1
|
Abohassan M, Khaleel AQ, Pallathadka H, Kumar A, Allela OQB, Hjazi A, Pramanik A, Mustafa YF, Hamzah HF, Mohammed BA. Circular RNA as a Biomarker for Diagnosis, Prognosis and Therapeutic Target in Acute and Chronic Lymphoid Leukemia. Cell Biochem Biophys 2024; 82:1979-1991. [PMID: 39136839 DOI: 10.1007/s12013-024-01404-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2024] [Indexed: 10/02/2024]
Abstract
Circular RNAs (circRNAs) are single-stranded RNAs that have received much attention in recent years. CircRNAs lack a 5' head and a 3' poly-A tail. The structure of this type of RNAs make them resistant to digestion by exonucleases. CircRNAs are expressed in different cells and have various functions. The function of circRNAs is done by sponging miRNAs, changing gene expression, and protein production. The expression of circRNAs changes in different types of cancers, which causes changes in cell growth, proliferation, differentiation, and apoptosis. Changes in the expression of circRNAs can cause the invasion and progression of tumors. Studies have shown that changes in the expression of circRNAs can be seen in acute lymphoid leukemia (ALL) and chronic lymphoid leukemia (CLL). The conducted studies aim to identify circRNAs whose expression has changed in these leukemias and their more precise function so that these circRNAs can be identified as biomarkers, prediction of patient prognosis, and treatment targets for ALL and CLL patients. In this study, we review the studies conducted on the role and function of circRNAs in ALL and CLL patients. The results of the studies show that there is a possibility of using circRNAs as biomarkers in the identification and treatment of patients in the future.
Collapse
MESH Headings
- Humans
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Prognosis
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- RNA/metabolism
- RNA/genetics
- MicroRNAs/genetics
- MicroRNAs/metabolism
Collapse
Affiliation(s)
- Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Abdulrahman Qais Khaleel
- Department of Medical Instruments Engineering, Al-Maarif University College, Al Anbar, 31001, Iraq.
| | | | - Ashwani Kumar
- Department of Life Sciences, School of Sciences, Jain (Deemed-to-be) University, Bengaluru, Karnataka, 560069, India
- Department of Pharmacy, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | | | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Ivison of Research and Innovation Uttaranchal University, Dehradun, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Hamza Fadhel Hamzah
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | | |
Collapse
|
2
|
Xu X, Yu C, He H, Pan X, Hou A, Feng J, Tan R, Gong L, Chen J, Ren J. MiR-337-3p improves metabolic-associated fatty liver disease through regulation of glycolipid metabolism. iScience 2023; 26:108352. [PMID: 38026196 PMCID: PMC10665915 DOI: 10.1016/j.isci.2023.108352] [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: 05/17/2023] [Revised: 07/26/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Epigenetic regulations play crucial roles in the pathogenesis of metabolic-associated fatty liver disease; therefore, elucidating the biological functions of differential miRNAs helps us to understand the pathogenesis. Herein, we discovered miR-337-3p was decreased in patients with NAFLD from Gene Expression Omnibus dataset, which was replicated in various cell and mouse models with lipid disorders. Subsequently, overexpression of miR-337-3p in vivo could ameliorate hepatic lipid accumulation, reduce fasting blood glucose, and improve insulin resistance. Meanwhile, we determined miR-337-3p might influence multiple genes involved in glycolipid metabolism through mass spectrometry detection, bioinformatics analysis, and experimental verification. Finally, we selected HMGCR as a representative example to investigate the molecular mechanism of miR-337-3p regulating these genes, where the seed region of miR-337-3p bound to 3'UTR of HMGCR to inhibit HMGCR translation. In conclusion, we discovered a new function of miR-337-3p in glycolipid metabolism and that might be a new therapeutic target of MAFLD.
Collapse
Affiliation(s)
- Xiaoding Xu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Chuwei Yu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Hongxiu He
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Xiangyu Pan
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, P.R. China
| | - Aijun Hou
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Jianxun Feng
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
| | - Rongrong Tan
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
| | - Likun Gong
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Jing Chen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Jin Ren
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| |
Collapse
|
3
|
Gannoun L, De Schrevel C, Belle M, Dauguet N, Achouri Y, Loriot A, Vanderaa C, Cordi S, Dili A, Heremans Y, Rooman I, Leclercq IA, Jacquemin P, Gatto L, Lemaigre FP. Axon guidance genes control hepatic artery development. Development 2023; 150:dev201642. [PMID: 37497580 DOI: 10.1242/dev.201642] [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: 02/03/2023] [Accepted: 07/19/2023] [Indexed: 07/28/2023]
Abstract
Earlier data on liver development demonstrated that morphogenesis of the bile duct, portal mesenchyme and hepatic artery is interdependent, yet how this interdependency is orchestrated remains unknown. Here, using 2D and 3D imaging, we first describe how portal mesenchymal cells become organised to form hepatic arteries. Next, we examined intercellular signalling active during portal area development and found that axon guidance genes are dynamically expressed in developing bile ducts and portal mesenchyme. Using tissue-specific gene inactivation in mice, we show that the repulsive guidance molecule BMP co-receptor A (RGMA)/neogenin (NEO1) receptor/ligand pair is dispensable for portal area development, but that deficient roundabout 2 (ROBO2)/SLIT2 signalling in the portal mesenchyme causes reduced maturation of the vascular smooth muscle cells that form the tunica media of the hepatic artery. This arterial anomaly does not impact liver function in homeostatic conditions, but is associated with significant tissular damage following partial hepatectomy. In conclusion, our work identifies new players in development of the liver vasculature in health and liver regeneration.
Collapse
Affiliation(s)
- Lila Gannoun
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| | - Catalina De Schrevel
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| | - Morgane Belle
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Department of Development, Rue Moreau 17, Paris 75012, France
| | - Nicolas Dauguet
- Flow cytometry CYTF platform, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Younes Achouri
- Transgene Technology Platform TRSG, Université Catholique de Louvain, Brussels, Avenue Hippocrate 75, Belgium 1200
| | - Axelle Loriot
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| | - Christophe Vanderaa
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| | - Sabine Cordi
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| | - Alexandra Dili
- HPB Surgery Unit, Centre Hospitalier Universitaire UCL Namur, Site Mont-Godinne, Avenue du Dr. Thérasse 1, Yvoir 5530, Belgium
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Avenue Mounier 53, Brussels 1200, Belgium
| | - Yves Heremans
- Visual & Spatial Tissue Analysis (VSTA) core facility, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels 1090, Belgium
| | - Ilse Rooman
- Laboratory of Medical and Molecular Oncology, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels 1090, Belgium
| | - Isabelle A Leclercq
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Avenue Mounier 53, Brussels 1200, Belgium
| | - Patrick Jacquemin
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| | - Laurent Gatto
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| | - Frédéric P Lemaigre
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| |
Collapse
|
4
|
Dynamics of hepatocyte-cholangiocyte cell-fate decisions during liver development and regeneration. iScience 2022; 25:104955. [PMID: 36060070 PMCID: PMC9437857 DOI: 10.1016/j.isci.2022.104955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/17/2022] [Accepted: 08/12/2022] [Indexed: 11/25/2022] Open
Abstract
The immense regenerative potential of the liver is attributed to the ability of its two key cell types – hepatocytes and cholangiocytes – to trans-differentiate to one another either directly or through intermediate progenitor states. However, the dynamic features of decision-making between these cell-fates during liver development and regeneration remains elusive. Here, we identify a core gene regulatory network comprising c/EBPα, TGFBR2, and SOX9 which is multistable in nature, enabling three distinct cell states – hepatocytes, cholangiocytes, and liver progenitor cells (hepatoblasts/oval cells) – and stochastic switching among them. Predicted expression signature for these three states are validated through multiple bulk and single-cell transcriptomic datasets collected across developmental stages and injury-induced liver repair. This network can also explain the experimentally observed spatial organization of phenotypes in liver parenchyma and predict strategies for efficient cellular reprogramming. Our analysis elucidates how the emergent dynamics of underlying regulatory networks drive diverse cell-fate decisions in liver development and regeneration. Identified minimal regulatory network to model liver development and regeneration Changes in phenotypic landscapes by in-silico perturbations of regulatory networks Ability to explain physiological spatial patterning of liver cell types Decoded strategies for efficient reprogramming among liver cell phenotypes
Collapse
|
5
|
Liang S, Huang G, Wu T, Peng Y, Liu X, Ji X, Sha W, Wang F, Shen L, Shen H. MIR337-3p Enhances Mycobacterial Pathogenicity Involving TLR4/MYD88 and STAT3 Signals, Impairing VDR Antimicrobial Response and Fast-Acting Immunity. Front Immunol 2021; 12:739219. [PMID: 34912331 PMCID: PMC8666424 DOI: 10.3389/fimmu.2021.739219] [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: 07/10/2021] [Accepted: 11/04/2021] [Indexed: 11/18/2022] Open
Abstract
Active form of vitamin D (VitD) enhances human innate immunity against Mycobacterium tuberculosis (Mtb) infection. Our previous studies showed that MIR337-3p was highly expressed in lymphocytes of tuberculosis (TB) patients. Here, we identified the mechanism of MIR337-3p in the regulation of fast-acting anti-TB immunity by inhibiting VitD-dependent antimicrobial response pathways. While high-level MIR337-3p expression was induced by mycobacterial infection in cellular models and mice, TB patients exhibited significantly increased MIR337-3p in CD14+ monocytes/macrophages, innate-like Vγ2+ T cells, and CD8+ lymphocytes containing natural killer (NK)/innate lymphoid cells. MIR337-3p promoted the mycobacterial entry/infection and replication/growth in host target cells: macrophages and lung epithelial cells. Such MIR337-3p-enhanced pathogenicity coincided with the MIR337-3p depression of VitD-dependent antimicrobial response of cytochrome P450, family 27, subfamily b, polypeptide 1 (CYP27B1)/Beta-defensin 4 (DEFB4A)/ cathelicidin antimicrobial peptide CAMP pathways. Surprisingly, single MIR337-3p species could specifically target both the Toll-like receptor 4 (TLR4) and signal transducer and activator of transcription 3 (STAT3) 3′-untranslated regions (UTRs) to depress the TLR4/MYD88 and STAT3 signals and impair either of the two signals inhibiting the VitD-dependent antimicrobial pathways in macrophages. Concurrently, human peripheral blood mononuclear cells (PBMCs) expressing high-level MIR337-3p exhibited a reduced ability of innate cell populations to mount fast-acting cellular immunity against intracellular mycobacterial infection. Furthermore, a higher expression of Mir337-3p after mycobacterial infection of mice coincided with much greater colony-forming unit (CFU) counts in lungs and even the death of infected animals, whereas Mir337-3p inhibitor treatment of infected mice reduced Mir337-3p levels and reversed Mir337-3p-mediated increases in CFU counts. Thus, TB-driven single MIR337-3p species could specifically target/impair both TLR4/MYD88 and STAT3 activation signals, inhibiting VitD-dependent antimicrobial response and fast-acting anti-TB immunity, leading to enhanced pathogenicity.
Collapse
Affiliation(s)
- Shanshan Liang
- Clinic and Research Center of Tuberculosis, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Guixian Huang
- Clinic and Research Center of Tuberculosis, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Tian Wu
- Clinic and Research Center of Tuberculosis, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Ying Peng
- Clinic and Research Center of Tuberculosis, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Xi Liu
- Clinic and Research Center of Tuberculosis, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Xuejiao Ji
- Clinic and Research Center of Tuberculosis, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Wei Sha
- Clinic and Research Center of Tuberculosis, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Feifei Wang
- Key Laboratory of Medical Molecular Virology (Ministry of Education of the people's Republic of China (MOE)/National Health Commission of the people's Republic of China (NHC)/Chinese Academy of Medical Sciences (CAMS)), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ling Shen
- Department of Microbiology & Immunology and Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, United States
| | - Hongbo Shen
- Clinic and Research Center of Tuberculosis, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Wu J, Nagy LE, Liangpunsakul S, Wang L. Non-coding RNA crosstalk with nuclear receptors in liver disease. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166083. [PMID: 33497819 PMCID: PMC7987766 DOI: 10.1016/j.bbadis.2021.166083] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 12/28/2020] [Accepted: 01/16/2021] [Indexed: 02/06/2023]
Abstract
The dysregulation of nuclear receptors (NRs) underlies the pathogenesis of a variety of liver disorders. Non-coding RNAs (ncRNAs) are defined as RNA molecules transcribed from DNA but not translated into proteins. MicroRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are two types of ncRNAs that have been extensively studied for regulating gene expression during diverse cellular processes. NRs as therapeutic targets in liver disease have been exemplified by the successful application of their pharmacological ligands in clinics. MiRNA-based reagents or drugs are emerging as flagship products in clinical trials. Advancing our understanding of the crosstalk between NRs and ncRNAs is critical to the development of diagnostic and therapeutic strategies. This review summarizes recent findings on the reciprocal regulation between NRs and ncRNAs (mainly on miRNAs and lncRNAs) and their implication in liver pathophysiology, which might be informative to the translational medicine of targeting NRs and ncRNAs in liver disease.
Collapse
Affiliation(s)
- Jianguo Wu
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America; Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, United States of America.
| | - Laura E Nagy
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America; Department of Gastroenterology and Hepatology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America; Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, United States of America
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States of America; Roudebush Veterans Administration Medical Center, Indianapolis, IN, United States of America; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Li Wang
- Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, CT, United States of America
| |
Collapse
|
7
|
Freeburg SH, Goessling W. Hepatobiliary Differentiation: Principles from Embryonic Liver Development. Semin Liver Dis 2020; 40:365-372. [PMID: 32526786 DOI: 10.1055/s-0040-1709679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Hepatocytes and biliary epithelial cells (BECs), the two endodermal cell types of the liver, originate from progenitor cells called hepatoblasts. Based principally on in vitro data, hepatoblasts are thought to be bipotent stem cells with the potential to produce both hepatocytes and BECs. However, robust in vivo evidence for this model has only recently emerged. We examine the molecular mechanisms that stimulate hepatoblast differentiation into hepatocytes or BECs. In the absence of extrinsic cues, the default fate of hepatoblasts is hepatocyte differentiation. Inductive cues from the hepatic portal vein, however, initiate transcription factor expression in hepatoblasts, driving biliary specification. Defining the mechanisms of hepatobiliary differentiation provides important insights into congenital disorders, such as Alagille syndrome, and may help to better characterize the poorly understood hepatic lineage relationships observed during regeneration from liver injury.
Collapse
Affiliation(s)
- Scott H Freeburg
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Wolfram Goessling
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts.,Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard-MIT Division of Health Science and Technology, Cambridge, Massachusetts.,Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
8
|
Wu W, Wu Z, Xia Y, Qin S, Li Y, Wu J, Liang J, Wang L, Zhu H, Fan L, Fu J, Xu W, Jin H, Li J. Downregulation of circ_0132266 in chronic lymphocytic leukemia promoted cell viability through miR-337-3p/PML axis. Aging (Albany NY) 2020; 11:3561-3573. [PMID: 31152142 PMCID: PMC6594798 DOI: 10.18632/aging.101997] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/23/2019] [Indexed: 12/26/2022]
Abstract
Circular RNAs (circRNAs) have recently been reported to play crucial roles in various regulatory processes and involved in cancer onset and progression. However, the potential mechanism of circRNAs in chronic lymphocytic leukemia (CLL) remains largely unknown. Here, we observed hsa_circ_0132266 (circ_0132266), a circRNA significantly decreased in the peripheral blood mononuclear cells (PBMCs) of CLL patients compared with healthy donors, could act as an endogenous sponge of hsa-miR-337-3p (miR-337-3p) and regulate its activity, which resulted in a downstream change of target-gene PML and a consequent influence on cell viability. Taken together, our data indicated the regulatory mechanism of circ_0132266 in CLL progression through circ_0132266/miR-337-3p/PML axis, suggesting that it may serve as a biomarker as well as an exploitable therapeutic target for CLL.
Collapse
Affiliation(s)
- Wei Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Zijuan Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Yi Xia
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Shuchao Qin
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Yue Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Jiazhu Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Jinhua Liang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Huayuan Zhu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Lei Fan
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Jianxin Fu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Wei Xu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Hui Jin
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Jianyong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| |
Collapse
|
9
|
Wei J, Liu X, Li T, Xing P, Zhang C, Yang J. The new horizon of liquid biopsy in sarcoma: the potential utility of circulating tumor nucleic acids. J Cancer 2020; 11:5293-5308. [PMID: 32742476 PMCID: PMC7391194 DOI: 10.7150/jca.42816] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
The diagnosis, treatment and prognosis of sarcoma are mainly dependent on tissue biopsy, which is limited in its ability to provide a panoramic view into the dynamics of tumor progression. In addition, effective biomarkers to monitor the progression and therapeutic response of sarcoma are lacking. Liquid biopsy, a recent technological breakthrough, has gained great attention in the last few decades. Nucleic acids (such as DNA, mRNAs, microRNAs, and long non-coding RNAs) that are released from tumors circulate in the blood of cancer patients and can be evaluated through liquid biopsy. Circulating tumor nucleic acids reflect the intertumoral and intratumoral heterogeneity, and thus liquid biopsy provides a noninvasive strategy to examine these molecules compared with traditional tissue biopsy. Over the past decade, a great deal of information on the potential utilization of circulating tumor nucleic acids in sarcoma screening, prognosis and therapy efficacy monitoring has emerged. Several specific gene mutations in sarcoma can be detected in peripheral blood samples from patients and can be found in circulating tumor DNA to monitor sarcoma. In addition, circulating tumor non-coding RNA may also be a promising biomarker in sarcoma. In this review, we discuss the clinical application of circulating tumor nucleic acids as blood-borne biomarkers in sarcoma.
Collapse
Affiliation(s)
- Junqiang Wei
- Department of bone and soft tissue tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin's Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- Department of Orthopedics, Affiliated Hospital of Chengde Medical College, Chengde, Hebei, 067000, China
| | - Xinyue Liu
- Department of bone and soft tissue tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin's Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Ting Li
- Department of bone and soft tissue tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin's Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Peipei Xing
- Department of bone and soft tissue tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin's Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Chao Zhang
- Department of bone and soft tissue tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin's Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Jilong Yang
- Department of bone and soft tissue tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin's Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| |
Collapse
|
10
|
Lemaigre FP. Development of the Intrahepatic and Extrahepatic Biliary Tract: A Framework for Understanding Congenital Diseases. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2019; 15:1-22. [PMID: 31299162 DOI: 10.1146/annurev-pathmechdis-012418-013013] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The involvement of the biliary tract in the pathophysiology of liver diseases and the increased attention paid to bile ducts in the bioconstruction of liver tissue for regenerative therapy have fueled intense research into the fundamental mechanisms of biliary development. Here, I review the molecular, cellular and tissular mechanisms driving differentiation and morphogenesis of the intrahepatic and extrahepatic bile ducts. This review focuses on the dynamics of the transcriptional and signaling modules that promote biliary development in human and mouse liver and discusses studies in which the use of zebrafish uncovered unexplored processes in mammalian biliary development. The review concludes by providing a framework for interpreting the mechanisms that may help us understand the origin of congenital biliary diseases.
Collapse
Affiliation(s)
- Frédéric P Lemaigre
- de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium;
| |
Collapse
|
11
|
Xia L, Wu L, Xia H, Bao J, Li Q, Chen X, Xia R. miR-337 suppresses cutaneous T-cell lymphoma via the STAT3 pathway. Cell Cycle 2019; 18:1635-1645. [PMID: 31213131 DOI: 10.1080/15384101.2019.1629789] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cutaneous T-cell lymphoma (CTCL) is associated with the downregulation of miR-337 expression, although the exact underlying mechanism is unknown. In the present work, we investigated the molecular mechanism and function of miR-337 in regulating CTCL cell viability and invasion. We observed that miR-337 expression was downregulated in both CTCL tumors and cell lines. Furthermore, CCK assay, BrdU incorporation assay, and flow cytometry revealed that transfection with the miR-337 mimic resulted in decreased proliferation and increased apoptotic levels in CTCL cells. Results of the Transwell migration assay indicated that the miR-337 mimic decreased CTCL cell invasion in vitro. Both bioinformatics prediction and the dual-luciferase reporter assay revealed that miR-337 targets the 3'-UTR of STAT3 for silencing. Overexpression of STAT3 counteracted the pro-apoptotic influence of miR-337 in CTCL cell lines and restored their invasion properties. The results thus indicate that the miR-337-STAT3 axis inhibits the proliferation of malignant T cells and that miR-337 may serve as a promising therapeutic target for CTCL.
Collapse
Affiliation(s)
- Liang Xia
- a Department of Hematology , The First Affiliated Hospital of Anhui Medical University , Hefei , China
| | - Linlin Wu
- b Department of Hematology , Anhui Provincial NO. 2 People's Hospital , Hefei , China
| | - Hailong Xia
- a Department of Hematology , The First Affiliated Hospital of Anhui Medical University , Hefei , China
| | - Jing Bao
- a Department of Hematology , The First Affiliated Hospital of Anhui Medical University , Hefei , China
| | - Qingsheng Li
- a Department of Hematology , The First Affiliated Hospital of Anhui Medical University , Hefei , China
| | - Xiaowen Chen
- a Department of Hematology , The First Affiliated Hospital of Anhui Medical University , Hefei , China
| | - Ruixiang Xia
- a Department of Hematology , The First Affiliated Hospital of Anhui Medical University , Hefei , China
| |
Collapse
|