (STAT3), which is regulated by miRNA-124 in HD. In conclusion, deregulation of 18 critical genes may contribute to the occurrence of HD. RNASE4, STAT3, and miRNA-124 may have a regulatory association with the pathological mechanisms in HD.
Collapse
Affiliation(s)
- Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Shuyan Cong
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
18
|
Nathan A, Reinhardt P, Kruspe D, Jörß T, Groth M, Nolte H, Habenicht A, Herrmann J, Holschbach V, Toth B, Krüger M, Wang ZQ, Platzer M, Englert C. The Wilms tumor protein Wt1 contributes to female fertility by regulating oviductal proteostasis. Hum Mol Genet 2017; 26:1694-1705. [PMID: 28334862 PMCID: PMC5411738 DOI: 10.1093/hmg/ddx075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/23/2017] [Indexed: 11/16/2022] Open
Abstract
Although the zinc finger transcription factor Wt1 has been linked to female fertility, its precise role in this process has not yet been understood. We have sequenced the WT1 exons in a panel of patients with idiopathic infertility and have identified a missense mutation in WT1 in one patient out of eight. This mutation leads to an amino acid change within the zinc finger domain and results in reduced DNA binding. We utilized Wt1+/- mice as a model to mechanistically pinpoint the consequences of reduced Wt1 levels for female fertility. Our results indicate that subfertility in Wt1+/- female mice is a maternal effect caused by the Wt1-dependent de-regulation of Prss29, encoding a serine protease. Notably, blocking Prss29 activity was sufficient to rescue subfertility in Wt1+/- mice indicating Prss29 as a critical factor in female fertility. Molecularly, Wt1 represses expression of Prss29. De-repression and precocious expression of Prss29 in the oviduct of Wt1+/- mice interferes with pre-implantation development. Our study reveals a novel role for Wt1 in early mammalian development and identifies proteases as critical mediators of the maternal-embryonic interaction. Our data also suggest that the role of Wt1 in regulating fertility is conserved in mammals.
Collapse
Affiliation(s)
| | | | | | | | - Marco Groth
- Genome Analysis Lab, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Hendrik Nolte
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Andreas Habenicht
- Institute for Vascular Medicine, Jena University Hospital, Jena, Germany
| | - Jörg Herrmann
- Department of Gynecology and Obstetrics, Hufeland Klinikum, 99425 Weimar, Germany
| | - Verena Holschbach
- Department of Gynecological Endocrinology and Fertility Disorders, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Bettina Toth
- Department of Gynecological Endocrinology and Fertility Disorders, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Marcus Krüger
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | | | - Matthias Platzer
- Genome Analysis Lab, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Christoph Englert
- Molecular Genetics Lab.,Institute of Biochemistry and Biophysics, Friedrich-Schiller-University Jena, 07745 Jena, Germany
| |
Collapse
|
19
|
Luo C, Balsa E, Thomas A, Hatting M, Jedrychowski M, Gygi SP, Widlund HR, Puigserver P. ERRα Maintains Mitochondrial Oxidative Metabolism and Constitutes an Actionable Target in PGC1α-Elevated Melanomas. Mol Cancer Res 2017; 15:1366-1375. [PMID: 28596418 PMCID: PMC5954239 DOI: 10.1158/1541-7786.mcr-17-0143] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/15/2017] [Accepted: 06/05/2017] [Indexed: 12/28/2022]
Abstract
The uncontrolled growth of tumors provides metabolic dependencies that can be harnessed for therapeutic benefit. Although tumor cells exhibit these increased metabolic demands due to their rapid proliferation, these metabolic processes are general to all cells, and furthermore, targeted therapeutic intervention can provoke compensatory adaptation that alters tumors' characteristics. As an example, a subset of melanomas depends on the transcriptional coactivator PGC1α function to sustain their mitochondrial energy-dependent survival. However, selective outgrowth of resistant PGC1α-independent tumor cells becomes endowed with an augmented metastatic phenotype. To find PGC1α-dependent components that would not affect metastasis in melanomas, an unbiased proteomic analyses was performed and uncovered the orphan nuclear receptor ERRα, which supports PGC1α's control of mitochondrial energetic metabolism, but does not affect the antioxidant nor antimetastatic regulatory roles. Specifically, genetic or pharmacologic inhibition of ERRα reduces the inherent bioenergetic capacity and decreases melanoma cell growth, but without altering the invasive characteristics. Thus, within this particularly aggressive subset of melanomas, which is characterized by heighted expression of PGC1α, ERRα specifically mediates prosurvival functions and represents a tangible therapeutic target.Implications: ERRα, a druggable protein, mediates the bioenergetic effects in melanomas defined by high PGC1α expression, suggesting a rational means for therapeutic targeting of this particularly aggressive melanoma subtype. Mol Cancer Res; 15(10); 1366-75. ©2017 AACR.
Collapse
Affiliation(s)
- Chi Luo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Eduardo Balsa
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Ajith Thomas
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Maximilian Hatting
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Mark Jedrychowski
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Hans R Widlund
- Brigham and Women's Hospital, Department of Dermatology, Harvard Medical School, Boston, Massachusetts
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
20
|
Hu B, Xu C, Cao P, Tian Y, Zhang Y, Shi C, Xu J, Yuan W, Chen H. TGF-β Stimulates Expression of Chondroitin Polymerizing Factor in Nucleus Pulposus Cells Through the Smad3, RhoA/ROCK1, and MAPK Signaling Pathways. J Cell Biochem 2017; 119:566-579. [PMID: 28608941 DOI: 10.1002/jcb.26215] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 06/12/2017] [Indexed: 01/01/2023]
Abstract
The enzyme chondroitin polymerizing factor (ChPF) is primarily involved in extension of the chondroitin sulfate backbone required for the synthesis of sulfated glycosaminoglycan (sGAG). Transforming growth factor beta (TGF-β) upregulates sGAG synthesis in nucleus pulposus cells; however, the mechanisms mediating this induction are incompletely understood. Our study demonstrated that ChPF expression was negatively correlated with the grade of degenerative intervertebral disc disease. Treatment of nucleus pulposus cells with TGF-β induced ChPF expression and enhanced Smad2/3, RhoA/ROCK activation, and the JNK, p38, and ERK1/2 MAPK signaling pathways. Selective inhibitors of Smad2/3, RhoA or ROCK1/2, and knockdown of Smad3 and ROCK1 attenuated ChPF expression and sGAG synthesis induced by TGF-β. In addition, we showed that RhoA/ROCK1 signaling upregulated ChPF via activation of the JNK pathway but not the p38 and ERK1/2 signaling pathways. Moreover, inhibitors of JNK, p38 and ERK1/2 activity also blocked ChPF expression and sGAG synthesis induced by TGF-β in a Smad3-independent manner. Collectively, our data suggest that TGF-β stimulated the expression of ChPF and sGAG synthesis in nucleus pulposus cells through Smad3, RhoA/ROCK1 and the three MAPK signaling pathways. J. Cell. Biochem. 119: 566-579, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bo Hu
- Department of Spinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Chen Xu
- Department of Spinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Peng Cao
- Department of Spinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Ye Tian
- Department of Spinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Ying Zhang
- Department of Spinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Changgui Shi
- Department of Spinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Jun Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Hei Longjiang Province, 150086, China
| | - Wen Yuan
- Department of Spinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Huajiang Chen
- Department of Spinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| |
Collapse
|
21
|
Li S, Chen Y, Sun D, Bai R, Gao X, Yang Y, Sheng J, Xu Z. Angiogenin Prevents Progranulin A9D Mutation-Induced Neuronal-Like Cell Apoptosis Through Cleaving tRNAs into tiRNAs. Mol Neurobiol 2017; 55:1338-1351. [DOI: 10.1007/s12035-017-0396-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 01/10/2017] [Indexed: 12/20/2022]
|
22
|
Solomon T, Smith EN, Matsui H, Braekkan SK, Wilsgaard T, Njølstad I, Mathiesen EB, Hansen JB, Frazer KA. Associations Between Common and Rare Exonic Genetic Variants and Serum Levels of 20 Cardiovascular-Related Proteins: The Tromsø Study. ACTA ACUST UNITED AC 2016; 9:375-83. [PMID: 27329291 PMCID: PMC4982757 DOI: 10.1161/circgenetics.115.001327] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 06/16/2016] [Indexed: 01/09/2023]
Abstract
Supplemental Digital Content is available in the text. Background— Genetic variation can be used to study causal relationships between biomarkers and diseases. Here, we identify new common and rare genetic variants associated with cardiovascular-related protein levels (protein quantitative trait loci [pQTLs]). We functionally annotate these pQTLs, predict and experimentally confirm a novel molecular interaction, and determine which pQTLs are associated with diseases and physiological phenotypes. Methods and Results— As part of a larger case–control study of venous thromboembolism, serum levels of 51 proteins implicated in cardiovascular diseases were measured in 330 individuals from the Tromsø Study. Exonic genetic variation near each protein’s respective gene (cis) was identified using sequencing and arrays. Using single site and gene-based tests, we identified 27 genetic associations between pQTLs and the serum levels of 20 proteins: 14 associated with common variation in cis, of which 6 are novel (ie, not previously reported); 7 associations with rare variants in cis, of which 4 are novel; and 6 associations in trans. Of the 20 proteins, 15 were associated with single sites and 7 with rare variants. cis-pQTLs for kallikrein and F12 also show trans associations for proteins (uPAR, kininogen) known to be cleaved by kallikrein and with NTproBNP. We experimentally demonstrate that kallikrein can cleave proBNP (NTproBNP precursor) in vitro. Nine of the pQTLs have previously identified associations with 17 disease and physiological phenotypes. Conclusions— We have identified cis and trans genetic variation associated with the serum levels of 20 proteins and utilized these pQTLs to study molecular mechanisms underlying disease and physiological phenotypes.
Collapse
Affiliation(s)
- Terry Solomon
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | - Erin N Smith
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | - Hiroko Matsui
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | - Sigrid K Braekkan
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | | | - Tom Wilsgaard
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | - Inger Njølstad
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | - Ellisiv B Mathiesen
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | - John-Bjarne Hansen
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | - Kelly A Frazer
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.).
| |
Collapse
|
23
|
Sheng J, Xu Z. Three decades of research on angiogenin: a review and perspective. Acta Biochim Biophys Sin (Shanghai) 2016; 48:399-410. [PMID: 26705141 DOI: 10.1093/abbs/gmv131] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/23/2015] [Indexed: 01/17/2023] Open
Abstract
As a member of the vertebrate-specific secreted ribonucleases, angiogenin (ANG) was first isolated and identified solely by its ability to induce new blood vessel formation, and now, it has been recognized to play important roles in various physiological and pathological processes through regulating cell proliferation, survival, migration, invasion, and/or differentiation. ANG exhibits very weak ribonucleolytic activity that is critical for its biological functions, and exerts its functions through activating different signaling transduction pathways in different target cells. A series of recent studies have indicated that ANG contributes to cellular nucleic acid metabolism. Here, we comprehensively review the results of studies regarding the structure, mechanism, and function of ANG over the past three decades. Moreover, current problems and future research directions of ANG are discussed. The understanding of the function and mechanism of ANG in a wide context will help to better delineate its roles in diseases, especially in cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Jinghao Sheng
- Institute of Environmental Health, Zhejiang University School of Public Health, Hangzhou 310058, China Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhengping Xu
- Institute of Environmental Health, Zhejiang University School of Public Health, Hangzhou 310058, China Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
24
|
Vanli N, Guo-Fu HU. Mechanism and Function of Angiogenin in Prostate Cancer. ZHONGGUO SHENG WU HUA XUE YU FEN ZI SHENG WU XUE BAO = CHINESE JOURNAL OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2015; 31:1261-1266. [PMID: 27175049 DOI: 10.13865/j.cnki.cjbmb.2015.12.06] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Angiogenin (ANG), the fifth member of the vertebrate-specific ribonuclease (RNase) A superfamily, is a secreted angiogenic ribonuclease strongly up-regulated in human prostate cancers. ANG is translocated to the nucleus in both prostate cancer epithelial cells and endothelial cells to exert its role in prostate cancer progression by mediating tumor angiogenesis, cancer cell survival and proliferation through rRNA biogenesis. ANG-stimulated rRNA is required not only for prostate intraepithelial neoplasia (PIN) formation, but also for androgen-independent growth of prostate cancer cells. Targeting ANG by various antagonists that inhibit its nuclear translocation, function and/or activity has proven to inhibit prostate cancer growth in animal models. Furthermore, the role of ANG in androgen independence has been firmly established, suggesting a strong rationale for therapeutically targeting ANG in the treatment of castration resistant prostate cancer.
Collapse
Affiliation(s)
- Nil Vanli
- Molecular Oncology Research Institute, Tufts Medical Center Graduate Program in Biochemistry, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - H U Guo-Fu
- Molecular Oncology Research Institute, Tufts Medical Center Graduate Program in Biochemistry, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| |
Collapse
|
25
|
Ribonuclease like 5 regulates zebrafish yolk extension by suppressing a p53-dependent DNA damage response pathway. Int J Biochem Cell Biol 2015; 65:12-9. [PMID: 25980932 DOI: 10.1016/j.biocel.2015.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 04/10/2015] [Accepted: 05/05/2015] [Indexed: 12/15/2022]
Abstract
Ribonuclease like 5 (Rnasel5) is a novel member of the zebrafish ribonuclease A family and its expression is increased during early embryogenesis. However, the in vivo biological function of Rnasel5 remains to be elucidated. Here, we report that knockdown of Rnasel5 by morhpolinos caused shrunken yolk extension as well as increased DNA damage at yolk syncytial layer and external tissue layers via the activation of p53 pathway. In addition, the morphological defects caused by Rnasel5 knockdown can be partially rescued by mRNA injection. Our findings provide the first functional characterization of Rnasel5 in zebrafish development and reveal its critical role in yolk extension by modulation of the p53 pathway.
Collapse
|