51
|
Epigenetic silencing of miR-125b is required for normal B-cell development. Blood 2018; 131:1920-1930. [PMID: 29555645 DOI: 10.1182/blood-2018-01-824540] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 03/05/2018] [Indexed: 11/20/2022] Open
Abstract
Deregulation of several microRNAs (miRs) can influence critical developmental checkpoints during hematopoiesis as well as cell functions, eventually leading to the development of autoimmune disease or cancer. We found that miR-125b is expressed in bone marrow multipotent progenitors and myeloid cells but shut down in the B-cell lineage, and the gene encoding miR-125b lacked transcriptional activation markers in B cells. To understand the biological importance of the physiological silencing of miR-125b expression in B cells, we drove its expression in the B-cell lineage and found that dysregulated miR-125b expression impaired egress of immature B cells from the bone marrow to peripheral blood. Such impairment appeared to be mediated primarily by inhibited expression of the sphingosine-1-phosphate receptor 1 (S1PR1). Enforced expression of S1PR1 or clustered regularly interspaced short palindromic repeats/Cas9-mediated genome editing of the miR-125b targeting site in the S1PR1 3' untranslated region rescued the miR-125b-mediated defect in B-cell egress. In addition to impaired B-cell egress, miR-125b dysregulation initially reduced pre-B-cell output but later induced pre-B-cell lymphoma/leukemia in mice. Genetic deletion of IRF4 was found in miR-125b-induced B-cell cancer, but its role in oncogenic miR-125b-induced B-cell transformation is still unknown. Here, we further demonstrated an interaction of the effects of miR-125b and IRF4 in cancer induction by showing that miR125b-induced B-cell leukemia was greatly accelerated in IRF4 homozygous mutant mice. Thus, we conclude that physiological silencing of miR-125b is required for normal B-cell development and also acts as a mechanism of cancer suppression.
Collapse
|
52
|
Liao Q, Wang B, Li X, Jiang G. miRNAs in acute myeloid leukemia. Oncotarget 2018; 8:3666-3682. [PMID: 27705921 PMCID: PMC5356910 DOI: 10.18632/oncotarget.12343] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 09/24/2016] [Indexed: 12/30/2022] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNAs found throughout the eukaryotes that control the expression of a number of genes involved in commitment and differentiation of hematopoietic stem cells and tumorigenesis. Widespread dysregulation of miRNAs have been found in hematological malignancies, including human acute myeloid leukemia (AML). A comprehensive understanding of the role of miRNAs within the complex regulatory networks that are disrupted in malignant AML cells is a prerequisite for the development of therapeutic strategies employing miRNA modulators. Herein, we review the roles of emerging miRNAs and the miRNAs regulatory networks in AML pathogenesis, prognosis, and miRNA-directed therapies.
Collapse
Affiliation(s)
- Qiong Liao
- Key Laboratory for Rare & Uncommon Dseases of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China.,School of Medicine and Life Sciences, Jinan University, Jinan, Shandong, P.R. China
| | - Bingping Wang
- Department of Hematology, Shengli Oilfield Central Hospital, Dongying, Shandong, P.R. China
| | - Xia Li
- Key Laboratory for Rare & Uncommon Dseases of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China.,Shandong University School of Medicine, Jinan, Shandong, P.R. China
| | - Guosheng Jiang
- Key Laboratory for Rare & Uncommon Dseases of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| |
Collapse
|
53
|
Liu Z, Smith KR, Khong HT, Huang J, Ahn EYE, Zhou M, Tan M. miR-125b regulates differentiation and metabolic reprogramming of T cell acute lymphoblastic leukemia by directly targeting A20. Oncotarget 2018; 7:78667-78679. [PMID: 27637078 PMCID: PMC5346668 DOI: 10.18632/oncotarget.12018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 09/02/2016] [Indexed: 11/25/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematopoietic malignancy. Although it has been reported that overexpression of miR-125b leads to T-ALL development, the underlying mechanisms of miR-125b action are still unclear. The goal of this study is to delineate the role of miR-125b in T-ALL development. We found that miR-125b is highly expressed in undifferentiated leukemic T cells (CD4-negative) while its expression is low in differentiated T cells (CD4-positive). Overexpression of miR-125b increased the CD4-negative population in T cells, whereas depletion of miR-125b by miR-125b-sponge decreased the CD4-negative cell population. We identified that A20 (TNFAIP3) is a direct target of miR-125b in T cells. Overexpression of miR-125b also increased glucose uptake and oxygen consumption in T cells through targeting A20. Furthermore, restoration of A20 in miR-125b-overexpressing cells decreased the CD4-negative population in T cell leukemia, and decreased glucose uptake and oxygen consumption to the basal level of T cells transfected with vector. In conclusion, our data demonstrate that miR-125b regulates differentiation and reprogramming of T cell glucose metabolism via targeting A20. Since both de-differentiation and dysregulated glucose metabolism contribute to the development of T-cell leukemia, these findings provide novel insights into the understanding and treatment of T-ALL.
Collapse
Affiliation(s)
- Zixing Liu
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Kelly R Smith
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Hung T Khong
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Jingshan Huang
- School of Computing, University of South Alabama, Mobile, AL, USA
| | | | - Ming Zhou
- Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Ming Tan
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA.,Department of Biochemistry & Molecular Biology, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
54
|
Trino S, Lamorte D, Caivano A, Laurenzana I, Tagliaferri D, Falco G, Del Vecchio L, Musto P, De Luca L. MicroRNAs as New Biomarkers for Diagnosis and Prognosis, and as Potential Therapeutic Targets in Acute Myeloid Leukemia. Int J Mol Sci 2018; 19:ijms19020460. [PMID: 29401684 PMCID: PMC5855682 DOI: 10.3390/ijms19020460] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 02/07/2023] Open
Abstract
Acute myeloid leukemias (AML) are clonal disorders of hematopoietic progenitor cells which are characterized by relevant heterogeneity in terms of phenotypic, genotypic, and clinical features. Among the genetic aberrations that control disease development there are microRNAs (miRNAs). miRNAs are small non-coding RNAs that regulate, at post-transcriptional level, translation and stability of mRNAs. It is now established that deregulated miRNA expression is a prominent feature in AML. Functional studies have shown that miRNAs play an important role in AML pathogenesis and miRNA expression signatures are associated with chemotherapy response and clinical outcome. In this review we summarized miRNA signature in AML with different cytogenetic, molecular and clinical characteristics. Moreover, we reviewed the miRNA regulatory network in AML pathogenesis and we discussed the potential use of cellular and circulating miRNAs as biomarkers for diagnosis and prognosis and as therapeutic targets.
Collapse
MESH Headings
- Animals
- Antagomirs/genetics
- Antagomirs/metabolism
- Antagomirs/therapeutic use
- Biomarkers, Tumor/agonists
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Chromosome Aberrations
- Extracellular Vesicles/metabolism
- Extracellular Vesicles/pathology
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/therapy
- Mice
- MicroRNAs/agonists
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Molecular Targeted Therapy
- Oligoribonucleotides/genetics
- Oligoribonucleotides/metabolism
- Oligoribonucleotides/therapeutic use
- Oncogene Proteins, Fusion/antagonists & inhibitors
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Prognosis
- Signal Transduction
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Stefania Trino
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Daniela Lamorte
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Antonella Caivano
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Ilaria Laurenzana
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Daniela Tagliaferri
- Biogem Scarl, Istituto di Ricerche Genetiche 'Gaetano Salvatore', 83031 Ariano Irpino, Italy.
| | - Geppino Falco
- Biogem Scarl, Istituto di Ricerche Genetiche 'Gaetano Salvatore', 83031 Ariano Irpino, Italy.
- Department of Biology, University of Naples Federico II, 80147 Naples, Italy.
| | - Luigi Del Vecchio
- CEINGE Biotecnologie Avanzate s.c.a r.l., 80147 Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80138 Naples, Italy.
| | - Pellegrino Musto
- Scientific Direction, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Potenza, Italy.
| | - Luciana De Luca
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| |
Collapse
|
55
|
Rossmann MP, Orkin SH, Chute JP. Hematopoietic Stem Cell Biology. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
56
|
miRNA regulation of Tip110 expression and self-renewal and differentiation of human CD34+ hematopoietic cells. Oncotarget 2017; 9:4823-4832. [PMID: 29435144 PMCID: PMC5797015 DOI: 10.18632/oncotarget.23572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 12/14/2017] [Indexed: 01/20/2023] Open
Abstract
Tip110 expression regulates hematopoiesis, but the regulatory mechanisms during hematopoiesis are not fully understood. There are a number of putative microRNA (miRNA) binding sites identified within the Tip110 3′ untranslated region (3′UTR). In this study, we determined the relationship among Tip110 miRNA, Tip110 expression and self-renewal and differentiation of human CD34+ hematopoietic cells. Using a Tip110 3UTR-based reporter gene assay, 11 miRNA showed the specific activity toward the Tip110 3′UTR and down-regulated constitutive Tip110 mRNA expression. When human cord blood CD34+ cells were differentiated, Tip110 mRNA expression showed significant decreases. Concurrently, five miRNA showed significant increases, five miRNA showed significant decreases, and one miRNA remained unchanged. To further assess the roles of miRNA in Tip110 expression and self-renewal and differentiation of human CD34+ hematopoietic cells, human cord blood CD34+ cells were transduced to express the full-length Tip110 3′UTR RNA. Expression of the Tip110 3′UTR RNA led to significant increases of Tip110 mRNA, and the number of hematopoietic stem cells and progenitor cells. Taken together, these results show important roles of Tip110 miRNA in Tip110 expression control and Tip110 regulation of hematopoiesis and offer a possibility of using Tip110 miRNA or 3′UTR as a strategy to maintain human CD34+ hematopoietic cells.
Collapse
|
57
|
Wang Y, Niu ZY, Guo YJ, Wang LH, Lin FR, Zhang JY. IL-11 promotes the treatment efficacy of hematopoietic stem cell transplant therapy in aplastic anemia model mice through a NF-κB/microRNA-204/thrombopoietin regulatory axis. Exp Mol Med 2017; 49:e410. [PMID: 29217821 PMCID: PMC5750475 DOI: 10.1038/emm.2017.217] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 04/30/2017] [Accepted: 06/07/2017] [Indexed: 12/26/2022] Open
Abstract
Hematopoietic stem cell (HSC) transplantation could be of therapeutic value for aplastic anemia (AA) patients, and immunosuppressants may facilitate the efficiency of the procedure. As anti-inflammatory cytokine interleukin-11 (IL-11) has a thrombopoietic effect, its use in cases of chronic bone marrow failure, such as AA, has been proposed to induce HSC function. However, the putative mechanisms that may support this process remain poorly defined. We found that decreased miR-204-5p levels were coincident with increased proliferation in mouse HSCs following exposure to IL-11 in vitro. Through inhibiting NF-кB activity, miR-204-5p repression was demonstrated to be a downstream effect of IL-11 signaling. miR-204-5p was shown to directly target thrombopoietin (TPO) via sequence-dependent 3′-UTR repression, indicating that this microRNA-dependent pathway could serve an essential role in supporting IL-11 functions in HSCs. Increased TPO expression in HSCs following IL-11 exposure could be mimicked or blocked by inhibiting or overexpressing miR-204-5p, respectively. Consistent with these in vitro findings, IL-11 promoted HSC engraftment in a mouse model of AA, an effect that was attenuated in cells overexpressing miR-204-5p. The reduction in miR-204-5p levels is an integral component of IL-11 signaling that may play an essential role in treating AA.
Collapse
Affiliation(s)
- Yan Wang
- Department of Hematology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Hematology, Shijiazhuang, China
| | - Zhi-Yun Niu
- Department of Hematology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Hematology, Shijiazhuang, China
| | - Yu-Jie Guo
- Department of Hematology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Hematology, Shijiazhuang, China
| | - Li-Hua Wang
- Department of Hematology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Hematology, Shijiazhuang, China
| | - Feng-Ru Lin
- Department of Hematology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Hematology, Shijiazhuang, China
| | - Jing-Yu Zhang
- Department of Hematology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Hematology, Shijiazhuang, China
| |
Collapse
|
58
|
Lim JE, Son Y. Endogenous Stem Cells in Homeostasis and Aging. Tissue Eng Regen Med 2017; 14:679-698. [PMID: 30603520 PMCID: PMC6171667 DOI: 10.1007/s13770-017-0097-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/06/2017] [Accepted: 11/07/2017] [Indexed: 12/22/2022] Open
Abstract
In almost all human tissues and organs, adult stem cells or tissue stem cells are present in a unique location, the so-called stem cell niche or its equivalent, continuously replenishing functional differentiated cells. Those endogenous stem cells can be expanded for cell therapeutics using ex vivo cell culture or recalled for tissue repair in situ through cell trafficking and homing. In the aging process, inefficiency in the endogenous stem cell-mediated healing mechanism can emerge from a variety of impairments that accumulate in the processes of stem cell self-renewal, function, differentiation capacity, and trafficking through cell autonomous intrinsic pathways (such as epigenetic alterations) or systemic extrinsic pathways. This review examines the homeostasis of endogenous stem cells, particularly bone marrow stem cells, and their dysregulation in disease and aging and discusses possible intervention strategies. Several systemic pro-aging and rejuvenating factors, recognized in heterochronic parabiosis or premature aging progeroid animal models, are reviewed as possible anti-aging pharmaceutical targets from the perspective of a healthy environment for endogenous stem cells. A variety of epigenetic modifications and chromosome architectures are reviewed as an intrinsic cellular pathway for aging and senescence. A gradual increase in inflammatory burden during aging is also reviewed. Finally, the tissue repair and anti-aging effects of Substance-P, a peptide stimulating stem cell trafficking from the bone marrow and modifying the inflammatory response, are discussed as a future anti-aging target.
Collapse
Affiliation(s)
- Ji Eun Lim
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104 Republic of Korea
| | - Youngsook Son
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104 Republic of Korea
- Kyung Hee Institute of Regenerative Medicine, Kyung Hee University Hospital, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02453 Republic of Korea
| |
Collapse
|
59
|
Djeghloul D, Kuranda K, Kuzniak I, Barbieri D, Naguibneva I, Choisy C, Bories JC, Dosquet C, Pla M, Vanneaux V, Socié G, Porteu F, Garrick D, Goodhardt M. Age-Associated Decrease of the Histone Methyltransferase SUV39H1 in HSC Perturbs Heterochromatin and B Lymphoid Differentiation. Stem Cell Reports 2017; 6:970-984. [PMID: 27304919 PMCID: PMC4911502 DOI: 10.1016/j.stemcr.2016.05.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 05/13/2016] [Accepted: 05/16/2016] [Indexed: 01/11/2023] Open
Abstract
The capacity of hematopoietic stem cells (HSC) to generate B lymphocytes declines with age, contributing to impaired immune function in the elderly. Here we show that the histone methyltransferase SUV39H1 plays an important role in human B lymphoid differentiation and that expression of SUV39H1 decreases with age in both human and mouse HSC, leading to a global reduction in H3K9 trimethylation and perturbed heterochromatin function. Further, we demonstrate that SUV39H1 is a target of microRNA miR-125b, a known regulator of HSC function, and that expression of miR-125b increases with age in human HSC. Overexpression of miR-125b and inhibition of SUV39H1 in young HSC induced loss of B cell potential. Conversely, both inhibition of miR-125 and enforced expression of SUV39H1 improved the capacity of HSC from elderly individuals to generate B cells. Our findings highlight the importance of heterochromatin regulation in HSC aging and B lymphopoiesis.
Collapse
Affiliation(s)
- Dounia Djeghloul
- INSERM UMRS-1126, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Klaudia Kuranda
- INSERM UMRS-1126, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Isabelle Kuzniak
- INSERM UMRS-1126, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Daniela Barbieri
- INSERM UMRS-1170, Gustave Roussy Cancer Campus, Université Paris Sud - Université Paris-Saclay, Villejuif, France
| | - Irina Naguibneva
- INSERM UMRS-967, Institut de Radiobiologie Cellulaire et Moléculaire, Commissariat à l'Energie Atomique, Fontenay-aux-Roses, France
| | - Caroline Choisy
- INSERM UMRS-1126, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Jean-Christophe Bories
- INSERM UMRS-1126, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Christine Dosquet
- INSERM UMRS-1131, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Marika Pla
- INSERM UMRS-1131, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Valérie Vanneaux
- AP-HP Unité de Thérapie Cellulaire, Centre d'Investigation Clinique en Biothérapie Cellulaire and INSERM UMRS-1160, Université Paris Diderot, Paris, France
| | - Gérard Socié
- AP-HP Hematology Transplantation and INSERM UMRS-1160, Université Paris Diderot, Paris, France
| | - Françoise Porteu
- INSERM UMRS-1170, Gustave Roussy Cancer Campus, Université Paris Sud - Université Paris-Saclay, Villejuif, France
| | - David Garrick
- INSERM UMRS-1126, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Michele Goodhardt
- INSERM UMRS-1126, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.
| |
Collapse
|
60
|
A carvedilol-responsive microRNA, miR-125b-5p protects the heart from acute myocardial infarction by repressing pro-apoptotic bak1 and klf13 in cardiomyocytes. J Mol Cell Cardiol 2017; 114:72-82. [PMID: 29122578 DOI: 10.1016/j.yjmcc.2017.11.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/22/2017] [Accepted: 11/05/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cardiac injury is accompanied by dynamic changes in the expression of microRNAs (miRs), small non-coding RNAs that post-transcriptionally regulate target genes. MiR-125b-5p is downregulated in patients with end-stage dilated and ischemic cardiomyopathy, and has been proposed as a biomarker of heart failure. We previously reported that the β-blocker carvedilol promotes cardioprotection via β-arrestin-biased agonism of β1-adrenergic receptor while stimulating miR-125b-5p processing in the mouse heart. We hypothesize that β1-adrenergic receptor/β-arrestin1-responsive miR-125b-5p confers the improvement of cardiac function and structure after acute myocardial infarction. METHODS AND RESULTS Using cultured cardiomyocyte (CM) and in vivo approaches, we show that miR-125b-5p is an ischemic stress-responsive protector against CM apoptosis. CMs lacking miR-125b-5p exhibit increased susceptibility to stress-induced apoptosis, while CMs overexpressing miR-125b-5p have increased phospho-AKT pro-survival signaling. Moreover, we demonstrate that loss-of-function of miR-125b-5p in the mouse heart causes abnormalities in cardiac structure and function after acute myocardial infarction. Mechanistically, the improvement of cardiac function and structure elicited by miR-125b-5p is in part attributed to repression of the pro-apoptotic genes Bak1 and Klf13 in CMs. CONCLUSIONS In conclusion, these findings reveal a pivotal role for miR-125b-5p in regulating CM survival during acute myocardial infarction.
Collapse
|
61
|
Jin C, Cheng L, Lu X, Xie T, Wu H, Wu N. Elevated expression of miR-155 is associated with the differentiation of CD8+ T cells in patients with HIV-1. Mol Med Rep 2017; 16:1584-1589. [PMID: 28627655 DOI: 10.3892/mmr.2017.6755] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 03/30/2017] [Indexed: 11/05/2022] Open
Abstract
The differentiation and response ofCD8+ T cells is vital in host defense against human immunodeficiency virus type 1 (HIV-1). MicroRNA (miR)‑155 is an important regulator of T cell differentiation. However, the profile of miR-155 in HIV‑1 infected individuals and its association with CD8+ T cell differentiation remain to be fully elucidated. The present cross‑sectional study was performed involving 63 HIV‑1‑infected patients undergoing highly active antiretroviral therapy (HAART), 31 HAART‑naïve patients and 35 healthy controls. The levels of miR‑155 in CD8+ T cells were detected using reverse transcription‑quantitative polymerase chain reaction analysis. Subsets of CD8+ T cell differentiation were detected using flow cytometry. The results revealed that the discord controllers and HAART‑naïve patients showed higher percentages of effector and effector memory cells, and lower percentages of naïve cells (P<0.05). The levels of miR‑155 in CD8+ T cells from the HIV‑1‑infected patients were higher, particularly in the discord controllers and HAART naïve patients (P<0.01). The expression levels of miR‑155 were positively correlated with the percentages of effector and effector memory CD8+ T cells, and negatively correlated with the percentages of naïve and central memory CD8+ T cells (P<0.01). Taken together, these findings suggested that the levels of miR‑155 in CD8+ T cells of patients with HIV-1 were increased and asso-ciated with CD8+ T cell differentiation.
Collapse
Affiliation(s)
- Changzhong Jin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Linfang Cheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Xiangyun Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Tiansheng Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Haibo Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Nanping Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
62
|
MicroRNAs and acute myeloid leukemia: therapeutic implications and emerging concepts. Blood 2017; 130:1290-1301. [PMID: 28751524 DOI: 10.1182/blood-2016-10-697698] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 07/24/2017] [Indexed: 02/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is a deadly hematologic malignancy characterized by the uncontrolled growth of immature myeloid cells. Over the past several decades, we have learned a tremendous amount regarding the genetic aberrations that govern disease development in AML. Among these are genes that encode noncoding RNAs, including the microRNA (miRNA) family. miRNAs are evolutionarily conserved small noncoding RNAs that display important physiological effects through their posttranscriptional regulation of messenger RNA targets. Over the past decade, studies have identified miRNAs as playing a role in nearly all aspects of AML disease development, including cellular proliferation, survival, and differentiation. These observations have led to the study of miRNAs as biomarkers of disease, and efforts to therapeutically manipulate miRNAs to improve disease outcome in AML are ongoing. Although much has been learned regarding the importance of miRNAs in AML disease initiation and progression, there are many unanswered questions and emerging facets of miRNA biology that add complexity to their roles in AML. Moving forward, answers to these questions will provide a greater level of understanding of miRNA biology and critical insights into the many translational applications for these small regulatory RNAs in AML.
Collapse
|
63
|
Hu J, Zheng L, Shen X, Zhang Y, Li C, Xi T. MicroRNA-125b inhibits AML cells differentiation by directly targeting Fes. Gene 2017; 620:1-9. [PMID: 28389358 DOI: 10.1016/j.gene.2017.04.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 02/09/2017] [Accepted: 04/03/2017] [Indexed: 01/05/2023]
Abstract
MicroRNA-125b (miR-125b) has been reported to be upregulated in several kinds of leukemia, suggesting that miR-125b plays a role in Leukemia development. In this study, it was shown that miR-125b expression level decreased in response to 1α, 25-dihydroxy-vitamin D3 (1,25D3) in a dose- and time-dependent manner and miR-125b blocked 1,25D3-induced monocytic differentiation of U937 cells. In addition, miR-125b decreased mRNA expression of myelomonocytic differentiation markers, including CD11c, CD18 and CD64 and arrested the cell cycle at the S phase in U937 and HL60 cells. Fes was identified as a novel direct target of miR-125b and miR-125b could also reduce the expression levels of PU.1 and macrophage colony-stimulating factor receptor (MCSFR). Furthermore, Fes was found to be involved in monocytic differentiation via upregulation of PU.1 and MCSFR and Fes siRNA could also inhibit 1,25D3-induced monocytic differentiation of U937 and HL60 cells and decrease mRNA expression of CD11c, CD18 and CD64. Importantly, the inhibition of Fes siRNA on 1,25D3-induced monocytic differentiation could be rescued by transfection with miR-125b inhibitor. Our data highlights an important role of miR-125b in AML progression, implying the potential application of miR-125b in AML therapy.
Collapse
Affiliation(s)
- Jinhang Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Lufeng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Xiao Shen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yan Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Cheng Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
64
|
Martiáñez Canales T, de Leeuw DC, Vermue E, Ossenkoppele GJ, Smit L. Specific Depletion of Leukemic Stem Cells: Can MicroRNAs Make the Difference? Cancers (Basel) 2017; 9:cancers9070074. [PMID: 28665351 PMCID: PMC5532610 DOI: 10.3390/cancers9070074] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/15/2017] [Accepted: 06/20/2017] [Indexed: 01/22/2023] Open
Abstract
For over 40 years the standard treatment for acute myeloid leukemia (AML) patients has been a combination of chemotherapy consisting of cytarabine and an anthracycline such as daunorubicin. This standard treatment results in complete remission (CR) in the majority of AML patients. However, despite these high CR rates, only 30–40% (<60 years) and 10–20% (>60 years) of patients survive five years after diagnosis. The main cause of this treatment failure is insufficient eradication of a subpopulation of chemotherapy resistant leukemic cells with stem cell-like properties, often referred to as “leukemic stem cells” (LSCs). LSCs co-exist in the bone marrow of the AML patient with residual healthy hematopoietic stem cells (HSCs), which are needed to reconstitute the blood after therapy. To prevent relapse, development of additional therapies targeting LSCs, while sparing HSCs, is essential. As LSCs are rare, heterogeneous and dynamic, these cells are extremely difficult to target by single gene therapies. Modulation of miRNAs and consequently the regulation of hundreds of their targets may be the key to successful elimination of resistant LSCs, either by inducing apoptosis or by sensitizing them for chemotherapy. To address the need for specific targeting of LSCs, miRNA expression patterns in highly enriched HSCs, LSCs, and leukemic progenitors, all derived from the same patients’ bone marrow, were determined and differentially expressed miRNAs between LSCs and HSCs and between LSCs and leukemic progenitors were identified. Several of these miRNAs are specifically expressed in LSCs and/or HSCs and associated with AML prognosis and treatment outcome. In this review, we will focus on the expression and function of miRNAs expressed in normal and leukemic stem cells that are residing within the AML bone marrow. Moreover, we will review their possible prospective as specific targets for anti-LSC therapy.
Collapse
Affiliation(s)
- Tania Martiáñez Canales
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - David C de Leeuw
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - Eline Vermue
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - Gert J Ossenkoppele
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - Linda Smit
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
65
|
Abstract
MicroRNAs (miRNAs) are crucial post-transcriptional regulators of haematopoietic cell fate decisions. They act by negatively regulating the expression of key immune development genes, thus contributing important logic elements to the regulatory circuitry. Deletion studies have made it increasingly apparent that they confer robustness to immune cell development, especially under conditions of environmental stress such as infectious challenge and ageing. Aberrant expression of certain miRNAs can lead to pathological consequences, such as autoimmunity and haematological cancers. In this Review, we discuss the mechanisms by which several miRNAs influence immune development and buffer normal haematopoietic output, first at the level of haematopoietic stem cells, then in innate and adaptive immune cells. We then discuss the pathological consequences of dysregulation of these miRNAs.
Collapse
|
66
|
A Macro View of MicroRNAs: The Discovery of MicroRNAs and Their Role in Hematopoiesis and Hematologic Disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 334:99-175. [PMID: 28838543 DOI: 10.1016/bs.ircmb.2017.03.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
MicroRNAs (MiRNAs) are a class of endogenously encoded ~22 nucleotide, noncoding, single-stranded RNAs that contribute to development, body planning, stem cell differentiation, and tissue identity through posttranscriptional regulation and degradation of transcripts. Given their importance, it is predictable that dysregulation of MiRNAs, which target a wide variety of transcripts, can result in malignant transformation. In this review, we explore the discovery of MiRNAs, their mechanism of action, and the tools that aid in their discovery and study. Strikingly, many of the studies that have expanded our understanding of the contributions of MiRNAs to normal physiology and in the development of diseases have come from studies in the hematopoietic system and hematologic malignancies, with some of the earliest identified functions for mammalian MiRNAs coming from observations made in leukemias. So, with a special focus on the hematologic system, we will discuss how MiRNAs contribute to differentiation of stem cells and how dysregulation of MiRNAs contributes to the development of malignancy, by providing examples of specific MiRNAs that function as oncogenes or tumor suppressors, as well as of defects in MiRNA processing. Finally, we will discuss the promise of MiRNA-based therapeutics and challenges for the future study of disease-causing MiRNAs.
Collapse
|
67
|
Ge FX, Li H, Yin X. Upregulation of microRNA-125b-5p is involved in the pathogenesis of osteoarthritis by downregulating SYVN1. Oncol Rep 2017; 37:2490-2496. [PMID: 28260078 DOI: 10.3892/or.2017.5475] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 10/31/2016] [Indexed: 11/05/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative disease characterized by deterioration of articular cartilage. The aim of this study was to identify and characterize the expression of microRNA-125b-5p (miR-125b-5p) in normal and OA synovial cells, and to determine its role in OA pathogenesis. First, the levels of miR-125b-5p and synoviolin 1 (SYVN1) were detected among normal, mild OA and severe OA groups with the use of quantitative PCR. Computational analysis was used to search for the target of the miR-125b-5p, and luciferase reporter assay system was used to validate SYVN1 as the target gene of miR-125b-5p. Then the SYVN1 expression level of cells transfected with miR-125b-5p mimics or inhibitors was estimated using quantitative PCR and western blotting. Finally, MTT assay was employed to estimate the effect of miR-125b-5p on apoptosis. We enrolled 36 participants consisting of 12 normal control, 12 mild OA and 12 severe OA, furthermore, we performed quantitative PCR to detect the levels of miR-125b-5p and SYVN1 among those groups, and found that miR-125b-5p was expressed at highest level in severe OA compared with normal control and mild OA groups, while SYVN1 was expressed at the lowest level in severe OA. Additionally, we identified that SYVN1 is a target of miR-125b-5p by using computational analysis and luciferase assay. Transfection with miR-125b-5p mimic or inhibitor was employed to investigate the effect of miR-125b-5p on expression of SYVN1 in synovial cells, and synovial cell viability and apoptosis, and the results showed that miR-125b-5p mimics significant decreased the expression of SYVN1, a substantially promoted apoptosis of synovial cells, while miR-125b-5p inhibitors remarkably increased the level of SYVN1, and substantially suppressed apoptosis of synovial cells. The data suggested that miR-125b-5p promoted apoptosis of synovial cells through targeting SYVN1 gene, with important implication for validating miR-125b-5p as a potential target for OA therapy.
Collapse
Affiliation(s)
- Feng-Xiao Ge
- Department of Orthopedics, The People's Hospital of Linyi, Linyi, Shandong 276000, P.R. China
| | - Haitao Li
- Department of Orthopedics, The People's Hospital of Linyi, Linyi, Shandong 276000, P.R. China
| | - Xin Yin
- Department of Orthopedics, The People's Hospital of Linyi, Linyi, Shandong 276000, P.R. China
| |
Collapse
|
68
|
Gagez AL, Duroux-Richard I, Leprêtre S, Orsini-Piocelle F, Letestu R, De Guibert S, Tuaillon E, Leblond V, Khalifa O, Gouilleux-Gruart V, Banos A, Tournilhac O, Dupuis J, Jorgensen C, Cartron G, Apparailly F. miR-125b and miR-532-3p predict the efficiency of rituximab-mediated lymphodepletion in chronic lymphocytic leukemia patients. A French Innovative Leukemia Organization study. Haematologica 2017; 102:746-754. [PMID: 28126961 PMCID: PMC5395115 DOI: 10.3324/haematol.2016.153189] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 01/23/2017] [Indexed: 12/22/2022] Open
Abstract
The underlying in vivo mechanisms of rituximab action remain incompletely understood in chronic lymphocytic leukemia. Recent data suggest that circulating micro-ribonucleic acids correlate with chronic lymphocytic leukemia progression and response to rituximab. Our study aimed at identifying circulating micro-ribonucleic acids that predict response to rituximab monotherapy in chronic lymphocytic leukemia patients. Using a hierarchical clustering of micro-ribonucleic acid expression profiles discriminating 10 untreated patients with low or high lymphocyte counts, we found 26 micro-ribonucleic acids significantly deregulated. Using individual real-time reverse transcription polymerase chain reaction, the expression levels of micro-ribonucleic acids representative of these two clusters were further validated in a larger cohort (n=61). MiR-125b and miR-532-3p were inversely correlated with rituximab-induced lymphodepletion (P=0.020 and P=0.001, respectively) and with the CD20 expression on CD19+ cells (P=0.0007 and P<0.0001, respectively). In silico analyses of genes putatively targeted by both micro-ribonucleic acids revealed a central role of the interleukin-10 pathway and CD20 (MS4A1) family members. Interestingly, both micro-ribonucleic acids were negatively correlated with MS4A1 expression, while they were positively correlated with MS4A3 and MSA47 Our results identify novel circulating predictive biomarkers for rituximab-mediated lymphodepletion efficacy in chronic lymphocytic leukemia, and suggest a novel molecular mechanism responsible for the rituximab mode of action that bridges miR-125b and miR-532-3p and CD20 family members. (clinicaltrials.gov Identifier: 01370772).
Collapse
Affiliation(s)
- Anne-Laure Gagez
- CNRS UMR 5235, University of Montpellier, France.,Department of Clinical Hematology, University Hospital Montpellier, France
| | - Isabelle Duroux-Richard
- INSERM, U1183, Institute of Regenerative Medicine and Biotherapy, University Hospital Montpellier, France
| | | | | | - Rémi Letestu
- Department of Biological Hematology, APHP, GHUPSSD, Avicenne Hospital, Bobigny, France
| | - Sophie De Guibert
- Department of Clinical Hematology, Pontchaillou Hospital, Rennes, France
| | - Edouard Tuaillon
- Department of Bacteriology-Virology, University Hospital Montpellier, France
| | - Véronique Leblond
- Department of Hematology, La Pitié Salpétrière Hospital, Paris, France
| | - Olfa Khalifa
- INSERM, U1183, Institute of Regenerative Medicine and Biotherapy, University Hospital Montpellier, France
| | | | - Anne Banos
- Department of Hematology, Cote Basque Hospital, Bayonne, France
| | - Olivier Tournilhac
- Department of Clinical Hematology, University Hospital Estaing, Clermont-Ferrand, France
| | - Jehan Dupuis
- Unit of Lymphoid Hematologic Malignancies, Henri Mondor Hospital, Créteil, France
| | - Christian Jorgensen
- INSERM, U1183, Institute of Regenerative Medicine and Biotherapy, University Hospital Montpellier, France.,Clinical department for Osteoarticular Diseases, University Hospital Lapeyronie, Montpellier, France
| | - Guillaume Cartron
- CNRS UMR 5235, University of Montpellier, France .,Department of Clinical Hematology, University Hospital Montpellier, France
| | - Florence Apparailly
- INSERM, U1183, Institute of Regenerative Medicine and Biotherapy, University Hospital Montpellier, France.,Clinical department for Osteoarticular Diseases, University Hospital Lapeyronie, Montpellier, France
| |
Collapse
|
69
|
miR-125b promotes MLL-AF9-driven murine acute myeloid leukemia involving a VEGFA-mediated non-cell-intrinsic mechanism. Blood 2017; 129:1491-1502. [PMID: 28053194 DOI: 10.1182/blood-2016-06-721027] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 12/28/2016] [Indexed: 12/14/2022] Open
Abstract
The hematopoietic stem cell-enriched miR-125 family microRNAs (miRNAs) are critical regulators of hematopoiesis. Overexpression of miR-125a or miR-125b is frequent in human acute myeloid leukemia (AML), and the overexpression of these miRNAs in mice leads to expansion of hematopoietic stem cells accompanied by perturbed hematopoiesis with mostly myeloproliferative phenotypes. However, whether and how miR-125 family miRNAs cooperate with known AML oncogenes in vivo, and how the resultant leukemia is dependent on miR-125 overexpression, are not well understood. We modeled the frequent co-occurrence of miR-125b overexpression and MLL translocations by examining functional cooperation between miR-125b and MLL-AF9 By generating a knock-in mouse model in which miR-125b overexpression is controlled by doxycycline induction, we demonstrated that miR-125b significantly enhances MLL-AF9-driven AML in vivo, and the resultant leukemia is partially dependent on continued overexpression of miR-125b Surprisingly, miR-125b promotes AML cell expansion and suppresses apoptosis involving a non-cell-intrinsic mechanism. MiR-125b expression enhances VEGFA expression and production from leukemia cells, in part by suppressing TET2 Recombinant VEGFA recapitulates the leukemia-promoting effects of miR-125b, whereas knockdown of VEGFA or inhibition of VEGF receptor 2 abolishes the effects of miR-125b In addition, significant correlation between miR-125b and VEGFA expression is observed in human AMLs. Our data reveal cooperative and dependent relationships between miR-125b and the MLL oncogene in AML leukemogenesis, and demonstrate a miR-125b-TET2-VEGFA pathway in mediating non-cell-intrinsic leukemia-promoting effects by an oncogenic miRNA.
Collapse
|
70
|
Yang Y, Zhang Y, Miao Z, Zou J, Luo J. miR-22Inhibits CD34+Cell Expansion Through Decreasing β-Catenin in Osteoblasts. Hum Gene Ther 2017; 28:135-145. [DOI: 10.1089/hum.2016.104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Yuxia Yang
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yanju Zhang
- Tianjin Central Hospital for Obstetrics and Gynecology, Tianjin, China
| | - Zhenchuan Miao
- Beijing Vitalstar Biotechnology Co. Ltd., Beijing, China
| | - Junhua Zou
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jianyuan Luo
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
71
|
Nizyaeva NV, Kulikova GV, Shchyogolev AI, Zemskov VM. The role of microRNA in regulation of the body’s immune responses. ACTA ACUST UNITED AC 2016. [DOI: 10.1134/s2079086416060050] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
72
|
miR-125b modulates megakaryocyte maturation by targeting the cell-cycle inhibitor p19 INK4D. Cell Death Dis 2016; 7:e2430. [PMID: 27763644 PMCID: PMC5133966 DOI: 10.1038/cddis.2016.288] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 08/16/2016] [Accepted: 08/17/2016] [Indexed: 12/16/2022]
Abstract
A better understanding of the mechanisms involved in megakaryocyte maturation will facilitate the generation of platelets in vitro and their clinical applications. A microRNA, miR-125b, has been suggested to have important roles in the self-renewal of megakaryocyte-erythroid progenitors and in platelet generation. However, miR-125b is also critical for hematopoietic stem cell self-renewal. Thus, the function of miR-125b and the complex signaling pathways regulating megakaryopoiesis remain to be elucidated. In this study, an attentive examination of the endogenous expression of miR-125b during megakaryocyte differentiation was performed. Accordingly, the differentiation of hematopoietic stem cells requires the downregulation of miR-125b, whereas megakaryocyte determination and maturation synchronize with miR-125b accumulation. The overexpression of miR-125b improves megakaryocytic differentiation of K562 and UT-7 cells. Furthermore, stage-specific overexpression of miR-125b in primary cells demonstrates that miR-125b mediates an enhancement of megakaryocytic differentiation after megakaryocyte determination, the stage at which megakaryocytes are negative for the expression of the hematopoietic progenitor marker CD34. The identification of miR-125b targets during megakaryopoiesis was focused on negative regulators of cell cycle because the transition of the G1/S phase has been associated with megakaryocyte polyploidization. Real-time PCR, western blot and luciferase reporter assay reveal that p19INK4D is a direct target of miR-125b. P19INK4D knockdown using small interfering RNA (siRNA) in megakaryocyte-induced K562 cells, UT-7 cells and CD61+ promegakaryocytes results in S-phase progression and increased polyploidy, as well as improved megakaryocyte differentiation, similarly to the effects of miR-125b overexpression. P19INK4D overexpression reverses these effects, as indicated by reduced expression of megakaryocyte markers, G1-phase arrest and polyploidy decrease. P19INK4D knockdown in miR-125b downregulated cells or p19INK4D overexpression in miR-125b upregulated cells rescued the effect of miR-125b. Taken together, these findings suggest that miR-125b expression positively regulates megakaryocyte development since the initial phases of megakaryocyte determination, and p19INK4D is one of the key mediators of miR-125b activity during the onset of megakaryocyte polyploidization.
Collapse
|
73
|
miR-125b controls monocyte adaptation to inflammation through mitochondrial metabolism and dynamics. Blood 2016; 128:3125-3136. [PMID: 27702798 DOI: 10.1182/blood-2016-02-697003] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 09/28/2016] [Indexed: 02/06/2023] Open
Abstract
Metabolic changes drive monocyte differentiation and fate. Although abnormal mitochondria metabolism and innate immune responses participate in the pathogenesis of many inflammatory disorders, molecular events regulating mitochondrial activity to control life and death in monocytes remain poorly understood. We show here that, in human monocytes, microRNA-125b (miR-125b) attenuates the mitochondrial respiration through the silencing of the BH3-only proapoptotic protein BIK and promotes the elongation of the mitochondrial network through the targeting of the mitochondrial fission process 1 protein MTP18, leading to apoptosis. Proinflammatory activation of monocyte-derived macrophages is associated with a concomitant increase in miR-125b expression and decrease in BIK and MTP18 expression, which lead to reduced oxidative phosphorylation and enhanced mitochondrial fusion. In a chronic inflammatory systemic disorder, CD14+ blood monocytes display reduced miR-125b expression as compared with healthy controls, inversely correlated with BIK and MTP18 messenger RNA expression. Our findings not only identify BIK and MTP18 as novel targets for miR-125b that control mitochondrial metabolism and dynamics, respectively, but also reveal a novel function for miR-125b in regulating metabolic adaptation of monocytes to inflammation. Together, these data unravel new molecular mechanisms for a proapoptotic role of miR-125b in monocytes and identify potential targets for interfering with excessive inflammatory activation of monocytes in inflammatory disorders.
Collapse
|
74
|
Biersack B. Non-coding RNA/microRNA-modulatory dietary factors and natural products for improved cancer therapy and prevention: Alkaloids, organosulfur compounds, aliphatic carboxylic acids and water-soluble vitamins. Noncoding RNA Res 2016; 1:51-63. [PMID: 30159411 PMCID: PMC6096427 DOI: 10.1016/j.ncrna.2016.09.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/01/2016] [Accepted: 09/01/2016] [Indexed: 02/06/2023] Open
Abstract
Non-coding small RNA molecules, the microRNAs (miRNAs), contribute decisively to the epigenetic regulation processes in cancer cells. Problematic pathogenic properties of cancer cells and the response of cancers towards anticancer drugs are highly influenced by miRNAs. Both increased drug activity and formation of tumor resistance are regulated by miRNAs. Further to this, the survival and proliferation of cancer cells and the formation of metastases is based on the modulated expression of certain miRNAs. In particular, drug-resistant cancer stem-like cells (CSCs) depend on the presence and absence of specific miRNAs. Fortunately, several small molecule natural compounds were discovered that target miRNAs involved in the modulation of tumor aggressiveness and drug resistance. This review gives an overview of the effects of a selection of naturally occurring small molecules (alkaloids, organosulfur compounds, aliphatic carboxylic acids and water-soluble vitamins) on miRNAs that are closely tangled with cancer diseases.
Collapse
Key Words
- AM, allyl mercaptan
- AOM, azoxymethane
- Aliphatic carboxylic acids
- Alkaloids
- Anticancer drugs
- CPT, camptothecin
- DADS, diallyl disulfide
- DHA, docosahexaenoic acid
- DIM, 3,3′-diindolylmethane
- EPA, eicosapentaenoic acid
- FA, folic acid
- GTC, green tea catechins
- I3C, indole-3-carbinol
- MiRNA
- NaB, sodium butyrate
- Organosulfur compounds
- PEITC, phenethylisothiocyanate
- PUFA, polyunsaturated fatty acid
- SAMC, S-allylmercaptocysteine
- SFN, sulforaphane
- TSA, trichostatin A
- Water-soluble vitamins
Collapse
|
75
|
MicroRNA-139-5p regulates proliferation of hematopoietic progenitors and is repressed during BCR-ABL-mediated leukemogenesis. Blood 2016; 128:2117-2129. [PMID: 27605510 DOI: 10.1182/blood-2016-02-702464] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 09/02/2016] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) have emerged as important regulators of the immune system. However, despite this prominence, our understanding of the function of miRNAs in the early hematopoietic stages is incomplete. In this study, we found that miR-139-5p negatively regulated the proliferation of hematopoietic stem cells and progenitor cells and that downregulation of miR-139-5p expression was associated with hematopoietic malignancy, such as chronic myeloid leukemia (CML). Knockdown of miR-139-5p resulted in myeloid-biased differentiation with expansion of myeloid progenitor cells. In contrast, miR-139-5p expression inhibited the proliferation of hematopoietic progenitors and resulted in the remission of a CML-like disease that is induced by breakpoint cluster region-Abelson (BCR-ABL) transformation. We also found that Brg1 is a functional target of miR-139-5p and that Brg1 is involved in BCR-ABL-induced leukemogenesis. Thus, our results identify miR-139-5p as a key regulator of cellular proliferation during early hematopoiesis and suggest that it is a potent antileukemic molecule.
Collapse
|
76
|
Roden C, Lu J. MicroRNAs in Control of Stem Cells in Normal and Malignant Hematopoiesis. CURRENT STEM CELL REPORTS 2016; 2:183-196. [PMID: 27547713 PMCID: PMC4988405 DOI: 10.1007/s40778-016-0057-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Studies on hematopoietic stem cells (HSCs) and leukemia stem cells (LSCs) have helped to establish the paradigms of normal and cancer stem cell concepts. For both HSCs and LSCs, specific gene expression programs endowed by their epigenome functionally distinguish them from their differentiated progenies. MicroRNAs (miRNAs), as a class of small non-coding RNAs, act to control post-transcriptional gene expression. Research in the past decade has yielded exciting findings elucidating the roles of miRNAs in control of multiple facets of HSC and LSC biology. Here we review recent progresses on the functions of miRNAs in HSC emergence during development, HSC switch from a fetal/neonatal program to an adult program, HSC self-renewal and quiescence, HSC aging, HSC niche, and malignant stem cells. While multiple different miRNAs regulate a diverse array of targets, two common themes emerge in HSC and LSC biology: miRNA mediated regulation of epigenetic machinery and cell signaling pathways. In addition, we propose that miRNAs themselves behave like epigenetic regulators, as they possess key biochemical and biological properties that can provide both stability and alterability to the epigenetic program. Overall, the studies of miRNAs in stem cells in the hematologic contexts not only provide key understandings to post-transcriptional gene regulation mechanisms in HSCs and LSCs, but also will lend key insights for other stem cell fields.
Collapse
Affiliation(s)
- Christine Roden
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Yale Stem Cell Center, Yale Cancer Center, New Haven, Connecticut, 06520, USA
- Graduate Program in Biological and Biomedical Sciences, Yale University, New Haven, Connecticut 06510, USA
| | - Jun Lu
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Yale Stem Cell Center, Yale Cancer Center, New Haven, Connecticut, 06520, USA
- Yale Center for RNA Science and Medicine, New Haven, Connecticut, 06520, USA
| |
Collapse
|
77
|
Wang B, Guo J, Feng L, Suen CW, Fu WM, Zhang JF, Li G. MiR124 suppresses collagen formation of human tendon derived stem cells through targeting egr1. Exp Cell Res 2016; 347:360-6. [PMID: 27569005 DOI: 10.1016/j.yexcr.2016.08.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/20/2016] [Accepted: 08/24/2016] [Indexed: 11/25/2022]
Abstract
Collagen formation is used as a crucial indicator of tenogenic differentiation of human tendon derived stem cell (hTDSC). Early growth response-1(egr1), a transcriptional factor, has been demonstrated to regulate tendon differentiation and promote tendon repair. Considering that the therapeutic options for tendon injuries remain limited, investigating the regulation of egr1 could facilitate the understanding of tendon development at molecular level so as to find a promising therapeutic target. MicroRNAs (miRNA) have been considered as epigenetic regulators to mediate multiple biological activities including stem cell differentiation. In the present study, biological experiments confirmed the prediction that miR124-3p (miR124) could have direct binding with egr1. We also found that miR124 suppressed collagen formation during the tendon differentiation of hTDSC while anti-miR124 promoted it. Furthermore, egr1 knockdown abolished the promotive effect of anti-miR124, suggesting that miR124 prevents tendon differentiation via suppressing egr1 expression. Therefore, miR124 may be a promising therapeutic target for tendon injury.
Collapse
Affiliation(s)
- Bin Wang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, PR China; The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
| | - Jia Guo
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, PR China
| | - Lu Feng
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, PR China
| | - Chun-Wai Suen
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, PR China
| | - Wei-Ming Fu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China.
| | - Jin-Fang Zhang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, PR China; The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
| | - Gang Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, PR China; The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China.
| |
Collapse
|
78
|
Elias HK, Khalaj M, Park CY. Divergent roles of miR-126 in normal and malignant stem cells. Transl Cancer Res 2016; 5:S328-S331. [PMID: 33088733 DOI: 10.21037/tcr.2016.07.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Harold K Elias
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, NY, USA
| | - Mona Khalaj
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, NY, USA.,Weill Graduate School of Medical Sciences, Cornell University, NY, USA
| | - Christopher Y Park
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, NY, USA.,Weill Graduate School of Medical Sciences, Cornell University, NY, USA.,Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, USA
| |
Collapse
|
79
|
Wojtowicz EE, Lechman ER, Hermans KG, Schoof EM, Wienholds E, Isserlin R, van Veelen PA, Broekhuis MJC, Janssen GMC, Trotman-Grant A, Dobson SM, Krivdova G, Elzinga J, Kennedy J, Gan OI, Sinha A, Ignatchenko V, Kislinger T, Dethmers-Ausema B, Weersing E, Alemdehy MF, de Looper HWJ, Bader GD, Ritsema M, Erkeland SJ, Bystrykh LV, Dick JE, de Haan G. Ectopic miR-125a Expression Induces Long-Term Repopulating Stem Cell Capacity in Mouse and Human Hematopoietic Progenitors. Cell Stem Cell 2016; 19:383-96. [PMID: 27424784 DOI: 10.1016/j.stem.2016.06.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 04/01/2016] [Accepted: 06/15/2016] [Indexed: 12/25/2022]
Abstract
Umbilical cord blood (CB) is a convenient and broadly used source of hematopoietic stem cells (HSCs) for allogeneic stem cell transplantation. However, limiting numbers of HSCs remain a major constraint for its clinical application. Although one feasible option would be to expand HSCs to improve therapeutic outcome, available protocols and the molecular mechanisms governing the self-renewal of HSCs are unclear. Here, we show that ectopic expression of a single microRNA (miRNA), miR-125a, in purified murine and human multipotent progenitors (MPPs) resulted in increased self-renewal and robust long-term multi-lineage repopulation in transplanted recipient mice. Using quantitative proteomics and western blot analysis, we identified a restricted set of miR-125a targets involved in conferring long-term repopulating capacity to MPPs in humans and mice. Our findings offer the innovative potential to use MPPs with enhanced self-renewal activity to augment limited sources of HSCs to improve clinical protocols.
Collapse
Affiliation(s)
- Edyta E Wojtowicz
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Antonius Deusinglaan 1, 9700 AV Groningen, the Netherlands
| | - Eric R Lechman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Karin G Hermans
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Erwin M Schoof
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Erno Wienholds
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Ruth Isserlin
- The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Peter A van Veelen
- Departments of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Mathilde J C Broekhuis
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Antonius Deusinglaan 1, 9700 AV Groningen, the Netherlands
| | - George M C Janssen
- Departments of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Aaron Trotman-Grant
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Stephanie M Dobson
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Gabriela Krivdova
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jantje Elzinga
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - James Kennedy
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Olga I Gan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Ankit Sinha
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Vladimir Ignatchenko
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Thomas Kislinger
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Bertien Dethmers-Ausema
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Antonius Deusinglaan 1, 9700 AV Groningen, the Netherlands
| | - Ellen Weersing
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Antonius Deusinglaan 1, 9700 AV Groningen, the Netherlands
| | - Mir Farshid Alemdehy
- Department of Hematology, Erasmus University Medical Center Cancer Institute, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | - Hans W J de Looper
- Department of Hematology, Erasmus University Medical Center Cancer Institute, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | - Gary D Bader
- The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Martha Ritsema
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Antonius Deusinglaan 1, 9700 AV Groningen, the Netherlands
| | - Stefan J Erkeland
- Department of Immunology, Erasmus University Medical Center, Wytemaweg 80, 3015CN Rotterdam, the Netherlands
| | - Leonid V Bystrykh
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Antonius Deusinglaan 1, 9700 AV Groningen, the Netherlands
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Gerald de Haan
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Antonius Deusinglaan 1, 9700 AV Groningen, the Netherlands.
| |
Collapse
|
80
|
Voronina N, Lemcke H, Wiekhorst F, Kühn JP, Rimmbach C, Steinhoff G, David R. Non-viral magnetic engineering of endothelial cells with microRNA and plasmid-DNA-An optimized targeting approach. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:2353-2364. [PMID: 27389150 DOI: 10.1016/j.nano.2016.06.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/27/2016] [Accepted: 06/23/2016] [Indexed: 12/18/2022]
Abstract
Genetic modulation of angiogenesis is a powerful tool for the treatment of multiple disorders. Here, we describe a strategy to produce modified endothelial cells, which can be efficiently magnetically guided. First, we defined optimal transfection conditions with both plasmid and microRNA, using a polyethyleneimine/magnetic nanoparticle-based vector (PEI/MNP), previously designed in our group. Further, two approaches were assessed in vitro: direct vector guidance and magnetic targeting of transfected cells. Due to its higher efficiency, including simulated dynamic conditions, production of miR/PEI/MNP-modified magnetically responsive cells was selected for further detailed investigation. In particular, we have studied internalization of transfection complexes, functional capacities and intercellular communication of engineered cells and delivery of therapeutic miR. Moreover, we demonstrated that 104 miRNA/PEI/MNP-modified magnetically responsive cells loaded with 0.37pg iron/cell are detectable with MRI. Taken together, our in vitro findings show that PEI/MNP is highly promising as a multifunctional tool for magnetically guided angiogenesis regulation.
Collapse
Affiliation(s)
- Natalia Voronina
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, University of Rostock, Rostock, Germany.
| | - Heiko Lemcke
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, University of Rostock, Rostock, Germany.
| | | | - Jens-Peter Kühn
- Department of Radiology and Neuroradiology, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany;.
| | - Christian Rimmbach
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, University of Rostock, Rostock, Germany
| | - Gustav Steinhoff
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, University of Rostock, Rostock, Germany.
| | - Robert David
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, University of Rostock, Rostock, Germany.
| |
Collapse
|
81
|
Nucera S, Giustacchini A, Boccalatte F, Calabria A, Fanciullo C, Plati T, Ranghetti A, Garcia-Manteiga J, Cittaro D, Benedicenti F, Lechman ER, Dick JE, Ponzoni M, Ciceri F, Montini E, Gentner B, Naldini L. miRNA-126 Orchestrates an Oncogenic Program in B Cell Precursor Acute Lymphoblastic Leukemia. Cancer Cell 2016; 29:905-921. [PMID: 27300437 DOI: 10.1016/j.ccell.2016.05.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 01/12/2016] [Accepted: 05/18/2016] [Indexed: 12/11/2022]
Abstract
MicroRNA (miRNA)-126 is a known regulator of hematopoietic stem cell quiescence. We engineered murine hematopoiesis to express miRNA-126 across all differentiation stages. Thirty percent of mice developed monoclonal B cell leukemia, which was prevented or regressed when a tetracycline-repressible miRNA-126 cassette was switched off. Regression was accompanied by upregulation of cell-cycle regulators and B cell differentiation genes, and downregulation of oncogenic signaling pathways. Expression of dominant-negative p53 delayed blast clearance upon miRNA-126 switch-off, highlighting the relevance of p53 inhibition in miRNA-126 addiction. Forced miRNA-126 expression in mouse and human progenitors reduced p53 transcriptional activity through regulation of multiple p53-related targets. miRNA-126 is highly expressed in a subset of human B-ALL, and antagonizing miRNA-126 in ALL xenograft models triggered apoptosis and reduced disease burden.
Collapse
Affiliation(s)
- Silvia Nucera
- San Raffaele Telethon Institute for Gene Therapy, 20132 Milan, Italy
| | - Alice Giustacchini
- San Raffaele Telethon Institute for Gene Therapy, 20132 Milan, Italy; Vita Salute San Raffaele University, 20132 Milan, Italy
| | - Francesco Boccalatte
- San Raffaele Telethon Institute for Gene Therapy, 20132 Milan, Italy; Vita Salute San Raffaele University, 20132 Milan, Italy
| | - Andrea Calabria
- San Raffaele Telethon Institute for Gene Therapy, 20132 Milan, Italy
| | - Cristiana Fanciullo
- San Raffaele Telethon Institute for Gene Therapy, 20132 Milan, Italy; Vita Salute San Raffaele University, 20132 Milan, Italy
| | - Tiziana Plati
- San Raffaele Telethon Institute for Gene Therapy, 20132 Milan, Italy
| | - Anna Ranghetti
- San Raffaele Telethon Institute for Gene Therapy, 20132 Milan, Italy
| | - Jose Garcia-Manteiga
- Centre for Translational Genomics and Bioinformatics, IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Davide Cittaro
- Centre for Translational Genomics and Bioinformatics, IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | | | - Eric R Lechman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Maurilio Ponzoni
- Vita Salute San Raffaele University, 20132 Milan, Italy; Pathology Unit, IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Fabio Ciceri
- Vita Salute San Raffaele University, 20132 Milan, Italy; Hematology and Bone Marrow Transplantation Unit, IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy, 20132 Milan, Italy
| | - Bernhard Gentner
- San Raffaele Telethon Institute for Gene Therapy, 20132 Milan, Italy; Hematology and Bone Marrow Transplantation Unit, IRCSS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy, 20132 Milan, Italy; Vita Salute San Raffaele University, 20132 Milan, Italy.
| |
Collapse
|
82
|
Zhang W, Hong S, Maniar KP, Cheng S, Jie C, Rademaker AW, Krensky AM, Clayberger C. KLF13 regulates the differentiation-dependent human papillomavirus life cycle in keratinocytes through STAT5 and IL-8. Oncogene 2016; 35:5565-5575. [PMID: 27041562 DOI: 10.1038/onc.2016.97] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 01/04/2016] [Accepted: 01/08/2016] [Indexed: 12/12/2022]
Abstract
High-risk strains of human papillomavirus (HPV) are the causative agents of cervical and anogenital cancers and are associated with 5% of all human cancers. Although prophylactic vaccines targeting a subset of HPV types are available, they are ineffective in HPV-infected individuals. Elucidation of the mechanisms controlling HPV replication may allow development of novel anti-HPV therapeutics. Infectious HPV virions are produced during terminal differentiation of host cells. The process of viral maturation requires synergistic interactions between viral and cellular proteins that leads to amplification of the viral genome and expression of late viral genes. Here we show that the transcription factor Kruppel-like factor 13 (KLF13) has a critical role in the HPV life cycle. KLF13 is overexpressed in HPV-positive keratinocytes and cervical cancer cell lines. Expression of KLF13 in normal cervical epithelium is low but increases significantly in cervical intraepithelial neoplasia and invasive squamous cervical cancer. After HPV infection, the E7 protein suppresses ubiquitin ligase FBW7 expression leading to an increase in KLF13 expression. Reduction of KLF13 with short hairpin RNA in differentiating HPV-positive cells resulted in diminished levels of viral gene expression and genome amplification. Knockdown of KLF13 also reduced the level of the transcription factor signal transducer and activator of transcription 5, which led to the downregulation of the ataxia-telangiectasia mutated DNA damage pathway and the chemokine interleukin-8 (IL-8). In addition, neutralization of IL-8 diminished viral genome amplification in differentiating HPV-positive cells. Thus, KLF13 is critical for the activation of the HPV productive life cycle and is likely involved in initiation and progression of cervical cancer.
Collapse
Affiliation(s)
- W Zhang
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - S Hong
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - K P Maniar
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - S Cheng
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - C Jie
- Department of Research Biostatistics, Des Moines University, Des Moines, IA, USA
| | - A W Rademaker
- Department of Preventive Medicine and the Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - A M Krensky
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - C Clayberger
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
83
|
Xu Z, Zhao L, Yang X, Ma S, Ge Y, Liu Y, Liu S, Shi J, Zheng D. Mmu-miR-125b overexpression suppresses NO production in activated macrophages by targeting eEF2K and CCNA2. BMC Cancer 2016; 16:252. [PMID: 27020049 PMCID: PMC4809031 DOI: 10.1186/s12885-016-2288-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 03/22/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND MicroRNAs have been shown to be important regulators of the immune response and the development of the immune system. It was reported that microRNA-125b (miR-125b) was down-regulated in macrophages challenged with endotoxin. However, little is known about the function and mechanism of action of miR-125b in macrophage activation. Macrophages use L-arginine to synthesize nitric oxide (NO) through inducible NO synthase (iNOS), and the released NO contributes to the tumoricidal activity of macrophages. METHODS Luciferase reporter assays were employed to validate regulation of a putative target of miR-125b. The effect of miR-125b on endogenous levels of this target were subsequently confirmed via Western blot. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was performed to determine the expression level of miR-125b in macrophage. MTS assays were conducted to explore the impact of miR-125b overexpression on the cell viability of 4T1 cells. RESULTS Here, we demonstrate that mmu-miR-125b overexpression suppresses NO production in activated macrophages and that LPS-activated macrophages with overexpressed mmu-miR-125b promote 4T1 tumor cell proliferation in vitro and 4T1 tumor growth in vivo. CCNA2 and eEF2K are the direct and functional targets of mmu-miR-125b in macrophages; CCNA2 and eEF2K expression was knocked down, which mimicked the mmu-miR-125b overexpression phenotype. CONCLUSIONS These data suggest that mmu-miR-125b decreases NO production in activated macrophages at least partially by suppressing eEF2K and CCNA2 expression.
Collapse
Affiliation(s)
- Zhenbiao Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Lianmei Zhao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Xin Yang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Sisi Ma
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Yehua Ge
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Yanxin Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Shilian Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Juan Shi
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
| | - Dexian Zheng
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
84
|
Abstract
Hematopoiesis is a dynamic process in which blood cells are continuously generated from hematopoietic stem cells (HSCs). The regulatory mechanisms controlling HSC fate have been studied extensively over the past several decades. Although many protein-coding genes have been shown to regulate hematopoietic differentiation, additional levels of HSC regulation are not well studied. Advances in deep sequencing have revealed many new classes of regulatory noncoding RNAs (ncRNAs), such as enhancer RNAs and antisense ncRNAs. Functional analysis of some of these ncRNAs has provided insights into the molecular mechanisms that regulate hematopoietic development and disease. In this review, we summarize recent advances in our understanding of functional regulatory ncRNAs associated with hematopoietic self-renewal and differentiation, as well as those dysregulated ncRNAs involved in hematologic malignancies.
Collapse
|
85
|
Lechman ER, Gentner B, Ng SWK, Schoof EM, van Galen P, Kennedy JA, Nucera S, Ciceri F, Kaufmann KB, Takayama N, Dobson SM, Trotman-Grant A, Krivdova G, Elzinga J, Mitchell A, Nilsson B, Hermans KG, Eppert K, Marke R, Isserlin R, Voisin V, Bader GD, Zandstra PW, Golub TR, Ebert BL, Lu J, Minden M, Wang JCY, Naldini L, Dick JE. miR-126 Regulates Distinct Self-Renewal Outcomes in Normal and Malignant Hematopoietic Stem Cells. Cancer Cell 2016; 29:214-28. [PMID: 26832662 PMCID: PMC4749543 DOI: 10.1016/j.ccell.2015.12.011] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 07/13/2015] [Accepted: 12/21/2015] [Indexed: 12/16/2022]
Abstract
To investigate miRNA function in human acute myeloid leukemia (AML) stem cells (LSC), we generated a prognostic LSC-associated miRNA signature derived from functionally validated subpopulations of AML samples. For one signature miRNA, miR-126, high bioactivity aggregated all in vivo patient sample LSC activity into a single sorted population, tightly coupling miR-126 expression to LSC function. Through functional studies, miR-126 was found to restrain cell cycle progression, prevent differentiation, and increase self-renewal of primary LSC in vivo. Compared with prior results showing miR-126 regulation of normal hematopoietic stem cell (HSC) cycling, these functional stem effects are opposite between LSC and HSC. Combined transcriptome and proteome analysis demonstrates that miR-126 targets the PI3K/AKT/MTOR signaling pathway, preserving LSC quiescence and promoting chemotherapy resistance.
Collapse
Affiliation(s)
- Eric R Lechman
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Bernhard Gentner
- San Raffaele Telethon Institute for Gene Therapy, San Raffaele Hospital, Milan 20132, Italy; Vita Salute San Raffaele University, San Raffaele Scientific Institute, San Raffaele Hospital, Milan 20132, Italy; Hematology and Bone Marrow Transplantation Unit, San Raffaele Hospital, Milan 20132, Italy
| | - Stanley W K Ng
- Department of Chemical Engineering and Applied Chemistry, Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5G 2M9, Canada; The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Erwin M Schoof
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Peter van Galen
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - James A Kennedy
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Medicine, University of Toronto, Toronto, ON M5G 2M9, Canada
| | - Silvia Nucera
- San Raffaele Telethon Institute for Gene Therapy, San Raffaele Hospital, Milan 20132, Italy; Vita Salute San Raffaele University, San Raffaele Scientific Institute, San Raffaele Hospital, Milan 20132, Italy
| | - Fabio Ciceri
- Vita Salute San Raffaele University, San Raffaele Scientific Institute, San Raffaele Hospital, Milan 20132, Italy; Hematology and Bone Marrow Transplantation Unit, San Raffaele Hospital, Milan 20132, Italy
| | - Kerstin B Kaufmann
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Naoya Takayama
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Stephanie M Dobson
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Aaron Trotman-Grant
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Gabriela Krivdova
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Janneke Elzinga
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Amanda Mitchell
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Björn Nilsson
- Department of Hematology and Transfusion Medicine, Lund University Hospital, Lund 221 84, Sweden
| | - Karin G Hermans
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Kolja Eppert
- Department of Pediatrics, McGill University and The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Rene Marke
- Laboratory of Pediatric Oncology, Radboud University Medical Center, Nijmegen, 6500 HB, Netherlands
| | - Ruth Isserlin
- The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Veronique Voisin
- The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Gary D Bader
- The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Peter W Zandstra
- Department of Chemical Engineering and Applied Chemistry, Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5G 2M9, Canada; The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Todd R Golub
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA
| | - Benjamin L Ebert
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jun Lu
- Yale Stem Cell Center, Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Mark Minden
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Medicine, University of Toronto, Toronto, ON M5G 2M9, Canada
| | - Jean C Y Wang
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Medicine, University of Toronto, Toronto, ON M5G 2M9, Canada
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy, San Raffaele Hospital, Milan 20132, Italy; Vita Salute San Raffaele University, San Raffaele Scientific Institute, San Raffaele Hospital, Milan 20132, Italy
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Research Tower, Room 8-301, 101 College Street, Toronto M5G 1L7, Canada.
| |
Collapse
|
86
|
Knackmuss U, Lindner SE, Aneichyk T, Kotkamp B, Knust Z, Villunger A, Herzog S. MAP3K11 is a tumor suppressor targeted by the oncomiR miR-125b in early B cells. Cell Death Differ 2016; 23:242-52. [PMID: 26138442 PMCID: PMC4678593 DOI: 10.1038/cdd.2015.87] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 05/05/2015] [Accepted: 05/22/2015] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small, non-coding RNAs that posttranscriptionally regulate gene expression and thereby control most, if not all, biological processes. Aberrant miRNA expression has been linked to a variety of human diseases including cancer, but the underlying molecular mechanism often remains unclear. Here we have screened a miRNA expression library in a growth factor-dependent mouse pre-B-cell system to identify miRNAs with oncogenic activity. We show that miR-125b is sufficient to render pre-B cells growth factor independent and demonstrate that continuous expression of miR-125b is necessary to keep these cells in a transformed state. Mechanistically, we find that the expression of miR-125b protects against apoptosis induced by growth factor withdrawal, and that it blocks the differentiation of pre-B to immature B cells. In consequence, miR-125b-transformed cells maintain expression of their pre-B-cell receptor that provides signals for continuous proliferation and survival even in the absence of growth factor. Employing microarray analysis, we identified numerous targets of miR-125b, but only reconstitution of MAP3K11, a critical regulator of mitogen- and stress-activated kinase signaling, interferes with the cellular fitness of the transformed cells. Together, this indicates that MAP3K11 might function as an important tumor suppressor neutralized by oncomiR-125b in B-cell leukemia.
Collapse
Affiliation(s)
- U Knackmuss
- Centre for Biological Signalling Studies (bioss), Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - S E Lindner
- Division of Developmental Immunology, Biocenter Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - T Aneichyk
- Division of Molecular Pathophysiology, Biocenter Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - B Kotkamp
- Centre for Biological Signalling Studies (bioss), Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Z Knust
- Centre for Biological Signalling Studies (bioss), Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - A Villunger
- Division of Developmental Immunology, Biocenter Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - S Herzog
- Centre for Biological Signalling Studies (bioss), Albert-Ludwigs-University Freiburg, Freiburg, Germany
- Division of Developmental Immunology, Biocenter Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
87
|
|
88
|
Function and significance of MicroRNAs in benign and malignant human stem cells. Semin Cancer Biol 2015; 35:200-11. [DOI: 10.1016/j.semcancer.2015.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 07/09/2015] [Accepted: 07/13/2015] [Indexed: 12/16/2022]
|
89
|
Diener Y, Jurk M, Kandil B, Choi YH, Wild S, Bissels U, Bosio A. RNA-based, transient modulation of gene expression in human haematopoietic stem and progenitor cells. Sci Rep 2015; 5:17184. [PMID: 26599627 PMCID: PMC4657003 DOI: 10.1038/srep17184] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 10/20/2015] [Indexed: 12/13/2022] Open
Abstract
Modulation of gene expression is a useful tool to study the biology of haematopoietic stem and progenitor cells (HSPCs) and might also be instrumental to expand these cells for therapeutic approaches. Most of the studies so far have employed stable gene modification by viral vectors that are burdensome when translating protocols into clinical settings. Our study aimed at exploring new ways to transiently modify HSPC gene expression using non-integrating, RNA-based molecules. First, we tested different methods to deliver these molecules into HSPCs. The delivery of siRNAs with chemical transfection methods such as lipofection or cationic polymers did not lead to target knockdown, although we observed more than 90% fluorescent cells using a fluorochrome-coupled siRNA. Confocal microscopic analysis revealed that despite extensive washing, siRNA stuck to or in the cell surface, thereby mimicking a transfection event. In contrast, electroporation resulted in efficient, siRNA-mediated protein knockdown. For transient overexpression of proteins, we used optimised mRNA molecules with modified 5'- and 3'-UTRs. Electroporation of mRNA encoding GFP resulted in fast, efficient and persistent protein expression for at least seven days. Our data provide a broad-ranging comparison of transfection methods for hard-to-transfect cells and offer new opportunities for DNA-free, non-integrating gene modulation in HSPCs.
Collapse
Affiliation(s)
| | - Marion Jurk
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | | | - Yeong-Hoon Choi
- Heart Center of the University of Cologne, Department of Cardiothoracic Surgery, Center of Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Stefan Wild
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - Ute Bissels
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | | |
Collapse
|
90
|
Epigenetic Control of Haematopoietic Stem Cell Aging and Its Clinical Implications. Stem Cells Int 2015; 2016:5797521. [PMID: 26681950 PMCID: PMC4670691 DOI: 10.1155/2016/5797521] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 07/01/2015] [Indexed: 01/16/2023] Open
Abstract
Aging, chronic inflammation, and environmental insults play an important role in a number of disease processes through alterations of the epigenome. In this review we explore how age-related changes in the epigenetic landscape can affect heterogeneity within the haematopoietic stem cell (HSC) compartment and the deriving clinical implications.
Collapse
|
91
|
Guglielmelli P, Bisognin A, Saccoman C, Mannarelli C, Coppe A, Vannucchi AM, Bortoluzzi S. Small RNA Sequencing Uncovers New miRNAs and moRNAs Differentially Expressed in Normal and Primary Myelofibrosis CD34+ Cells. PLoS One 2015; 10:e0140445. [PMID: 26468945 PMCID: PMC4607157 DOI: 10.1371/journal.pone.0140445] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/25/2015] [Indexed: 12/20/2022] Open
Abstract
Myeloproliferative neoplasms (MPN) are chronic myeloid cancers thought to arise at the level of CD34+ hematopoietic stem/progenitor cells. They include essential thrombocythemia (ET), polycythemia vera (PV) and primary myelofibrosis (PMF). All can progress to acute leukemia, but PMF carries the worst prognosis. Increasing evidences indicate that deregulation of microRNAs (miRNAs) might plays an important role in hematologic malignancies, including MPN. To attain deeper knowledge of short RNAs (sRNAs) expression pattern in CD34+ cells and of their possible role in mediating post-transcriptional regulation in PMF, we sequenced with Illumina HiSeq2000 technology CD34+ cells from healthy subjects and PMF patients. We detected the expression of 784 known miRNAs, with a prevalence of miRNA up-regulation in PMF samples, and discovered 34 new miRNAs and 99 new miRNA-offset RNAs (moRNAs), in CD34+ cells. Thirty-seven small RNAs were differentially expressed in PMF patients compared with healthy subjects, according to microRNA sequencing data. Five miRNAs (miR-10b-5p, miR-19b-3p, miR-29a-3p, miR-379-5p, and miR-543) were deregulated also in PMF granulocytes. Moreover, 3’-moR-128-2 resulted consistently downregulated in PMF according to RNA-seq and qRT-PCR data both in CD34+ cells and granulocytes. Target predictions of these validated small RNAs de-regulated in PMF and functional enrichment analyses highlighted many interesting pathways involved in tumor development and progression, such as signaling by FGFR and DAP12 and Oncogene Induced Senescence. As a whole, data obtained in this study deepened the knowledge of miRNAs and moRNAs altered expression in PMF CD34+ cells and allowed to identify and validate a specific small RNA profile that distinguishes PMF granulocytes from those of normal subjects. We thus provided new information regarding the possible role of miRNAs and, specifically, of new moRNAs in this disease.
Collapse
Affiliation(s)
- Paola Guglielmelli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Andrea Bisognin
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | - Carmela Mannarelli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandro Coppe
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | - Stefania Bortoluzzi
- Department of Molecular Medicine, University of Padova, Padova, Italy
- * E-mail:
| |
Collapse
|
92
|
Hong SH, Kim KS, Oh IH. Concise review: Exploring miRNAs--toward a better understanding of hematopoiesis. Stem Cells 2015; 33:1-7. [PMID: 25132287 DOI: 10.1002/stem.1810] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 07/14/2014] [Indexed: 12/12/2022]
Abstract
Hematopoiesis is governed by a multidimensional regulatory network involving both intrinsic and extrinsic factors that control self-renewal and differentiation of hematopoietic stem cells (HSCs) through the coordination of influences that affect cell fate. Increasing evidence indicates that microRNAs (miRNAs), short noncoding RNAs of approximately 22 nucleotides, play a central role in orchestrating these regulatory mechanisms to modulate the multiple entities of hematopoietic function in a cell-type specific manner, including self-renewal, lineage commitment, and survival of HSCs as well as their microenvironmental crosstalk. Here, we summarize the current understanding regarding the regulatory effects of miRNA on hematopoietic cells, thus enlightening their role in fine-tuning HSC function and hematopoietic homeostasis.
Collapse
Affiliation(s)
- Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Stem Cell Institute, Kangwon National University, Chuncheon, South Korea
| | | | | |
Collapse
|
93
|
Yang Y, Wang S, Miao Z, Ma W, Zhang Y, Su L, Hu M, Zou J, Yin Y, Luo J. miR-17 promotes expansion and adhesion of human cord blood CD34(+) cells in vitro. Stem Cell Res Ther 2015; 6:168. [PMID: 26345634 PMCID: PMC4562375 DOI: 10.1186/s13287-015-0159-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 07/14/2015] [Accepted: 08/17/2015] [Indexed: 01/11/2023] Open
Abstract
Introduction We have recently found that miR-17 is necessary in the cell-extrinsic control of cord blood (CB) CD34+ cell function. Here, we demonstrated that the proper level of miR-17 is also necessary in the cell-intrinsic control of the hematopoietic properties of CB CD34+ cells. Methods The miR-17 overexpression and knockdown models were created using primary CB CD34+ cells transfected by the indicated vectors. Long-term culture, colony forming, adhesion and trans-well migration assays were carried out to investigate the function of miR-17 on CB CD34+ cells in vitro. NOD prkdcscid Il2rgnull mice were used in a SCID repopulating cell assay to investigate the function of miR-17 on CB CD34+ cells in vivo. A two-tailed Student’s t-test was used for statistical comparisons. Results In vitro assays revealed that ectopic expression of miR-17 promoted long-term expansion, especially in the colony-forming of CB CD34+ cells and CD34+CD38− cells. Conversely, downregulation of miR-17 inhibited the expansion of CB CD34+ cells. However, the overexpression of miR-17 in vivo reduced the hematopoietic reconstitution potential of CB CD34+ cells compared to that of control cells. The increased expression of major adhesion molecules in miR-17 overexpressed CB CD34+ cells suggests that the adhesion between miR-17 overexpressed CB CD34+ cells and their niche in vivo is regulated abnormally, which may further lead to the reduced hematopoietic reconstitution capability of 17/OE cells in engrafted mice. Conclusion We conclude that the proper expression of miR-17 is required, at least partly, for normal hematopoietic stem cell–niche interaction and for the regulation of adult hematopoiesis. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0159-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuxia Yang
- Department of Medical & Research Technology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA.
| | - Saifeng Wang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Zhenchuan Miao
- Beijing Vitalstar Biotechnology Co., Ltd., Beijing, China.
| | - Wei Ma
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| | - Yanju Zhang
- Tianjin Central Hospital for Obstetrics and Gynecology, Tianjin, China.
| | - Li Su
- Center of Medical and Health Analysis, Peking University, Beijing, China.
| | - Mengyu Hu
- Department of Medical & Research Technology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA.
| | - Junhua Zou
- Department of Medical & Research Technology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA.
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Jianyuan Luo
- Department of Medical & Research Technology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA. .,Department of Medical & Research Technology, Department of Pathology, School of Medicine, University of Maryland, College Park, USA.
| |
Collapse
|
94
|
Pulecio J, Nivet E, Sancho-Martinez I, Vitaloni M, Guenechea G, Xia Y, Kurian L, Dubova I, Bueren J, Laricchia-Robbio L, Belmonte JCI. Conversion of human fibroblasts into monocyte-like progenitor cells. Stem Cells 2015; 32:2923-2938. [PMID: 25175072 DOI: 10.1002/stem.1800] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 06/12/2014] [Accepted: 06/19/2014] [Indexed: 01/02/2023]
Abstract
Reprogramming technologies have emerged as a promising approach for future regenerative medicine. Here, we report on the establishment of a novel methodology allowing for the conversion of human fibroblasts into hematopoietic progenitor-like cells with macrophage differentiation potential. SOX2 overexpression in human fibroblasts, a gene found to be upregulated during hematopoietic reconstitution in mice, induced the rapid appearance of CD34+ cells with a concomitant upregulation of mesoderm-related markers. Profiling of cord blood hematopoietic progenitor cell populations identified miR-125b as a factor facilitating commitment of SOX2-generated CD34+ cells to immature hematopoietic-like progenitor cells with grafting potential. Further differentiation toward the monocytic lineage resulted in the appearance of CD14+ cells with functional phagocytic capacity. In vivo transplantation of SOX2/miR-125b-generated CD34+ cells facilitated the maturation of the engrafted cells toward CD45+ cells and ultimately the monocytic/macrophage lineage. Altogether, our results indicate that strategies combining lineage conversion and further lineage specification by in vivo or in vitro approaches could help to circumvent long-standing obstacles for the reprogramming of human cells into hematopoietic cells with clinical potential.
Collapse
Affiliation(s)
- Julian Pulecio
- Center of Regenerative Medicine in Barcelona, Dr. Aiguader, 88, 08003 Barcelona
| | - Emmanuel Nivet
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Ignacio Sancho-Martinez
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Marianna Vitaloni
- Center of Regenerative Medicine in Barcelona, Dr. Aiguader, 88, 08003 Barcelona
| | - Guillermo Guenechea
- Hematopoiesis and Gene Therapy Division. Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)/Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER). Madrid, Spain
| | - Yun Xia
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Leo Kurian
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Ilir Dubova
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Juan Bueren
- Hematopoiesis and Gene Therapy Division. Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)/Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER). Madrid, Spain
| | | | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| |
Collapse
|
95
|
Andl T, Botchkareva NV. MicroRNAs (miRNAs) in the control of HF development and cycling: the next frontiers in hair research. Exp Dermatol 2015; 24:821-6. [PMID: 26121602 DOI: 10.1111/exd.12785] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2015] [Indexed: 12/17/2022]
Abstract
Hair follicle development and its postnatal regeneration are characterized by dramatic changes in its microanatomy and cellular activity, which are controlled by multiple signalling pathways, transcription factors and epigenetic regulators, including microRNAs (miRNAs). miRNAs and their targets form remarkably diverse regulatory networks, playing a key role in the execution of gene expression programmes in the different cell lineages of the hair follicle. This review summarizes the roles of miRNAs in the control of hair follicle development, cycling and hair pigmentation, emphasizes the remaining problems/unanswered questions, and provides future directions in this rapidly growing and exciting area of research.
Collapse
Affiliation(s)
- Thomas Andl
- Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Natalia V Botchkareva
- Centre for Skin Sciences, Faculty of Life Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
96
|
Raval A, Behbehani GK, Nguyen LXT, Thomas D, Kusler B, Garbuzov A, Ramunas J, Holbrook C, Park CY, Blau H, Nolan GP, Artandi SE, Mitchell BS. Reversibility of Defective Hematopoiesis Caused by Telomere Shortening in Telomerase Knockout Mice. PLoS One 2015; 10:e0131722. [PMID: 26133370 PMCID: PMC4489842 DOI: 10.1371/journal.pone.0131722] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 06/04/2015] [Indexed: 01/08/2023] Open
Abstract
Telomere shortening is common in bone marrow failure syndromes such as dyskeratosis congenita (DC), aplastic anemia (AA) and myelodysplastic syndromes (MDS). However, improved knowledge of the lineage-specific consequences of telomere erosion and restoration of telomere length in hematopoietic progenitors is required to advance therapeutic approaches. We have employed a reversible murine model of telomerase deficiency to compare the dependence of erythroid and myeloid lineage differentiation on telomerase activity. Fifth generation Tert-/- (G5 Tert-/-) mice with shortened telomeres have significant anemia, decreased erythroblasts and reduced hematopoietic stem cell (HSC) populations associated with neutrophilia and increased myelopoiesis. Intracellular multiparameter analysis by mass cytometry showed significantly reduced cell proliferation and increased sensitivity to activation of DNA damage checkpoints in erythroid progenitors and in erythroid-biased CD150hi HSC, but not in myeloid progenitors. Strikingly, Cre-inducible reactivation of telomerase activity restored hematopoietic stem and progenitor cell (HSPC) proliferation, normalized the DNA damage response, and improved red cell production and hemoglobin levels. These data establish a direct link between the loss of TERT activity, telomere shortening and defective erythropoiesis and suggest that novel strategies to restore telomerase function may have an important role in the treatment of the resulting anemia.
Collapse
Affiliation(s)
- Aparna Raval
- Stanford Cancer Institute and Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, 94305, United States of America
| | - Gregory K. Behbehani
- Stanford Cancer Institute and Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, 94305, United States of America
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology Stanford University, School of Medicine, Stanford, CA, 94305, United States of America
| | - Le Xuan Truong Nguyen
- Stanford Cancer Institute and Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, 94305, United States of America
| | - Daniel Thomas
- Stanford Cancer Institute and Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, 94305, United States of America
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94035, United States of America
| | - Brenda Kusler
- Stanford Cancer Institute and Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, 94305, United States of America
| | - Alina Garbuzov
- Department of Genetics, Stanford University, Stanford, CA, 94305, United States of America
| | - John Ramunas
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology Stanford University, School of Medicine, Stanford, CA, 94305, United States of America
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94035, United States of America
| | - Colin Holbrook
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology Stanford University, School of Medicine, Stanford, CA, 94305, United States of America
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94035, United States of America
| | - Christopher Y. Park
- Human Oncology and Pathogenesis Program and Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, United States of America
| | - Helen Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology Stanford University, School of Medicine, Stanford, CA, 94305, United States of America
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94035, United States of America
| | - Garry P. Nolan
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology Stanford University, School of Medicine, Stanford, CA, 94305, United States of America
| | - Steven E. Artandi
- Departments of Medicine and Biochemistry, Stanford University, Stanford, CA, 94305, United States of America
| | - Beverly S. Mitchell
- Stanford Cancer Institute and Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, 94305, United States of America
- * E-mail:
| |
Collapse
|
97
|
Ferdowsi S, Atarodi K, Amirizadeh N, Toogeh G, Azarkeivan A, Shirkoohi R, Faranoush M, Vaezi M, Alimoghaddam K, Ghavamzadeh A, Naghadeh HT, Ghaffari SH. Expression analysis of microRNA-125 in patients with polycythemia vera and essential thrombocythemia and correlation with JAK2 allele burden and laboratory findings. Int J Lab Hematol 2015; 37:661-7. [PMID: 26011312 DOI: 10.1111/ijlh.12381] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 04/07/2015] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The JAK2V617F mutation has emerged in recent years as a diagnostic as well as a treatment target in patients with polycythemia vera (PV) and essential thrombocythemia (ET). The disease phenotype is also influenced by other factors such as microRNA (miRNA) deregulation. The aim of this study was to investigate miR-125 expression level in these patients with those obtained from healthy control subjects and its correlation with JAK2 allele burden and laboratory findings. METHODS In total, forty patients with a clinical diagnosis of PV and ET were examined at the time of diagnosis. Ten healthy subjects were checked as controls. We performed JAK2 V617F allele burdens measurement and expression analysis of miR-125b-5p, miR-125b-3p, miR-125a-5p, and miR-125a-3p in leukocytes isolated from peripheral blood by quantitative real-time polymerase chain reaction. RESULTS MiR-125b-5p and miR-125a-5p were upregulated in both patients with PV (P = 0.00 and P = 0.003, respectively) and ET (P = 0.02 and P = 0.002, respectively). In PV group, a significant correlation was observed between miR-125a-5p and platelet counts (P = 0.01, r = 0.531). The correlation between miRNA and JAk2 allele burden was not significant. CONCLUSION In conclusion, our data indicate that other factors such as aberrant miR-125 expression may influence on the disease phenotype in patients with PV and ET.
Collapse
Affiliation(s)
- S Ferdowsi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - K Atarodi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - N Amirizadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - G Toogeh
- Hematology-Oncology and BMT Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - A Azarkeivan
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - R Shirkoohi
- Molecular Genetics, Cancer Research Center, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - M Faranoush
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - M Vaezi
- Hematology-Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - K Alimoghaddam
- Hematology-Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - A Ghavamzadeh
- Hematology-Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - H Teimori Naghadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - S H Ghaffari
- Hematology-Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
98
|
Akhavan Rahnama M, Movassaghpour AA, Soleimani M, Atashi A, Anbarlou A, Shams Asenjan K. MicroRNA-15b target Sall4 and diminish in vitro UCB-derived HSCs expansion. EXCLI JOURNAL 2015; 14:601-10. [PMID: 26648817 PMCID: PMC4669904 DOI: 10.17179/excli2014-687] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 12/16/2014] [Indexed: 12/13/2022]
Abstract
Hematopoietic Stem Cells (HSCs) are cells that have the ability to self-renewal and differentiate into all of hematopoietic lineages. The lack of donors and unavailable efficient protocols for ex vivo expansion of HSCs, are obstacles in successful cell therapies. MicroRNAs (also refer as miRNAs or miRs) have significant roles in hematopoiesis; they can effect on HSCs expansion, maintaining undifferentiated state, self-renewal and differentiation. Recently attentions have been given to these small regulatory molecules to utilize them in order to expand HSCs. Using bioinformatics analysis we identified Sall4 as putative target of miR-15b and miR-219-5p. Relative expression levels of miRNAs and Sall4 were evaluated by qRT-PCR. Here we show 247-fold and 4.2-fold increasing Sall4 expression level compared to control group in CD34+ cells nucleofected by anti-miR-15b and anti-miR-219-5p, respectively. These data showed that anti-miR-15b can promote clonogenic capacity of HSCs and also we found that miR-15b alone was able to increase the number of CD34+HSCs in vitro by more than 2 fold by targeting Sall4. Moreover, level of CD34 marker in HSCs nucleofected by anti-miR-15b increased more than 50 %. Our analysis showed no statistically difference in mRNA level of Sall4 after nucleofection of anti-miR-219-5p. Sall4 is a factor capable of enhancing HSC expansion significantly. We demonstrated that inhibition of miR-15b can enhance ex vivo expansion of UCB-derived HSCs and also expression of Sall4 allowed expansion and preserve self- renewal of CD34+ HSCs.
Collapse
Affiliation(s)
| | | | - Masoud Soleimani
- Department of Hematology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Atashi
- Department of Hematology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Azadeh Anbarlou
- Department of Hematology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Karim Shams Asenjan
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tabriz, Iran
| |
Collapse
|
99
|
Cheng N, Chen X, Kim J, Shi AH, Nguyen C, Wersto R, Weng N. MicroRNA-125b modulates inflammatory chemokine CCL4 expression in immune cells and its reduction causes CCL4 increase with age. Aging Cell 2015; 14:200-8. [PMID: 25620312 PMCID: PMC4364832 DOI: 10.1111/acel.12294] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2014] [Indexed: 12/21/2022] Open
Abstract
Chemokines play a pivotal role in regulating the immune response through a tightly controlled expression. Elevated levels of inflammatory chemokines commonly occur with aging but the mechanism underlying this age-associated change is not fully understood. Here, we report the role of microRNA-125b (miR-125b) in regulating inflammatory CC chemokine 4 (CCL4) expression in human immune cells and its altered expression with aging. We first analyzed the mRNA level of CCL4 in eight different types of immune cells including CD4 and CD8 T-cell subsets (naïve, central and effector memory), B cells and monocytes in blood from both young (≤42 years) and old (≥70 years) adults. We observed that monocytes and naïve CD8 T cells expressed higher levels of CCL4 and exhibited an age-related increase in CCL4. We then found the level of miR-125b was inversely correlated with the level of CCL4 in these cells, and the level of miR-125b was reduced in monocytes and naïve CD8 T cells of the old compared to the young adults. Knock-down of miR-125b by shRNA in monocytes and naïve CD8 T cells led to an increase of CCL4 protein, whereas enhanced miR-125b expression by transfection in naïve CD8 T cells resulted in a reduction of the CCL4 mRNA and protein in response to stimulation. Finally, we demonstrated that miR-125b action requires the 'seed' sequence in 3'UTR of CCL4. Together these findings demonstrated that miR-125b is a negative regulator of CCL4 and its reduction is partially responsible for the age-related increase of CCL4.
Collapse
Affiliation(s)
- Nai‐Lin Cheng
- Laboratory of Molecular Biology & Immunology National Institute on Aging National Institutes of Health 251 Bayview Blvd. Baltimore MD 21224 USA
| | - Xiaochun Chen
- Laboratory of Molecular Biology & Immunology National Institute on Aging National Institutes of Health 251 Bayview Blvd. Baltimore MD 21224 USA
| | - Jiewan Kim
- Laboratory of Molecular Biology & Immunology National Institute on Aging National Institutes of Health 251 Bayview Blvd. Baltimore MD 21224 USA
| | - Alvin H. Shi
- Laboratory of Molecular Biology & Immunology National Institute on Aging National Institutes of Health 251 Bayview Blvd. Baltimore MD 21224 USA
| | - Cuong Nguyen
- Flow Cytometry Unit National Institute on Aging National Institutes of Health 251 Bayview Blvd. Baltimore MD 21224 USA
| | - Robert Wersto
- Flow Cytometry Unit National Institute on Aging National Institutes of Health 251 Bayview Blvd. Baltimore MD 21224 USA
| | - Nan‐ping Weng
- Laboratory of Molecular Biology & Immunology National Institute on Aging National Institutes of Health 251 Bayview Blvd. Baltimore MD 21224 USA
| |
Collapse
|
100
|
Kroesen BJ, Teteloshvili N, Smigielska-Czepiel K, Brouwer E, Boots AMH, van den Berg A, Kluiver J. Immuno-miRs: critical regulators of T-cell development, function and ageing. Immunology 2015; 144:1-10. [PMID: 25093579 DOI: 10.1111/imm.12367] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 07/30/2014] [Accepted: 07/31/2014] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are instrumental to many aspects of immunity, including various levels of T-cell immunity. Over the last decade, crucial immune functions were shown to be regulated by specific miRNAs. These 'immuno-miRs' regulate generic cell biological processes in T cells, such as proliferation and apoptosis, as well as a number of T-cell-specific features that are fundamental to the development, differentiation and function of T cells. In this review, we give an overview of the current literature with respect to the role of miRNAs at various stages of T-cell development, maturation, differentiation, activation and ageing. Little is known about the involvement of miRNAs in thymic T-cell development, although miR-181a and miR-150 have been implicated herein. In contrast, several broadly expressed miRNAs including miR-21, miR-155 and miR-17~92, have now been shown to regulate T-cell activation. Other miRNAs, including miR-146a, show a more T-cell-subset-specific expression pattern and are involved in the regulation of processes unique to that specific T-cell subset. Importantly, differences in the miRNA target gene repertoires of different T-cell subsets allow similar miRNAs to control different T-cell-subset-specific functions. Interestingly, several of the here described immuno-miRs have also been implicated in T-cell ageing and there are clear indications for causal involvement of miRNAs in immunosenescence. It is concluded that immuno-miRs have a dynamic regulatory role in many aspects of T-cell differentiation, activation, function and ageing. An important notion when studying miRNAs in relation to T-cell biology is that specific immuno-miRs may have quite unrelated functions in closely related T-cell subsets.
Collapse
Affiliation(s)
- Bart-Jan Kroesen
- Department of Laboratory Medicine, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|