1
|
Wu Z, Wang D, Fu D, Ning D, Gu S. Rituximab-Chidamide combination chemotherapy enhances autophagy to overcome drug resistance in diffuse large B-cell lymphoma. Int Immunopharmacol 2025; 156:114578. [PMID: 40258315 DOI: 10.1016/j.intimp.2025.114578] [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: 12/13/2024] [Revised: 03/10/2025] [Accepted: 03/27/2025] [Indexed: 04/23/2025]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a challenging malignancy, particularly when resistance to standard therapies such as Rituximab develops. This study investigates the combined therapeutic effects of Rituximab and Chidamide on DLBCL, focusing on drug resistance mechanisms and autophagy regulation. Using high-throughput proteomics and transcriptomic analyses, key proteins and signaling pathways were identified. BTG1 emerged as a signature gene, while autophagy-related genes such as BECN1, ATG5, HSPA8, PTEN, and MAPK8 were highlighted as pivotal players. In vitro experiments using Rituximab-sensitive and -resistant DLBCL cell lines (Raji and Raji-4RH) demonstrated that Chidamide significantly inhibited cell proliferation in a dose- and time-dependent manner, induced G0/G1 phase arrest, and enhanced autophagy. Mechanistically, Chidamide upregulated histone acetylation and autophagy-related proteins while reducing p62 levels, synergistically promoting autophagy with Rituximab. In vivo mouse models confirmed the combined treatment's efficacy in suppressing tumor growth. These findings suggest that the BTG1/BECN1/ATG5 signaling axis plays a critical role in enhancing autophagy and reversing Rituximab resistance. The combination of Chidamide and Rituximab presents a promising therapeutic strategy, offering new insights into overcoming drug resistance in DLBCL.
Collapse
Affiliation(s)
- Zelai Wu
- Department of Hematology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Dongni Wang
- Department of Hematology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Di Fu
- Department of General Practice, The Affiliated Center Hospital of Shenyang Medical College, Shenyang 110036, China
| | - Daohua Ning
- Department of Hematology, Anshan Central Hospital, Anshan 114000, China
| | - Shanshan Gu
- Department of Hematology, General Hospital of Northern Theater Command, Shenyang 110016, China.
| |
Collapse
|
2
|
Glass BH, Ye AC, Hemphill CN, Jones KG, Dworetzky AG, Barott KL. Hypoxia Disrupts Sex-Specific Physiology and Gene Expression Leading to Decreased Fitness in the Estuarine Sea Anemone Nematostella vectensis. Mol Ecol 2025; 34:e17755. [PMID: 40192436 PMCID: PMC12010470 DOI: 10.1111/mec.17755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/07/2025] [Accepted: 03/27/2025] [Indexed: 04/22/2025]
Abstract
Coastal seawater hypoxia is increasing in temperate estuaries under global climate change, yet it is unknown how low oxygen conditions affect most estuarine species. We found that hypoxia has increased since the 1990s in an estuary hosting the sea anemone Nematostella vectensis (Jacques Cousteau National Estuarine Research Reserve, New Jersey, USA). Adult N. vectensis bred from anemones collected in this estuary exposed to three consecutive nights of hypoxia (dissolved oxygen = 0.5-1.5 mg L-1 for ~12 h night-1) during gametogenesis displayed decreased aerobic respiration rates and biomass, indicating metabolic disruption. Physiological declines were correlated with changes in the expression of genes related to oxygen-dependent metabolic processes, many of which are targets of hypoxia-inducible factor 1α (HIF1α), demonstrating the activity of this transcription factor for the first time in this early-diverging metazoan. The upregulation of genes involved in the unfolded protein response and endoplasmic reticulum and Golgi apparatus homeostasis suggested that misfolded proteins contributed to disrupted physiology. Notably, these responses were more pronounced in females, demonstrating sex-specific sensitivity that was also observed in reproductive outcomes, with declines in female but not male fecundity following hypoxia exposure. However, sperm from exposed males had higher mitochondrial membrane potential, indicating altered spermatogenesis. Further, crosses performed with gametes from hypoxia-exposed adults yielded strikingly low developmental success (~2%), yet larvae that did develop displayed similar respiration rates and accelerated settlement compared to controls. Overall, hypoxia depressed fitness in N. vectensis by over 95%, suggesting that even stress-tolerant estuarine species may be threatened by coastal deoxygenation.
Collapse
Affiliation(s)
- Benjamin H. Glass
- Department of Biology, School of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Angela C. Ye
- Department of Biology, School of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Cassidy N. Hemphill
- Department of Biology, School of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental Biology, Epigenetics Institute, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Katelyn G. Jones
- Department of Biology, School of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Anna G. Dworetzky
- Department of Biology, School of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Katie L. Barott
- Department of Biology, School of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
3
|
Mount HO, Urbanus ML, Zangari F, Gingras AC, Ensminger AW. The Legionella pneumophila effector PieF modulates mRNA stability through association with eukaryotic CCR4-NOT. mSphere 2025; 10:e0089124. [PMID: 39699231 PMCID: PMC11774319 DOI: 10.1128/msphere.00891-24] [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: 10/23/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
The eukaryotic CCR4-NOT deadenylase complex is a highly conserved regulator of mRNA metabolism that influences the expression of the complete transcriptome, representing a prime target for a generalist bacterial pathogen. We show that a translocated bacterial effector protein, PieF (Lpg1972) of Legionella pneumophila, directly interacts with the CNOT7/8 nuclease module of CCR4-NOT, with a dissociation constant in the low nanomolar range. PieF is a robust in vitro inhibitor of the DEDD-type nuclease, CNOT7, acting in a stoichiometric, dose-dependent manner. Heterologous expression of PieF phenocopies knockout of the CNOT7 ortholog (POP2) in Saccharomyces cerevisiae, resulting in 6-azauracil sensitivity. In mammalian cells, expression of PieF leads to a variety of quantifiable phenotypes: PieF silences gene expression and reduces mRNA steady-state levels when artificially tethered to a reporter transcript, and its overexpression results in the nuclear exclusion of CNOT7. PieF expression also disrupts the association between CNOT6/6L EEP-type nucleases and CNOT7. Adding to the complexities of PieF activity in vivo, we identified a separate domain of PieF responsible for binding to eukaryotic kinases. Unlike what we observe for CNOT6/6L, we show that these interactions can occur concomitantly with PieF's binding to CNOT7. Collectively, this work reveals a new, highly conserved target of L. pneumophila effectors and suggests a mechanism by which the pathogen may be modulating host mRNA stability and expression during infection. IMPORTANCE The intracellular bacterial pathogen Legionella pneumophila targets conserved eukaryotic pathways to establish a replicative niche inside host cells. With a host range that spans billions of years of evolution (from protists to humans), the interaction between L. pneumophila and its hosts frequently involves conserved eukaryotic pathways (protein translation, ubiquitination, membrane trafficking, autophagy, and the cytoskeleton). Here, we present the identification of a new, highly conserved host target of L. pneumophila effectors: the CCR4-NOT complex. CCR4-NOT modulates mRNA stability in eukaryotes from yeast to humans, making it an attractive target for a generalist pathogen, such as L. pneumophila. We show that the uncharacterized L. pneumophila effector PieF specifically targets one component of this complex, the deadenylase subunit CNOT7/8. We show that the interaction between PieF and CNOT7 is direct, occurs with high affinity, and reshapes the catalytic activity, localization, and composition of the complex across evolutionarily diverse eukaryotic cells.
Collapse
Affiliation(s)
| | - Malene L. Urbanus
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Francesco Zangari
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Anne-Claude Gingras
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Alexander W. Ensminger
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Pichiri G, Piludu M, Congiu T, Grandi N, Coni P, Piras M, Jaremko M, Lachowicz JI. Kojic Acid Derivative as an Antimitotic Agent That Selectively Kills Tumour Cells. Pharmaceuticals (Basel) 2024; 18:11. [PMID: 39861074 PMCID: PMC11768441 DOI: 10.3390/ph18010011] [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: 10/08/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: The primary method used to pharmacologically arrest cancer development and its metastasis is to disrupt the cell division process. There are a few approaches that may be used to meet this objective, mainly through inhibiting DNA replication or mitosis. Despite intensive studies on new chemotherapeutics, the biggest problem remains the side effects associated with the inhibition of cell division in non-tumoural host cells. Methods: The efficacy and selectivity of the kojic acid derivative (L1) was studied in vitro with the use of tumoural (Caco2, SW480, HT29, T98G) and non- tumoural (HEK293T, RAW) cell lines. Light and electron microscopy observations were supported by the next generation sequencing (NGS), cytoflow, and spectroscopy analysis of mRNA and biomolecules, respectively. Results: The light and electron microscopy observations showed that L1 treatment leads to significant morphological changes in Caco2 cells, which are characteristic of mitosis arrest. Moreover, the fluorescent tubulin staining revealed the formation of tubulin ring structure associated with the apoptotic stage. Mitotic exit into apoptosis was further conformed by the cytoflow of early/late apoptosis stages and caspase-3 analysis. NGS investigation showed differentiated expressions of genes involved in mitosis and apoptosis processes. The observed IC50 in tumoural cell lines were as follows: Caco2 (IC50 = 68.2 mM), SW480 (IC50 = 15.5 mM), and HT29 (IC50 = 4.7 mM). Conclusions: The findings presented here suggest that L1 could be a valid candidate for oral prevention and/or chemotherapy in colorectal cancer. Considering high selectivity of L1 versus tumoural cell lines, more in-depth mechanistic studies could reveal unknown stages in carcinogenesis.
Collapse
Affiliation(s)
- Giuseppina Pichiri
- Department of Medical Sciences and Public Health, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (G.P.); (T.C.); (P.C.); (M.P.)
| | - Marco Piludu
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy;
| | - Terenzio Congiu
- Department of Medical Sciences and Public Health, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (G.P.); (T.C.); (P.C.); (M.P.)
| | - Nicole Grandi
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy;
| | - Pierpaolo Coni
- Department of Medical Sciences and Public Health, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (G.P.); (T.C.); (P.C.); (M.P.)
| | - Monica Piras
- Department of Medical Sciences and Public Health, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (G.P.); (T.C.); (P.C.); (M.P.)
| | - Mariusz Jaremko
- Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia;
| | - Joanna Izabela Lachowicz
- Department of Medical Sciences and Public Health, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (G.P.); (T.C.); (P.C.); (M.P.)
- Department of Population Health, Division of Environmental Health, Occupational Medicine and Epidemiology, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368 Wroclaw, Poland
| |
Collapse
|
5
|
Jia L, Meng Q, Xu X. Autophagy-related miRNAs, exosomal miRNAs, and circRNAs in tumor progression and drug-and radiation resistance in colorectal cancer. Pathol Res Pract 2024; 263:155597. [PMID: 39426141 DOI: 10.1016/j.prp.2024.155597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 07/16/2024] [Accepted: 09/20/2024] [Indexed: 10/21/2024]
Abstract
Targeted therapies are often more tolerable than traditional cytotoxic ones. Nurses play a critical role in providing patients and caregivers with information about the disease, available therapies, and the kind, severity, and identification of any potential adverse events. By doing this, it may be possible to ensure that any adverse effects are managed quickly, maximizing the therapeutic benefit. In colorectal cancer (CRC), autophagy-related activities are significantly influenced by miRNAs and exosomal miRNAs. CRC development and treatment resistance have been associated with the cellular process of autophagy. miRNAs, which are short non-coding RNA molecules, have the ability to control the expression of genes by binding to the 3' untranslated region (UTR) of target mRNAs and either preventing or suppressing translation. It has been discovered that several miRNAs are significant regulators of CRC autophagy. By preventing autophagy, these miRNAs enhance the survival and growth of cancer cells. Exosomes are small membrane vesicles that are released by cells and include miRNAs among other bioactive compounds. Exosomes have the ability to modify recipient cells' biological processes by delivering their cargo, which includes miRNAs. It has been demonstrated that exosomal miRNAs control autophagy in CRC in both autocrine and paracrine ways. We will discuss the potential roles of miRNAs, exosomal miRNAs, and circRNAs in CRC autophagy processes and how nursing care can reduce unfavorable outcomes.
Collapse
Affiliation(s)
- Liting Jia
- Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing 102413, China
| | - Qingyun Meng
- Gastroenterology Department, Qingdao Municipal Hospital, Qingdao 266000, China
| | - Xiaofeng Xu
- Thoracic Surgery, Qingdao Municipal Hospital, Qingdao 266000, China.
| |
Collapse
|
6
|
Xia CH, Lin W, Li R, Xing X, Shang GJ, Zhang H, Gong X. Altered Cell Clusters and Upregulated Aqp1 in Connexin 50 Knockout Lens Epithelium. Invest Ophthalmol Vis Sci 2024; 65:27. [PMID: 39287589 PMCID: PMC11412383 DOI: 10.1167/iovs.65.11.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Purpose To characterize the heterogeneity and cell clusters of postnatal lens epithelial cells (LECs) and to investigate the downstream targets of connexin 50 (Cx50) in the regulation of lens homeostasis and lens growth. To determine differentially expressed genes (DEGs) in the connexin 50 knockout (Cx50KO) lens epithelial cells that shed light on novel mechanism underlying the cataract and small size of the Cx50KO lenses. Methods Single-cell RNA sequencing (scRNA-seq) of lens epithelial cells isolated from one-month-old Cx50KO and wild-type (WT) mice were performed. Differentially expressed genes were identified, and selected DEGs were further studied by quantitative real-time PCR (RT-qPCR) analysis and Western blot analysis. Results The expression profiles of several thousand genes were identified by scRNA-seq data analysis. In comparison to the WT control, many DEGs were identified in the Cx50KO lens epithelial cells, including growth regulating transcriptional factors and genes encoding water channels. Significantly upregulated aquaporin 1 (Aqp1) gene expression was confirmed by RT-qPCR, and upregulated AQP1 protein expression was confirmed by Western blot analysis and immunostaining both in vivo and in vitro. Conclusions Lens epithelial cells exhibit an intrinsic heterogeneity of different cell clusters in regulating lens homeostasis and lens growth. Upregulated Aqp1 in Cx50KO lens epithelial cells suggests that both connexin 50 and AQP1 likely play important roles in regulating water homeostasis in lens epithelial cells.
Collapse
Affiliation(s)
- Chun-Hong Xia
- Herbert Wertheim School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, California, United States
| | - William Lin
- Herbert Wertheim School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, California, United States
| | - Rachel Li
- Herbert Wertheim School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, California, United States
| | - Xinfang Xing
- Herbert Wertheim School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, California, United States
| | - Guangdu Jack Shang
- Herbert Wertheim School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, California, United States
| | - Haiwei Zhang
- Herbert Wertheim School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, California, United States
| | - Xiaohua Gong
- Herbert Wertheim School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, California, United States
| |
Collapse
|
7
|
Lee SM, Yoon BH, Lee JW, Jeong IJY, Kim I, Pack CG, Kim YH, Ha CH. Circulating miRNA-4701-3p as a predictive biomarker of cardiovascular disease which induces angiogenesis by inhibition of TOB2. Microvasc Res 2024; 155:104698. [PMID: 38801943 DOI: 10.1016/j.mvr.2024.104698] [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: 08/24/2023] [Revised: 05/07/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
Angiogenesis is mainly regulated by the delivery of VEGF-dependent signaling to cells. However, the angiogenesis mechanism regulated by VEGF-induced miRNA is still not understood. After VEGF treatment in HUVECs, we screened the changed miRNAs through small-RNA sequencing and found VEGF-induced miR-4701-3p. Furthermore, the GFP reporter gene was used to reveal that TOB2 expression was regulated by miR-4701-3p, and it was found that TOB2 and miR-4701-3p modulation could cause angiogenesis in an in-vitro angiogenic assay. Through the luciferase assay, it was confirmed that the activation of the angiogenic transcription factor MEF2 was regulated by the suppression and overexpression of TOB2 and miR-4701-3p. As a result, MEF2 downstream gene mRNAs that induce angiogenic function were regulated. We used the NCBI GEO datasets to reveal that the expression of TOB2 and MEF2 was significantly changed in cardiovascular disease. Finally, it was confirmed that the expression of circulating miR-4701-3p in the blood of myocardial infarction patients was remarkably increased. In patients with myocardial infarction, circulating miR-4701-3p was increased regardless of age, BMI, and sex, and showed high AUC levels in specificity and sensitivity analysis (AUROC) (AUC = 0.8451, 95 % CI 0.78-0.90). Our data showed TOB2-mediated modulation of MEF2 and its angiogenesis by VEGF-induced miR-4701-3p in vascular endothelial cells. In addition, through bioinformatics analysis using GEO data, changes in TOB2 and MEF2 were revealed in cardiovascular disease. We suggest that circulating miR-4701-3p has high potential as a biomarker for myocardial infarction.
Collapse
Affiliation(s)
- Seung Min Lee
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Bo Hyun Yoon
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin Woo Lee
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - I Jin-Yong Jeong
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Inki Kim
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; ConveRgence mEDIcine research cenTer (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Chan-Gi Pack
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; ConveRgence mEDIcine research cenTer (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Young-Hak Kim
- Cardiology Division, Asan Medical Center and University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Chang Hoon Ha
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Liu M, Jiang H, Momeni MR. Epigenetic regulation of autophagy by non-coding RNAs and exosomal non-coding RNAs in colorectal cancer: A narrative review. Int J Biol Macromol 2024; 273:132732. [PMID: 38823748 DOI: 10.1016/j.ijbiomac.2024.132732] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/16/2024] [Accepted: 05/19/2024] [Indexed: 06/03/2024]
Abstract
One of the major diseases affecting people globally is colorectal cancer (CRC), which is primarily caused by a lack of effective medical treatment and a limited understanding of its underlying mechanisms. Cellular autophagy functions to break down and eliminate superfluous proteins and substances, thereby facilitating the continual replacement of cellular elements and generating vital energy for cell processes. Non-coding RNAs and exosomal ncRNAs have a crucial impact on regulating gene expression and essential cellular functions such as autophagy, metastasis, and treatment resistance. The latest research has indicated that specific ncRNAs and exosomal ncRNA to influence the process of autophagy in CRC cells, which could have significant consequences for the advancement and treatment of this disease. It has been determined that a variety of ncRNAs have a vital function in regulating the genes essential for the formation and maturation of autophagosomes. Furthermore, it has been confirmed that ncRNAs have a considerable influence on the signaling pathways associated with autophagy, such as those involving AMPK, AKT, and mTOR. Additionally, numerous ncRNAs have the potential to affect specific genes involved in autophagy. This study delves into the control mechanisms of ncRNAs and exosomal ncRNAs and examines how they simultaneously influence autophagy in CRC.
Collapse
Affiliation(s)
- Minghua Liu
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning, China
| | - Hongfang Jiang
- Department of Geriatrics, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning, China.
| | - Mohammad Reza Momeni
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States.
| |
Collapse
|
9
|
Wang M, He B, Hao Y, Srinivasan D, Shrinet J, Fraser P. Cellular reprogramming is driven by widespread rewiring of promoter-enhancer interactions. BMC Biol 2023; 21:264. [PMID: 37981682 PMCID: PMC10658794 DOI: 10.1186/s12915-023-01766-0] [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: 04/17/2023] [Accepted: 11/09/2023] [Indexed: 11/21/2023] Open
Abstract
BACKGROUND Long-range interactions between promoters and cis-regulatory elements, such as enhancers, play critical roles in gene regulation. However, the role of three-dimensional (3D) chromatin structure in orchestrating changes in transcriptional regulation during direct cell reprogramming is not fully understood. RESULTS Here, we performed integrated analyses of chromosomal architecture, epigenetics, and gene expression using Hi-C, promoter Capture Hi-C (PCHi-C), ChIP-seq, and RNA-seq during trans-differentiation of Pre-B cells into macrophages with a β-estradiol inducible C/EBPαER transgene. Within 1h of β-estradiol induction, C/EBPα translocated from the cytoplasm to the nucleus, binding to thousands of promoters and putative regulatory elements, resulting in the downregulation of Pre-B cell-specific genes and induction of macrophage-specific genes. Hi-C results were remarkably consistent throughout trans-differentiation, revealing only a small number of TAD boundary location changes, and A/B compartment switches despite significant changes in the expression of thousands of genes. PCHi-C revealed widespread changes in promoter-anchored loops with decreased interactions in parallel with decreased gene expression, and new and increased promoter-anchored interactions in parallel with increased expression of macrophage-specific genes. CONCLUSIONS Overall, our data demonstrate that C/EBPα-induced trans-differentiation involves few changes in genome architecture at the level of TADs and A/B compartments, in contrast with widespread reorganization of thousands of promoter-anchored loops in association with changes in gene expression and cell identity.
Collapse
Affiliation(s)
- Miao Wang
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Bing He
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Yueling Hao
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Divyaa Srinivasan
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Jatin Shrinet
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Peter Fraser
- Department of Biological Science, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
10
|
Sulic AM, Das Roy R, Papagno V, Lan Q, Saikkonen R, Jernvall J, Thesleff I, Mikkola ML. Transcriptomic landscape of early hair follicle and epidermal development. Cell Rep 2023; 42:112643. [PMID: 37318953 DOI: 10.1016/j.celrep.2023.112643] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 04/04/2023] [Accepted: 05/29/2023] [Indexed: 06/17/2023] Open
Abstract
Morphogenesis of ectodermal organs, such as hair, tooth, and mammary gland, starts with the formation of local epithelial thickenings, or placodes, but it remains to be determined how distinct cell types and differentiation programs are established during ontogeny. Here, we use bulk and single-cell transcriptomics and pseudotime modeling to address these questions in developing hair follicles and epidermis and produce a comprehensive transcriptomic profile of cellular populations in the hair placode and interplacodal epithelium. We report previously unknown cell populations and marker genes, including early suprabasal and genuine interfollicular basal markers, and propose the identity of suprabasal progenitors. By uncovering four different hair placode cell populations organized in three spatially distinct areas, with fine gene expression gradients between them, we posit early biases in cell fate establishment. This work is accompanied by a readily accessible online tool to stimulate further research on skin appendages and their progenitors.
Collapse
Affiliation(s)
- Ana-Marija Sulic
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Rishi Das Roy
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Verdiana Papagno
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Qiang Lan
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Riikka Saikkonen
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Jukka Jernvall
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland; Department of Geosciences and Geography, University of Helsinki, 00014 Helsinki, Finland
| | - Irma Thesleff
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Marja L Mikkola
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland.
| |
Collapse
|
11
|
Velayutham N, Calderon MU, Alfieri CM, Padula SL, van Leeuwen FN, Scheijen B, Yutzey KE. Btg1 and Btg2 regulate neonatal cardiomyocyte cell cycle arrest. J Mol Cell Cardiol 2023; 179:30-41. [PMID: 37062247 PMCID: PMC10192094 DOI: 10.1016/j.yjmcc.2023.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 03/21/2023] [Accepted: 03/29/2023] [Indexed: 04/18/2023]
Abstract
Rodent cardiomyocytes undergo mitotic arrest in the first postnatal week. Here, we investigate the role of transcriptional co-regulator Btg2 (B-cell translocation gene 2) and functionally-similar homolog Btg1 in postnatal cardiomyocyte cell cycling and maturation. Btg1 and Btg2 (Btg1/2) are expressed in neonatal C57BL/6 mouse left ventricles coincident with cardiomyocyte cell cycle arrest. Btg1/2 constitutive double knockout (DKO) mouse hearts exhibit increased pHH3+ mitotic cardiomyocytes compared to Wildtype at postnatal day (P)7, but not at P30. Similarly, neonatal AAV9-mediated Btg1/2 double knockdown (DKD) mouse hearts exhibit increased EdU+ mitotic cardiomyocytes compared to Scramble AAV9-shRNA controls at P7, but not at P14. In neonatal rat ventricular myocyte (NRVM) cultures, siRNA-mediated Btg1/2 single and double knockdown cohorts showed increased EdU+ cardiomyocytes compared to Scramble siRNA controls, without increase in binucleation or nuclear DNA content. RNAseq analyses of Btg1/2-depleted NRVMs support a role for Btg1/2 in inhibiting cell proliferation, and in modulating reactive oxygen species response pathways, implicated in neonatal cardiomyocyte cell cycle arrest. Together, these data identify Btg1 and Btg2 as novel contributing factors in mammalian cardiomyocyte cell cycle arrest after birth.
Collapse
Affiliation(s)
- Nivedhitha Velayutham
- Molecular and Developmental Biology Graduate Program, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA; The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Maria Uscategui Calderon
- Molecular and Developmental Biology Graduate Program, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA; The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Christina M Alfieri
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Stephanie L Padula
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | | | - Katherine E Yutzey
- Molecular and Developmental Biology Graduate Program, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA; The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
12
|
Wang S, Xu J, Zhao X, Feng Y, Xu W, Xue H, Wu M, Xu L. Small RNA-seq and hormones in the testes of dwarf hamsters ( Cricetulus barabensis) reveal the potential pathways in photoperiod regulated reproduction. Heliyon 2023; 9:e15687. [PMID: 37144180 PMCID: PMC10151367 DOI: 10.1016/j.heliyon.2023.e15687] [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: 05/07/2022] [Revised: 03/26/2023] [Accepted: 04/18/2023] [Indexed: 05/06/2023] Open
Abstract
Photoperiod regulates the functions and development of gonadal organs of seasonally breeding animals, resulting in breeding peaks in specific seasons. miRNA plays an important role in the regulation of testicular physiological functions. However, the relationship between photoperiods and miRNA levels in testes has yet to be conclusively determined. We investigated testicular miRNA of striped dwarf hamster (Cricetulus barabensis) responses to different photoperiods (long daylength [LD], moderate daylength [MD], and short daylength [SD]) and the potential pathways involved in photoperiod regulated reproduction. Testicular weights and reproductive hormone levels were measured in each of photoperiod treatments after 30 days. The concentrations of testosterone (T) and dihydrogen testosterone (DHT) in testes and Gonadotropin-releasing hormone (GnRH), follicle-stimulating hormone (FSH), and luteinizing hormone (LH) in serum were higher in MD than in the other two groups. Testicular weights were heaviest in MD. Small RNA-seq was performed for the testes of hamsters in three groups. A total of 769 miRNAs were identified, of which 83 were differentially expressed between LD, MD, and SD. GO and KEGG analysis of target genes revealed that some miRNAs influence testicular activities by regulating the pathways related to cell apoptosis and metabolism. Gene expression pattern analysis showed that the MAPK signaling pathway may be the core pathway for photoperiodic regulation of reproduction. These results suggest that moderate daylength is more suitable for hamster reproduction while long daylength and short daylength may regulate reproduction through different molecular pathways.
Collapse
|
13
|
Zhang X, Zhai Y, Zhang D, Che C, Zhang Y, Li Q, Zhang X, Zhao L. RNAseq analysis of the drug jian-yan-ling (JYL) using both in vivo and in vitro models. Heliyon 2023; 9:e16143. [PMID: 37251843 PMCID: PMC10213199 DOI: 10.1016/j.heliyon.2023.e16143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 04/21/2023] [Accepted: 05/06/2023] [Indexed: 05/31/2023] Open
Abstract
Ethnopharmacological relevance Jian-yan-ling (JYL) is a drug used in traditional Chinese medicine (TCM) prescriptions for the treatment of tumors after radiotherapy and chemotherapy, to effectively alleviate leukocytopenia. However, the genetic mechanisms underlying the function of JYL remain unclear. Aim of the study This study aimed to explore the RNA changes and potential biological processes related to the anti-aging or life-extending effects of JYL treatments. Materials and methods In vivo treatments were performed using Canton-S Drosophila corresponding to three groups: control, low-concentration (low-conc.), and high-concentration (high-conc.) groups. The low-conc. And the high-conc. Groups were treated with 4 mg/mL JYL and 8 mg/mL JYL, respectively. Thirty Drosophila eggs were placed in each vial, and the third instar larvae and adults 7 and 21 days post-eclosion were collected for RNA sequencing, irrespective of the gender.In vitro treatments were conducted using humanized immune cell lines HL60 and Jurkat, which were divided into 3 groups: control (0 μg/mL JYL), low-concentration (40 μg/mL JYL), and high-concentration (80 μg/mL JYL). The cells were collected after 48 h of each JYL drug treatment. Both the Drosophila and cell samples were analyzed using RNA sequencing. Results The in vivo experiments revealed 74 upregulated genes in the low-concentration group, and CG13078 was a commonly downregulated differential gene, which is involved in ascorbate iron reductase activity. Further analysis of the co-expression map identified the key genes: regulatory particle non-ATPase (RPN), regulatory particle triple-A ATPase (RPT), and tripeptidyl-peptidase II (TPP II). For the in vitro experiments, 19 co-differential genes were compared between different concentrations of the HL 60 cell line, of which three genes were upregulated: LOC107987457 (phostensin-like gene), HSPA1A (heat shock protein family A member 1 A), and H2AC19 (H2A clustered histone 19). In the HL 60 cell line, JYL activated proteasome-related functions. In the Jurkat cell line, there were no common differential genes despite the presence of a dosage-dependent trend. Conclusions The RNA-seq results showed that the traditional Chinese medicine JYL has longevity and anti-aging effects, indicating that further investigation is required.
Collapse
Affiliation(s)
- Xiaobo Zhang
- School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yunliang Zhai
- Lei Yun Shang Pharmaceutical Group Co.,Ltd., Suzhou, 215009, China
| | - Dandan Zhang
- Lei Yun Shang Pharmaceutical Group Co.,Ltd., Suzhou, 215009, China
| | - Chang Che
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, China
| | - Yayun Zhang
- Lei Yun Shang Pharmaceutical Group Co.,Ltd., Suzhou, 215009, China
| | - Quan Li
- Lei Yun Shang Pharmaceutical Group Co.,Ltd., Suzhou, 215009, China
| | - Xue Zhang
- Lei Yun Shang Pharmaceutical Group Co.,Ltd., Suzhou, 215009, China
| | - Lingrui Zhao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, China
| |
Collapse
|
14
|
Chowdhury RR, Valainis JR, Dubey M, von Boehmer L, Sola E, Wilhelmy J, Guo J, Kask O, Ohanyan M, Sun M, Huang H, Huang X, Nguyen PK, Scriba TJ, Davis MM, Bendall SC, Chien YH. NK-like CD8 + γδ T cells are expanded in persistent Mycobacterium tuberculosis infection. Sci Immunol 2023; 8:eade3525. [PMID: 37000856 PMCID: PMC10408713 DOI: 10.1126/sciimmunol.ade3525] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 03/09/2023] [Indexed: 04/03/2023]
Abstract
The response of gamma delta (γδ) T cells in the acute versus chronic phases of the same infection is unclear. How γδ T cells function in acute Mycobacterium tuberculosis (Mtb) infection is well characterized, but their response during persistent Mtb infection is not well understood, even though most infections with Mtb manifest as a chronic, clinically asymptomatic state. Here, we analyze peripheral blood γδ T cells from a South African adolescent cohort and show that a unique CD8+ γδ T cell subset with features of "memory inflation" expands in chronic Mtb infection. These cells are hyporesponsive to T cell receptor (TCR)-mediated signaling but, like NK cells, can mount robust CD16-mediated cytotoxic responses. These CD8+ γδ T cells comprise a highly focused TCR repertoire, with clonotypes that are Mycobacterium specific but not phosphoantigen reactive. Using multiparametric single-cell pseudo-time trajectory analysis, we identified the differentiation paths that these CD8+ γδ T cells follow to develop into effectors in this infection state. Last, we found that circulating CD8+ γδ T cells also expand in other chronic inflammatory conditions, including cardiovascular disease and cancer, suggesting that persistent antigenic exposure may drive similar γδ T cell effector programs and differentiation fates.
Collapse
Affiliation(s)
- Roshni Roy Chowdhury
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Program in Immunology, Stanford University, Stanford, CA, USA
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, IL, USA
| | | | - Megha Dubey
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Lotta von Boehmer
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, USA
| | - Elsa Sola
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, USA
| | - Julie Wilhelmy
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, USA
| | - Jing Guo
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Oliver Kask
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Mane Ohanyan
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Meng Sun
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, USA
| | - Huang Huang
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, USA
| | - Xianxi Huang
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
- The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Patricia K. Nguyen
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Mark M. Davis
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Program in Immunology, Stanford University, Stanford, CA, USA
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, USA
- The Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Sean C. Bendall
- Program in Immunology, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Yueh-hsiu Chien
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Program in Immunology, Stanford University, Stanford, CA, USA
| |
Collapse
|
15
|
Delage L, Lambert M, Bardel É, Kundlacz C, Chartoire D, Conchon A, Peugnet AL, Gorka L, Auberger P, Jacquel A, Soussain C, Destaing O, Delecluse HJ, Delecluse S, Merabet S, Traverse-Glehen A, Salles G, Bachy E, Billaud M, Ghesquières H, Genestier L, Rouault JP, Sujobert P. BTG1 inactivation drives lymphomagenesis and promotes lymphoma dissemination through activation of BCAR1. Blood 2023; 141:1209-1220. [PMID: 36375119 DOI: 10.1182/blood.2022016943] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 10/11/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022] Open
Abstract
Understanding the functional role of mutated genes in cancer is required to translate the findings of cancer genomics into therapeutic improvement. BTG1 is recurrently mutated in the MCD/C5 subtype of diffuse large B-cell lymphoma (DLBCL), which is associated with extranodal dissemination. Here, we provide evidence that Btg1 knock out accelerates the development of a lethal lymphoproliferative disease driven by Bcl2 overexpression. Furthermore, we show that the scaffolding protein BCAR1 is a BTG1 partner. Moreover, after BTG1 deletion or expression of BTG1 mutations observed in patients with DLBCL, the overactivation of the BCAR1-RAC1 pathway confers increased migration ability in vitro and in vivo. These modifications are targetable with the SRC inhibitor dasatinib, which opens novel therapeutic opportunities in BTG1 mutated DLBCL.
Collapse
Affiliation(s)
- Lorric Delage
- Centre International de Recherche en Infectiologie (Team LIB), Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
| | - Mireille Lambert
- Université de Paris, Institut Cochin, INSERM U1016, Plateforme BioMecan'IC, Biomécanique de la cellule, Paris, France
| | - Émilie Bardel
- Centre International de Recherche en Infectiologie (Team LIB), Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
| | - Cindy Kundlacz
- Institut de Génomique Fonctionnelle de Lyon, Centre National de la Recherche Scientifique UMR5242, Université Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Dimitri Chartoire
- Centre International de Recherche en Infectiologie (Team LIB), Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
| | - Axel Conchon
- Centre International de Recherche en Infectiologie (Team LIB), Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
| | - Anne-Laure Peugnet
- Centre International de Recherche en Infectiologie (Team LIB), Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
| | - Lucas Gorka
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
| | - Patrick Auberger
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM U1065, Nice, France
| | - Arnaud Jacquel
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM U1065, Nice, France
| | - Carole Soussain
- Institut Curie, Site de Saint-Cloud, Hematologie, et INSERM U932 Institut Curie, PSL Research University, Paris, France
| | - Olivier Destaing
- Centre de Recherche UGA, INSERM U1209, Institute for Advanced Biosciences, Grenoble, France
| | | | | | - Samir Merabet
- Institut de Génomique Fonctionnelle de Lyon, Centre National de la Recherche Scientifique UMR5242, Université Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Alexandra Traverse-Glehen
- Centre International de Recherche en Infectiologie (Team LIB), Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
| | - Gilles Salles
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Emmanuel Bachy
- Centre International de Recherche en Infectiologie (Team LIB), Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
| | - Marc Billaud
- INSERM Unité Mixte de Recherche (UMR)-U1052, Centre National de la Recherche UMR 5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Hervé Ghesquières
- Centre International de Recherche en Infectiologie (Team LIB), Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
| | - Laurent Genestier
- Centre International de Recherche en Infectiologie (Team LIB), Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
| | - Jean-Pierre Rouault
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
- INSERM Unité Mixte de Recherche (UMR)-U1052, Centre National de la Recherche UMR 5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Pierre Sujobert
- Centre International de Recherche en Infectiologie (Team LIB), Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
| |
Collapse
|
16
|
Mlynarczyk C, Teater M, Pae J, Chin CR, Wang L, Arulraj T, Barisic D, Papin A, Hoehn KB, Kots E, Ersching J, Bandyopadhyay A, Barin E, Poh HX, Evans CM, Chadburn A, Chen Z, Shen H, Isles HM, Pelzer B, Tsialta I, Doane AS, Geng H, Rehman MH, Melnick J, Morgan W, Nguyen DTT, Elemento O, Kharas MG, Jaffrey SR, Scott DW, Khelashvili G, Meyer-Hermann M, Victora GD, Melnick A. BTG1 mutation yields supercompetitive B cells primed for malignant transformation. Science 2023; 379:eabj7412. [PMID: 36656933 PMCID: PMC10515739 DOI: 10.1126/science.abj7412] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 12/12/2022] [Indexed: 01/21/2023]
Abstract
Multicellular life requires altruistic cooperation between cells. The adaptive immune system is a notable exception, wherein germinal center B cells compete vigorously for limiting positive selection signals. Studying primary human lymphomas and developing new mouse models, we found that mutations affecting BTG1 disrupt a critical immune gatekeeper mechanism that strictly limits B cell fitness during antibody affinity maturation. This mechanism converted germinal center B cells into supercompetitors that rapidly outstrip their normal counterparts. This effect was conferred by a small shift in MYC protein induction kinetics but resulted in aggressive invasive lymphomas, which in humans are linked to dire clinical outcomes. Our findings reveal a delicate evolutionary trade-off between natural selection of B cells to provide immunity and potentially dangerous features that recall the more competitive nature of unicellular organisms.
Collapse
Affiliation(s)
- Coraline Mlynarczyk
- Division of Hematology and Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Matt Teater
- Division of Hematology and Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Juhee Pae
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Christopher R. Chin
- Division of Hematology and Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional PhD Program in Computational Biomedicine, New York, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Ling Wang
- Division of Hematology and Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Theinmozhi Arulraj
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology (BRICS), Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Darko Barisic
- Division of Hematology and Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Antonin Papin
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Kenneth B. Hoehn
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Ekaterina Kots
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Jonatan Ersching
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Arnab Bandyopadhyay
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology (BRICS), Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ersilia Barin
- Department of Pharmacology and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Hui Xian Poh
- Department of Pharmacology and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Chiara M. Evans
- Molecular Pharmacology Program and Center for Cell Engineering, Center for Stem Cell Biology, Center for Experimental Therapeutics, and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Amy Chadburn
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Zhengming Chen
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Hao Shen
- Division of Hematology and Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Hannah M. Isles
- Division of Hematology and Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Benedikt Pelzer
- Division of Hematology and Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Ioanna Tsialta
- Division of Hematology and Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Ashley S. Doane
- Division of Hematology and Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Huimin Geng
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Muhammad Hassan Rehman
- Division of Hematology and Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell Medicine–Qatar, Doha, Qatar
| | - Jonah Melnick
- Division of Hematology and Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Wyatt Morgan
- Division of Hematology and Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Diu T. T. Nguyen
- Molecular Pharmacology Program and Center for Cell Engineering, Center for Stem Cell Biology, Center for Experimental Therapeutics, and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Olivier Elemento
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine and Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Michael G. Kharas
- Molecular Pharmacology Program and Center for Cell Engineering, Center for Stem Cell Biology, Center for Experimental Therapeutics, and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samie R. Jaffrey
- Department of Pharmacology and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - David W. Scott
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
| | - George Khelashvili
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology (BRICS), Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Gabriel D. Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Ari Melnick
- Division of Hematology and Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
17
|
Grigoryan A, Zacharaki D, Balhuizen A, Côme CR, Garcia AG, Hidalgo Gil D, Frank AK, Aaltonen K, Mañas A, Esfandyari J, Kjellman P, Englund E, Rodriguez C, Sime W, Massoumi R, Kalantari N, Prithiviraj S, Li Y, Dupard SJ, Isaksson H, Madsen CD, Porse BT, Bexell D, Bourgine PE. Engineering human mini-bones for the standardized modeling of healthy hematopoiesis, leukemia, and solid tumor metastasis. Sci Transl Med 2022; 14:eabm6391. [PMID: 36223446 DOI: 10.1126/scitranslmed.abm6391] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The bone marrow microenvironment provides indispensable factors to sustain blood production throughout life. It is also a hotspot for the progression of hematologic disorders and the most frequent site of solid tumor metastasis. Preclinical research relies on xenograft mouse models, but these models preclude the human-specific functional interactions of stem cells with their bone marrow microenvironment. Instead, human mesenchymal cells can be exploited for the in vivo engineering of humanized niches, which confer robust engraftment of human healthy and malignant blood samples. However, mesenchymal cells are associated with major reproducibility issues in tissue formation. Here, we report the fast and standardized generation of human mini-bones by a custom-designed human mesenchymal cell line. These resulting humanized ossicles (hOss) consist of fully mature bone and bone marrow structures hosting a human mesenchymal niche with retained stem cell properties. As compared to mouse bones, we demonstrate superior engraftment of human cord blood hematopoietic cells and primary acute myeloid leukemia samples and also validate hOss as a metastatic site for breast cancer cells. We further report the engraftment of neuroblastoma patient-derived xenograft cells in a humanized model, recapitulating clinically described osteolytic lesions. Collectively, our human mini-bones constitute a powerful preclinical platform to model bone-developing tumors using patient-derived materials.
Collapse
Affiliation(s)
- Ani Grigoryan
- Cell, Tissue & Organ engineering laboratory, Biomedical Centre (BMC) B11, Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Dimitra Zacharaki
- Cell, Tissue & Organ engineering laboratory, Biomedical Centre (BMC) B11, Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Alexander Balhuizen
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.,Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark.,Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christophe Rm Côme
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.,Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark.,Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Alejandro Garcia Garcia
- Cell, Tissue & Organ engineering laboratory, Biomedical Centre (BMC) B11, Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - David Hidalgo Gil
- Cell, Tissue & Organ engineering laboratory, Biomedical Centre (BMC) B11, Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Anne-Katrine Frank
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.,Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark.,Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kristina Aaltonen
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Adriana Mañas
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Javanshir Esfandyari
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Pontus Kjellman
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Emelie Englund
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Carmen Rodriguez
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Wondossen Sime
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Ramin Massoumi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Nasim Kalantari
- Cell, Tissue & Organ engineering laboratory, Biomedical Centre (BMC) B11, Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Sujeethkumar Prithiviraj
- Cell, Tissue & Organ engineering laboratory, Biomedical Centre (BMC) B11, Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Yuan Li
- Cell, Tissue & Organ engineering laboratory, Biomedical Centre (BMC) B11, Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Steven J Dupard
- Cell, Tissue & Organ engineering laboratory, Biomedical Centre (BMC) B11, Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Hanna Isaksson
- Department of Biomedical Engineering, Lund University, 221 85 Lund, Sweden
| | - Chris D Madsen
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Bo T Porse
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.,Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark.,Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Daniel Bexell
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Paul E Bourgine
- Cell, Tissue & Organ engineering laboratory, Biomedical Centre (BMC) B11, Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| |
Collapse
|
18
|
Kots E, Mlynarczyk C, Melnick A, Khelashvili G. Conformational transitions in BTG1 antiproliferative protein and their modulation by disease mutants. Biophys J 2022; 121:3753-3764. [PMID: 35459639 PMCID: PMC9617077 DOI: 10.1016/j.bpj.2022.04.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/01/2022] [Accepted: 04/15/2022] [Indexed: 12/01/2022] Open
Abstract
B cell translocation gene 1 (BTG1) protein belongs to the BTG/transducer of ERBB2 (TOB) family of antiproliferative proteins whose members regulate various key cellular processes such as cell cycle progression, apoptosis, and differentiation. Somatic missense mutations in BTG1 are found in ∼70% of a particularly malignant and disseminated subtype of diffuse large B cell lymphoma (DLBCL). Antiproliferative activity of BTG1 has been linked to its ability to associate with transcriptional cofactors and various enzymes. However, molecular mechanisms underlying these functional interactions and how the disease-linked mutations in BTG1 affect these mechanisms are currently unknown. To start filling these knowledge gaps, here, using atomistic molecular dynamics (MD) simulations, we explored structural, dynamic, and kinetic characteristics of BTG1 protein, and studied how various DLBCL mutations affect these characteristics. We focused on the protein region formed by α2 and α4 helices, as this interface has been reported not only to serve as a binding hotspot for several cellular partners but also to harbor sites for the majority of known DLBCL mutations. Markov state modeling analysis of extensive MD simulations revealed that the α2-α4 interface in the wild-type (WT) BTG1 undergoes conformational transitions between closed and open metastable states. Importantly, we show that some of the mutations in this region that are observed in DLBCL, such as Q36H, F40C, Q45P, E50K (in α2), and A83T and A84E (in α4), either overstabilize one of these two metastable states or give rise to new conformations in which these helices are distorted (i.e., kinked or unfolded). Based on these results, we conclude that the rapid interconversion between the closed and open conformations of the α2-α4 interface is an essential component of the BTG1 functional dynamics that can prime the protein for functional associations with its binding partners. Disruption of the native dynamic equilibrium by DLBCL mutants leads to the ensemble of conformations in BTG1 that are unlikely structurally and/or kinetically to enable productive functional interactions with the binding proteins.
Collapse
Affiliation(s)
- Ekaterina Kots
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - Coraline Mlynarczyk
- Division of Hematology/Oncology, Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Ari Melnick
- Division of Hematology/Oncology, Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - George Khelashvili
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
19
|
Affiliation(s)
- Sang Hyeon Kim
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Severance Biomedical Science Institute and Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
| | - In Ryeong Jung
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Severance Biomedical Science Institute and Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Soo Seok Hwang
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Severance Biomedical Science Institute and Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
- Chronic Intractable Disease Systems Medicine Research Center, Institute of Genetic Science, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
20
|
Luan SH, Yang YQ, Ye MP, Liu H, Rao QF, Kong JL, Wu FR. ASIC1a promotes hepatic stellate cell activation through the exosomal miR-301a-3p/BTG1 pathway. Int J Biol Macromol 2022; 211:128-139. [PMID: 35561854 DOI: 10.1016/j.ijbiomac.2022.05.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 12/11/2022]
Abstract
Activation of hepatic stellate cells (HSCs) is a key cause of liver fibrosis. However, the mechanisms leading to the activation of HSCs are not fully understood. In the pathological process, acid-sensing ion channel 1a (ASIC1a) is widely involved in the development of inflammatory diseases, suggesting that ASIC1a may play an important role in liver fibrosis. We found that in an acidic environment, ASIC1a leads to HSC-T6 cell activation. Meanwhile, exosomes produced by activated HSC-T6 cells (HSC-EXOs) can be reabsorbed by quiescent HSC-T6 cells to promote their activation. Exosomes mainly carry miRNAs involved in intercellular information exchange. We performed exosome miRNA whole transcriptome sequencing. The results indicated that the acidic environment could alter the miRNA expression profile in the exosomes of HSC-T6 cells. Further studies revealed that ASIC1a promotes the activation of HSCs by regulating miR-301a-3p targeting B-cell translocation gene 1 (BTG1). In conclusion, our study found that ASIC1a may affect HSC activation through the exosomal miR-301a-3p/BTG1 axis, and inhibiting ASIC1a may be a promising treatment strategy for liver fibrosis.
Collapse
Affiliation(s)
- Shao-Hua Luan
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | | | - Man-Ping Ye
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Hui Liu
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Qiu-Fan Rao
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Jin-Ling Kong
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Fan-Rong Wu
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China.
| |
Collapse
|
21
|
Amanda S, Tan TK, Ong JZL, Theardy MS, Wong RWJ, Huang XZ, Ali MZ, Li Y, Gong Z, Inagaki H, Foo EY, Pang B, Tan SY, Iida S, Sanda T. IRF4 drives clonal evolution and lineage choice in a zebrafish model of T-cell lymphoma. Nat Commun 2022; 13:2420. [PMID: 35504924 PMCID: PMC9065160 DOI: 10.1038/s41467-022-30053-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 04/13/2022] [Indexed: 12/15/2022] Open
Abstract
IRF4 is a master regulator of immunity and is also frequently overexpressed in mature lymphoid neoplasms. Here, we demonstrate the oncogenicity of IRF4 in vivo, its potential effects on T-cell development and clonal evolution using a zebrafish model. IRF4-transgenic zebrafish develop aggressive tumors with massive infiltration of abnormal lymphocytes that spread to distal organs. Many late-stage tumors are mono- or oligoclonal, and tumor cells can expand in recipient animals after transplantation, demonstrating their malignancy. Mutation of p53 accelerates tumor onset, increases penetrance, and results in tumor heterogeneity. Surprisingly, single-cell RNA-sequencing reveals that the majority of tumor cells are double-negative T-cells, many of which express tcr-γ that became dominant as the tumors progress, whereas double-positive T-cells are largely diminished. Gene expression and epigenetic profiling demonstrates that gata3, mycb, lrrn1, patl1 and psip1 are specifically activated in tumors, while genes responsible for T-cell differentiation including id3 are repressed. IRF4-driven tumors are sensitive to the BRD inhibitor.
Collapse
Affiliation(s)
- Stella Amanda
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Tze King Tan
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Jolynn Zu Lin Ong
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | | | - Regina Wan Ju Wong
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Muhammad Zulfaqar Ali
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Yan Li
- Department of Biological Sciences, National University of Singapore, 117543, Singapore, Singapore
| | - Zhiyuan Gong
- Department of Biological Sciences, National University of Singapore, 117543, Singapore, Singapore
| | - Hiroshi Inagaki
- Department of Pathology and Molecular Diagnostics, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Ee Yong Foo
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - Brendan Pang
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - Soo Yong Tan
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Takaomi Sanda
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore.
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore.
| |
Collapse
|
22
|
Studniberg SI, Ioannidis LJ, Utami RAS, Trianty L, Liao Y, Abeysekera W, Li‐Wai‐Suen CSN, Pietrzak HM, Healer J, Puspitasari AM, Apriyanti D, Coutrier F, Poespoprodjo JR, Kenangalem E, Andries B, Prayoga P, Sariyanti N, Smyth GK, Cowman AF, Price RN, Noviyanti R, Shi W, Garnham AL, Hansen DS. Molecular profiling reveals features of clinical immunity and immunosuppression in asymptomatic P. falciparum malaria. Mol Syst Biol 2022; 18:e10824. [PMID: 35475529 PMCID: PMC9045086 DOI: 10.15252/msb.202110824] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 01/12/2023] Open
Abstract
Clinical immunity to P. falciparum malaria is non-sterilizing, with adults often experiencing asymptomatic infection. Historically, asymptomatic malaria has been viewed as beneficial and required to help maintain clinical immunity. Emerging views suggest that these infections are detrimental and constitute a parasite reservoir that perpetuates transmission. To define the impact of asymptomatic malaria, we pursued a systems approach integrating antibody responses, mass cytometry, and transcriptional profiling of individuals experiencing symptomatic and asymptomatic P. falciparum infection. Defined populations of classical and atypical memory B cells and a TH2 cell bias were associated with reduced risk of clinical malaria. Despite these protective responses, asymptomatic malaria featured an immunosuppressive transcriptional signature with upregulation of pathways involved in the inhibition of T-cell function, and CTLA-4 as a predicted regulator in these processes. As proof of concept, we demonstrated a role for CTLA-4 in the development of asymptomatic parasitemia in infection models. The results suggest that asymptomatic malaria is not innocuous and might not support the induction of immune processes to fully control parasitemia or efficiently respond to malaria vaccines.
Collapse
Affiliation(s)
- Stephanie I Studniberg
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,Department of Medical BiologyThe University of MelbourneParkvilleVic.Australia
| | - Lisa J Ioannidis
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,Department of Medical BiologyThe University of MelbourneParkvilleVic.Australia
| | - Retno A S Utami
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,Department of Medical BiologyThe University of MelbourneParkvilleVic.Australia,Eijkman Institute for Molecular BiologyJakartaIndonesia
| | - Leily Trianty
- Eijkman Institute for Molecular BiologyJakartaIndonesia
| | - Yang Liao
- Olivia Newton‐John Cancer Research InstituteHeidelbergVic.Australia
| | - Waruni Abeysekera
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,School of Mathematics and StatisticsThe University of MelbourneParkvilleVic.Australia
| | - Connie S N Li‐Wai‐Suen
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,School of Mathematics and StatisticsThe University of MelbourneParkvilleVic.Australia
| | - Halina M Pietrzak
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,Department of Medical BiologyThe University of MelbourneParkvilleVic.Australia
| | - Julie Healer
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,Department of Medical BiologyThe University of MelbourneParkvilleVic.Australia
| | | | - Dwi Apriyanti
- Eijkman Institute for Molecular BiologyJakartaIndonesia
| | | | | | | | | | - Pak Prayoga
- Papuan Health and Community FoundationPapuaIndonesia
| | | | - Gordon K Smyth
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,School of Mathematics and StatisticsThe University of MelbourneParkvilleVic.Australia
| | - Alan F Cowman
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,Department of Medical BiologyThe University of MelbourneParkvilleVic.Australia
| | - Ric N Price
- Global and Tropical Health DivisionMenzies School of Health Research and Charles Darwin UniversityDarwinNTAustralia,Centre for Tropical Medicine and Global HealthNuffield Department of MedicineUniversity of OxfordOxfordUK,Mahidol‐Oxford Tropical Medicine Research UnitMahidol UniversityBangkokThailand
| | | | - Wei Shi
- Olivia Newton‐John Cancer Research InstituteHeidelbergVic.Australia
| | - Alexandra L Garnham
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,School of Mathematics and StatisticsThe University of MelbourneParkvilleVic.Australia
| | - Diana S Hansen
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,Department of Medical BiologyThe University of MelbourneParkvilleVic.Australia
| |
Collapse
|
23
|
Xu Y, Peng Y, Shen M, Liu L, Lei J, Gao S, Wang Y, Lan A, Li H, Liu S. Construction and Validation of Angiogenesis-Related Prognostic Risk Signature to Facilitate Survival Prediction and Biomarker Excavation of Breast Cancer Patients. JOURNAL OF ONCOLOGY 2022; 2022:1525245. [PMID: 35498539 PMCID: PMC9045999 DOI: 10.1155/2022/1525245] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/05/2022] [Indexed: 02/06/2023]
Abstract
This study is aimed at exploring the potential mechanism of angiogenesis, a biological process-related gene in breast cancer (BRCA), and constructing a risk model related to the prognosis of BRCA patients. We used multiple bioinformatics databases and multiple bioinformatics analysis methods to complete our exploration in this research. First, we use the RNA-seq transcriptome data in the TCGA database to conduct a preliminary screening of angiogenesis-related genes through univariate Cox curve analysis and then use LASSO regression curve analysis for secondary screening. We successfully established a risk model consisting of seven angiogenesis-related genes in BRCA. The results of ROC curve analysis show that the risk model has good prediction accuracy. We can successfully divide BRCA patients into the high-risk and low-risk groups with significant prognostic differences based on this risk model. In addition, we used angiogenesis-related genes to perform cluster analysis in BRCA patients and successfully divided BRCA patients into three clusters with significant prognostic differences, namely, cluster 1, cluster 2, and cluster 3. Subsequently, we combined the clinical-pathological data for correlation analysis, and there is a significant correlation between the risk model and the patient's T and stage. Multivariate Cox regression curve analysis showed that the age of BRCA patients and the risk score of the risk model could be used as independent risk factors in the progression of BRCA. In particular, based on this angiogenesis-related risk model, we have drawn a matching nomogram that can predict the 5-, 7-, and 10-year overall survival rates of BRCA patients. Subsequently, we performed a series of pan-cancer analyses of CNV, SNV, OS, methylation, and immune infiltration for this risk model gene and used GDSC data to explore drug sensitivity. Subsequently, to gain insight into the protein expression of these risk model genes in BRCA, we used the immunohistochemical data in the THPA database for verification. The results showed that the protein expressions of IL18, RUNX1, SCG2, and THY1 molecules in BRCA tissues were significantly higher than those in normal breast tissues, while the protein expressions of PF4 and TNFSF12 molecules in BRCA tissues were significantly lower than those in normal breast tissues. Finally, we conducted multiple GSEA analyses to explore the biological pathways these risk model genes can cross in cancer progression. In summary, we believe that this study can provide valuable data and clues for future studies on angiogenesis in BRCA.
Collapse
Affiliation(s)
- Yingkun Xu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Yang Peng
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Meiying Shen
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Li Liu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Jinwei Lei
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Shun Gao
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Yuan Wang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Ailin Lan
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Han Li
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Shengchun Liu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| |
Collapse
|
24
|
Feng J. The p53 Pathway Related Genes Predict the Prognosis of Colon Cancer. Int J Gen Med 2022; 15:169-177. [PMID: 35023955 PMCID: PMC8747760 DOI: 10.2147/ijgm.s346280] [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: 11/04/2021] [Accepted: 12/22/2021] [Indexed: 12/14/2022] Open
Abstract
Background Colon cancer is a common gastrointestinal malignancy. This study aimed to explore the relationship between p53 pathway-related genes and prognosis of colon cancer. Methods The mRNA datasets of colon cancer and adjacent tissues were downloaded from The Cancer Genome Atlas (TCGA) database, and the differential expression of genes in two groups was analyzed. Then, P53 pathway-related genes were intersected with differentially expressed genes (DEGs) to obtain P53 pathway-related differentially expressed genes. Then, overall survival (OS), disease-specific survival (DSS), and progression-free survival (PFS) in clusters were compared by consistent cluster analysis. Univariate and multivariate Cox regression analysis of DEGs was performed to obtain survival-related DEGs. Risk scores were calculated for each sample based on survival-related DEGs, and patients were divided into high/low risk scores. Prognostic differences, tumor immune cell infiltration levels, and immune pathway activation status were compared between the two groups. Results We identified 28 DEGs and two clusters. There are significant differences in PFS between the two clusters (P=0.011), and no significant difference between OS and DSS. We obtained 3 DEGs (CDKN2A, BAK1, BTG1) that were significantly related to PFS, and CDKN2A was considered an independent prognostic factor. PFS showed statistically significant difference between high/low risk score groups (P=0.015). There were significant differences in immune cell infiltration level and immune pathway activity between two groups. Conclusion The p53 pathway-related genes are significantly related to PFS in colon cancer patients and play an important role in regulating the tumor immune microenvironment.
Collapse
Affiliation(s)
- Jinggao Feng
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, Yongzhou City, Hunan Province, 425100, People's Republic of China
| |
Collapse
|
25
|
Xue K, Wu JC, Li XY, Li R, Zhang QL, Chang JJ, Liu YZ, Xu CH, Zhang JY, Sun XJ, Gu JJ, Guo WJ, Wang L. Chidamide triggers BTG1-mediated autophagy and reverses the chemotherapy resistance in the relapsed/refractory B-cell lymphoma. Cell Death Dis 2021; 12:900. [PMID: 34599153 PMCID: PMC8486747 DOI: 10.1038/s41419-021-04187-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/10/2021] [Accepted: 09/15/2021] [Indexed: 11/08/2022]
Abstract
Rituximab/chemotherapy relapsed and refractory B cell lymphoma patients have a poor overall prognosis, and it is urgent to develop novel drugs for improving the therapy outcomes. Here, we examined the therapeutic effects of chidamide, a new histone deacetylase (HDAC) inhibitor, on the cell and mouse models of rituximab/chemotherapy resistant B-cell lymphoma. In Raji-4RH/RL-4RH cells, the rituximab/chemotherapy resistant B-cell lymphoma cell lines (RRCL), chidamide treatment induced growth inhibition and G0/G1 cell cycle arrest. The primary B-cell lymphoma cells from Rituximab/chemotherapy relapsed patients were sensitive to chidamide. Interestingly, chidamide triggered the cell death with the activation of autophagy in RRCLs, likely due to the lack of the pro-apoptotic proteins. Based on the RNA-seq and chromatin immunoprecipitation (ChIP) analysis, we identified BTG1 and FOXO1 as chidamide target genes, which control the autophagy and the cell cycle, respectively. Moreover, the combination of chidamide with the chemotherapy drug cisplatin increased growth inhibition on the RRCL in a synergistic manner, and significantly reduced the tumor burden of a mouse lymphoma model established with engraftment of RRCL. Taken together, these results provide a theoretic and mechanistic basis for further evaluation of the chidamide-based treatment in rituximab/chemotherapy relapsed and refractory B-cell lymphoma patients.
Collapse
Affiliation(s)
- Kai Xue
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Ji-Chuan Wu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xi-Ya Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ran Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qun-Ling Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jin-Jia Chang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Yi-Zhen Liu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Chun-Hui Xu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jia-Ying Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiao-Jian Sun
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan J Gu
- Department of Medicine & Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Wei-Jian Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Lan Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
26
|
Amine H, Ripin N, Sharma S, Stoecklin G, Allain FH, Séraphin B, Mauxion F. A conserved motif in human BTG1 and BTG2 proteins mediates interaction with the poly(A) binding protein PABPC1 to stimulate mRNA deadenylation. RNA Biol 2021; 18:2450-2465. [PMID: 34060423 PMCID: PMC8632095 DOI: 10.1080/15476286.2021.1925476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Antiproliferative BTG/Tob proteins interact directly with the CAF1 deadenylase subunit of the CCR4-NOT complex. This binding requires the presence of two conserved motifs, boxA and boxB, characteristic of the BTG/Tob APRO domain. Consistently, these proteins were shown to stimulate mRNA deadenylation and decay in several instances. Two members of the family, BTG1 and BTG2, were reported further to associate with the protein arginine methyltransferase PRMT1 through a motif, boxC, conserved only in this subset of proteins. We recently demonstrated that BTG1 and BTG2 also contact the first RRM domain of the cytoplasmic poly(A) binding protein PABPC1. To decipher the mode of interaction of BTG1 and BTG2 with partners, we performed nuclear magnetic resonance experiments as well as mutational and biochemical analyses. Our data demonstrate that, in the context of an APRO domain, the boxC motif is necessary and sufficient to allow interaction with PABPC1 but, unexpectedly, that it is not required for BTG2 association with PRMT1. We show further that the presence of a boxC motif in an APRO domain endows it with the ability to stimulate deadenylation in cellulo and in vitro. Overall, our results identify the molecular interface allowing BTG1 and BTG2 to activate deadenylation, a process recently shown to be necessary for maintaining T-cell quiescence.
Collapse
Affiliation(s)
- Hamza Amine
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Centre National de Recherche Scientifique (CNRS) UMR 7104, Illkirch, France.,Institut National de Santé et de Recherche Médicale (INSERM) U1258, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Nina Ripin
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zürich, Switzerland
| | - Sahil Sharma
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,German Cancer Research Center (DKFZ)-ZMBH Alliance, Center for Molecular Biology of Heidelberg University (ZMBH), Heidelberg, Germany
| | - Georg Stoecklin
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,German Cancer Research Center (DKFZ)-ZMBH Alliance, Center for Molecular Biology of Heidelberg University (ZMBH), Heidelberg, Germany
| | - Frédéric H Allain
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zürich, Switzerland.,Department of Biology, Institute of Biochemistry, ETH Zürich, Switzerland
| | - Bertrand Séraphin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Centre National de Recherche Scientifique (CNRS) UMR 7104, Illkirch, France.,Institut National de Santé et de Recherche Médicale (INSERM) U1258, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Fabienne Mauxion
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Centre National de Recherche Scientifique (CNRS) UMR 7104, Illkirch, France.,Institut National de Santé et de Recherche Médicale (INSERM) U1258, Illkirch, France.,Université de Strasbourg, Illkirch, France
| |
Collapse
|
27
|
Ierardi JL, Veloso A, Mancia A. Transcriptome analysis of cadmium exposure in kidney fibroblast cells of the North Atlantic Right Whale (Eubalaena glacialis). Comp Biochem Physiol C Toxicol Pharmacol 2021; 242:108946. [PMID: 33285320 DOI: 10.1016/j.cbpc.2020.108946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/20/2020] [Accepted: 11/29/2020] [Indexed: 11/16/2022]
Abstract
An 8X15k oligonucleotide microarray was developed consisting of 2334 Eubalaena glacialis probes and 2166 Tursiops truncatus probes and used to measure the effects, at transcriptomic level, of cadmium exposure in right whale kidney fibroblast cells. Cells were exposed to three concentrations (1 μM, 0.1 μM, and 0.01 μM) of cadmium chloride (CdCl2) for three exposure times (1, 4, and 24 h). Cells exposed to 1 μM CdCl2 for 4 h and 24 h showed upregulated genes involved in protection from metal toxicity and oxidative stress, protein renaturation, apoptosis inhibition, as well as several regulators of cellular processes. Downregulated genes represented a suite of functions including cell proliferation, transcription regulation, actin polymerization, and stress fiber synthesis. The collection of differentially expressed genes in this study support proposed mechanisms of cadmium-induced apoptosis such as ubiquitin proteasome system disruption, Ca2+ homeostasis interference, mitochondrial membrane potential collapse, reactive oxygen species (ROS) production, and cell cycle arrest. The results also have confirmed the right whale microarray as a reproducible tool in measuring differentiated gene expression that could be a valuable asset for transcriptome analysis of other baleen whales and potential health assessment protocols.
Collapse
Affiliation(s)
- Jessalyn L Ierardi
- Graduate Program of Marine Biology, College of Charleston, 205 Fort Johnson Rd, Charleston, SC 29412, USA
| | - Artur Veloso
- Graduate Program of Marine Biology, College of Charleston, 205 Fort Johnson Rd, Charleston, SC 29412, USA
| | - Annalaura Mancia
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 171 Ashley Ave, Charleston, SC 29425, USA; Marine Biomedicine and Environmental Sciences Center, Medical University of South Carolina, 331 Fort Johnson Rd, Charleston, SC 29412, USA; Department of Life Sciences and Biotechnology, University of Ferrara, via L. Borsari 46, Ferrara 44121, Italy.
| |
Collapse
|
28
|
Iyer AA, Groves AK. Transcription Factor Reprogramming in the Inner Ear: Turning on Cell Fate Switches to Regenerate Sensory Hair Cells. Front Cell Neurosci 2021; 15:660748. [PMID: 33854418 PMCID: PMC8039129 DOI: 10.3389/fncel.2021.660748] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/08/2021] [Indexed: 12/15/2022] Open
Abstract
Non-mammalian vertebrates can restore their auditory and vestibular hair cells naturally by triggering the regeneration of adjacent supporting cells. The transcription factor ATOH1 is a key regulator of hair cell development and regeneration in the inner ear. Following the death of hair cells, supporting cells upregulate ATOH1 and give rise to new hair cells. However, in the mature mammalian cochlea, such natural regeneration of hair cells is largely absent. Transcription factor reprogramming has been used in many tissues to convert one cell type into another, with the long-term hope of achieving tissue regeneration. Reprogramming transcription factors work by altering the transcriptomic and epigenetic landscapes in a target cell, resulting in a fate change to the desired cell type. Several studies have shown that ATOH1 is capable of reprogramming cochlear non-sensory tissue into cells resembling hair cells in young animals. However, the reprogramming ability of ATOH1 is lost with age, implying that the potency of individual hair cell-specific transcription factors may be reduced or lost over time by mechanisms that are still not clear. To circumvent this, combinations of key hair cell transcription factors have been used to promote hair cell regeneration in older animals. In this review, we summarize recent findings that have identified and studied these reprogramming factor combinations for hair cell regeneration. Finally, we discuss the important questions that emerge from these findings, particularly the feasibility of therapeutic strategies using reprogramming factors to restore human hearing in the future.
Collapse
Affiliation(s)
- Amrita A. Iyer
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Program in Genetics & Genomics, Houston, TX, United States
| | - Andrew K. Groves
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Program in Genetics & Genomics, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
29
|
Ebrahimi M, Yaghoobi MM. Effects of aqueous and methanolic extracts of Astragalus Longistylus on growth and proliferation of human dental pulp stem cells. ADVANCES IN TRADITIONAL MEDICINE 2021. [DOI: 10.1007/s13596-020-00519-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
30
|
Almasmoum HA, Airhihen B, Seedhouse C, Winkler GS. Frequent loss of BTG1 activity and impaired interactions with the Caf1 subunit of the Ccr4-Not deadenylase in non-Hodgkin lymphoma. Leuk Lymphoma 2020; 62:281-290. [PMID: 33021411 DOI: 10.1080/10428194.2020.1827243] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mutations in the highly similar genes B-cell translocation gene 1 (BTG1) and BTG2 are identified in approximately 10-15% of non-Hodgkin lymphoma cases, which may suggest a direct involvement of BTG1 and BTG2 in malignant transformation. However, it is unclear whether or how disease-associated mutations impair the function of these genes. Therefore, we selected 16 BTG1 variants based on in silico analysis. We then evaluated (i) the ability of these variants to interact with the known protein-binding partners CNOT7 and CNOT8, which encode the Caf1 catalytic subunit of the Ccr4-Not deadenylase complex; (ii) the activity of the variant proteins in cell cycle progression; (iii) translational repression; and (iv) mRNA degradation. Based on these analyses, we conclude that mutations in BTG1 may contribute to malignant transformation and tumor cell proliferation by interfering with its anti-proliferative activity and ability to interact with CNOT7 and CNOT8.
Collapse
Affiliation(s)
- Hibah Ali Almasmoum
- School of Pharmacy, The University of Nottingham, University Park, Nottingham, UK.,Department of Haematology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK.,Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Blessing Airhihen
- School of Pharmacy, The University of Nottingham, University Park, Nottingham, UK
| | - Claire Seedhouse
- Department of Haematology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | | |
Collapse
|
31
|
Long J, He Q, Yin Y, Lei X, Li Z, Zhu W. The effect of miRNA and autophagy on colorectal cancer. Cell Prolif 2020; 53:e12900. [PMID: 32914514 PMCID: PMC7574865 DOI: 10.1111/cpr.12900] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/29/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) has become a concern because of its high recurrence rate and metastasis rate, low early diagnosis rate and poor therapeutic effect. At present, various studies have shown that autophagy is closely connected with the occurrence and progression of CRC. Autophagy is a highly cytosolic catabolic process involved in lysosomes in biological evolution. Cells degrade proteins and damaged organelles by autophagy to achieve material circulation and maintain cell homeostasis. Moreover, microRNAs are key regulators of autophagy, and their mediated regulation of transcriptional and post-transcriptional levels plays an important role in autophagy in CRC cells. This review focuses on the recent research advances of how autophagy and related microRNAs are involved in affecting occurrence and progression of CRC and provides a new perspective for the study of CRC treatment strategies.
Collapse
Affiliation(s)
- Jiali Long
- Department of PathologyGuangdong Medical UniversityDongguanChina
- Department of Pathologythe Eighth Affiliated HospitalSun Yat‐Sen UniversityShenzhenChina
| | - Qinglian He
- Department of PathologyGuangdong Medical UniversityDongguanChina
| | - Yuting Yin
- Department of PathologyGuangdong Medical UniversityDongguanChina
| | - Xue Lei
- Department of PathologyGuangdong Medical UniversityDongguanChina
| | - Ziqi Li
- Department of PathologyGuangdong Medical UniversityDongguanChina
| | - Wei Zhu
- Department of PathologyGuangdong Medical UniversityDongguanChina
| |
Collapse
|
32
|
Müller S, Wedler A, Breuer J, Glaß M, Bley N, Lederer M, Haase J, Misiak C, Fuchs T, Ottmann A, Schmachtel T, Shalamova L, Ewe A, Aigner A, Rossbach O, Hüttelmaier S. Synthetic circular miR-21 RNA decoys enhance tumor suppressor expression and impair tumor growth in mice. NAR Cancer 2020; 2:zcaa014. [PMID: 34316687 PMCID: PMC8210135 DOI: 10.1093/narcan/zcaa014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 01/07/2023] Open
Abstract
Naturally occurring circular RNAs efficiently impair miRNA functions. Synthetic circular RNAs may thus serve as potent agents for miRNA inhibition. Their therapeutic effect critically relies on (i) the identification of optimal miRNA targets, (ii) the optimization of decoy structures and (iii) the development of efficient formulations for their use as drugs. In this study, we extensively explored the functional relevance of miR-21-5p in cancer cells. Analyses of cancer transcriptomes reveal that miR-21-5p is the by far most abundant miRNA in human cancers. Deletion of the MIR21 locus in cancer-derived cells identifies several direct and indirect miR-21-5p targets, including major tumor suppressors with prognostic value across cancers. To impair miR-21-5p activities, we evaluate synthetic, circular RNA decoys containing four repetitive binding elements. In cancer cells, these decoys efficiently elevate tumor suppressor expression and impair tumor cell vitality. For their in vivo delivery, we for the first time evaluate the formulation of decoys in polyethylenimine (PEI)-based nanoparticles. We demonstrate that PEI/decoy nanoparticles lead to a significant inhibition of tumor growth in a lung adenocarcinoma xenograft mouse model via the upregulation of tumor suppressor expression. These findings introduce nanoparticle-delivered circular miRNA decoys as a powerful potential therapeutic strategy in cancer treatment.
Collapse
Affiliation(s)
- Simon Müller
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Alice Wedler
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Janina Breuer
- Institute of Biochemistry, Faculty of Biology and Chemistry, Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Markus Glaß
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Nadine Bley
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Marcell Lederer
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Jacob Haase
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Claudia Misiak
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Tommy Fuchs
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Alina Ottmann
- Institute of Biochemistry, Faculty of Biology and Chemistry, Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Tessa Schmachtel
- Institute of Biochemistry, Faculty of Biology and Chemistry, Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Lyudmila Shalamova
- Institute of Biochemistry, Faculty of Biology and Chemistry, Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Alexander Ewe
- Department of Clinical Pharmacology, Rudolf Boehm Institute for Pharmacology and Toxicology, Faculty of Medicine, Leipzig University, 04107 Leipzig, Germany
| | - Achim Aigner
- Department of Clinical Pharmacology, Rudolf Boehm Institute for Pharmacology and Toxicology, Faculty of Medicine, Leipzig University, 04107 Leipzig, Germany
| | - Oliver Rossbach
- Institute of Biochemistry, Faculty of Biology and Chemistry, Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Stefan Hüttelmaier
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| |
Collapse
|
33
|
Yaghoobi MM, Sheikoleslami M, Ebrahimi M. Effects of hydrogen peroxide, doxorubicin and ultraviolet irradiation on senescence of human dental pulp stem cells. Arch Oral Biol 2020; 117:104819. [PMID: 32592933 DOI: 10.1016/j.archoralbio.2020.104819] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/19/2020] [Accepted: 06/11/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The objective of this study was to evaluate the ability of three distinct agents on the induction of senescence in human dental pulp stem cells (DPSCs). DESIGN DPSCs from three separate donors were treated with H2O2, doxorubicin and ultraviolet (UV) irradiation. The response of the cells to the three agents was assayed by specific staining for SA-βGal, RT-qPCR and flow cytometry. RESULTS The results showed that incubation with 100 μM H2O2 and 20 nM Doxorubicin for seven days led to senescence in all donors' cells equally. Interestingly, UV irradiation for just one minute was sufficient to induce senescence in the cells. The SA-βGal positive senescent cells were arrested in G1 phase and their S phase was significantly reduced as analyzed by flow cytometry. Significant increment in p21 and BTG1 expression and decrement in CCND1 expression also confirmed the cells have been arrested and get senescent via p53-p21 pathway. CONCLUSION All three agents successfully triggered senescence in the cells. There was no significant difference in the capacity of the three donor's cells for senescence. To avoid premature senescence in stem cell in vitro, it is recommended to avoid unnecessary exposure of the cell to fluorescent and UV light. Moreover, to prevent ROS production, we recommend using a separate incubator with low oxygen content for cell culture, if possible.
Collapse
Affiliation(s)
- Mohammad Mehdi Yaghoobi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran.
| | - Mozhgan Sheikoleslami
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Maryam Ebrahimi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| |
Collapse
|
34
|
Chen XQ, Meng FQ, Xiong H, Wang YL, Tang WH, Zou YM. Identification of BTG1 Status in Solid Cancer for Future Researches Using a System Review and Meta-analysis. Curr Med Sci 2020; 40:85-94. [PMID: 32166669 DOI: 10.1007/s11596-020-2150-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 12/10/2019] [Indexed: 12/24/2022]
Abstract
Abundant studies have been conducted to identify how B-cell translocation gene 1 protein (BTG1) gene affects the differentiation, proliferation, metastasis of cancer cells, and how it further regulates the generation or development of diseases to influence the prognosis of patients. However, the data from single research were not powerful enough. The correlations between BTG1 expression and mechanisms of tumorigenesis or prognosis of patients are still in controversial. Our system review and meta-analysis provided a complete explanation about the association between BTG1 expression and clinicopathological features or prognosis of patients, which further laid a foundation for future research on BTG1. Fifteen eligible studies consisting of 1992 participants were included. We uncovered that BTG1 expression in solid tumors was associated with lymph node status (RR=0.66, 95% CI: 0.58-0.75, P=0.142), TMN stage status (RR=2.13, 95% CI: 1.71-2.65, P=0.001), T category (RR=1.90, 95% CI: 1.20-3.00, P=0.000), histological differentiation (RR=1.91, 95% CI: 1.55-2.37, P=0.012), vascular invasion (RR=0.90, 95% CI: 0.57-1.41, P=0.001). BTG1 low expression was significantly associated with overall survival (OS) (HR=0.47, 95% CI: 0.38-0.67, P=0.000). It concluded that BTG1 possessed the potential value for future research and could be recommended as a significant biomarker in solid tumor.
Collapse
Affiliation(s)
- Xiu-Qiong Chen
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | | | - Hua Xiong
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ya-Li Wang
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wen-Hua Tang
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan-Mei Zou
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
35
|
Ceccarelli M, D'Andrea G, Micheli L, Tirone F. Deletion of Btg1 Induces Prmt1-Dependent Apoptosis and Increased Stemness in Shh-Type Medulloblastoma Cells Without Affecting Tumor Frequency. Front Oncol 2020; 10:226. [PMID: 32231994 PMCID: PMC7082329 DOI: 10.3389/fonc.2020.00226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/10/2020] [Indexed: 12/16/2022] Open
Abstract
About 30% of medulloblastomas (MBs), a tumor of the cerebellum, arise from cerebellar granule cell precursors (GCPs) undergoing transformation following activation of the Sonic hedgehog (Shh) pathway. To study this process, we generated a new MB model by crossing Patched1 heterozygous (Ptch1+/−) mice, which develop spontaneous Shh-type MBs, with mice lacking B-cell translocation gene 1 (Btg1), a regulator of cerebellar development. In MBs developing in Ptch1+/− mice, deletion of Btg1 does not alter tumor and lesion frequencies, nor affect the proliferation of neoplastic precursor cells. However, in both tumors and lesions arising in Ptch1+/− mice, ablation of Btg1 increases by about 25% the apoptotic neoplastic precursor cells, as judged by positivity to activated caspase-3. Moreover, although Btg1 ablation in early postnatal GCPs, developing in the external granule cell layer, leads to a significant increase of proliferation, and decrease of differentiation, relative to wild-type, no synergy occurs with the Ptch1+/− mutation. However, Btg1 deletion greatly increases apoptosis in postnatal GCPs, with strong synergy between Btg1-null and Ptch1+/− mutations. That pronounced increase of apoptosis observed in Ptch1+/−/Btg1 knockout young or neoplastic GCPs may be responsible for the lack of effect of Btg1 ablation on tumorigenesis. This increased apoptosis may be a consequence of increased expression of protein arginine methyltransferase 1 (Prmt1) protein that we observe in Btg1 knockout/Ptch1+/− MBs. In fact, apoptotic genes, such as BAD, are targets of Prmt1. Moreover, in Btg1-null MBs, we observed a two-fold increase of cells positive to CD15, which labels tumor stem cells, raising the possibility of activation of quiescent tumor cells, known for their role in long-term resistance to treatment and relapses. Thus, Btg1 appears to play a role in cerebellar tumorigenesis by regulating the balance between apoptosis and proliferation during MB development, also influencing the number of tumor stem cells.
Collapse
Affiliation(s)
- Manuela Ceccarelli
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC-CNR), Rome, Italy
| | - Giorgio D'Andrea
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC-CNR), Rome, Italy
| | - Laura Micheli
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC-CNR), Rome, Italy
| | - Felice Tirone
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC-CNR), Rome, Italy
| |
Collapse
|
36
|
ElTanbouly MA, Zhao Y, Nowak E, Li J, Schaafsma E, Le Mercier I, Ceeraz S, Lines JL, Peng C, Carriere C, Huang X, Day M, Koehn B, Lee SW, Silva Morales M, Hogquist KA, Jameson SC, Mueller D, Rothstein J, Blazar BR, Cheng C, Noelle RJ. VISTA is a checkpoint regulator for naïve T cell quiescence and peripheral tolerance. Science 2020; 367:eaay0524. [PMID: 31949051 PMCID: PMC7391053 DOI: 10.1126/science.aay0524] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/30/2019] [Accepted: 12/02/2019] [Indexed: 12/18/2022]
Abstract
Negative checkpoint regulators (NCRs) temper the T cell immune response to self-antigens and limit the development of autoimmunity. Unlike all other NCRs that are expressed on activated T lymphocytes, V-type immunoglobulin domain-containing suppressor of T cell activation (VISTA) is expressed on naïve T cells. We report an unexpected heterogeneity within the naïve T cell compartment in mice, where loss of VISTA disrupted the major quiescent naïve T cell subset and enhanced self-reactivity. Agonistic VISTA engagement increased T cell tolerance by promoting antigen-induced peripheral T cell deletion. Although a critical player in naïve T cell homeostasis, the ability of VISTA to restrain naïve T cell responses was lost under inflammatory conditions. VISTA is therefore a distinctive NCR of naïve T cells that is critical for steady-state maintenance of quiescence and peripheral tolerance.
Collapse
Affiliation(s)
- Mohamed A ElTanbouly
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Yanding Zhao
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Elizabeth Nowak
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | | | - Evelien Schaafsma
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | | | - Sabrina Ceeraz
- Immunology Discovery, Janssen Research and Development LLC, Spring House, PA, USA
| | - J Louise Lines
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Changwei Peng
- Division of Rheumatic and Autoimmune Diseases, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- The Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | | | - Xin Huang
- ImmuNext Corporation, Lebanon, NH, USA
| | - Maria Day
- ImmuNext Corporation, Lebanon, NH, USA
| | - Brent Koehn
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Sam W Lee
- Yale University School of Medicine, New Haven, CT, USA
| | - Milagros Silva Morales
- Division of Rheumatic and Autoimmune Diseases, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Kristin A Hogquist
- Division of Rheumatic and Autoimmune Diseases, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- The Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Stephen C Jameson
- Division of Rheumatic and Autoimmune Diseases, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- The Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Daniel Mueller
- Division of Rheumatic and Autoimmune Diseases, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- The Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | | | - Bruce R Blazar
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Chao Cheng
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Randolph J Noelle
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA.
- ImmuNext Corporation, Lebanon, NH, USA
| |
Collapse
|
37
|
Li E, Han K, Zhou X. microRNA-27a-3p Down-regulation Inhibits Malignant Biological Behaviors of Ovarian Cancer by Targeting BTG1. Open Med (Wars) 2019; 14:577-585. [PMID: 31410369 PMCID: PMC6689206 DOI: 10.1515/med-2019-0065] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 05/20/2019] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer is the most deadly malignant tumor. MicroRNA-27a-3p (miR-27a-3p) was a tumor oncogene in various cancers. However, the role and mechanism of miR-27a-3p in ovarian cancer are still unknown. In this study, we found that miR-27a-3p over-expression could significantly promote the viability of SK-OV-3 cells, enhance cell migration and invasion, and reduce cell apoptosis. Besides, results from western blot assay showed that miR-27a-3p over-expression could increase Bcl-2 protein expression and decrease Bax protein expression. Furthermore, TargetScan and the dual luciferase reporter gene assay revealed that BTG anti-proliferation factor 1 (BTG1) was a direct target of miR-27a-3p. In addition, we found that miR-27a-3p down-regulation suppressed SK-OV-3 cell viability, migration and invasion, and promoted cell apoptosis. All the effects of miR-27a-3p down-regulation on SK-OV-3 cells were reversed by BTG1-siRNA. Therefore, miR-27a-3p/BTG1 axis may be a new potential target for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Enfang Li
- Department of Obstetrics and Gynecology, Taikang Xianlin Gulou Hospital, No. 188 Lingshan North Road, Qixia District, Nanjing 210000, China
| | - Ke Han
- Department of Obstetrics and Gynecology, Taikang Xianlin Gulou Hospital, No. 188 Lingshan North Road, Qixia District, Nanjing 210000, China
| | - Xuan Zhou
- Department of Obstetrics and Gynecology, Taikang Xianlin Gulou Hospital, No. 188 Lingshan North Road, Qixia District, Nanjing 210000, China
| |
Collapse
|
38
|
Su C, Huang DP, Liu JW, Liu WY, Cao YO. miR-27a-3p regulates proliferation and apoptosis of colon cancer cells by potentially targeting BTG1. Oncol Lett 2019; 18:2825-2834. [PMID: 31452761 PMCID: PMC6676402 DOI: 10.3892/ol.2019.10629] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 05/13/2019] [Indexed: 01/16/2023] Open
Abstract
microRNA (miR/miRNA)-27a-3p has been reported to be abnormally expressed in various types of cancer, including colorectal cancer (CRC). B-cell translocation gene 1 (BTG1) has also been implicated with CRC. However, the association between miR-27a-3p and BTG1 in CRC, to the best of our knowledge, has not been investigated. In order to assess whether miR-27a-3p is associated with CRC, reverse transcription-quantitative PCR was performed on 20 paired CRC and paracancerous tissues for miRNA analysis. For the screening and validation of miR-27a-3p expression in colon cancer, several colon cancer cell lines (HCT-116, HCT8, SW480, HT29, LOVO and Caco2) and the normal colorectal epithelial cell line NCM460 were examined. The highest expression levels of miR-27a-3p were detected in the HCT-116, which was selected for further experimentation. The HCT-116 cells were divided into control, miR-27a-3p mimic and inhibitor groups, and cell proliferation was tested using an MTT assay. Additionally, miR-27a-3p inhibitor/mimic or BTG1 plasmid were transfected into the HCT-116 cells, and flow cytometry was performed to analyze cell cycle distributions. TUNEL analysis was performed to detect apoptosis. Protein levels of factors in the downstream signaling pathway mediated by miR-27a-3p [ERK/mitogen-activated extracellular signal-regulated kinase (MEK)] were detected. miR-27a-3p was revealed to be overexpressed in human CRC tissues and colon cancer cell lines. Knockdown of miR-27a-3p suppressed proliferation of HCT-116 cells and apoptosis was increased. It further markedly upregulated expression levels of BTG1 and inhibited activation of proteins of the ERK/MEK signaling pathway. In addition, overexpression of BTG1 in HCT-116 cells triggered G1/S phase cell cycle arrest and increased apoptosis via the ERK/MEK signaling pathway. In conclusion, the present study demonstrated that the effects of miR-27a-3p on colon cancer cell proliferation and apoptosis were similar to those of the tumor suppressor gene BTG1. The miR-27a-3p/BTG1 axis may have potential implications for diagnostic and therapeutic approaches in CRC.
Collapse
Affiliation(s)
- Chang Su
- Department of Surgery, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Dong-Ping Huang
- Department of Surgery, People's Hospital of Putuo District, Shanghai 200060, P.R. China
| | - Jian-Wen Liu
- Department of Molecular and Cellular Pharmacology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P.R. China
| | - Wei-Yan Liu
- Department of Surgery, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Yi-Ou Cao
- Department of Surgery, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai 201199, P.R. China
| |
Collapse
|
39
|
Role of B-Cell Translocation Gene 1 in the Pathogenesis of Endometriosis. Int J Mol Sci 2019; 20:ijms20133372. [PMID: 31324015 PMCID: PMC6651142 DOI: 10.3390/ijms20133372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 07/05/2019] [Accepted: 07/05/2019] [Indexed: 01/03/2023] Open
Abstract
Estrogen affects endometrial cellular proliferation by regulating the expression of the c-myc gene. B-cell translocation gene 1 (BTG1), a translocation partner of the c-myc, is a tumor suppressor gene that promotes apoptosis and negatively regulates cellular proliferation and cell-to-cell adhesion. The aim of this study was to determine the role of BTG1 in the pathogenesis of endometriosis. BTG1 mRNA and protein expression was evaluated in eutopic and ectopic endometrium of 30 patients with endometriosis (endometriosis group), and in eutopic endometrium of 22 patients without endometriosis (control group). The effect of BTG1 downregulation on cellular migration, proliferation, and apoptosis was evaluated using transfection of primarily cultured human endometrial stromal cells (HESCs) with BTG1 siRNA. BTG1 mRNA expression level of eutopic and ectopic endometrium of endometriosis group were significantly lower than that of the eutopic endometrium of the control group. Migration and wound healing assays revealed that BTG1 downregulation resulted in a significant increase in migration potential of HESCs, characterized by increased expression of matrix metalloproteinase 2 (MMP2) and MMP9. Downregulation of BTG1 in HESCs significantly reduced Caspase 3 expression, indicating a decrease in apoptotic potential. In conclusion, our data suggest that downregulation of BTG1 plays an important role in the pathogenesis of endometriosis.
Collapse
|
40
|
Deng J, Guo Y, Yuan F, Chen S, Yin H, Jiang X, Jiao F, Wang F, Ji H, Hu G, Ying H, Chen Y, Zhai Q, Xiao F, Guo F. Autophagy inhibition prevents glucocorticoid-increased adiposity via suppressing BAT whitening. Autophagy 2019; 16:451-465. [PMID: 31184563 DOI: 10.1080/15548627.2019.1628537] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The mechanisms underlying glucocorticoid (GC)-increased adiposity are poorly understood. Brown adipose tissue (BAT) acquires white adipose tissue (WAT) cell features defined as BAT whitening under certain circumstances. The aim of our current study was to investigate the possibility and mechanisms of GC-induced BAT whitening. Here, we showed that one-week dexamethasone (Dex) treatment induced BAT whitening, characterized by lipid droplet accumulation, in vitro and in vivo. Furthermore, autophagy and ATG7 (autophagy related 7) expression was induced in BAT by Dex, and treatment with the autophagy inhibitor chloroquine or adenovirus-mediated ATG7 knockdown prevented Dex-induced BAT whitening and fat mass gain. Moreover, Dex-increased ATG7 expression and autophagy was mediated by enhanced expression of BTG1 (B cell translocation gene 1, anti-proliferative) that stimulated activity of CREB1 (cAMP response element binding protein 1). The importance of BTG1 in this regulation was further demonstrated by the observed BAT whitening in adipocyte-specific BTG1-overexpressing mice and the attenuated Dex-induced BAT whitening and fat mass gain in mice with BTG1 knockdown in BAT. Taken together, we showed that Dex induces a significant whitening of BAT via BTG1- and ATG7-dependent autophagy, which might contribute to Dex-increased adiposity. These results provide new insights into the mechanisms underlying GC-increased adiposity and possible strategy for preventing GC-induced side effects via the combined use of an autophagy inhibitor.Abbreviations: ACADL: acyl-Coenzyme A dehydrogenase, long-chain; ACADM: acyl-Coenzyme A dehydrogenase, medium-chain; ACADS: acyl-Coenzyme A dehydrogenase, short-chain; ADIPOQ: adiponectin; AGT: angiotensinogen; Atg: autophagy-related; BAT: brown adipose tissue; BTG1: B cell translocation gene 1, anti-proliferative; CEBPA: CCAAT/enhancer binding protein (C/EBP), alpha; CIDEA: cell death-inducing DNA fragmentation factor, alpha subunit-like effector A; CPT1B: carnitine palmitoyltransferase 1b, muscle; CPT2: carnitine palmitoyltransferase 2; CQ: chloroquine; Dex: dexamethasone; eWAT: epididymal white adipose tissue; FABP4: fatty acid binding protein 4, adipocyte; FFAs: free fatty acids; GCs: glucocorticoids; NRIP1: nuclear receptor interacting protein 1; OCR: oxygen consumption rate; PBS: phosphate-buffered saline; PPARA: peroxisome proliferator activated receptor alpha; PPARG: peroxisome proliferator activated receptor gamma; PPARGC1A: peroxisome proliferator activated receptor, gamma, coactivator 1 alpha; PRDM16: PR domain containing 16; PSAT1: phosphoserine aminotransferase 1; RB1: RB transcriptional corepressor 1; RBL1/p107: RB transcriptional corepressor like 1; SQSTM1: sequestosome 1; sWAT: subcutaneous white adipose tissue; TG: triglycerides; UCP1: uncoupling protein 1 (mitochondrial, proton carrier); WT: wild-type.
Collapse
Affiliation(s)
- Jiali Deng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yajie Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Feixiang Yuan
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shanghai Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hanrui Yin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoxue Jiang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Fuxin Jiao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Fenfen Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hongbin Ji
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guohong Hu
- The Key Laboratory of Stem Cell Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Ying
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiwei Zhai
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Fei Xiao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Feifan Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
41
|
Zhang ZZ, Qin XH, Zhang J. MicroRNA-183 inhibition exerts suppressive effects on diabetic retinopathy by inactivating BTG1-mediated PI3K/Akt/VEGF signaling pathway. Am J Physiol Endocrinol Metab 2019; 316:E1050-E1060. [PMID: 30835506 DOI: 10.1152/ajpendo.00444.2018] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Diabetic retinopathy (DR) is a serious diabetic complication caused by both environmental and genetic factors. Molecular mechanisms of DR may lead to the discovery of reliable prognostic indicators. The current study aimed to clarify the mechanism of microRNA-183 (miR-183) in DR in relation to the PI3K/Akt/VEGF signaling pathway. Microarray-based gene expression profiling of DR was used to identify the differentially expressed genes. Sprague-Dawley rats were used for the establishment of DR models, and then miR-183 was altered by mimic or inhibitor or BTG1 was downregulated by siRNA to explore the regulatory mechanism of miR-183 in DR. Furthermore, the expression of miR-183, CD34, endothelial nitric oxide synthase (eNOS), BTG1 and the PI3K/Akt/VEGF signaling pathway-related genes as well as reactive oxygen species (ROS) level was determined, and the relationship between miR-183 and BTG1 was also verified. Cell growth, cell apoptosis, and angiogenesis were determined. Microarray analysis revealed the involvement of miR-183 in DR via the PI3K/Akt/VEGF signaling pathway by targeting BTG1. Upregulated miR-183 and downregulated BTG1 were observed in retinal tissues of DR rats. miR-183 overexpression activated the PI3K/Akt/VEGF signaling pathway, upregulated CD34, eNOS, and ROS, and inhibited BTG1. BTG1 was confirmed as a target gene of miR-183. miR-183 overexpression or BTG1 knockdown promoted cell growth and tube formation while it suppressed cell apoptosis of vascular endothelial cells in DR rats. In this study, we demonstrated that miR-183 silencing inhibiting cell growth and tube formation in vascular endothelial cells of DR rats via the PI3K/Akt/VEGF signaling pathway by upregulating BTG1.
Collapse
Affiliation(s)
- Zhen-Zhen Zhang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , People's Republic of China
| | - Xiu-Hong Qin
- Department of Ophthalmology, The First Affiliated Hospital of Dalian Medical University , Dalian , People's Republic of China
| | - Jing Zhang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , People's Republic of China
| |
Collapse
|
42
|
Qian Y, Lu X, Li Q, Zhao S, Fang D. The treatment effects and the underlying mechanism of B cell translocation gene 1 on the oncogenesis of brain glioma. J Cell Biochem 2019; 120:13310-13320. [DOI: 10.1002/jcb.28605] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Yu Qian
- Department of Neurosurgery Jiangsu University Affiliated People's Hospital
| | - Xinyu Lu
- Department of Neurosurgery Jiangsu University Affiliated People's Hospital
| | - Qiaoyu Li
- Department of Neurosurgery Jiangsu University Affiliated People's Hospital
| | - Su Zhao
- Department of Neurosurgery Jiangsu University Affiliated People's Hospital
| | - Dazhao Fang
- Department of Neurosurgery The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University Huaiyin China
| |
Collapse
|
43
|
Yan W, Li SX, Gao H, Yang W. Identification of B-cell translocation gene 1-controlled gene networks in diffuse large B-cell lymphoma: A study based on bioinformatics analysis. Oncol Lett 2019; 17:2825-2835. [PMID: 30854058 PMCID: PMC6365947 DOI: 10.3892/ol.2019.9900] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 11/16/2018] [Indexed: 02/06/2023] Open
Abstract
B-cell translocation gene 1 (BTG1) is a member of the BTG/transducer of Erb family. The present study evaluated the impact of BTG1 gene expression on the clinical outcome of diffuse large B-cell lymphoma (DLBCL) and investigated potential mechanisms using the Gene Expression Omnibus (GEO) database. The gene expression profile datasets GSE31312, GSE10846, GSE65420 and GSE87371 were downloaded from the GEO database. BTG1 expression and clinicopathological data were obtained from the GSE31312 dataset. In 498 cases, the expression of BTG1 in DLBCL was associated with treatment response (χ2=19.020; P<0.001) and International Prognostic Index score (χ2=5.320; P=0.025). Using the Kaplan-Meier method, it was identified that the expression of BTG1 was associated with overall survival (OS) and progression-free survival (PFS) times. Univariate and multivariate Cox regression analysis demonstrated that BTG1 was an independent predictive factor for OS and PFS. From the overlapping analysis of 407 BTG1-associated genes and 22,187 DLBCL-associated genes, 401 genes were identified as BTG1-associated DLBCL genes. Pathway analysis revealed that BTG1-associated DLBCL genes were associated with cancer progression and DLBCL signaling pathways. Subsequently, a protein-protein interaction network was constructed of the BTG1-associated genes, which consisted of 235 genes and 601 interactions. Additionally, 24 genes with high degrees in the network were identified as hub genes, which included genes associated with ‘ribosome’ [ribosomal protein (RP) L11, RPL3, RPS29, RPL19, RPL15 and RPL12], ‘cell cycle’ (ubiquitin carboxyl extension protein 52, ATM and Ras homolog family member H), ‘mitogen-activated protein kinase pathway’ (mitogen-activated protein kinase 1), ‘histone modification’ (ASH1-like protein) and ‘transcription/translation’ (eukaryotic translation initiation factor 3 subunit E, eukaryotic translation elongation factor 1 δ, transcription termination factor 1, cAMP responsive element binding protein 1 and RNA polymerase II subunit F). In conclusion, BTG1 may serve as a predictive biomarker for DLBCL prognosis. Additionally, bioinformatics analysis indicated that BTG1 may exhibit key functions in the progression and development of DLBCL.
Collapse
Affiliation(s)
- Wei Yan
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Shawn Xiang Li
- International College, China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Hongyu Gao
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Wei Yang
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| |
Collapse
|
44
|
Yuniati L, Scheijen B, van der Meer LT, van Leeuwen FN. Tumor suppressors BTG1 and BTG2: Beyond growth control. J Cell Physiol 2018; 234:5379-5389. [PMID: 30350856 PMCID: PMC6587536 DOI: 10.1002/jcp.27407] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 08/22/2018] [Indexed: 01/21/2023]
Abstract
Since the identification of B‐cell translocation gene 1 (BTG1) and BTG2 as antiproliferation genes more than two decades ago, their protein products have been implicated in a variety of cellular processes including cell division, DNA repair, transcriptional regulation and messenger RNA stability. In addition to affecting differentiation during development and in the adult, BTG proteins play an important role in maintaining homeostasis under conditions of cellular stress. Genomic profiling of B‐cell leukemia and lymphoma has put BTG1 and BTG2 in the spotlight, since both genes are frequently deleted or mutated in these malignancies, pointing towards a role as tumor suppressors. Moreover, in solid tumors, reduced expression of BTG1 or BTG2 is often correlated with malignant cell behavior and poor treatment outcome. Recent studies have uncovered novel roles for BTG1 and BTG2 in genotoxic and integrated stress responses, as well as during hematopoiesis. This review summarizes what is currently known about the roles of BTG1 and BTG2 in these and other cellular processes. In addition, we will highlight the molecular mechanisms and biological consequences of BTG1 and BTG2 deregulation during cancer progression and elaborate on the potential clinical implications of these findings.
Collapse
Affiliation(s)
- Laurensia Yuniati
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands.,Hubrecht Institute-KNAW, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Blanca Scheijen
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Laurens T van der Meer
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frank N van Leeuwen
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
45
|
Copy number abnormality of acute lymphoblastic leukemia cell lines based on their genetic subtypes. Int J Hematol 2018; 108:312-318. [DOI: 10.1007/s12185-018-2474-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 05/13/2018] [Accepted: 05/14/2018] [Indexed: 11/29/2022]
|
46
|
Zhao S, Chen SR, Yang XF, Shen DF, Takano Y, Su RJ, Zheng HC. BTG1 might be employed as a biomarker for carcinogenesis and a target for gene therapy in colorectal cancers. Oncotarget 2018; 8:7502-7520. [PMID: 27447746 PMCID: PMC5352338 DOI: 10.18632/oncotarget.10649] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 06/03/2016] [Indexed: 12/17/2022] Open
Abstract
Here, BTG1 overexpression inhibited proliferation, induced differentiation, autophagy, and apoptosis in colorectal cancer cells (p2 arrest might be related to Cyclin B1 and Cdc25B hypoexpression in HCT-15 transfectants, while G1 arrest in HCT-116 transfectants overexpressing p21 and p27. BTG1 overexpression decreased the expression of Bcl-2, Bcl-xL, XIAP, Akt1 or survivin and increased the expression of Bax or p53 in colorectal cancer cells. BTG1-induced autophagy was dependent on Beclin-1 expression. BTG1 overexpression might weaken β-catenin pathway in colorectal cancer cells. The chemosensitivity of BTG1 transfectants to paclitaxel, cisplatin, MG132 or SAHA was positively correlated with its apoptotic induction. There was a lower expression level of BTG1 in cancer than matched non-neoplastic mucosa by RT-PCR (p
Collapse
Affiliation(s)
- Shuang Zhao
- Cancer Center, Key Laboratory of Brain and Spinal Cord Injury of Liaoning Province, and Animal Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Shu-Rui Chen
- Department of Science and Technology, Jinzhou Medical University, Jinzhou, China
| | - Xue-Feng Yang
- Cancer Center, Key Laboratory of Brain and Spinal Cord Injury of Liaoning Province, and Animal Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Dao-Fu Shen
- Cancer Center, Key Laboratory of Brain and Spinal Cord Injury of Liaoning Province, and Animal Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yasuo Takano
- School of Health Science, Tokyo University of Technology, Nishi-Kamata, Ohta-ku, Tokyo, Japan
| | - Rong-Jian Su
- Life Science Institute of Jinzhou Medical University, Jinzhou, China
| | - Hua-Chuan Zheng
- Cancer Center, Key Laboratory of Brain and Spinal Cord Injury of Liaoning Province, and Animal Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China.,Life Science Institute of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
47
|
Juszczak GR, Stankiewicz AM. Glucocorticoids, genes and brain function. Prog Neuropsychopharmacol Biol Psychiatry 2018; 82:136-168. [PMID: 29180230 DOI: 10.1016/j.pnpbp.2017.11.020] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 10/18/2017] [Accepted: 11/23/2017] [Indexed: 01/02/2023]
Abstract
The identification of key genes in transcriptomic data constitutes a huge challenge. Our review of microarray reports revealed 88 genes whose transcription is consistently regulated by glucocorticoids (GCs), such as cortisol, corticosterone and dexamethasone, in the brain. Replicable transcriptomic data were combined with biochemical and physiological data to create an integrated view of the effects induced by GCs. The most frequently reported genes were Errfi1 and Ddit4. Their up-regulation was associated with the altered transcription of genes regulating growth factor and mTORC1 signaling (Gab1, Tsc22d3, Dusp1, Ndrg2, Ppp5c and Sesn1) and progression of the cell cycle (Ccnd1, Cdkn1a and Cables1). The GC-induced reprogramming of cell function involves changes in the mRNA level of genes responsible for the regulation of transcription (Klf9, Bcl6, Klf15, Tle3, Cxxc5, Litaf, Tle4, Jun, Sox4, Sox2, Sox9, Irf1, Sall2, Nfkbia and Id1) and the selective degradation of mRNA (Tob2). Other genes are involved in the regulation of metabolism (Gpd1, Aldoc and Pdk4), actin cytoskeleton (Myh2, Nedd9, Mical2, Rhou, Arl4d, Osbpl3, Arhgef3, Sdc4, Rdx, Wipf3, Chst1 and Hepacam), autophagy (Eva1a and Plekhf1), vesicular transport (Rhob, Ehd3, Vps37b and Scamp2), gap junctions (Gjb6), immune response (Tiparp, Mertk, Lyve1 and Il6r), signaling mediated by thyroid hormones (Thra and Sult1a1), calcium (Calm2), adrenaline/noradrenaline (Adcy9 and Adra1d), neuropeptide Y (Npy1r) and histamine (Hdc). GCs also affected genes involved in the synthesis of polyamines (Azin1) and taurine (Cdo1). The actions of GCs are restrained by feedback mechanisms depending on the transcription of Sgk1, Fkbp5 and Nr3c1. A side effect induced by GCs is increased production of reactive oxygen species. Available data show that the brain's response to GCs is part of an emergency mode characterized by inactivation of non-core activities, restrained inflammation, restriction of investments (growth), improved efficiency of energy production and the removal of unnecessary or malfunctioning cellular components to conserve energy and maintain nutrient supply during the stress response.
Collapse
Affiliation(s)
- Grzegorz R Juszczak
- Department of Animal Behavior, Institute of Genetics and Animal Breeding, Jastrzebiec, ul. Postepu 36A, 05-552 Magdalenka, Poland.
| | - Adrian M Stankiewicz
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, Jastrzebiec, ul. Postepu 36A, 05-552 Magdalenka, Poland
| |
Collapse
|
48
|
BTG1 low expression in pancreatic ductal adenocarcinoma is associated with a poorer prognosis. Int J Biol Markers 2017; 33:189-194. [PMID: 29076521 DOI: 10.5301/ijbm.5000310] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE BTG1 is a member of the TOB/BTG protein family, which is a transducer of ErbB-2 and TOB2. It is known to inhibit tumor genesis, but its role in pancreatic ductal adenocarcinoma (PDAC) is still unknown. The purpose of this study is to investigate the expression of BTG1 protein in PDAC and to determine its prognostic significance. METHODS Immunohistochemistry is used to determine the protein expression of the BTG1 gene in 79 surgically resected PDAC. The association of BTG1 expression with all the patients' clinicopathologic parameters, including survival, was analyzed using statistical software. RESULTS High BTG1 expression was observed in 27.8% (22/79) of the PDAC tissues, which was significantly lower than the 58.2% (46/79) of corresponding normal adjacent noncancerous tissues by immunohistochemical staining (p<0.001).Through the stratified analysis, we found a significant difference of BTG1 expression in peri-neural invasion (p = 0.002), T stage (p = 0.000), N stage (p = 0.018), and tumor, node, and metastasis stage (p = 0.000). Univariate and multivariate Cox analysis revealed that BTG1 expression status was an independent prognostic factor in PDAC (p = 0.027). Moreover, overall survival was better in PDAC cases with higher rather than lower BTG1 expression (p = 0.027). CONCLUSIONS This study demonstrated for the first time that lower expression of BTG1 might be involved in the progression of PDAC, suggesting that BTG1 might be a novel prognostic marker and a target for therapy.
Collapse
|
49
|
He C, Yu T, Shi Y, Ma C, Yang W, Fang L, Sun M, Wu W, Xiao F, Guo F, Chen M, Yang H, Qian J, Cong Y, Liu Z. MicroRNA 301A Promotes Intestinal Inflammation and Colitis-Associated Cancer Development by Inhibiting BTG1. Gastroenterology 2017; 152:1434-1448.e15. [PMID: 28193514 DOI: 10.1053/j.gastro.2017.01.049] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 01/15/2017] [Accepted: 01/24/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Intestinal tissues from patients with inflammatory bowel disease (IBD) and colorectal cancer have increased expression of microRNA-301a (MIR301A) compared with tissues from patients without IBD. We studied the mechanisms of MIR301A in the progression of IBD in human tissues and mice. METHODS We isolated intestinal epithelial cells (IECs) from biopsy samples of the colon from 153 patients with different stages of IBD activity, 6 patients with colitis-associated cancer (CAC), and 35 healthy individuals (controls), enrolled in the study in Shanghai, China. We measured expression of MIR301A and BTG anti-proliferation factor 1 (BTG1) by IECs using quantitative reverse-transcription polymerase chain reaction. Human colon cancer cell lines (HCT-116 and SW480) were transfected with a lentivirus that expresses MIR301A; expression of cytokines and tight junction proteins were measured by quantitative reverse transcription polymerase chain reaction, flow cytometry, and immunofluorescence staining. We generated mice with disruption of the microRNA-301A gene (MIR301A-knockout mice), and also studied mice that express a transgene-encoding BTG1. Colitis was induced in knockout, transgenic, and control (C57BL/B6) mice by administration of dextran sulfate sodium (DSS), and mice were given azoxymethane to induce colorectal carcinogenesis. Colons were collected and analyzed histologically and by immunohistochemistry; tumor nodules were counted and tumor size was measured. SW480 cells expressing the MIR301A transgene were grown as xenograft tumors in nude mice. RESULTS Expression of MIR301A increased in IECs from patients with IBD and CAC compared with controls. MIR301A-knockout mice were resistant to the development of colitis following administration of DSS; their colon tissues expressed lower levels of interleukin 1β (IL1β), IL6, IL8, and tumor necrosis factor than colons of control mice. Colon tissues from MIR301A-knockout mice had increased epithelial barrier integrity and formed fewer tumors following administration of azoxymethane than control mice. Human IECs expressing transgenic MIR301A down-regulated expression of cadherin 1 (also called E-cadherin or CDH1). We identified BTG1 mRNA as a target of MIR301A; levels of BTG1 mRNA were reduced in inflamed mucosa from patients with active IBD compared with controls. There was an inverse correlation between levels of BTG1 mRNA and levels of MIR301A in inflamed mucosal tissues from patients with active IBD. Human colon cancer cell lines that expressed a MIR301A transgene increased proliferation; they had increased permeability and decreased expression of CDH1 compared with cells transfected with a control vector, indicating reduced intestinal barrier function. BTG1 transgenic mice developed less severe colitis than control mice following administration of DSS. SW480 cells expressing anti-MIR301A formed fewer xenograft tumors in nude mice than cells expressing a control vector. CONCLUSIONS Levels of MIR301A are increased in IECs from patients with active IBD. MIR301A reduces expression of BTG1 to reduce epithelial integrity and promote inflammation in mouse colon and promotes tumorigenesis. Strategies to decrease levels of MIR301A in colon tissues might be developed to treat patients with IBD and CAC.
Collapse
Affiliation(s)
- Chong He
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Tianming Yu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Yan Shi
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Caiyun Ma
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Wenjing Yang
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Leilei Fang
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Mingming Sun
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Wei Wu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Fei Xiao
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, the Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Feifan Guo
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, the Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hong Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiaming Qian
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX.
| | - Zhanju Liu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
| |
Collapse
|
50
|
Kakaradov B, Arsenio J, Widjaja CE, He Z, Aigner S, Metz PJ, Yu B, Wehrens EJ, Lopez J, Kim SH, Zuniga EI, Goldrath AW, Chang JT, Yeo GW. Early transcriptional and epigenetic regulation of CD8 + T cell differentiation revealed by single-cell RNA sequencing. Nat Immunol 2017; 18:422-432. [PMID: 28218746 PMCID: PMC5360497 DOI: 10.1038/ni.3688] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 01/13/2017] [Indexed: 12/13/2022]
Abstract
During microbial infection, responding CD8+ T lymphocytes differentiate into heterogeneous subsets that together provide immediate and durable protection. To elucidate the dynamic transcriptional changes that underlie this process, we applied a single-cell RNA-sequencing approach and analyzed individual CD8+ T lymphocytes sequentially throughout the course of a viral infection in vivo. Our analyses revealed a striking transcriptional divergence among cells that had undergone their first division and identified previously unknown molecular determinants that controlled the fate specification of CD8+ T lymphocytes. Our findings suggest a model for the differentiation of terminal effector cells initiated by an early burst of transcriptional activity and subsequently refined by epigenetic silencing of transcripts associated with memory lymphocytes, which highlights the power and necessity of single-cell approaches.
Collapse
Affiliation(s)
- Boyko Kakaradov
- Department of Cellular and Molecular Medicine, University of California, San Diego, California, USA
| | - Janilyn Arsenio
- Department of Medicine, University of California, San Diego, California, USA
| | | | - Zhaoren He
- Department of Cellular and Molecular Medicine, University of California, San Diego, California, USA
| | - Stefan Aigner
- Department of Cellular and Molecular Medicine, University of California, San Diego, California, USA
| | - Patrick J Metz
- Department of Medicine, University of California, San Diego, California, USA
| | - Bingfei Yu
- Division of Biological Sciences, University of California, San Diego, California, USA
| | - Ellen J Wehrens
- Division of Biological Sciences, University of California, San Diego, California, USA
| | - Justine Lopez
- Department of Medicine, University of California, San Diego, California, USA
| | - Stephanie H Kim
- Department of Medicine, University of California, San Diego, California, USA
| | - Elina I Zuniga
- Division of Biological Sciences, University of California, San Diego, California, USA
| | - Ananda W Goldrath
- Division of Biological Sciences, University of California, San Diego, California, USA
| | - John T Chang
- Department of Medicine, University of California, San Diego, California, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California, San Diego, California, USA.,Institute for Genomic Medicine, University of California, San Diego, California, USA.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|