1
|
Aloliqi AA, Alnuqaydan AM, Albutti A, Alharbi BF, Rahmani AH, Khan AA. Current updates regarding biogenesis, functions and dysregulation of microRNAs in cancer: Innovative approaches for detection using CRISPR/Cas13‑based platforms (Review). Int J Mol Med 2025; 55:90. [PMID: 40242952 PMCID: PMC12021393 DOI: 10.3892/ijmm.2025.5531] [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: 01/15/2025] [Accepted: 03/04/2025] [Indexed: 04/18/2025] Open
Abstract
MicroRNAs (miRNAs) are short non‑coding RNAs, which perform a key role in cellular differentiation and development. Most human diseases, particularly cancer, are linked to miRNA functional dysregulation implicated in the expression of tumor‑suppressive or oncogenic targets. Cancer hallmarks such as continued proliferative signaling, dodging growth suppressors, invasion and metastasis, triggering angiogenesis, and avoiding cell death have all been demonstrated to be affected by dysregulated miRNAs. Thus, for the treatment of different cancer types, the detection and quantification of this type of RNA is significant. The classical and current methods of RNA detection, including northern blotting, reverse transcription‑quantitative PCR, rolling circle amplification and next‑generation sequencing, may be effective but differ in efficiency and accuracy. Furthermore, these approaches are expensive, and require special instrumentation and expertise. Thus, researchers are constantly looking for more innovative approaches for miRNA detection, which can be advantageous in all aspects. In this regard, an RNA manipulation tool known as the CRISPR and CRISPR‑associated sequence 13 (CRISPR/Cas13) system has been found to be more advantageous in miRNA detection. The Cas13‑based miRNA detection approach is cost effective and requires no special instrumentation or expertise. However, more research and validation are required to confirm the growing body of CRISPR/Cas13‑based research that has identified miRNAs as possible cancer biomarkers for diagnosis and prognosis, and as targets for treatment. In the present review, current updates regarding miRNA biogenesis, structural and functional aspects, and miRNA dysregulation during cancer are described. In addition, novel approaches using the CRISPR/Cas13 system as a next‑generation tool for miRNA detection are discussed. Furthermore, challenges and prospects of CRISPR/Cas13‑based miRNA detection approaches are described.
Collapse
Affiliation(s)
- Abdulaziz A. Aloliqi
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Abdullah M. Alnuqaydan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Aqel Albutti
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Basmah F. Alharbi
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| |
Collapse
|
2
|
Bahmani F, Shayanmanesh M, Safari M, Alaei A, Yasaman Pouriafar, Rasti Z, Zaker F, Rostami S, Damerchiloo F, Safa M. Bone marrow microenvironment in myelodysplastic neoplasms: insights into pathogenesis, biomarkers, and therapeutic targets. Cancer Cell Int 2025; 25:175. [PMID: 40349084 PMCID: PMC12065391 DOI: 10.1186/s12935-025-03793-z] [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: 12/26/2024] [Accepted: 04/17/2025] [Indexed: 05/14/2025] Open
Abstract
Myelodysplastic neoplasms (MDS) represent a heterogeneous group of malignant hematopoietic stem and progenitor cell (HSPC) disorders characterized by cytopenia, ineffective hematopoiesis, as well as the potential to progress to acute myeloid leukemia (AML). The pathogenesis of MDS is influenced by intrinsic factors, such as genetic insults, and extrinsic factors, including altered bone marrow microenvironment (BMM) composition and architecture. BMM is reprogrammed in MDS, initially to prevent the development of the disease but eventually to provide a survival advantage to dysplastic cells. Recently, inflammation or age-related inflammation in the bone marrow has been identified as a key pathogenic mechanism for MDS. Inflammatory signals trigger stress hematopoiesis, causing HSPCs to emerge from quiescence and resulting in MDS development. A better understanding of the role of the BMM in the pathogenesis of MDS has opened up new avenues for improving diagnosis, prognosis, and treatment of the disease. This article provides a comprehensive review of the current knowledge regarding the significance of the BMM to MDS pathophysiology and highlights recent advances in developing innovative therapies.
Collapse
Affiliation(s)
- Forouzan Bahmani
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Shayanmanesh
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahdi Safari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirarsalan Alaei
- Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Yasaman Pouriafar
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Rasti
- Department of Hematology, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Zaker
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shahrbano Rostami
- Research Institute for Oncology, Hematology and Cell Therapy, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Damerchiloo
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Gharaee N, Wegrzyn-Woltosz J, Jiang J, Akhade VS, Bridgers J, Stubbins RJ, Hiwase D, Kutyna MM, Chan O, Komrokji R, Padron E, Deng Y, Cole G, Umlandt P, Fuller M, Kim A, Karsan A. Haploinsufficiency of miR-143 and miR-145 reveal targetable dependencies in resistant del(5q) myelodysplastic neoplasm. Leukemia 2025; 39:917-928. [PMID: 40000845 PMCID: PMC11976265 DOI: 10.1038/s41375-025-02537-2] [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: 04/28/2024] [Revised: 12/21/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
Myelodysplastic neoplasms (MDS) are stem cell disorders characterized by ineffective hematopoiesis and risk of transformation to acute myeloid leukemia (AML). Chromosomal alterations are frequent in MDS, with interstitial deletion of chromosome 5q (del(5q)) being the most common. Lenalidomide is the current first-line treatment for del(5q) MDS and its efficacy relies on degradation of CK1α which is encoded by the CSNK1A1 gene located in the commonly deleted region (CDR) of chromosome 5q. However, lenalidomide-resistance is common, often secondary to loss-of-function mutations in TP53 or RUNX1. The CDR in del(5q) harbors several genes, including noncoding miRNAs, the loss of which contribute to disease phenotypes. miR-143 and miR-145 are located within the del(5q) CDR, but precise understanding of their role in human hematopoiesis and in the pathogenesis of del(5q) MDS is lacking. Here we provide evidence that deficiency of miR-143 and miR-145 plays a role in clonal expansion of del(5q) MDS. We show that insulin-like growth factor 1 receptor (IGF-1R) is a direct target of both miR-143 and miR-145. Our data demonstrate that IGF-1R inhibition reduces proliferation and viability of del(5q) cells in vitro and in vivo, and that lenalidomide-resistant del(5q) MDS cells depleted of either TP53 or RUNX1 are sensitive to IGF-1R inhibition. Resistant del(5q) MDS-L cells, as well as primary MDS marrow cells, are also sensitive to targeting of IGF-1R-related dependencies in del(5q) MDS, which include the Abl and MAPK signaling pathways. This work thus provides potential new therapeutic avenues for lenalidomide-resistant del(5q) MDS.
Collapse
Affiliation(s)
- Nadia Gharaee
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Joanna Wegrzyn-Woltosz
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jihong Jiang
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Vijay Suresh Akhade
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Joshua Bridgers
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Ryan J Stubbins
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
- Leukemia/BMT Program of BC, BC Cancer and Vancouver Coastal Health, Vancouver, BC, Canada
- Division of Hematology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Devendra Hiwase
- Department of Haematology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Monika M Kutyna
- Department of Haematology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | | | | | | | - Yu Deng
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gary Cole
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Patricia Umlandt
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Megan Fuller
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Ada Kim
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Aly Karsan
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada.
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
4
|
Stubbins RJ, Cherniawsky H, Karsan A. Cellular and immunotherapies for myelodysplastic syndromes. Semin Hematol 2024; 61:397-408. [PMID: 39426936 DOI: 10.1053/j.seminhematol.2024.09.006] [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: 08/21/2024] [Accepted: 09/17/2024] [Indexed: 10/21/2024]
Abstract
In this review article, we outline the current landscape of immune and cell therapy-based approaches for patients with myelodysplastic syndromes (MDS). Given the well characterized graft-versus-leukemia (GVL) effect observed with allogeneic hematopoietic cell transplantation, and the known immune escape mechanisms observed in MDS cells, significant interest exists in developing immune-based approaches to treat MDS. These attempts have included antibody-based drugs that block immune escape molecules, such as inhibitors of the PD-1/PD-L1 and TIM-3/galectin-9 axes that mediate interactions between MDS cells and T-lymphocytes, as well as antibodies that block the CD47/SIRPα interaction, which mediates macrophage phagocytosis. Unfortunately, these approaches have been largely unsuccessful. There is significant potential for T-cell engaging therapies and chimeric antigen receptor T (CAR-T) cells, but there are also several limitations to these approaches that are unique to MDS. However, many of these limitations may be overcome by the next generation of cellular therapies, including those with engineered T-cell receptors or natural killer (NK)-cell based platforms. Regardless of the approach, all these immune cells are subject to the complex bone marrow microenvironment in MDS, which harbours a variable and heterogeneous mix of pro-inflammatory cytokines and immunosuppressive elements. Understanding this interaction will be paramount to ensuring the success of immune and cellular therapies in MDS.
Collapse
Affiliation(s)
- Ryan J Stubbins
- Leukemia/BMT Program of BC, BC Cancer, Vancouver V5Z 1M9, BC, Canada; Division of Hematology, Department of Medicine, University of British Columbia, Vancouver V5Z 1M9, BC, Canada.
| | - Hannah Cherniawsky
- Leukemia/BMT Program of BC, BC Cancer, Vancouver V5Z 1M9, BC, Canada; Division of Hematology, Department of Medicine, University of British Columbia, Vancouver V5Z 1M9, BC, Canada
| | - Aly Karsan
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 1L3, Canada.
| |
Collapse
|
5
|
Mohanraj L, Carter C, Liu J, Swift-Scanlan T. MicroRNA Profiles in Hematopoietic Stem Cell Transplant Recipients. Biol Res Nurs 2024; 26:559-568. [PMID: 38819871 DOI: 10.1177/10998004241257847] [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] [Indexed: 06/01/2024]
Abstract
Background: Hematopoietic Stem Cell Transplant (HCT) is a potentially curative treatment for hematologic malignancies, including multiple myeloma. Biomarker investigation can guide identification of HCT recipients at-risk for poor outcomes. MicroRNAs (miRNAs) are a class of non-coding RNAs involved in the modulation and regulation of pathological processes and are emerging as prognostic and predictive biomarkers for multiple health conditions. This pilot study aimed to examine miRNA profiles associated with HCT-related risk factors and outcomes in patients undergoing autologous HCT. Methods: Patients eligible for autologous HCT were recruited and blood samples and HCT-related variables were collected. Differential expression analysis of miRNA was conducted on 24 patient samples to compare changes in miRNA profile in HCT eligible patients before and after transplant. Results: Unsupervised clustering of differentially expressed (p < .05) miRNAs pre- and post- HCT identified clusters of up- and down-regulated miRNAs. Four miRNAs (miR-125a-5p, miR-99b-5p, miR-382-5p, miR-145-5p) involved in hematopoiesis (differentiation of progenitor cells, granulocyte function, thrombopoiesis, and tumor suppression) were significantly downregulated post-HCT. Correlation analyses identified select miRNAs associated with risk factors (such as frailty, fatigue, cognitive decline) and quality of life pre- and post-HCT. Select miRNAs were correlated with platelet engraftment. Conclusion: Future studies should examine miRNA signatures in larger cohorts in association with HCT outcomes; and expand investigations in patients receiving allogeneic transplants. This will lead to identification of biomarkers for risk stratification of HCT recipients.
Collapse
Affiliation(s)
- Lathika Mohanraj
- Department of Adult Health and Nursing Systems, School of Nursing, Virginia Commonwealth University, Richmond, VA, USA
| | - Christiane Carter
- Bioinformatics Shared Resource, Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Jinze Liu
- Department of Biostatistics, School of Population Health, Virginia Commonwealth University, Richmond, VA, USA
| | | |
Collapse
|
6
|
Liu M, Liu S, Qin L, Lv D, Wang G, Liu Q, Huang B, Zhang D. Global changes of miRNA expression indicates an increased reprogramming efficiency of induced mammary epithelial cells by repression of miR-222-3p in fibroblasts. PeerJ 2024; 12:e17657. [PMID: 39011384 PMCID: PMC11249016 DOI: 10.7717/peerj.17657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/08/2024] [Indexed: 07/17/2024] Open
Abstract
Background Our previous studies have successfully reported the reprogramming of fibroblasts into induced mammary epithelial cells (iMECs). However, the regulatory relationships and functional roles of MicroRNAs (miRNAs) in the progression of fibroblasts achieving the cell fate of iMECs are insufficiently understood. Methods First, we performed pre-and post-induction miRNAs sequencing analysis by using high-throughput sequencing. Following that, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment studies were used to determine the primary roles of the significantly distinct miRNAs and targeted genes. Finally, the effect of miR-222-3p on iMECs fate reprogramming in vitro by transfecting. Results As a result goat ear fibroblasts (GEFs) reprogramming into iMECs activates a regulatory program, involving 79 differentially expressed miRNAs. Besides, the programming process involved changes in multiple signaling pathways such as adherens junction, TGF-β signaling pathway, GnRH secretion and the prolactin signaling pathway, etc. Furthermore, it was discovered that the expression of miR-222-3p downregulation by miR-222-3p inhibitor significantly increase the reprogramming efficiency and promoted lipid accumulation of iMECs.
Collapse
Affiliation(s)
- Mingxing Liu
- Guangxi Key Laboratory of Eye Health, Guangxi Academy of Medical Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
- Guangxi University, School of Animal Science and Technology, Nanning, Guangxi, China
| | - Siyi Liu
- Guangxi University, School of Animal Science and Technology, Nanning, Guangxi, China
| | - Liangshan Qin
- Guangxi University, School of Animal Science and Technology, Nanning, Guangxi, China
| | - Danwei Lv
- Guangxi University, School of Animal Science and Technology, Nanning, Guangxi, China
| | - Guodong Wang
- Guangxi University, School of Animal Science and Technology, Nanning, Guangxi, China
| | - Quanhui Liu
- Guangxi University, School of Animal Science and Technology, Nanning, Guangxi, China
| | - Ben Huang
- Guangxi Key Laboratory of Eye Health, Guangxi Academy of Medical Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
- Guangxi University, School of Animal Science and Technology, Nanning, Guangxi, China
| | - Dandan Zhang
- Guangxi Key Laboratory of Eye Health, Guangxi Academy of Medical Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| |
Collapse
|
7
|
Wang Y, Yu J, Ou C, Zhao Y, Chen L, Cai W, Wang H, Huang S, Hu J, Sun G, Li L. miRNA-146a-5p Inhibits Hypoxia-Induced Myocardial Fibrosis Through EndMT. Cardiovasc Toxicol 2024; 24:133-145. [PMID: 38180639 DOI: 10.1007/s12012-023-09818-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024]
Abstract
Cardiac Vascular disease particularly myocardial infarction (MI) is a threat to health worldwide. microRNAs (miRNAs) have been shown to regulate myocardial fibrosis. Therefore, it is potential to investigate the mechanism of miRNA and fibrosis following myocardial infarction. Hypoxia human cardiac microvascular endothelial cells (HCMECs) were selected for the vitro experimental model. The miR-146a-5p expression was tested via RT-qPCR. The level of endothelial-to-mesenchymal transition (EndMT) and fibrosis markers were detected by Western blotting and immunofluorescence. Then, the inflammation, cell viability and apoptosis were investigated. The target was predicted by an online database and verified by a dual-luciferase activity assay. An MI mouse model was created to validate that miR-146a-5p regulates cardiac fibrosis in vivo. MI mouse was transfected with miR-146a-5p lentivirus. Subsequently, its effect on cardiac fibrosis of infarcted hearts was assessed by In situ hybridization (ISH), Immunohistochemistry (IHC), Triphenylterazolium chloride (TTC) staining and Masson staining. Herein, we confirmed that miR-146a-5p was down-regulated in hypoxia HCMECs. Overexpression of miR-146a-5p inhibited hypoxia-induced cardiac fibrosis following myocardial infarction by inhibiting EndMT in HCMECs. Thioredoxin-interacting protein (TXNIP) was a target that was negatively regulated by miR-146a-5p. Up-regulation of miR-146a-5p inhibited cardiac fibrosis via regulating EndMT by targeting TXNIP, and it also regulated EndMT to inhibit cardiac fibrosis in vivo.
Collapse
Affiliation(s)
- Yan Wang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Rd, Kunming, 650032, Yunnan, China.
| | - Jie Yu
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, No. 212 Daguan Rd, Kunming, 650032, Yunnan, China.
| | - Chunxia Ou
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Rd, Kunming, 650032, Yunnan, China
| | - Yue Zhao
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Rd, Kunming, 650032, Yunnan, China
| | - Lixing Chen
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Rd, Kunming, 650032, Yunnan, China
| | - Wenke Cai
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, No. 212 Daguan Rd, Kunming, 650032, Yunnan, China
| | - Huawei Wang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Rd, Kunming, 650032, Yunnan, China
| | - Shiying Huang
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Rd, Kunming, 650032, Yunnan, China
| | - Jie Hu
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Rd, Kunming, 650032, Yunnan, China
| | - Guihu Sun
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Rd, Kunming, 650032, Yunnan, China
| | - Longjun Li
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Rd, Kunming, 650032, Yunnan, China
| |
Collapse
|
8
|
Muto T, Walker CS, Agarwal P, Vick E, Sampson A, Choi K, Niederkorn M, Ishikawa C, Hueneman K, Varney M, Starczynowski DT. Inactivation of p53 provides a competitive advantage to del(5q) myelodysplastic syndrome hematopoietic stem cells during inflammation. Haematologica 2023; 108:2715-2729. [PMID: 37102608 PMCID: PMC10542836 DOI: 10.3324/haematol.2022.282349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 04/17/2023] [Indexed: 04/28/2023] Open
Abstract
Inflammation is associated with the pathogenesis of myelodysplastic syndromes (MDS) and emerging evidence suggests that MDS hematopoietic stem and progenitor cells (HSPC) exhibit an altered response to inflammation. Deletion of chromosome 5 (del(5q)) is the most common chromosomal abnormality in MDS. Although this MDS subtype contains several haploinsufficient genes that impact innate immune signaling, the effects of inflammation on del(5q) MDS HSPC remains undefined. Utilizing a model of del(5q)-like MDS, inhibiting the IRAK1/4-TRAF6 axis improved cytopenias, suggesting that activation of innate immune pathways contributes to certain clinical features underlying the pathogenesis of low-risk MDS. However, low-grade inflammation in the del(5q)-like MDS model did not contribute to more severe disease but instead impaired the del(5q)-like HSPC as indicated by their diminished numbers, premature attrition and increased p53 expression. Del(5q)-like HSPC exposed to inflammation became less quiescent, but without affecting cell viability. Unexpectedly, the reduced cellular quiescence of del(5q) HSPC exposed to inflammation was restored by p53 deletion. These findings uncovered that inflammation confers a competitive advantage of functionally defective del(5q) HSPC upon loss of p53. Since TP53 mutations are enriched in del(5q) AML following an MDS diagnosis, increased p53 activation in del(5q) MDS HSPC due to inflammation may create a selective pressure for genetic inactivation of p53 or expansion of a pre-existing TP53-mutant clone.
Collapse
Affiliation(s)
- Tomoya Muto
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Hematology, Chiba University Hospital, Chiba.
| | - Callum S Walker
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Puneet Agarwal
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Eric Vick
- Division of Hematology and Oncology, University of Cincinnati, Cincinnati, OH
| | - Avery Sampson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Kwangmin Choi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Madeline Niederkorn
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Chiharu Ishikawa
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Cancer Biology, University of Cincinnati, Cincinnati, OH
| | - Kathleen Hueneman
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Melinda Varney
- Department of Pharmaceutical Science and Research, Marshall University, Huntington, WV
| | - Daniel T Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Cancer Biology, University of Cincinnati, Cincinnati, OH; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; UC Cancer Center, Cincinnati, OH.
| |
Collapse
|
9
|
Georgoulis V, Koumpis E, Hatzimichael E. The Role of Non-Coding RNAs in Myelodysplastic Neoplasms. Cancers (Basel) 2023; 15:4810. [PMID: 37835504 PMCID: PMC10571949 DOI: 10.3390/cancers15194810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Myelodysplastic syndromes or neoplasms (MDS) are a heterogeneous group of myeloid clonal disorders characterized by peripheral blood cytopenias, blood and marrow cell dysplasia, and increased risk of evolution to acute myeloid leukemia (AML). Non-coding RNAs, especially microRNAs and long non-coding RNAs, serve as regulators of normal and malignant hematopoiesis and have been implicated in carcinogenesis. This review presents a comprehensive summary of the biology and role of non-coding RNAs, including the less studied circRNA, siRNA, piRNA, and snoRNA as potential prognostic and/or predictive biomarkers or therapeutic targets in MDS.
Collapse
Affiliation(s)
- Vasileios Georgoulis
- Department of Haematology, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, 45 500 Ioannina, Greece; (V.G.); (E.K.)
| | - Epameinondas Koumpis
- Department of Haematology, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, 45 500 Ioannina, Greece; (V.G.); (E.K.)
| | - Eleftheria Hatzimichael
- Department of Haematology, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, 45 500 Ioannina, Greece; (V.G.); (E.K.)
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19 107, USA
| |
Collapse
|
10
|
Ortiz GGR, Mohammadi Y, Nazari A, Ataeinaeini M, Kazemi P, Yasamineh S, Al-Naqeeb BZT, Zaidan HK, Gholizadeh O. A state-of-the-art review on the MicroRNAs roles in hematopoietic stem cell aging and longevity. Cell Commun Signal 2023; 21:85. [PMID: 37095512 PMCID: PMC10123996 DOI: 10.1186/s12964-023-01117-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/25/2023] [Indexed: 04/26/2023] Open
Abstract
Aging is a biological process determined through time-related cellular and functional impairments, leading to a decreased standard of living for the organism. Recently, there has been an unprecedented advance in the aging investigation, especially the detection that the rate of senescence is at least somewhat regulated via evolutionarily preserved genetic pathways and biological processes. Hematopoietic stem cells (HSCs) maintain blood generation over the whole lifetime of an organism. The senescence process influences many of the natural features of HSC, leading to a decline in their capabilities, independently of their microenvironment. New studies show that HSCs are sensitive to age-dependent stress and gradually lose their self-renewal and regeneration potential with senescence. MicroRNAs (miRNAs) are short, non-coding RNAs that post-transcriptionally inhibit translation or stimulate target mRNA cleavage of target transcripts via the sequence-particular connection. MiRNAs control various biological pathways and processes, such as senescence. Several miRNAs are differentially expressed in senescence, producing concern about their use as moderators of the senescence process. MiRNAs play an important role in the control of HSCs and can also modulate processes associated with tissue senescence in specific cell types. In this review, we display the contribution of age-dependent alterations, including DNA damage, epigenetic landscape, metabolism, and extrinsic factors, which affect HSCs function during aging. In addition, we investigate the particular miRNAs regulating HSCs senescence and age-associated diseases. Video Abstract.
Collapse
Affiliation(s)
- Geovanny Genaro Reivan Ortiz
- Laboratory of Basic Psychology, Behavioral Analysis and Programmatic Development (PAD-LAB), Catholic University of Cuenca, Cuenca, Ecuador
| | - Yasaman Mohammadi
- Faculty of Dentistry, Islamic Azad University, Shiraz Branch, Shiraz, Iran
| | - Ahmad Nazari
- Tehran University of Medical Sciences, Tehran, Iran
| | | | - Parisa Kazemi
- Faculty of Dentistry, Ilam University of Medical Sciences, Ilam, Iran
| | - Saman Yasamineh
- Stem Cell Research Center at, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Haider Kamil Zaidan
- Department of Medical Laboratories Techniques, Al-Mustaqbal University College, Hillah, Babylon, Iraq
| | - Omid Gholizadeh
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Wang F, Cai R, Tan W. Self-Powered Biosensor for a Highly Efficient and Ultrasensitive Dual-Biomarker Assay. Anal Chem 2023; 95:6046-6052. [PMID: 36976790 DOI: 10.1021/acs.analchem.3c00097] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
A dual-biomarker, self-powered biosensor was fabricated for the ultrasensitive detection of microRNA-21 (miRNA-21) and miRNA-155 based on enzymatic biofuel cells (EBFCs), catalytic hairpin assembly (CHA), and DNA hybridization chain reaction (HCR), with a capacitor and digital multimeter (DMM). In the presence of miRNA-21, the CHA and HCR are triggered and lead to the generation of a double-helix chain, which stimulates [Ru(NH3)6]3+ to move to the biocathode surface due to electrostatic interaction. Subsequently, the biocathode obtains electrons from the bioanode and reduces [Ru(NH3)6]3+ to [Ru(NH3)6]2+, which significantly increases the open-circuit voltage (E1OCV). When miRNA-155 is present, CHA and HCR cannot be completed, resulting in a low E2OCV. The self-powered biosensor allows for the simultaneous ultrasensitive detection of miRNA-21 and miRNA-155 with detection limits of 0.15 and 0.66 fM, respectively. Moreover, this self-powered biosensor exhibits the highly sensitive detection for miRNA-21 and miRNA-155 assay in human serum samples.
Collapse
Affiliation(s)
- Futing Wang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Material Science and Engineering, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha 410082, China
| | - Ren Cai
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Material Science and Engineering, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Material Science and Engineering, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha 410082, China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
12
|
Gonzalez-Lugo JD, Verma A. Targeting inflammation in lower-risk MDS. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:382-387. [PMID: 36485128 PMCID: PMC9821551 DOI: 10.1182/hematology.2022000350] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The myelodysplastic syndromes (MDS) are a heterogeneous group of malignant hematopoietic stem cell disorders characterized by ineffective growth and differentiation of hematopoietic progenitors leading to peripheral blood cytopenias, dysplasia, and a variable risk of transformation to acute myelogenous leukemia. As most patients present with lower-risk disease, understanding the pathogenesis of ineffective hematopoiesis is important for developing therapies that will increase blood counts in patients with MDS. Various inflammatory cytokines are elevated in MDS and contribute to dysplastic differentiation. Inflammatory pathways mediated by interleukin (IL) 1b, IL-6, IL-1RAP, IL-8, and others lead to growth of aberrant MDS stem and progenitors while inhibiting healthy hematopoiesis. Spliceosome mutations can lead to missplicing of genes such as IRAK4, CASP8, and MAP3K, which lead to activation of proinflammatory nuclear factor κB-driven pathways. Therapeutically, targeting of ligands of the transforming growth factor β (TGF-β) pathway has led to approval of luspatercept in transfusion-dependent patients with MDS. Presently, various clinical trials are evaluating inhibitors of cytokines and their receptors in low-risk MDS. Taken together, an inflammatory microenvironment can support the pathogenesis of clonal hematopoiesis and low-risk MDS, and clinical trials are evaluating anti-inflammatory strategies in these diseases.
Collapse
Affiliation(s)
- Jesus D Gonzalez-Lugo
- Division of Hemato-Oncology, Department of Oncology, Montefiore-Einstein Cancer Center, Blood Cancer Institute, Bronx, NY
| | - Amit Verma
- Division of Hemato-Oncology, Department of Oncology, Montefiore-Einstein Cancer Center, Blood Cancer Institute, Bronx, NY
| |
Collapse
|
13
|
Kalaimani L, Devarajan B, Namperumalsamy VP, Veerappan M, Daniels JT, Chidambaranathan GP. Hsa-miR-143-3p inhibits Wnt-β-catenin and MAPK signaling in human corneal epithelial stem cells. Sci Rep 2022; 12:11432. [PMID: 35794158 PMCID: PMC9259643 DOI: 10.1038/s41598-022-15263-x] [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: 02/18/2022] [Accepted: 06/21/2022] [Indexed: 11/09/2022] Open
Abstract
Our previous study demonstrated hsa-miR-143-3p as one of the highly expressed miRNAs in enriched corneal epithelial stem cells (CESCs). Hence this study aims to elucidate the regulatory role of hsa-miR-143-3p in the maintenance of stemness in CESCs. The target genes of hsa-miR-143-3p were predicted and subjected to pathway analysis to select the targets for functional studies. Primary cultured limbal epithelial cells were transfected with hsa-miR-143-3p mimic, inhibitor or scrambled sequence using Lipofectamine 3000. The transfected cells were analysed for (i) colony forming potential, (ii) expression of stem cell (SC) markers/ transcription factors (ABCG2, NANOG, OCT4, KLF4, ΔNp63), (iii) differentiation marker (Cx43), (iv) predicted five targets of hsa-miR-143-3p (DVL3, MAPK1, MAPK14, KRAS and KAT6A), (v) MAPK signaling regulators and (vi) Wnt-β-catenin signaling regulators by qPCR, immunofluorescence staining and/or Western blotting. High expression of hsa-miR-143-3p increased the colony forming potential (10.04 ± 1.35%, p < 0.001) with the ability to form holoclone-like colonies in comparison to control (3.33 ± 0.71%). The mimic treated cells had increased expression of SC markers but reduced expression of Cx43 and hsa-miR-143-3p targets involved in Wnt-β-catenin and MAPK signaling pathways. The expression of β-catenin, active β-catenin and ERK2 in hsa-miR-143-3p inhibitor transfected cells were higher than the control cells and the localized nuclear expression indicated the activation of Wnt and MAPK signaling. Thus, the probable association of hsa-miR-143-3p in the maintenance of CESCs through inhibition of Wnt and MAPK signaling pathways was thus indicated.
Collapse
Affiliation(s)
- Lavanya Kalaimani
- Department of Immunology and Stem Cell Biology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, 625020, India.,Department of Biotechnology, Aravind Medical Research Foundation-Affiliated to Alagappa University, Karaikudi, Tamil Nadu, India.,Institute of Ophthalmology, University College London, London, UK
| | - Bharanidharan Devarajan
- Department of Bioinformatics, Aravind Medical Research Foundation, Madurai, Tamil Nadu, India
| | | | - Muthukkaruppan Veerappan
- Department of Immunology and Stem Cell Biology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, 625020, India
| | - Julie T Daniels
- Institute of Ophthalmology, University College London, London, UK
| | - Gowri Priya Chidambaranathan
- Department of Immunology and Stem Cell Biology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, 625020, India. .,Department of Biotechnology, Aravind Medical Research Foundation-Affiliated to Alagappa University, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
14
|
Crisafulli L, Ficara F. Micro-RNAs: A safety net to protect hematopoietic stem cell self-renewal. WILEY INTERDISCIPLINARY REVIEWS. RNA 2022; 13:e1693. [PMID: 34532984 PMCID: PMC9285953 DOI: 10.1002/wrna.1693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 11/05/2022]
Abstract
The hematopoietic system is sustained over time by a small pool of hematopoietic stem cells (HSCs). They reside at the apex of a complex hierarchy composed of cells with progressively more restricted lineage potential, regenerative capacity, and with different proliferation characteristics. Like other somatic stem cells, HSCs are endowed with long-term self-renewal and multipotent differentiation ability, to sustain the high turnover of mature cells such as erythrocytes or granulocytes, and to rapidly respond to acute peripheral stresses including bleeding, infections, or inflammation. Maintenance of both attributes over time, and of the proper balance between these opposite features, is crucial to ensure the homeostasis of the hematopoietic system. Micro-RNAs (miRNAs) are short non-coding RNAs that regulate gene expression posttranscriptionally upon binding to specific mRNA targets. In the past 10 years they have emerged as important players for preserving the HSC pool by acting on several biological mechanisms, such as maintenance of the quiescent state while preserving proliferation ability, prevention of apoptosis, premature differentiation, lineage skewing, excessive expansion, or retention within the BM niche. miRNA-mediated posttranscriptional fine-tuning of all these processes constitutes a safety mechanism to protect HSCs, by complementing the action of transcription factors and of other regulators and avoiding unwanted expansion or aplasia. The current knowledge of miRNAs function in different aspects of HSC biology, including consequences of aberrant miRNA expression, will be reviewed; yet unsolved issues will be discussed. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA in Disease and Development > RNA in Development.
Collapse
Affiliation(s)
- Laura Crisafulli
- UOS Milan Unit, Istituto di Ricerca Genetica e Biomedica (IRGB), CNRMilanItaly
- IRCCS Humanitas Research HospitalMilanItaly
| | - Francesca Ficara
- UOS Milan Unit, Istituto di Ricerca Genetica e Biomedica (IRGB), CNRMilanItaly
- IRCCS Humanitas Research HospitalMilanItaly
| |
Collapse
|
15
|
Inflammation and myeloid malignancy: Quenching the flame. Blood 2022; 140:1067-1074. [PMID: 35468199 DOI: 10.1182/blood.2021015162] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/22/2022] [Indexed: 01/19/2023] Open
Abstract
Chronic inflammation with aging ("inflammaging") plays a prominent role in the pathogenesis of myeloid malignancies. Aberrant inflammatory activity impacts many different cells in the marrow, including normal blood and stromal marrow elements and leukemic cells, in unique and distinct ways. Inflammation can promote selective clonal expansion through differential immune-mediated suppression of normal hematopoietic cells and malignant clones. We review these complex roles, how they can be understood by separating cell-intrinsic from extrinsic effects, and how this informs future clinical trials.
Collapse
|
16
|
Gopal A, Ibrahim R, Fuller M, Umlandt P, Parker J, Tran J, Chang L, Wegrzyn-Woltosz J, Lam J, Li J, Lu M, Karsan A. TIRAP drives myelosuppression through an Ifnγ-Hmgb1 axis that disrupts the endothelial niche in mice. J Exp Med 2022; 219:212987. [PMID: 35089323 PMCID: PMC8932532 DOI: 10.1084/jem.20200731] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 09/15/2021] [Accepted: 12/16/2021] [Indexed: 12/31/2022] Open
Abstract
Inflammation is associated with bone marrow failure syndromes, but how specific molecules impact the bone marrow microenvironment is not well elucidated. We report a novel role for the miR-145 target, Toll/interleukin-1 receptor domain containing adaptor protein (TIRAP), in driving bone marrow failure. We show that TIRAP is overexpressed in various types of myelodysplastic syndromes (MDS) and suppresses all three major hematopoietic lineages. TIRAP expression promotes up-regulation of Ifnγ, leading to myelosuppression through Ifnγ-Ifnγr–mediated release of the alarmin, Hmgb1, which disrupts the bone marrow endothelial niche. Deletion of Ifnγ blocks Hmgb1 release and is sufficient to reverse the endothelial defect and restore myelopoiesis. Contrary to current dogma, TIRAP-activated Ifnγ-driven bone marrow suppression is independent of T cell function or pyroptosis. In the absence of Ifnγ, TIRAP drives myeloproliferation, implicating Ifnγ in suppressing the transformation of MDS to acute leukemia. These findings reveal novel, noncanonical roles of TIRAP, Hmgb1, and Ifnγ in the bone marrow microenvironment and provide insight into the pathophysiology of preleukemic syndromes.
Collapse
Affiliation(s)
- Aparna Gopal
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Rawa Ibrahim
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Megan Fuller
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Patricia Umlandt
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Jeremy Parker
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Jessica Tran
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Linda Chang
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joanna Wegrzyn-Woltosz
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jeffrey Lam
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Jenny Li
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Melody Lu
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Aly Karsan
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
17
|
Hu M, Lu Y, Wang S, Zhang Z, Qi Y, Chen N, Shen M, Chen F, Chen M, Yang L, Chen S, Zeng D, Wang F, Su Y, Xu Y, Wang J. CD63 acts as a functional marker in maintaining hematopoietic stem cell quiescence through supporting TGFβ signaling in mice. Cell Death Differ 2022; 29:178-191. [PMID: 34363017 PMCID: PMC8738745 DOI: 10.1038/s41418-021-00848-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023] Open
Abstract
Hematopoietic stem cell (HSC) fate is tightly controlled by various regulators, whereas the underlying mechanism has not been fully uncovered due to the high heterogeneity of these populations. In this study, we identify tetraspanin CD63 as a novel functional marker of HSCs in mice. We show that CD63 is unevenly expressed on the cell surface in HSC populations. Importantly, HSCs with high CD63 expression (CD63hi) are more quiescent and have more robust self-renewal and myeloid differentiation abilities than those with negative/low CD63 expression (CD63-/lo). On the other hand, using CD63 knockout mice, we find that loss of CD63 leads to reduced HSC numbers in the bone marrow. In addition, CD63-deficient HSCs exhibit impaired quiescence and long-term repopulating capacity, accompanied by increased sensitivity to irradiation and 5-fluorouracil treatment. Further investigations demonstrate that CD63 is required to sustain TGFβ signaling activity through its interaction with TGFβ receptors I and II, thereby playing an important role in regulating the quiescence of HSCs. Collectively, our data not only reveal a previously unrecognized role of CD63 but also provide us with new insights into HSC heterogeneity.
Collapse
Affiliation(s)
- Mengjia Hu
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Yukai Lu
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Song Wang
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Zihao Zhang
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Yan Qi
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Naicheng Chen
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Mingqiang Shen
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Fang Chen
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Mo Chen
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Lijing Yang
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Shilei Chen
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Dongfeng Zeng
- grid.410570.70000 0004 1760 6682Department of Hematology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Fengchao Wang
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Yongping Su
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Yang Xu
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Junping Wang
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| |
Collapse
|
18
|
Qi S, Wang C, Li L, Li T, Chen Q, Wang J. Association Between miR-143/145 rs4705343 Polymorphism and Risk of Congenital Heart Disease in a Chinese Tibetan Population. Genet Test Mol Biomarkers 2021; 25:735-740. [PMID: 34918978 DOI: 10.1089/gtmb.2021.0154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Objective: Congenital heart disease (CHD) is the most common birth defect worldwide and is caused by both genetic and environmental factors. The microRNA (miR)-143/145 cluster is involved in various biological processes related to cardiovascular development. The functional single nucleotide polymorphism (SNP) rs4705343 of miR-143/145 may influence the expression of these miRNAs. In this study, we aimed to estimate the association between miR-143/145 rs4705343 and the risk of CHD in a Chinese Tibetan population. Methods: Matrix-assisted laser desorption ionization time-of-flight mass spectrometry assays were performed to genotype the miRNA-143/145 rs4705343 SNP in 510 CHD Tibetan patients and 681 unrelated Tibetan healthy controls. The associations between the SNP frequencies and the CHD risk were analyzed by χ2 test/Fisher's test and assessed by odds ratios (ORs) and 95% confidence intervals (95% CIs). Results: We successfully genotyped 1165 subjects with a SNP call rate of 97.8%. Under the allelic model we found that rs4705343 was not associated with the risk of CHD (p = 0.082), but under the recessive model the CC genotype at this locus was associated with a significantly increased risk of CHD compared with the other genotypes (CC vs TT+TC: OR = 1.60, 95% CI = 1.08-2.37, p = 0.017). Conclusion: The present study suggests that the rs4705343 CC genotype of miR-143/145 is associated with CHD risk in a Chinese Tibetan population.
Collapse
Affiliation(s)
- Shenggui Qi
- Cardiovascular Center, Qinghai High Altitude Medical Research Institute, Xining, China
| | - Chunyan Wang
- Graduate School of Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Center for Genetics, National Research Institute for Family Planning, Beijing, China
| | - Li Li
- Cardiovascular Center, Qinghai High Altitude Medical Research Institute, Xining, China
| | - Tengyan Li
- Center for Genetics, National Research Institute for Family Planning, Beijing, China
| | - Qiuhong Chen
- Cardiovascular Center, Qinghai High Altitude Medical Research Institute, Xining, China
| | - Jing Wang
- Department of Medical Genetics, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Differentiation therapy for myeloid malignancies: beyond cytotoxicity. Blood Cancer J 2021; 11:193. [PMID: 34864823 PMCID: PMC8643352 DOI: 10.1038/s41408-021-00584-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 02/07/2023] Open
Abstract
Blocked cellular differentiation is a central pathologic feature of the myeloid malignancies, myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Treatment regimens promoting differentiation have resulted in incredible cure rates in certain AML subtypes, such as acute promyelocytic leukemia. Over the past several years, we have seen many new therapies for MDS/AML enter clinical practice, including epigenetic therapies (e.g., 5-azacitidine), isocitrate dehydrogenase (IDH) inhibitors, fms-like kinase 3 (FLT3) inhibitors, and lenalidomide for deletion 5q (del5q) MDS. Despite not being developed with the intent of manipulating differentiation, induction of differentiation is a major mechanism by which several of these novel agents function. In this review, we examine the new therapeutic landscape for these diseases, focusing on the role of hematopoietic differentiation and the impact of inflammation and aging. We review how current therapies in MDS/AML promote differentiation as a part of their therapeutic effect, and the cellular mechanisms by which this occurs. We then outline potential novel avenues to achieve differentiation in the myeloid malignancies for therapeutic purposes. This emerging body of knowledge about the importance of relieving differentiation blockade with anti-neoplastic therapies is important to understand how current novel agents function and may open avenues to developing new treatments that explicitly target cellular differentiation. Moving beyond cytotoxic agents has the potential to open new and unexpected avenues in the treatment of myeloid malignancies, hopefully providing more efficacy with reduced toxicity.
Collapse
|
20
|
Darden DB, Mira JC, Lopez MC, Stortz JA, Fenner BP, Kelly LS, Nacionales DC, Budharaju A, Loftus TJ, Baker HV, Moore FA, Brakenridge SC, Moldawer LL, Mohr AM, Efron PA. Identification of unique microRNA expression patterns in bone marrow hematopoietic stem and progenitor cells after hemorrhagic shock and multiple injuries in young and old adult mice. J Trauma Acute Care Surg 2021; 91:692-699. [PMID: 34252063 PMCID: PMC8463436 DOI: 10.1097/ta.0000000000003350] [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] [Indexed: 11/26/2022]
Abstract
BACKGROUND After severe trauma, the older host experiences more dysfunctional hematopoiesis of bone marrow (BM) hematopoietic stem and progenitor cells (HSPCs), and dysfunctional differentiation of circulating myeloid cells into effective innate immune cells. Our main objective was to compare BM HSPC microRNA (miR) responses of old and young mice in a clinically relevant model of severe trauma and shock. METHODS C57BL/6 adult male mice aged 8 to 12 weeks (young) and 18 to 24 months (old) underwent multiple injuries and hemorrhagic shock (polytrauma [PT]) that engenders the equivalent of major trauma (Injury Severity Score, >15). Pseudomonas pneumonia (PNA) was induced in some young and old adult mice 24 hours after PT. MicroRNA expression patterns were determined from lineage-negative enriched BM HSPCs isolated from PT and PT-PNA mice at 24 and 48 hours postinjury, respectively. Genome-wide expression and pathway analyses were also performed on bronchoalveolar lavage (BAL) leukocytes from both mouse cohorts. RESULTS MicroRNA expression significantly differed among all experimental conditions (p < 0.05), except for old-naive versus old-injured (PT or PT-PNA) mice, suggesting an inability of old mice to mount a robust early miR response to severe shock and injury. In addition, young adult mice had significantly more leukocytes obtained from their BAL, and there were greater numbers of polymorphonuclear cells compared with old mice (59.8% vs. 2.2%, p = 0.0069). Despite increased gene expression changes, BAL leukocytes from old mice demonstrated a more dysfunctional transcriptomic response to PT-PNA than young adult murine BAL leukocytes, as reflected in predicted upstream functional pathway analysis. CONCLUSION The miR expression pattern in BM HSPCs after PT (+/-PNA) is dissimilar in old versus young adult mice. In the acute postinjury phase, old adult mice are unable to mount a robust miR HSPC response. Hematopoietic stem and progenitor cell miR expression in old PT mice reflects a diminished functional status and a blunted capacity for terminal differentiation of myeloid cells.
Collapse
Affiliation(s)
- Dijoia B. Darden
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Juan C. Mira
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Maria-Cecilia Lopez
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL
| | - Julie A. Stortz
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Brittany P. Fenner
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Lauren S. Kelly
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Dina C. Nacionales
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Ashrita Budharaju
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Tyler J. Loftus
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Henry V. Baker
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL
| | - Frederick A. Moore
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Scott C. Brakenridge
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Lyle L. Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Alicia M. Mohr
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Philip A. Efron
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| |
Collapse
|
21
|
Xin Z, Zhang W, Gong S, Zhu J, Li Y, Zhang Z, Fang X. Mapping Human Pluripotent Stem Cell-derived Erythroid Differentiation by Single-cell Transcriptome Analysis. GENOMICS PROTEOMICS & BIOINFORMATICS 2021; 19:358-376. [PMID: 34284135 PMCID: PMC8864192 DOI: 10.1016/j.gpb.2021.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 01/22/2021] [Accepted: 03/06/2021] [Indexed: 10/28/2022]
Abstract
There is an imbalance between the supply and demand of functional red blood cells (RBCs) in clinical applications. This imbalance can be addressed by regenerating RBCs using several in vitro methods. Induced pluripotent stem cells (iPSCs) can handle the low supply of cord blood and the ethical issues in embryonic stem cell research and provide a promising strategy to eliminate immune rejection. However, no complete single-cell level differentiation pathway exists for the iPSC-derived RBC differentiation system. In this study, we used iPSC line BC1 to establish a RBCs regeneration system. The 10× genomics single-cell transcriptome platform was used to map the cell lineage and differentiation trajectories on day 14 of the regeneration system. We observed that iPSCs differentiation was not synchronized during embryoid body (EB) culture. The cells (day 14) mainly consisted of mesodermal and various blood cells, similar to the yolk sac hematopoiesis. We identified six cell classifications and characterized the regulatory transcription factors (TFs) networks and cell-cell contacts underlying the system. iPSCs undergo two transformations during the differentiation trajectory, accompanied by the dynamic expression of cell adhesion molecules and estrogen-responsive genes. We identified different stages of erythroid cells, such as burst-forming unit erythroid (BFU-E) and orthochromatic erythroblasts (ortho-E), and found that the regulation of TFs (e.g., TFDP1 and FOXO3) is erythroid-stage specific. Immune erythroid cells were identified in our system. This study provides systematic theoretical guidance for optimizing the iPSCs-derived RBCs differentiation system, and this system is a useful model for simulating in vivo hematopoietic development and differentiation.
Collapse
Affiliation(s)
- Zijuan Xin
- CAS Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center of Bioinformation, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Zhang
- CAS Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center of Bioinformation, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shangjin Gong
- CAS Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center of Bioinformation, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junwei Zhu
- CAS Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center of Bioinformation, Beijing 100101, China; Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, 100101, China
| | - Yanming Li
- CAS Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center of Bioinformation, Beijing 100101, China
| | - Zhaojun Zhang
- CAS Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center of Bioinformation, Beijing 100101, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100190, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, 100101, China.
| | - Xiangdong Fang
- CAS Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center of Bioinformation, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100190, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, 100101, China.
| |
Collapse
|
22
|
Trowbridge JJ, Starczynowski DT. Innate immune pathways and inflammation in hematopoietic aging, clonal hematopoiesis, and MDS. J Exp Med 2021; 218:212382. [PMID: 34129017 PMCID: PMC8210621 DOI: 10.1084/jem.20201544] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/23/2022] Open
Abstract
With a growing aged population, there is an imminent need to develop new therapeutic strategies to ameliorate disorders of hematopoietic aging, including clonal hematopoiesis and myelodysplastic syndrome (MDS). Cell-intrinsic dysregulation of innate immune- and inflammatory-related pathways as well as systemic inflammation have been implicated in hematopoietic defects associated with aging, clonal hematopoiesis, and MDS. Here, we review and discuss the role of dysregulated innate immune and inflammatory signaling that contribute to the competitive advantage and clonal dominance of preleukemic and MDS-derived hematopoietic cells. We also propose how emerging concepts will further reveal critical biology and novel therapeutic opportunities.
Collapse
Affiliation(s)
| | - Daniel T Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH.,Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
23
|
Altered microRNA expression links IL6 and TNF-induced inflammaging with myeloid malignancy in humans and mice. Blood 2021; 135:2235-2251. [PMID: 32384151 DOI: 10.1182/blood.2019003105] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/20/2020] [Indexed: 12/14/2022] Open
Abstract
Aging is associated with significant changes in the hematopoietic system, including increased inflammation, impaired hematopoietic stem cell (HSC) function, and increased incidence of myeloid malignancy. Inflammation of aging ("inflammaging") has been proposed as a driver of age-related changes in HSC function and myeloid malignancy, but mechanisms linking these phenomena remain poorly defined. We identified loss of miR-146a as driving aging-associated inflammation in AML patients. miR-146a expression declined in old wild-type mice, and loss of miR-146a promoted premature HSC aging and inflammation in young miR-146a-null mice, preceding development of aging-associated myeloid malignancy. Using single-cell assays of HSC quiescence, stemness, differentiation potential, and epigenetic state to probe HSC function and population structure, we found that loss of miR-146a depleted a subpopulation of primitive, quiescent HSCs. DNA methylation and transcriptome profiling implicated NF-κB, IL6, and TNF as potential drivers of HSC dysfunction, activating an inflammatory signaling relay promoting IL6 and TNF secretion from mature miR-146a-/- myeloid and lymphoid cells. Reducing inflammation by targeting Il6 or Tnf was sufficient to restore single-cell measures of miR-146a-/- HSC function and subpopulation structure and reduced the incidence of hematological malignancy in miR-146a-/- mice. miR-146a-/- HSCs exhibited enhanced sensitivity to IL6 stimulation, indicating that loss of miR-146a affects HSC function via both cell-extrinsic inflammatory signals and increased cell-intrinsic sensitivity to inflammation. Thus, loss of miR-146a regulates cell-extrinsic and -intrinsic mechanisms linking HSC inflammaging to the development of myeloid malignancy.
Collapse
|
24
|
The miRNA Profile in Non-Hodgkin's Lymphoma Patients with Secondary Myelodysplasia. Cells 2020; 9:cells9102318. [PMID: 33086588 PMCID: PMC7656297 DOI: 10.3390/cells9102318] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/07/2020] [Accepted: 10/16/2020] [Indexed: 12/15/2022] Open
Abstract
Myelodysplastic syndromes are a group of clonal diseases of hematopoietic stem cells and are characterized by multilineage dysplasia, ineffective hematopoiesis, peripheral blood cytopenias, genetic instability and a risk of transformation to acute myeloid leukemia. Some patients with non-Hodgkin lymphomas (NHLs) may have developed secondary myelodysplasia before therapy. Bone marrow (BM) hematopoiesis is regulated by a spectrum of epigenetic factors, among which microRNAs (miRNAs) are special. The aim of this work is to profile miRNA expression in BM cells in untreated NHL patients with secondary myelodysplasia. A comparative analysis of miRNA expression levels between the NHL and non-cancer blood disorders samples revealed that let-7a-5p was upregulated, and miR-26a-5p, miR-199b-5p, miR-145-5p and miR-150-5p were downregulated in NHL with myelodysplasia (p < 0.05). We for the first time developed a profile of miRNA expression in BM samples in untreated NHL patients with secondary myelodysplasia. It can be assumed that the differential diagnosis for blood cancers and secondary BM conditions based on miRNA expression profiles will improve the accuracy and relevance of the early diagnosis of cancerous and precancerous lesions in BM.
Collapse
|
25
|
McElhinney JMWR, Hasan A, Sajini AA. The epitranscriptome landscape of small noncoding RNAs in stem cells. Stem Cells 2020; 38:1216-1228. [PMID: 32598085 PMCID: PMC7586957 DOI: 10.1002/stem.3233] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 05/04/2020] [Accepted: 05/11/2020] [Indexed: 12/14/2022]
Abstract
Stem cells (SCs) are unique cells that have an inherent ability to self‐renew or differentiate. Both fate decisions are strongly regulated at the molecular level via intricate signaling pathways. The regulation of signaling networks promoting self‐renewal or differentiation was thought to be largely governed by the action of transcription factors. However, small noncoding RNAs (ncRNAs), such as vault RNAs, and their post‐transcriptional modifications (the epitranscriptome) have emerged as additional regulatory layers with essential roles in SC fate decisions. RNA post‐transcriptional modifications often modulate RNA stability, splicing, processing, recognition, and translation. Furthermore, modifications on small ncRNAs allow for dual regulation of RNA activity, at both the level of biogenesis and RNA‐mediated actions. RNA post‐transcriptional modifications act through structural alterations and specialized RNA‐binding proteins (RBPs) called writers, readers, and erasers. It is through SC‐context RBPs that the epitranscriptome coordinates specific functional roles. Small ncRNA post‐transcriptional modifications are today exploited by different mechanisms to facilitate SC translational studies. One mechanism readily being studied is identifying how SC‐specific RBPs of small ncRNAs regulate fate decisions. Another common practice of using the epitranscriptome for regenerative applications is using naturally occurring post‐transcriptional modifications on synthetic RNA to generate induced pluripotent SCs. Here, we review exciting insights into how small ncRNA post‐transcriptional modifications control SC fate decisions in development and disease. We hope, by illustrating how essential the epitranscriptome and their associated proteome are in SCs, they would be considered as novel tools to propagate SCs for regenerative medicine.
Collapse
Affiliation(s)
- James M W R McElhinney
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Ayesha Hasan
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Abdulrahim A Sajini
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW MiRNAs are critical regulators for gene expression. Numerous studies have revealed how miRNAs contribute to the pathogenesis of hematologic malignancies. RECENT FINDINGS The identification of novel miRNA regulatory factors and pathways crucial for miRNA dysregulation has been linked to hematologic malignancies. miRNA expression profiling has shown their potential to predict outcomes and treatment responses. Recently, targeting miRNA biogenesis or pathways has become a promising therapeutic strategy with recent miRNA-therapeutics being developed. SUMMARY We provide a comprehensive overview of the role of miRNAs for diagnosis, prognosis, and therapeutic potential in hematologic malignancies.
Collapse
Affiliation(s)
- Zhen Han
- Division of Dermatology, City of Hope, Duarte, CA, USA
- Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Steven T. Rosen
- Dept of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA
- Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Christiane Querfeld
- Division of Dermatology, City of Hope, Duarte, CA, USA
- Department of Pathology, City of Hope, Duarte, CA, USA
- Beckman Research Institute, City of Hope, Duarte, CA, USA
| |
Collapse
|
27
|
Song H, He S, Li S, Wu J, Yin W, Shao Z, Du G, Wu J, Li J, Weisel RD, Verma S, Xie J, Li R. Knock-out of MicroRNA 145 impairs cardiac fibroblast function and wound healing post-myocardial infarction. J Cell Mol Med 2020; 24:9409-9419. [PMID: 32628810 PMCID: PMC7417705 DOI: 10.1111/jcmm.15597] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/09/2020] [Accepted: 06/16/2020] [Indexed: 12/18/2022] Open
Abstract
Prevention of infarct scar thinning and dilatation and stimulation of scar contracture can prevent progressive heart failure. Since microRNA 145 (miR-145) plays an important role in cardiac fibroblast response to wound healing and cardiac repair after an myocardial infarction (MI), using a miR-145 knock-out (KO) mouse model, we evaluated contribution of down-regulation of miR-145 to cardiac fibroblast and myofibroblast function during adverse cardiac remodelling. Cardiac function decreased more and the infarct size was larger in miR-145 KO than that in WT mice after MI and this phenomenon was accompanied by a decrease in cardiac fibroblast-to-myofibroblast differentiation. Quantification of collagen I and α-SMA protein levels as well as wound contraction revealed that transdifferentiation of cardiac fibroblasts into myofibroblasts was lower in KO than WT mice. In vitro restoration of miR-145 induced more differentiation of fibroblasts to myofibroblasts and this effect involved the target genes Klf4 and myocardin. MiR-145 contributes to infarct scar contraction in the heart and the absence of miR-145 contributes to dysfunction of cardiac fibroblast, resulting in greater infarct thinning and dilatation. Augmentation of miR-145 could be an attractive target to prevent adverse cardiac remodelling after MI by enhancing the phenotypic switch of cardiac fibroblasts to myofibroblasts.
Collapse
Affiliation(s)
- Hui‐Fang Song
- Department of AnatomyShanxi Medical UniversityTaiyuanChina
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationShanxi Medical UniversityTaiyuanChina
- Toronto General Research InstituteUniversity Health NetworkTorontoONCanada
| | - Sheng He
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationShanxi Medical UniversityTaiyuanChina
- Toronto General Research InstituteUniversity Health NetworkTorontoONCanada
| | - Shu‐Hong Li
- Toronto General Research InstituteUniversity Health NetworkTorontoONCanada
| | - Jun Wu
- Toronto General Research InstituteUniversity Health NetworkTorontoONCanada
| | - Wenjuan Yin
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationShanxi Medical UniversityTaiyuanChina
- Toronto General Research InstituteUniversity Health NetworkTorontoONCanada
| | - Zhengbo Shao
- Toronto General Research InstituteUniversity Health NetworkTorontoONCanada
| | - Guo‐qing Du
- Toronto General Research InstituteUniversity Health NetworkTorontoONCanada
| | - Jie Wu
- Toronto General Research InstituteUniversity Health NetworkTorontoONCanada
| | - Jiao Li
- Toronto General Research InstituteUniversity Health NetworkTorontoONCanada
| | - Richard D. Weisel
- Toronto General Research InstituteUniversity Health NetworkTorontoONCanada
- Division of Cardiac SurgeryDepartment of SurgeryUniversity of TorontoTorontoONCanada
| | - Subodh Verma
- Division of Cardiac SurgeryLi Ka Shing Knowledge Institute of St Michael's HospitalDepartment of SurgeryUniversity of TorontoTorontoONCanada
| | - Jun Xie
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationShanxi Medical UniversityTaiyuanChina
| | - Ren‐Ke Li
- Toronto General Research InstituteUniversity Health NetworkTorontoONCanada
- Division of Cardiac SurgeryDepartment of SurgeryUniversity of TorontoTorontoONCanada
| |
Collapse
|
28
|
Nousbeck J, McAleer M, Hurault G, Kenny E, Harte K, Kezic S, Tanaka R, Irvine A. MicroRNA analysis of childhood atopic dermatitis reveals a role for miR‐451a*. Br J Dermatol 2020; 184:514-523. [DOI: 10.1111/bjd.19254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2020] [Indexed: 12/13/2022]
Affiliation(s)
- J. Nousbeck
- National Children’s Research Centre Crumlin, Dublin Ireland
- Clinical Medicine Trinity College Dublin Dublin Ireland
| | - M.A. McAleer
- National Children’s Research Centre Crumlin, Dublin Ireland
- Paediatric Dermatology Our Lady’s Children’s Hospital Crumlin, Dublin Ireland
| | - G. Hurault
- Department of Bioengineering Imperial College London London UK
| | - E. Kenny
- Department of Psychiatry Trinity Translational Medicine Institute St James’s Hospital Dublin Ireland
| | - K. Harte
- Department of Psychiatry Trinity Translational Medicine Institute St James’s Hospital Dublin Ireland
| | - S. Kezic
- Coronel Institute of Occupational Health Academic Medical Center Amsterdam the Netherlands
| | - R.J. Tanaka
- Department of Bioengineering Imperial College London London UK
| | - A.D. Irvine
- National Children’s Research Centre Crumlin, Dublin Ireland
- Clinical Medicine Trinity College Dublin Dublin Ireland
- Paediatric Dermatology Our Lady’s Children’s Hospital Crumlin, Dublin Ireland
| |
Collapse
|
29
|
MicroRNAs in hematopoietic stem cell aging. Mech Ageing Dev 2020; 189:111281. [PMID: 32512019 DOI: 10.1016/j.mad.2020.111281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/17/2020] [Accepted: 06/01/2020] [Indexed: 12/23/2022]
Abstract
The functional decline that is observed in HSCs upon aging is attributed mainly to cell intrinsic factors that regulate quiescence, self-renewal and differentiation. MicroRNAs (miRs) have an indispensable role in the regulation of HSCs and have been shown to also regulate processes related to tissue aging in specific cell types. Here we discuss the role of miRs in the regulation of HSC self-renewal and differentiation throughout life and its implications for future research.
Collapse
|
30
|
The Microrna-143/145 Cluster in Tumors: A Matter of Where and When. Cancers (Basel) 2020; 12:cancers12030708. [PMID: 32192092 PMCID: PMC7140083 DOI: 10.3390/cancers12030708] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/13/2020] [Accepted: 03/15/2020] [Indexed: 01/06/2023] Open
Abstract
The establishment and spreading of cancer involve the acquirement of many biological functions including resistance to apoptosis, enhanced proliferation and the ability to invade the surrounding tissue, extravasate from the primary site, survive in circulating blood, and finally extravasate and colonize distant organs giving origin to metastatic lesions, the major cause of cancer deaths. Dramatic changes in the expression of protein coding genes due to altered transcription factors activity or to epigenetic modifications orchestrate these events, intertwining with a microRNA regulatory network that is often disrupted in cancer cells. microRNAs-143 and -145 represent puzzling players of this game, with apparently contradictory functions. They were at first classified as tumor suppressive due to their frequently reduced levels in tumors, correlating with cell survival, proliferation, and migration. More recently, pro-oncogenic roles of these microRNAs have been described, challenging their simplistic definition as merely tumor-suppressive. Here we review their known activities in tumors, whether oncogenic or onco-suppressive, and highlight how their expression and functions are strongly dependent on their complex regulation downstream and upstream of cytokines and growth factors, on the cell type of expression and on the specific tumor stage.
Collapse
|
31
|
Tu H, Chen D, Cai C, Du Q, Lin H, Pan T, Sheng L, Xu Y, Teng T, Tu J, Lin Z, Wang X, Wang R, Xu L, Chen Y. microRNA-143-3p attenuated development of hepatic fibrosis in autoimmune hepatitis through regulation of TAK1 phosphorylation. J Cell Mol Med 2020; 24:1256-1267. [PMID: 31808606 PMCID: PMC6991639 DOI: 10.1111/jcmm.14750] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 09/12/2019] [Accepted: 09/24/2019] [Indexed: 12/11/2022] Open
Abstract
Autoimmune hepatitis (AIH) is a chronic liver disease due to autoimmune system attacks hepatocytes and causes inflammation and fibrosis. Intracellular signalling and miRNA may play an important role in regulation of liver injury. This study aimed to investigate the potential roles of microRNA 143 in a murine AIH model and a hepatocyte injury model. Murine AIH model was induced by hepatic antigen S100, and hepatocyte injury model was induced by LPS. Mice and AML12 cells were separated into six groups with or without the treatment of miRNA-143. Inflammation and fibrosis as well as gene expression were examined by different cellular and molecular techniques. The model was successfully established with the elevation of ALT and AST as well as inflammatory and fibrotic markers. Infection or transfection of mir-143 in mice or hepatocytes significantly attenuated the development of alleviation of hepatocyte injury. Moreover, the study demonstrated phosphorylation of TAK1-mediated miRNA-143 regulation of hepatic inflammation and fibrosis as well as hepatocyte injury. Our studies demonstrated a significant role of miRNA-143 in attenuation of liver injury in AIH mice and hepatocytes. miRNA-143 regulates inflammation and fibrosis through its regulation of TAK1 phosphorylation, which warrants TAK1 as a target for the development of new therapeutic strategy of autoimmune hepatitis.
Collapse
Affiliation(s)
- Hanxiao Tu
- Department of Infectious DiseasesWenzhou Key Laboratory of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityHepatology Institute of Wenzhou Medical UniversityWenzhouChina
| | - Dazhi Chen
- Department of GastroenterologyThe First Hospital of Peking UniversityBeiJingChina
| | - Chao Cai
- Department of Infectious DiseasesWenzhou Key Laboratory of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityHepatology Institute of Wenzhou Medical UniversityWenzhouChina
| | - Qianjing Du
- Department of Infectious DiseasesWenzhou Key Laboratory of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityHepatology Institute of Wenzhou Medical UniversityWenzhouChina
| | - Hongwei Lin
- Department of Infectious DiseasesWenzhou Key Laboratory of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityHepatology Institute of Wenzhou Medical UniversityWenzhouChina
| | - Tongtong Pan
- Department of Infectious DiseasesWenzhou Key Laboratory of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityHepatology Institute of Wenzhou Medical UniversityWenzhouChina
| | - Lina Sheng
- Department of Infectious DiseasesWenzhou Key Laboratory of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityHepatology Institute of Wenzhou Medical UniversityWenzhouChina
- Department of Infectious DiseasesThe Affiliated Yiwu Central Hospital of Wenzhou Medical UniversityYiwuChina
| | - Yuedong Xu
- Department of Infectious DiseasesWenzhou Key Laboratory of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityHepatology Institute of Wenzhou Medical UniversityWenzhouChina
| | - Teng Teng
- Department of Infectious DiseasesWenzhou Key Laboratory of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityHepatology Institute of Wenzhou Medical UniversityWenzhouChina
| | - Jingjing Tu
- Department of Infectious DiseasesWenzhou Key Laboratory of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityHepatology Institute of Wenzhou Medical UniversityWenzhouChina
| | - Zhuo Lin
- Department of Infectious DiseasesWenzhou Key Laboratory of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityHepatology Institute of Wenzhou Medical UniversityWenzhouChina
| | - Xiaodong Wang
- Department of Infectious DiseasesWenzhou Key Laboratory of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityHepatology Institute of Wenzhou Medical UniversityWenzhouChina
| | - Rui Wang
- Department of Infectious DiseasesWenzhou Key Laboratory of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityHepatology Institute of Wenzhou Medical UniversityWenzhouChina
| | - Lanman Xu
- Department of Infectious DiseasesWenzhou Key Laboratory of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityHepatology Institute of Wenzhou Medical UniversityWenzhouChina
- Department of Infectious Diseases and Liver DiseasesNingbo Medical Center Lihuili HospitalNingboChina
- Department of Infectious Diseases and Liver DiseasesThe Affiliated Lihuili Hospital of Ningbo UniversityNingboChina
| | - Yongping Chen
- Department of Infectious DiseasesWenzhou Key Laboratory of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityHepatology Institute of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
32
|
Pinweha P, Phillips CA, Gregory PA, Li X, Chuayboonya P, Mongkolsiri P, Goodall GJ, Jitrapakdee S. MicroRNA-143-3p targets pyruvate carboxylase expression and controls proliferation and migration of MDA-MB-231 cells. Arch Biochem Biophys 2019; 677:108169. [DOI: 10.1016/j.abb.2019.108169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/31/2019] [Accepted: 11/02/2019] [Indexed: 01/06/2023]
|
33
|
Wang L, Qiu JG, He J, Liu WJ, Ge X, Zhou FM, Huang YX, Jiang BH, Liu LZ. Suppression of miR-143 contributes to overexpression of IL-6, HIF-1α and NF-κB p65 in Cr(VI)-induced human exposure and tumor growth. Toxicol Appl Pharmacol 2019; 378:114603. [PMID: 31152816 DOI: 10.1016/j.taap.2019.114603] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/22/2019] [Accepted: 05/28/2019] [Indexed: 01/18/2023]
Abstract
Hexavalent chromium [Cr(VI)] is a known occupational and environmental contaminant and carcinogen, but new mechanisms of Cr(VI)-induced carcinogenesis remain to be elucidated. In this study, we found that expression of miR-143 is decreased, whereas that of Interleukin 6 (IL-6) is increased in blood samples of Cr(VI)-exposing workers compared with corresponding unexposed workers. In addition, IL-6 was increased in human bronchial epithelial cells (BEAS-Cr) exposed to Cr(VI) compared with unexposed BEAS-2B cells. To further investigate the mechanisms by which Cr(VI) promotes these changes, we assessed the effects of miR-143 on gene expression and found that miR-143 suppressed expression of IL-6, HIF-1α and NF-κB p65, and that inhibiting miR-143 promoted expression of IL-6, HIF-1α and NF-κB p65. Interestingly, IL-6 regulated expression of HIF-1α, and HIF-1α transcriptionally regulated expression of IL-6. Experiments in animals showed that miR-143 inhibited tumor growth and angiogenesis by regulating IL-6/HIF-1α and downstream signaling pathways in vivo. These outcomes support the hypothesis that the miR-143/IL-6/HIF-1α pathway functions to regulate Cr(VI)-induced carcinogenesis.
Collapse
Affiliation(s)
- Lin Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jian-Ge Qiu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jun He
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, USA
| | - Wen-Jing Liu
- Department of Oncology, Henan Cancer Hospital, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Xin Ge
- Department of Pathology, Nanjing Medical University, Nanjing 210000, China
| | - Feng-Mei Zhou
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Ying-Xue Huang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Bing-Hua Jiang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan, China; Department of Pathology, Carver College of Medicine, the University of Iowa, IA 52242, USA.
| | - Ling-Zhi Liu
- Department of Pathology, Carver College of Medicine, the University of Iowa, IA 52242, USA.
| |
Collapse
|
34
|
Transforming growth factor (TGF)-β pathway as a therapeutic target in lower risk myelodysplastic syndromes. Leukemia 2019; 33:1303-1312. [PMID: 30962581 DOI: 10.1038/s41375-019-0448-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/17/2019] [Accepted: 03/01/2019] [Indexed: 01/08/2023]
Abstract
The transforming growth factor (TGF)-β superfamily comprises more than 30 soluble growth factors that play a central role in erythropoiesis and are part of a tightly regulated myelosuppressive negative feedback loop under physiologic conditions. TGF-β receptor activation and phosphorylation trigger a regulatory circuit of activating and inhibitory SMAD proteins and increased activation of the TGF-β signaling pathway either by a loss of negative feedback or constitutive activation has been associated with the myelosuppression and ineffective erythropoiesis in myelodysplastic syndromes (MDS). Anemia is the predominant cause of morbidity and quality of life impairment in patients with lower-risk (LR)-MDS, and there are very limited therapy options for these patients after failure of erythropoiesis stimulating agents (ESAs). Targeting the aberrant TGF-ß signaling pathway has therefore been investigated as a promising therapeutic approach to resolve the ineffective erythropoiesis in LR-MDS. In this article, we provide a brief overview of the TGF-β signaling cascade in hematopoiesis under physiologic conditions and its role in MDS pathogenesis. We also review preclinical and clinical data for the activin receptor type IIA ligand traps sotatercept and luspatercept that have recently shown promising results in overcoming the myelosuppressive effects of TGF-β signaling alterations to improve hematopoiesis in transfusion-dependent, non-del(5q) LR-MDS patients. Additional potential targets within the TGF-β pathway have also been identified in preclinical experiments and may provide further therapeutic options. Finally, combining different TGF-β pathway inhibitors or using them in combination with ESAs or the immunomodulator lenalidomide might have synergistic effects as well.
Collapse
|
35
|
Muench DE, Ferchen K, Velu CS, Pradhan K, Chetal K, Chen X, Weirauch MT, Colmenares C, Verma A, Salomonis N, Grimes HL. SKI controls MDS-associated chronic TGF-β signaling, aberrant splicing, and stem cell fitness. Blood 2018; 132:e24-e34. [PMID: 30249787 PMCID: PMC6251005 DOI: 10.1182/blood-2018-06-860890] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/18/2018] [Indexed: 01/19/2023] Open
Abstract
The transforming growth factor beta (TGF-β) signaling pathway controls hematopoietic stem cell (HSC) behavior in the marrow niche; however, TGF-β signaling becomes chronic in early-stage myelodysplastic syndrome (MDS). Although TGF-β signaling normally induces negative feedback, in early-stage MDS, high levels of microRNA-21 (miR-21) contribute to chronic TGF-β signaling. We found that a TGF-β signal-correlated gene signature is sufficient to identify an MDS patient population with abnormal RNA splicing (eg, CSF3R) independent of splicing factor mutations and coincident with low HNRNPK activity. Levels of SKI messenger RNA (mRNA) encoding a TGF-β antagonist are sufficient to identify these patients. However, MDS patients with high SKI mRNA and chronic TGF-β signaling lack SKI protein because of miR-21 activity. To determine the impact of SKI loss, we examined murine Ski -/- HSC function. First, competitive HSC transplants revealed a profound defect in stem cell fitness (competitive disadvantage) but not specification, homing, or multilineage production. Aged recipients of Ski -/- HSCs exhibited mild phenotypes similar to phenotypes in those with macrocytic anemia. Second, blastocyst complementation revealed a dramatic block in Ski -/- hematopoiesis in the absence of transplantation. Similar to SKI-high MDS patient samples, Ski -/- HSCs strikingly upregulated TGF-β signaling and deregulated expression of spliceosome genes (including Hnrnpk). Moreover, novel single-cell splicing analyses demonstrated that Ski -/- HSCs and high levels of SKI expression in MDS patient samples share abnormal alternative splicing of common genes (including those that encode splicing factors). We conclude that miR-21-mediated loss of SKI activates TGF-β signaling and alternative splicing to impair the competitive advantage of normal HSCs (fitness), which could contribute to selection of early-stage MDS-genic clones.
Collapse
Affiliation(s)
- David E Muench
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Kyle Ferchen
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Chinavenmeni S Velu
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Kith Pradhan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | | | | | - Matthew T Weirauch
- Division of Biomedical Informatics
- Center for Autoimmune Genomics and Etiology, and
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Clemencia Colmenares
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Amit Verma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY; and
| | | | - H Leighton Grimes
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
36
|
Chronic immune response dysregulation in MDS pathogenesis. Blood 2018; 132:1553-1560. [PMID: 30104218 DOI: 10.1182/blood-2018-03-784116] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 08/03/2018] [Indexed: 12/18/2022] Open
Abstract
Chronic innate immune signaling in hematopoietic cells is widely described in myelodysplastic syndromes (MDS), and innate immune pathway activation, predominantly via pattern recognition receptors, increases the risk of developing MDS. An inflammatory component to MDS has been reported for many years, but only recently has evidence supported a more direct role of chronic innate immune signaling and associated inflammatory pathways in the pathogenesis of MDS. Here we review recent findings and discuss relevant questions related to chronic immune response dysregulation in MDS.
Collapse
|