1
|
Garofano K, Mariani V, Rashid K, Suwunnakorn S, Sidahmed A, Horvath A, Maggirwar SB, O’Brien TJ, Perera MA, Whalen M, Lee NH. Transcriptomic and functional characterization of megakaryocytic-derived platelet-like particles: impaired aggregation and prominent anti-tumor effects. Platelets 2025; 36:2449344. [PMID: 39812346 PMCID: PMC11890189 DOI: 10.1080/09537104.2024.2449344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025]
Abstract
Platelet-like particles (PLPs), derived from megakaryocytic cell lines MEG-01 and K-562, are widely used as a surrogate to study platelet formation and function. We demonstrate by RNA-Seq that PLPs are transcriptionally distinct from platelets. Expression of key genes in signaling pathways promoting platelet activation/aggregation, such as the PI3K/AKT, protein kinase A, phospholipase C, and α-adrenergic and GP6 receptor pathways, was missing or under-expressed in PLPs. Functionally, PLPs do not aggregate following epinephrine, collagen, or ADP stimulation. While PLPs aggregated in response to thrombin, they did not display enhanced expression of surface markers P-selectin and activated α2bβ3, in contrast to platelets. We have previously demonstrated that platelets physically couple to MDA-PCa-2b and RC77T/E prostate cancer (PCa) cells via specific ligand-receptor interactions, leading to platelet-stimulated cell invasiveness and apoptotic resistance, and reciprocal cell-induced platelet aggregation. In contrast, PLP interactions with PCa cells inhibited both cell invasion and apoptotic resistance while failing to promote PLP aggregation. Moreover, PLPs reduced platelet-PCa cell interactions and antagonized platelet-stimulated oncogenic effects in PCa cells. RNA-Seq analysis identified candidate ligand-transmembrane protein combinations involved in anti-tumorigenic signaling of PLPs to PCa cells. Antibody neutralization of the TIMP3-MMP15 and VEGFB-FGFR1 signaling axes reversed PLP-mediated anti-invasion and apoptotic sensitization, respectively. In summary, PLPs lack many transcriptomic, molecular and functional features of platelets and possess novel anti-tumorigenic properties. These findings indicate that PLPs may have a potential therapeutic role in targeting and disrupting the oncogenic signaling between platelets and cancer cells, offering a new avenue for anti-cancer strategies.
Collapse
Affiliation(s)
- Kaitlin Garofano
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, 20037
| | - Vera Mariani
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, 20037
| | - Kameron Rashid
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, 20037
| | - Sumanun Suwunnakorn
- Department of Microbiology Immunology and Tropical Medicine, The George Washington University, Washington, DC, 20037
| | - Alfateh Sidahmed
- Department of Medicine, George Washington University, Washington, DC, 20037
| | - Anelia Horvath
- Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC, 20037
| | - Sanjay B. Maggirwar
- Department of Microbiology Immunology and Tropical Medicine, The George Washington University, Washington, DC, 20037
| | - Travis J. O’Brien
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, 20037
| | - Minoli A. Perera
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University, Chicago, IL, 60611
| | - Michael Whalen
- GW Cancer Center, George Washington University, Washington, DC, 20037
| | - Norman H Lee
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, 20037
- GW Cancer Center, George Washington University, Washington, DC, 20037
| |
Collapse
|
2
|
Gwag T, Lee S, Li Z, Newcomb A, Otuagomah J, Weinman SA, Liang Y, Zhou C, Wang S. Platelet-derived thrombospondin 1 promotes immune cell liver infiltration and exacerbates diet-induced steatohepatitis. JHEP Rep 2024; 6:101019. [PMID: 38455470 PMCID: PMC10918562 DOI: 10.1016/j.jhepr.2024.101019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/09/2024] [Accepted: 01/16/2024] [Indexed: 03/09/2024] Open
Abstract
Background & Aims Recent studies have implicated platelets, particularly α-granules, in the development of non-alcoholic steatohepatitis (NASH). However, the specific mechanisms involved have yet to be determined. Notably, thrombospondin 1 (TSP1) is a major component of the platelet α-granules released during platelet activation. Hence, we aimed to determine the role of platelet-derived TSP1 in NASH. Methods Platelet-specific Tsp1 knockout mice (TSP1Δpf4) and their wild-type littermates (TSP1F/F) were used. NASH was induced by feeding the mice with a diet enriched in fat, sucrose, fructose, and cholesterol (AMLN diet). A human liver NASH organoid model was also employed. Results Although TSP1 deletion in platelets did not affect diet-induced steatosis, TSP1Δpf4 mice exhibited attenuated NASH and liver fibrosis, accompanied by improvements in plasma glucose and lipid homeostasis. Furthermore, TSP1Δpf4 mice showed reduced intrahepatic platelet accumulation, activation, and chemokine production, correlating with decreased immune cell infiltration into the liver. Consequently, this diminished proinflammatory signaling in the liver, thereby mitigating the progression of NAFLD. Moreover, in vitro data revealed that co-culturing TSP1-deficient platelets in a human liver NASH organoid model attenuated hepatic stellate cell activation and NASH progression. Additionally, TSP1-deficient platelets play a role in regulating brown fat endocrine function, specifically affecting Nrg4 (neuregulin 4) production. Crosstalk between brown fat and the liver may also influence the progression of NAFLD. Conclusions These data suggest that platelet α-granule-derived TSP1 is a significant contributor to diet-induced NASH and fibrosis, potentially serving as a new therapeutic target for this severe liver disease. Impact and implications Recent studies have implicated platelets, specifically α-granules, in the development of non-alcoholic steatohepatitis, yet the precise mechanisms remain unknown. In this study, through the utilization of a tissue-specific knockout mouse model and human 3D liver organoid, we demonstrated that platelet α-granule-derived TSP1 significantly contributes to diet-induced non-alcoholic steatohepatitis and fibrosis. This contribution is, in part, attributed to the regulation of intrahepatic immune cell infiltration and potential crosstalk between fat and the liver. These findings suggest that platelet-derived TSP1 may represent a novel therapeutic target in non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Taesik Gwag
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, United States
- Lexington Veterans Affairs Medical Center, Lexington, KY 40502, United States
| | - Sangderk Lee
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, United States
| | - Zhenyu Li
- Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX, 77843, United States
| | - Alana Newcomb
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, United States
| | - Josephine Otuagomah
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, United States
| | - Steven A. Weinman
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, United States
- Research Service, Kansas City VA Medical Center, Kansas City, MO 64128, United States
| | - Ying Liang
- New York Blood Center, 310 East 72 Street, New York, NY 10065, United States
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA92521, United States
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, United States
- Lexington Veterans Affairs Medical Center, Lexington, KY 40502, United States
| |
Collapse
|
3
|
Petito E, Gresele P. Immune attack on megakaryocytes in immune thrombocytopenia. Res Pract Thromb Haemost 2024; 8:102345. [PMID: 38525349 PMCID: PMC10960061 DOI: 10.1016/j.rpth.2024.102345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/04/2024] [Indexed: 03/26/2024] Open
Abstract
A State of the Art lecture titled "Immune Attack on Megakaryocytes in ITP: The Role of Megakaryocyte Impairment" was presented at the International Society on Thrombosis and Haemostasis Congress in 2023. Immune thrombocytopenia (ITP) is an acquired autoimmune disorder caused by autoantibodies against platelet surface glycoproteins that provoke increased clearance of circulating platelets, leading to reduced platelet number. However, there is also evidence of a direct effect of antiplatelet autoantibodies on bone marrow megakaryocytes. Indeed, immunologic cells responsible for autoantibody production reside in the bone marrow; megakaryocytes progressively express during their maturation the same glycoproteins against which ITP autoantibodies are directed, and platelet autoantibodies have been detected in the bone marrow of patients with ITP. In vitro studies using ITP sera or monoclonal antibodies against platelet and megakaryocyte surface glycoproteins have shown an impairment of many steps of megakaryopoiesis and thrombopoiesis, such as megakaryocyte differentiation and maturation, migration from the osteoblastic to the vascular niche, adhesion to extracellular matrix proteins, and proplatelet formation, resulting in impaired and ectopic platelet production in the bone marrow and diminished platelet release in the bloodstream. Moreover, cytotoxic T cells may target bone marrow megakaryocytes, resulting in megakaryocyte destruction. Altogether, these findings suggest that antiplatelet autoantibodies and cellular immunity against bone marrow megakaryocytes may significantly contribute to thrombocytopenia in some patients with ITP. Finally, we summarize relevant new data on this topic presented during the 2023 International Society on Thrombosis and Haemostasis Congress. The complete unraveling of the mechanisms of immune attack-induced impairment of megakaryopoiesis and thrombopoiesis may open the way to new therapeutic approaches.
Collapse
Affiliation(s)
- Eleonora Petito
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Paolo Gresele
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
4
|
Karam-Palos S, Andrés-Blasco I, Campos-Borges C, Zanón-Moreno V, Gallego-Martínez A, Alegre-Ituarte V, García-Medina JJ, Pastor-Idoate S, Sellés-Navarro I, Vila-Arteaga J, Lleó-Perez AV, Pinazo-Durán MD. Oxidative Stress Mediates Epigenetic Modifications and the Expression of miRNAs and Genes Related to Apoptosis in Diabetic Retinopathy Patients. J Clin Med 2023; 13:74. [PMID: 38202081 PMCID: PMC10780047 DOI: 10.3390/jcm13010074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Knowledge on the underlying mechanisms and molecular targets for managing the ocular complications of type 2 diabetes mellitus (T2DM) remains incomplete. Diabetic retinopathy (DR) is a major cause of irreversible visual disability worldwide. By using ophthalmological and molecular-genetic approaches, we gathered specific information to build a data network for deciphering the crosslink of oxidative stress (OS) and apoptosis (AP) processes, as well as to identify potential epigenetic modifications related to noncoding RNAs in the eyes of patients with T2DM. A total of 120 participants were recruited, being classified into two groups: individuals with T2MD (T2MDG, n = 67), divided into a group of individuals with (+DR, n = 49) and without (-DR, n = 18) DR, and a control group (CG, n = 53). Analyses of compiled data reflected significantly higher plasma levels of malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GPx) and significantly lower total antioxidant capacity (TAC) in the +DR patients compared with the -DR and the CG groups. Furthermore, the plasma caspase-3 (CAS3), highly involved in apoptosis (AP), showed significantly higher values in the +DR group than in the -DR patients. The microRNAs (miR) hsa-miR 10a-5p and hsa-miR 15b-5p, as well as the genes BCL2L2 and TP53 involved in these pathways, were identified in relation to DR clinical changes. Our data suggest an interaction between OS and the above players in DR pathogenesis. Furthermore, potential miRNA-regulated target genes were identified in relation to DR. In this concern, we may raise new diagnostic and therapeutic challenges that hold the potential to significantly improve managing the diabetic eye.
Collapse
Affiliation(s)
- Sarah Karam-Palos
- Ophthalmic Research Unit “Santiago Grisolía”/FISABIO, 46017 Valencia, Spain; (S.K.-P.); (I.A.-B.); (C.C.-B.); (V.A.-I.); (A.V.L.-P.)
- Cellular and Molecular Ophthalmo-Biology Group, Department of Surgery, University of Valencia, 46010 Valencia, Spain
- Department of Ophthalmology, University Hospital “Arnau de Vilanova”, 25196 Valencia, Spain
| | - Irene Andrés-Blasco
- Ophthalmic Research Unit “Santiago Grisolía”/FISABIO, 46017 Valencia, Spain; (S.K.-P.); (I.A.-B.); (C.C.-B.); (V.A.-I.); (A.V.L.-P.)
- Cellular and Molecular Ophthalmo-Biology Group, Department of Surgery, University of Valencia, 46010 Valencia, Spain
- Net of Research in Inflammatory Diseases and Immunopathology of Organs and Systems “REI-RICORS” RD, Institute of Health Carlos III, 28029 Madrid, Spain; (J.J.G.-M.); (S.P.-I.); (I.S.-N.)
| | - Cristina Campos-Borges
- Ophthalmic Research Unit “Santiago Grisolía”/FISABIO, 46017 Valencia, Spain; (S.K.-P.); (I.A.-B.); (C.C.-B.); (V.A.-I.); (A.V.L.-P.)
- Cellular and Molecular Ophthalmo-Biology Group, Department of Surgery, University of Valencia, 46010 Valencia, Spain
- Institute of Biotechnology, University of Porto, 4169-007 Porto, Portugal
| | - Vicente Zanón-Moreno
- Ophthalmic Research Unit “Santiago Grisolía”/FISABIO, 46017 Valencia, Spain; (S.K.-P.); (I.A.-B.); (C.C.-B.); (V.A.-I.); (A.V.L.-P.)
- Cellular and Molecular Ophthalmo-Biology Group, Department of Surgery, University of Valencia, 46010 Valencia, Spain
- Net of Research in Inflammatory Diseases and Immunopathology of Organs and Systems “REI-RICORS” RD, Institute of Health Carlos III, 28029 Madrid, Spain; (J.J.G.-M.); (S.P.-I.); (I.S.-N.)
- Department of Preventive Medicine and Public Health, University of Valencia, 46010 Valencia, Spain
| | - Alex Gallego-Martínez
- Ophthalmic Research Unit “Santiago Grisolía”/FISABIO, 46017 Valencia, Spain; (S.K.-P.); (I.A.-B.); (C.C.-B.); (V.A.-I.); (A.V.L.-P.)
- Cellular and Molecular Ophthalmo-Biology Group, Department of Surgery, University of Valencia, 46010 Valencia, Spain
| | - Victor Alegre-Ituarte
- Ophthalmic Research Unit “Santiago Grisolía”/FISABIO, 46017 Valencia, Spain; (S.K.-P.); (I.A.-B.); (C.C.-B.); (V.A.-I.); (A.V.L.-P.)
- Cellular and Molecular Ophthalmo-Biology Group, Department of Surgery, University of Valencia, 46010 Valencia, Spain
| | - Jose J. García-Medina
- Net of Research in Inflammatory Diseases and Immunopathology of Organs and Systems “REI-RICORS” RD, Institute of Health Carlos III, 28029 Madrid, Spain; (J.J.G.-M.); (S.P.-I.); (I.S.-N.)
- Department of Ophthalmology, University Hospital “Morales Meseguer”, 30008 Murcia, Spain
- Department of Surgery, Pediatrics, Obstetrics and Ginecology, Faculty of Medicine, University of Murcia, 30100 Murcia, Spain
| | - Salvador Pastor-Idoate
- Net of Research in Inflammatory Diseases and Immunopathology of Organs and Systems “REI-RICORS” RD, Institute of Health Carlos III, 28029 Madrid, Spain; (J.J.G.-M.); (S.P.-I.); (I.S.-N.)
- Institute of Applied Ophthalmobiology “IOBA”, University of Valladolid, 47002 Valladolid, Spain
- Department of Ophthalmology, University Clinic Hospital of Valladolid, 47003 Valladolid, Spain
| | - Inmaculada Sellés-Navarro
- Net of Research in Inflammatory Diseases and Immunopathology of Organs and Systems “REI-RICORS” RD, Institute of Health Carlos III, 28029 Madrid, Spain; (J.J.G.-M.); (S.P.-I.); (I.S.-N.)
- Department of Surgery, Pediatrics, Obstetrics and Ginecology, Faculty of Medicine, University of Murcia, 30100 Murcia, Spain
- Department of Ophthalmology, University Hospital “Reina Sofia”, 30003 Murcia, Spain
| | - Jorge Vila-Arteaga
- Department of Ophthalmology, University and Polyclinic Hospital “La Fé”, 46026 Valencia, Spain;
- Innova Ocular Vila Clinic, 46004 Valencia, Spain
| | - Antonio V. Lleó-Perez
- Ophthalmic Research Unit “Santiago Grisolía”/FISABIO, 46017 Valencia, Spain; (S.K.-P.); (I.A.-B.); (C.C.-B.); (V.A.-I.); (A.V.L.-P.)
- Cellular and Molecular Ophthalmo-Biology Group, Department of Surgery, University of Valencia, 46010 Valencia, Spain
- Department of Ophthalmology, University Hospital “Arnau de Vilanova”, 25196 Valencia, Spain
| | - Maria D. Pinazo-Durán
- Ophthalmic Research Unit “Santiago Grisolía”/FISABIO, 46017 Valencia, Spain; (S.K.-P.); (I.A.-B.); (C.C.-B.); (V.A.-I.); (A.V.L.-P.)
- Cellular and Molecular Ophthalmo-Biology Group, Department of Surgery, University of Valencia, 46010 Valencia, Spain
- Net of Research in Inflammatory Diseases and Immunopathology of Organs and Systems “REI-RICORS” RD, Institute of Health Carlos III, 28029 Madrid, Spain; (J.J.G.-M.); (S.P.-I.); (I.S.-N.)
| |
Collapse
|
5
|
Seetharam SM, Liu Y, Wu J, Fechter L, Murugesan K, Maecker H, Gotlib J, Zehnder J, Paulmurugan R, Krishnan A. Enkurin: a novel marker for myeloproliferative neoplasms from platelet, megakaryocyte, and whole blood specimens. Blood Adv 2023; 7:5433-5445. [PMID: 37315179 PMCID: PMC10509670 DOI: 10.1182/bloodadvances.2022008939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 05/10/2023] [Accepted: 05/30/2023] [Indexed: 06/16/2023] Open
Abstract
Impaired protein homeostasis, though well established in age-related disorders, has been recently linked with the pathogenesis of myeloproliferative neoplasms (MPNs). However, little is known about MPN-specific modulators of proteostasis, thus impeding our ability for increased mechanistic understanding and discovery of additional therapeutic targets. Loss of proteostasis, in itself, is traced to dysregulated mechanisms in protein folding and intracellular calcium signaling at the endoplasmic reticulum (ER). Here, using ex vivo and in vitro systems (including CD34+ cultures from patient bone marrow and healthy cord/peripheral blood specimens), we extend our prior data from platelet RNA sequencing in patients with MPN and discover select proteostasis-associated markers at RNA and/or protein levels in each of platelet, parent megakaryocyte, and whole blood specimens. Importantly, we identify a novel role in MPNs for enkurin (ENKUR), a calcium mediator protein originally implicated only in spermatogenesis. Our data reveal consistent ENKUR downregulation at both RNA and protein levels across specimens from patients with MPN and experimental models (including upon treatment with thapsigargin, an agent that causes protein misfolding in the ER by selective loss of calcium), with a concomitant upregulation of a cell cycle marker, CDC20. Silencing of ENKUR using short hairpin RNA in CD34+-derived megakaryocytes further confirms this association with CDC20 at both RNA and protein levels and indicates a likely role for the PI3K/Akt pathway. Together, our work sheds light on enkurin as a novel marker of MPN pathogenesis and indicates further mechanistic investigation into a role for dysregulated calcium homeostasis and ER and protein folding stress in MPN transformation.
Collapse
Affiliation(s)
| | - Yi Liu
- Department of Radiology, Stanford University, Stanford, CA
| | - Jason Wu
- High-Throughput Bioscience Center and Stanford Genomics, Stanford University School of Medicine, Stanford, CA
| | - Lenn Fechter
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | | | - Holden Maecker
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA
| | - Jason Gotlib
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - James Zehnder
- Department of Pathology, Stanford University, Stanford, CA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Anandi Krishnan
- Department of Pathology, Stanford University, Stanford, CA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
6
|
Sun Y, Wang T, Lv Y, Li J, Jiang X, Jiang J, Zhang D, Bian W, Zhang C. MALAT1 promotes platelet activity and thrombus formation through PI3k/Akt/GSK-3β signalling pathway. Stroke Vasc Neurol 2023; 8:181-192. [PMID: 36241224 PMCID: PMC10359792 DOI: 10.1136/svn-2022-001498] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 09/21/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Ischaemic stroke and other cardiovascular illnesses are characterised by abnormalities in the processes of thrombosis and haemostasis, which rely on platelet activity. In platelets, a wide variety of microRNAs (long non-coding RNA, lncRNAs) is found. Due to the absence of nuclear DNA in platelets, lncRNAs may serve as critical post-transcriptional regulators of platelet activities. However, research into the roles of lncRNAs in platelets is limited. OBJECTIVE The purpose of this study is to learn more about the molecular mechanism by which MALAT1 affects platelet activity and thrombus formation. METHODS/RESULTS The CD34+ megakaryocytes used in this research as an in vitro model for human megakaryocytes and platelets. Cell adhesion and spreading are enhanced in the absence and presence of agonists in CD34+ megakaryocytes subjected to MALAT1 knockdown (KD). The adhesion and activity of platelet-like particles produced by MALAT1 KD cells are significantly enhanced at rest and after thrombin activation. Thrombus development on a collagen matrix is also greatly enhanced in the microfluidic whole-blood perfusion model: platelets lacking MALAT1 exhibit elevated accumulation, distributing area and activity. In addition, MALAT1-deficient mice bleed less and form a stable occlusive thrombus more quickly than wild-type mice. PTEN and PDK1 regulated the activity of MALAT1 in platelets to carry out its PI3k/Akt/GSK-3β signalling pathway-related function. CONCLUSION The suppression of MALAT1 expression significantly increases platelet adhesion, spreading, platelet activity, and thrombus formation. lncRNAs may constitute a unique class of platelet function modulators.
Collapse
Affiliation(s)
- Yeying Sun
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Tao Wang
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Yan Lv
- College of Life Sciences, Yantai University, Yantai, Shandong, China
| | - Jiahua Li
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Xiaoli Jiang
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Jing Jiang
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Daolai Zhang
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Weihua Bian
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Chunxiang Zhang
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
- Department of Cardiology, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
7
|
Huang YF, Su SC, Chuang HY, Chen HH, Twu YC. Histone deacetylation-regulated cell surface Siglec-7 expression promoted megakaryocytic maturation and enhanced platelet-like particle release. J Thromb Haemost 2023; 21:329-343. [PMID: 36700509 DOI: 10.1016/j.jtha.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Functioning as important hematologic cells for hemostasis, wound healing and immune defense platelets are produced before being released into the blood by cytoplasmic fragmentation at the end of the megakaryocyte (MK) differentiation, during which the involvement of both apoptosis and autophagy has been reported. Inhibitory sialic acid-binding immunoglobulin-like lectin-7 gene (Siglec-7) can be expressed on platelets and induce apoptosis on activation for uncharacterized function. OBJECTIVE We aimed to investigate the regulatory mechanism for Siglec-7 activation along MK differentiation and its physiologic role during the MK maturation and platelet formation. METHODS By using 2 well-established MK differentiation models (HEL and K562) and human primary CD34+ cell, we examined the upregulations of transcript and protein levels of Siglec-7 during MK differentiation, and the effect of Siglec-7 surface presence on MK differentiation and platelet-like particles (PLPs) release. RESULTS We show that both transcripts and surface Siglec-7 were elevated during MK differentiation, and the histone deacetylase 1 (HDAC1) acted as a negative regulator for Siglec-7 activation. By increasing Siglec-7 surface expression, we found that increased presence of Siglec-7 not only enhanced MK maturation but also the release of PLPs by activating caspase 3-dependent signaling, as evidenced in the observation of more CD41, polyploidy, and platelet factor 4 transcript formations. CONCLUSION In this study, we demonstrated that Siglec-7 activation was subjected to epigenetic regulation, and the resulting induced expression of surface Siglec-7 played an important regulatory role in promoting MK differentiation, maturation, and PLP formation.
Collapse
Affiliation(s)
- Yun-Fei Huang
- Department of Biotechnology and Laboratory Science in Medicine, School of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Chi Su
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Hui-Yu Chuang
- Department of Biotechnology and Laboratory Science in Medicine, School of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiao-Han Chen
- Department of Biotechnology and Laboratory Science in Medicine, School of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yuh-Ching Twu
- Department of Biotechnology and Laboratory Science in Medicine, School of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
8
|
Mosale Seetharam S, Liu Y, Wu J, Fechter L, Murugesan K, Maecker H, Gotlib J, Zehnder J, Paulmurugan R, Krishnan A. Enkurin: A novel marker for myeloproliferative neoplasms from platelet, megakaryocyte, and whole blood specimens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.07.523111. [PMID: 36712071 PMCID: PMC9881897 DOI: 10.1101/2023.01.07.523111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Impaired protein homeostasis, though well established in age-related disorders, has been linked in recent research with the pathogenesis of myeloproliferative neoplasms (MPNs). As yet, however, little is known about MPN-specific modulators of proteostasis, thus impeding our ability for increased mechanistic understanding and discovery of additional therapeutic targets. Loss of proteostasis, in itself, is traced to dysregulated mechanisms in protein folding and intracellular calcium signaling at the endoplasmic reticulum (ER). Here, using ex vivo and in vitro systems (including CD34 + cultures from patient bone marrow, and healthy cord/peripheral blood specimens), we extend our prior data from MPN patient platelet RNA sequencing, and discover select proteostasis-associated markers at RNA and/or protein levels in each of platelets, parent megakaryocytes, and whole blood specimens. Importantly, we identify a novel role in MPNs for enkurin ( ENKUR ), a calcium mediator protein, implicated originally only in spermatogenesis. Our data reveal consistent ENKUR downregulation at both RNA and protein levels across MPN patient specimens and experimental models, with a concomitant upregulation of a cell cycle marker, CDC20 . Silencing of ENKUR by shRNA in CD34 + derived megakaryocytes further confirm this association with CDC20 at both RNA and protein levels; and indicate a likely role for the PI3K/Akt pathway. The inverse association of ENKUR and CDC20 expression was further confirmed upon treatment with thapsigargin (an agent that causes protein misfolding in the ER by selective loss of calcium) in both megakaryocyte and platelet fractions at RNA and protein levels. Together, our work sheds light on enkurin as a novel marker of MPN pathogenesis beyond the genetic alterations; and indicates further mechanistic investigation into a role for dysregulated calcium homeostasis, and ER and protein folding stress in MPN transformation. VISUAL ABSTRACT Key Points Enkurin, a calcium adaptor protein, is identified as a novel marker of pathogenesis in MPNs.MPN megakaryocyte and platelet expression of enkurin at RNA and protein levels is inversely associated with a cell differentiation cycle gene, CDC20.Likely role for dysregulated calcium homeostasis, and ER and protein folding stress in MPN transformation.
Collapse
Affiliation(s)
| | - Yi Liu
- Department of Radiology, Stanford University, Stanford, CA
| | - Jason Wu
- High-Throughput Bioscience Center (HTBC), Stanford University School of Medicine, Stanford, CA
| | - Lenn Fechter
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | | | - Holden Maecker
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA
| | - Jason Gotlib
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - James Zehnder
- Department of Pathology, Stanford University, Stanford, CA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Anandi Krishnan
- Department of Pathology, Stanford University, Stanford, CA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
9
|
Yang S, Wang L, Wu Y, Wu A, Huang F, Tang X, Kantawong F, Anuchapreeda S, Qin D, Mei Q, Chen J, Huang X, Zhang C, Wu J. Apoptosis in megakaryocytes: Safeguard and threat for thrombopoiesis. Front Immunol 2023; 13:1025945. [PMID: 36685543 PMCID: PMC9845629 DOI: 10.3389/fimmu.2022.1025945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023] Open
Abstract
Platelets, generated from precursor megakaryocytes (MKs), are central mediators of hemostasis and thrombosis. The process of thrombopoiesis is extremely complex, regulated by multiple factors, and related to many cellular events including apoptosis. However, the role of apoptosis in thrombopoiesis has been controversial for many years. Some researchers believe that apoptosis is an ally of thrombopoiesis and platelets production is apoptosis-dependent, while others have suggested that apoptosis is dispensable for thrombopoiesis, and is even inhibited during this process. In this review, we will focus on this conflict, discuss the relationship between megakaryocytopoiesis, thrombopoiesis and apoptosis. In addition, we also consider why such a vast number of studies draw opposite conclusions of the role of apoptosis in thrombopoiesis, and try to figure out the truth behind the mystery. This review provides more comprehensive insights into the relationship between megakaryocytopoiesis, thrombopoiesis, and apoptosis and finds some clues for the possible pathological mechanisms of platelet disorders caused by abnormal apoptosis.
Collapse
Affiliation(s)
- Shuo Yang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Long Wang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yuesong Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Anguo Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
| | - Feihong Huang
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
| | - Xiaoqin Tang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Fahsai Kantawong
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Songyot Anuchapreeda
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Dalian Qin
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qibing Mei
- School of Pharmacy, Southwest Medical University, Luzhou, China
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jianping Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xinwu Huang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chunxiang Zhang
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
| | - Jianming Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
10
|
Sequence and expression regulation of the BCL2L2 gene in pigs. Gene 2023; 851:146992. [DOI: 10.1016/j.gene.2022.146992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 09/11/2022] [Accepted: 10/14/2022] [Indexed: 11/27/2022]
|
11
|
Freitag M, Schwertz H. A New Role of NAP1L1 in Megakaryocytes and Human Platelets. Int J Mol Sci 2022; 23:ijms232314694. [PMID: 36499021 PMCID: PMC9737020 DOI: 10.3390/ijms232314694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/26/2022] Open
Abstract
Platelets (PLTs) are anucleate and considered incapable of nuclear functions. Contrastingly, nuclear proteins were detected in human PLTs. For most of these proteins, it is unclear if nuclear or alternatively assigned functions are performed, a question we wanted to address for nuclear assembly protein 1like 1 (NAP1L1). Using a wide array of molecular methods, including RNAseq, co-IP, overexpression and functional assays, we explored expression pattern and functionality of NAP1L1 in PLTs, and CD34+-derived megakaryocytes (MKs). NAP1L1 is expressed in PLTs and MKs. Co-IP experiments revealed that dihydrolipolylysine-residue acetyltransferase (DLAT encoded protein PDC-E2, ODP2) dynamically interacts with NAP1L1. PDC-E2 is part of the mitochondrial pyruvate-dehydrogenase (PDH) multi-enzyme complex, playing a crucial role in maintaining cellular respiration, and promoting ATP-synthesis via the respiratory chain. Since altered mitochondrial function is a hallmark of infectious syndromes, we analyzed PDH activity in PLTs from septic patients demonstrating increased activity, paralleling NAP1L1 expression levels. MKs PDH activity decreased following an LPS-challenge. Furthermore, overexpression of NAP1L1 significantly altered the ability of MKs to form proplatelet extensions, diminishing thrombopoiesis. These results indicate that NAP1L1 performs in other than nucleosome-assembly functions in PTLs and MKs, binding a key mitochondrial protein as a potential chaperone, and gatekeeper, influencing PDH activity and thrombopoiesis.
Collapse
Affiliation(s)
- Martin Freitag
- Department of Cardiac Surgery, Heart Center Leipzig-University Hospital, 04289 Leipzig, Germany
| | - Hansjörg Schwertz
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
- Division of Occupational Medicine, University of Utah, Salt Lake City, UT 84112, USA
- Occupational Medicine at Billings Clinic Bozeman, Bozeman, MT 59715, USA
- Correspondence: or
| |
Collapse
|
12
|
Kumar B, Afshar-Kharghan V, Mendt M, Sackstein R, Tanner MR, Popat U, Ramdial J, Daher M, Jimenez J, Basar R, Melo Garcia L, Shanley M, Kaplan M, Wan X, Nandivada V, Reyes Silva F, Woods V, Gilbert A, Gonzalez-Delgado R, Acharya S, Lin P, Rafei H, Banerjee PP, Shpall EJ. Engineered cord blood megakaryocytes evade killing by allogeneic T-cells for refractory thrombocytopenia. Front Immunol 2022; 13:1018047. [PMID: 36203567 PMCID: PMC9530569 DOI: 10.3389/fimmu.2022.1018047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/05/2022] [Indexed: 11/23/2022] Open
Abstract
The current global platelet supply is often insufficient to meet all the transfusion needs of patients, in particular for those with alloimmune thrombocytopenia. To address this issue, we have developed a strategy employing a combination of approaches to achieve more efficient production of functional megakaryocytes (MKs) and platelets collected from cord blood (CB)-derived CD34+ hematopoietic cells. This strategy is based on ex-vivo expansion and differentiation of MKs in the presence of bone marrow niche-mimicking mesenchymal stem cells (MSCs), together with two other key components: (1) To enhance MK polyploidization, we used the potent pharmacological Rho-associated coiled-coil kinase (ROCK) inhibitor, KD045, resulting in liberation of increased numbers of functional platelets both in-vitro and in-vivo; (2) To evade HLA class I T-cell-driven killing of these expanded MKs, we employed CRISPR-Cas9-mediated β-2 microglobulin (β2M) gene knockout (KO). We found that coculturing with MSCs and MK-lineage-specific cytokines significantly increased MK expansion. This was further increased by ROCK inhibition, which induced MK polyploidization and platelet production. Additionally, ex-vivo treatment of MKs with KD045 resulted in significantly higher levels of engraftment and donor chimerism in a mouse model of thrombocytopenia. Finally, β2M KO allowed MKs to evade killing by allogeneic T-cells. Overall, our approaches offer a novel, readily translatable roadmap for producing adult donor-independent platelet products for a variety of clinical indications.
Collapse
Affiliation(s)
- Bijender Kumar
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Vahid Afshar-Kharghan
- Section of Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mayela Mendt
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Robert Sackstein
- Department of Translational Medicine, Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Mark R. Tanner
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jeremy Ramdial
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - May Daher
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Juan Jimenez
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rafet Basar
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Luciana Melo Garcia
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mayra Shanley
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mecit Kaplan
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xinhai Wan
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Vandana Nandivada
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Francia Reyes Silva
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Vernikka Woods
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - April Gilbert
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ricardo Gonzalez-Delgado
- Section of Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sunil Acharya
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Paul Lin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hind Rafei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Pinaki Prosad Banerjee
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Elizabeth J. Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: Elizabeth J. Shpall,
| |
Collapse
|
13
|
Toward in Vitro Production of Platelet from Induced Pluripotent Stem Cells. Stem Cell Rev Rep 2022; 18:2376-2387. [PMID: 35397051 DOI: 10.1007/s12015-022-10366-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 10/18/2022]
Abstract
Platelets (PLTs) are small anucleate blood cells that release from polyploidy megakaryocytes(MKs). PLT transfusion is standard therapy to prevent hemorrhage. PLT transfusion is donor-dependent way which have limitations including the inadequate donor blood supply, poor quality, and issues related to infection and immunity. Overcoming these obstacles is possible with in vitro production of human PLTs. Currently several cells have been considered as source to in vitro production of PLTs such as hematopoietic stem cells (HSCs), embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). However, HSCs are a limited source for PLT production and large-scale expansion of HSC-derived PLT remains difficult. Alternative sources can be ESCs which have unlimited expansion capacity. But ESCs have ethical issues related to destroying human embryos. iPSCs are considered as an ideal unlimited source for PLT production. They are able to differentiate into any cells and have the capacity of self-renewal. Moreover, iPSCs can be acquired from any donor and easily manipulated. Due to new advances in development of MK cell lines, bioreactors, feeder cell-free production and the ability of large scale generation, iPSC-based PLTs are moving toward clinical applicability and considering the minimal risk of alloimmunization and tumorigenesis of these products, there is great hopefulness they will become the standard source for blood transfusions in the future. This review will focus on how to progress of in vitro generation of PLT from stem cell especially iPSCs and some of the successful strategies that can be easily used in clinic will be described.
Collapse
|
14
|
Zehnle PMA, Wu Y, Pommerening H, Erlacher M. Stayin‘ alive: BCL-2 proteins in the hematopoietic system. Exp Hematol 2022; 110:1-12. [PMID: 35315320 DOI: 10.1016/j.exphem.2022.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 11/04/2022]
|
15
|
Bhatlekar S, Jacob S, Tugolukova E, Manne BK, Kosaka Y, Loher P, O'Connell RM, Planelles V, Rondina MT, Rigoutsos I, Bray PF. Interferon α-induced SAMHD1 regulates human cultured megakaryocyte apoptosis and proplatelet formation. Haematologica 2022; 107:558-561. [PMID: 34758609 PMCID: PMC8804568 DOI: 10.3324/haematol.2021.279864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/28/2021] [Indexed: 11/16/2022] Open
Affiliation(s)
- Seema Bhatlekar
- Program in Molecular Medicine and Department of Internal Medicine, University of Utah, Salt Lake City
| | - Shancy Jacob
- Program in Molecular Medicine and Department of Internal Medicine, University of Utah, Salt Lake City
| | - Emilia Tugolukova
- Program in Molecular Medicine and Department of Internal Medicine, University of Utah, Salt Lake City
| | - Bhanu K Manne
- Program in Molecular Medicine and Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Yasuhiro Kosaka
- Program in Molecular Medicine and Department of Internal Medicine, University of Utah, Salt Lake City
| | - Phillipe Loher
- Computational Medicine Center, Thomas Jefferson University, Philadelphia
| | - Ryan M O'Connell
- Division of Microbiology and Immunology, Department of Pathology, and Huntsman Cancer Institute, University of Utah Health Sciences Center, University of Utah, Salt Lake City
| | - Vicente Planelles
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City
| | - Matthew T Rondina
- Program in Molecular Medicine and Department of Internal Medicine, University of Utah, Salt Lake City, UT; Department of Pathology, University of Utah, Salt Lake City, UT and the George E. Wahlen VAMC Department of Medicine and George E. Wahlen VAMC GRECC, Salt Lake City
| | - Isidore Rigoutsos
- Department of Pathology, University of Utah, Salt Lake City, UT and the George E. Wahlen VAMC Department of Medicine and George E. Wahlen VAMC GRECC, Salt Lake City
| | - Paul F Bray
- Program in Molecular Medicine and Department of Internal Medicine, University of Utah, Salt Lake City, UT; Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City.
| |
Collapse
|
16
|
Yu T, Yu J, Lu L, Zhang Y, Zhou Y, Zhou Y, Huang F, Sun L, Guo Z, Hou G, Dong Z, Wang B. MT1JP-mediated miR-24-3p/BCL2L2 axis promotes Lenvatinib resistance in hepatocellular carcinoma cells by inhibiting apoptosis. Cell Oncol (Dordr) 2021; 44:821-834. [PMID: 33974236 PMCID: PMC8338827 DOI: 10.1007/s13402-021-00605-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2021] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Lenvatinib is a long-awaited alternative to Sorafenib for first-line targeted therapy of patients with advanced hepatocellular carcinoma (HCC). However, resistance to Lenvatinib results in tumor progression and has become a major obstacle to improving the prognosis of HCC patients. Exploring the mechanisms underlying Lenvatinib resistance is considered essential for the treatment of advanced HCC. METHODS Lenvatinib resistant HCC (LR-HCC) cells were generated and potential long non-coding RNAs (Lnc-RNAs) upregulated in LR-HCC cells were identified by RNA sequencing. The effects of upregulated Lnc-RNAs were evaluated in vitro in cell models and in vivo in experimental animals using quantitative cell viability and apoptosis assays. RESULTS We found that Lnc-RNA MT1JP (MT1JP) was upregulated in LR-HCC cells and inhibited the apoptosis signaling pathway. In addition, we found that sponging of microRNA-24-3p by MT1JP released Bcl-2 like 2 (BCL2L2), an anti-apoptotic protein, thereby forming a positive-feedback loop. The role of this feedback loop was validated using rescue assays. Additionally, we found that upregulation of MT1JP and BCL2L2 impaired the sensitivity of HCC cells to Lenvatinib both vitro and vivo. CONCLUSIONS Our results suggest a novel molecular feedback loop between MT1JP and apoptosis signaling in Lenvatinib sensitive HCC cells.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Apoptosis/drug effects
- Apoptosis/genetics
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/metabolism
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Cell Line, Tumor
- Cell Survival/drug effects
- Cell Survival/genetics
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Mice, Inbred BALB C
- Mice, Inbred NOD
- Mice, Knockout
- Mice, Nude
- Mice, SCID
- MicroRNAs/genetics
- Phenylurea Compounds/therapeutic use
- Quinolines/therapeutic use
- RNA, Long Noncoding/genetics
- Xenograft Model Antitumor Assays/methods
- Mice
Collapse
Affiliation(s)
- Ting Yu
- Department of Hepatobiliary, Fuling Central Hospital of Chongqing City, Chongqing, China
| | - Jiajian Yu
- Department of Hepatobiliary, Fuling Central Hospital of Chongqing City, Chongqing, China
| | - Lu Lu
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Yize Zhang
- Precision Medicine Center, Gene Hospital of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yadong Zhou
- Department of Hepatobiliary, Fuling Central Hospital of Chongqing City, Chongqing, China
| | - Yong Zhou
- Department of Hepatobiliary, Fuling Central Hospital of Chongqing City, Chongqing, China
| | - Fengling Huang
- Department of Radiology, Fuling Central Hospital of Chongqing City, Chongqing, China
| | - Lu Sun
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhixian Guo
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guojun Hou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China.
| | - Zihui Dong
- Precision Medicine Center, Gene Hospital of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Bibo Wang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China.
| |
Collapse
|
17
|
Montenont E, Bhatlekar S, Jacob S, Kosaka Y, Manne BK, Lee O, Parra-Izquierdo I, Tugolukova E, Tolley ND, Rondina MT, Bray PF, Rowley JW. CRISPR-edited megakaryocytes for rapid screening of platelet gene functions. Blood Adv 2021; 5:2362-2374. [PMID: 33944898 PMCID: PMC8114553 DOI: 10.1182/bloodadvances.2020004112] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/09/2021] [Indexed: 01/07/2023] Open
Abstract
Human anucleate platelets cannot be directly modified using traditional genetic approaches. Instead, studies of platelet gene function depend on alternative models. Megakaryocytes (the nucleated precursor to platelets) are the nearest cell to platelets in origin, structure, and function. However, achieving consistent genetic modifications in primary megakaryocytes has been challenging, and the functional effects of induced gene deletions on human megakaryocytes for even well-characterized platelet genes (eg, ITGA2B) are unknown. Here we present a rapid and systematic approach to screen genes for platelet functions in CD34+ cell-derived megakaryocytes called CRIMSON (CRISPR-edited megakaryocytes for rapid screening of platelet gene functions). By using CRISPR/Cas9, we achieved efficient nonviral gene editing of a panel of platelet genes in megakaryocytes without compromising megakaryopoiesis. Gene editing induced loss of protein in up to 95% of cells for platelet function genes GP6, RASGRP2, and ITGA2B; for the immune receptor component B2M; and for COMMD7, which was previously associated with cardiovascular disease and platelet function. Gene deletions affected several select responses to platelet agonists in megakaryocytes in a manner largely consistent with those expected for platelets. Deletion of B2M did not significantly affect platelet-like responses, whereas deletion of ITGA2B abolished agonist-induced integrin activation and spreading on fibrinogen without affecting the translocation of P-selectin. Deletion of GP6 abrogated responses to collagen receptor agonists but not thrombin. Deletion of RASGRP2 impaired functional responses to adenosine 5'-diphosphate (ADP), thrombin, and collagen receptor agonists. Deletion of COMMD7 significantly impaired multiple responses to platelet agonists. Together, our data recommend CRIMSON for rapid evaluation of platelet gene phenotype associations.
Collapse
Affiliation(s)
- Emilie Montenont
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Seema Bhatlekar
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Shancy Jacob
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Yasuhiro Kosaka
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Bhanu K Manne
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Olivia Lee
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | | | - Emilia Tugolukova
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Neal D Tolley
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Matthew T Rondina
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
- Department of Internal Medicine
- George E. Wahlen Department of Veterans Affairs Medical Center
- Department of Internal Medicine and Geriatric Research and Education Clinical Center, and
- Department of Pathology, The University of Utah, Salt Lake City, UT
| | - Paul F Bray
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
- Department of Internal Medicine
| | - Jesse W Rowley
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
- Department of Internal Medicine
| |
Collapse
|
18
|
Di Buduo CA, Aguilar A, Soprano PM, Bocconi A, Miguel CP, Mantica G, Balduini A. Latest culture techniques: cracking the secrets of bone marrow to mass-produce erythrocytes and platelets ex vivo. Haematologica 2021; 106:947-957. [PMID: 33472355 PMCID: PMC8017859 DOI: 10.3324/haematol.2020.262485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Since the dawn of medicine, scientists have carefully observed, modeled and interpreted the human body to improve healthcare. At the beginning there were drawings and paintings, now there is three-dimensional modeling. Moving from two-dimensional cultures and towards complex and relevant biomaterials, tissue-engineering approaches have been developed in order to create three-dimensional functional mimics of native organs. The bone marrow represents a challenging organ to reproduce because of its structure and composition that confer it unique biochemical and mechanical features to control hematopoiesis. Reproducing the human bone marrow niche is instrumental to answer the growing demand for human erythrocytes and platelets for fundamental studies and clinical applications in transfusion medicine. In this review, we discuss the latest culture techniques and technological approaches to obtain functional platelets and erythrocytes ex vivo. This is a rapidly evolving field that will define the future of targeted therapies for thrombocytopenia and anemia, but also a long-term promise for new approaches to the understanding and cure of hematologic diseases.
Collapse
Affiliation(s)
| | - Alicia Aguilar
- Department of Molecular Medicine, University of Pavia, Pavia
| | - Paolo M Soprano
- Department of Molecular Medicine, University of Pavia, Pavia
| | - Alberto Bocconi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; Department of Chemistry, Materials and Chemical Engineering G. Natta, Politecnico di Milano, Milano
| | | | | | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; Department of Biomedical Engineering, Tufts University, Medford, MA
| |
Collapse
|
19
|
Campbell RA, Boilard E, Rondina MT. Is there a role for the ACE2 receptor in SARS-CoV-2 interactions with platelets? J Thromb Haemost 2021; 19:46-50. [PMID: 33119197 PMCID: PMC7899240 DOI: 10.1111/jth.15156] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/12/2020] [Accepted: 10/26/2020] [Indexed: 01/11/2023]
Abstract
There is an urgent need to understand the underlying mechanisms contributing to thrombotic and inflammatory complications during COVID-19. Data from independent groups have identified that platelets are hyperreactive during COVID-19. Platelet hyperreactivity is accompanied by changes in platelet gene expression, and enhanced interactions between platelets and leukocytes. In some patients, SARS-CoV-2 mRNA has been detected in platelets. Together, this suggests that SARS-CoV-2 may interact with platelets. However, controversy remains on which receptors mediate SARS-CoV-2 platelet interactions. Most, but not all, transcriptomic and proteomic analyses fail to observe the putative SARS-CoV-2 receptor, angiotensin converting enzyme-2, or the cellular serine protease necessary for viral entry, TMPRSS2, on platelets and megakaryocytes. Interestingly, platelets express other known SARS-CoV-2 receptors, which induce similar patterns of activation to those observed when platelets are incubated with SARS-CoV-2. This article explores these findings and discusses ongoing areas of controversy and uncertainty with regard to SARS-CoV-2 platelet interactions.
Collapse
Affiliation(s)
- Robert A Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, UT, USA
- Departments of Internal Medicine & Pathology, University of Utah, Salt Lake City, UT, USA
| | - Eric Boilard
- Centre de Recherche du Centre Hospitalier,, Universitaire de Québec- Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'immunologie, Université Laval, Québec, QC, Canada
| | - Matthew T Rondina
- University of Utah Molecular Medicine Program, Salt Lake City, UT, USA
- Departments of Internal Medicine & Pathology, University of Utah, Salt Lake City, UT, USA
- George E. Wahlen VAMC Department of Internal Medicine and GRECC, Salt Lake City, UT, USA
| |
Collapse
|
20
|
Di Buduo CA, Soprano PM, Miguel CP, Perotti C, Del Fante C, Balduini A. A Gold Standard Protocol for Human Megakaryocyte Culture Based on the Analysis of 1,500 Umbilical Cord Blood Samples. Thromb Haemost 2020; 121:538-542. [PMID: 33160288 DOI: 10.1055/s-0040-1719028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Christian A Di Buduo
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy
| | - Paolo M Soprano
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy
| | - Carolina P Miguel
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy
| | - Cesare Perotti
- Immunohematology and Transfusion Service and Cell Therapy Unit, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy
| | - Claudia Del Fante
- Immunohematology and Transfusion Service and Cell Therapy Unit, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy.,Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States
| |
Collapse
|
21
|
Bhatlekar S, Manne BK, Basak I, Edelstein LC, Tugolukova E, Stoller ML, Cody MJ, Morley SC, Nagalla S, Weyrich AS, Rowley JW, O'Connell RM, Rondina MT, Campbell RA, Bray PF. miR-125a-5p regulates megakaryocyte proplatelet formation via the actin-bundling protein L-plastin. Blood 2020; 136:1760-1772. [PMID: 32844999 PMCID: PMC7544541 DOI: 10.1182/blood.2020005230] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/24/2020] [Indexed: 12/17/2022] Open
Abstract
There is heritability to interindividual variation in platelet count, and better understanding of the regulating genetic factors may provide insights for thrombopoiesis. MicroRNAs (miRs) regulate gene expression in health and disease, and megakaryocytes (MKs) deficient in miRs have lower platelet counts, but information about the role of miRs in normal human MK and platelet production is limited. Using genome-wide miR profiling, we observed strong correlations among human bone marrow MKs, platelets, and differentiating cord blood-derived MK cultures, and identified MK miR-125a-5p as associated with human platelet number but not leukocyte or hemoglobin levels. Overexpression and knockdown studies showed that miR-125a-5p positively regulated human MK proplatelet (PP) formation in vitro. Inhibition of miR-125a-5p in vivo lowered murine platelet counts. Analyses of MK and platelet transcriptomes identified LCP1 as a miR-125a-5p target. LCP1 encodes the actin-bundling protein, L-plastin, not previously studied in MKs. We show that miR-125a-5p directly targets and reduces expression of MK L-plastin. Overexpression and knockdown studies show that L-plastin promotes MK progenitor migration, but negatively correlates with human platelet count and inhibits MK PP formation (PPF). This work provides the first evidence for the actin-bundling protein, L-plastin, as a regulator of human MK PPF via inhibition of the late-stage MK invagination system, podosome and PPF, and PP branching. We also provide resources of primary and differentiating MK transcriptomes and miRs associated with platelet counts. miR-125a-5p and L-plastin may be relevant targets for increasing in vitro platelet manufacturing and for managing quantitative platelet disorders.
Collapse
Affiliation(s)
- Seema Bhatlekar
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
| | - Bhanu K Manne
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
| | - Indranil Basak
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
| | - Leonard C Edelstein
- Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA
| | - Emilia Tugolukova
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
| | | | - Mark J Cody
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
| | - Sharon C Morley
- Division of Infectious Diseases, Department of Pediatrics and
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Srikanth Nagalla
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Andrew S Weyrich
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
- Division of Pulmonary, Department of Internal Medicine
| | - Jesse W Rowley
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
- Division of Pulmonary, Department of Internal Medicine
| | - Ryan M O'Connell
- Division of Microbiology and Immunology, Department of Pathology, and
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Matthew T Rondina
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
- Geriatric Research, Education and Clinical Center, George E. Wahlen VAMC GRECC, Salt Lake City, UT; and
- Division of General Internal Medicine and
| | - Robert A Campbell
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
- Division of General Internal Medicine and
| | - Paul F Bray
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT
| |
Collapse
|
22
|
BCL-w: apoptotic and non-apoptotic role in health and disease. Cell Death Dis 2020; 11:260. [PMID: 32317622 PMCID: PMC7174325 DOI: 10.1038/s41419-020-2417-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 12/17/2022]
Abstract
The BCL-2 family of proteins integrates signals that trigger either cell survival or apoptosis. The balance between pro-survival and pro-apoptotic proteins is important for tissue development and homeostasis, while impaired apoptosis contributes to several pathologies and can be a barrier against effective treatment. BCL-w is an anti-apoptotic protein that shares a sequence similarity with BCL-XL, and exhibits a high conformational flexibility. BCL-w level is controlled by a number of signaling pathways, and the repertoire of transcriptional regulators largely depends on the cellular and developmental context. As only a few disease-relevant genetic alterations of BCL2L2 have been identified, increased levels of BCL-w might be a consequence of abnormal activation of signaling cascades involved in the regulation of BCL-w expression. In addition, BCL-w transcript is a target of a plethora of miRNAs. Besides its originally recognized pro-survival function during spermatogenesis, BCL-w has been envisaged in different types of normal and diseased cells as an anti-apoptotic protein. BCL-w contributes to survival of senescent and drug-resistant cells. Its non-apoptotic role in the promotion of cell migration and invasion has also been elucidated. Growing evidence indicates that a high BCL-w level can be therapeutically relevant in neurodegenerative disorders, neuron dysfunctions and after small intestinal resection, whereas BCL-w inhibition can be beneficial for cancer patients. Although several drugs and natural compounds can bi-directionally affect BCL-w level, agents that selectively target BCL-w are not yet available. This review discusses current knowledge on the role of BCL-w in health, non-cancerous diseases and cancer.
Collapse
|
23
|
Melchinger H, Jain K, Tyagi T, Hwa J. Role of Platelet Mitochondria: Life in a Nucleus-Free Zone. Front Cardiovasc Med 2019; 6:153. [PMID: 31737646 PMCID: PMC6828734 DOI: 10.3389/fcvm.2019.00153] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/08/2019] [Indexed: 12/19/2022] Open
Abstract
Platelets are abundant, small, anucleate circulating cells, serving many emerging pathophysiological roles beyond hemostasis; including active critical roles in thrombosis, injury response, and immunoregulation. In the absence of genomic DNA transcriptional regulation (no nucleus), platelets require strategic prepackaging of all the needed RNA and organelles from megakaryocytes, to sense stress (e.g., hyperglycemia), to protect themselves from stress (e.g., mitophagy), and to communicate a stress response to other cells (e.g., granule and microparticle release). Distinct from avian thrombocytes that have a nucleus, the absence of a nucleus allows the mammalian platelet to maintain its small size, permits morphological flexibility, and may improve speed and efficiency of protein expression in response to stress. In the absence of a nucleus, platelet lifespan of 7–10 days, is largely determined by the mitochondria. The packaging of 5–8 mitochondria is critical in aerobic respiration and yielding metabolic substrates needed for function and survival. Mitochondria damage or dysfunction, as observed with several disease processes, results in greatly attenuated platelet survival and increased risk for thrombovascular events. Here we provide insights into the emerging roles of platelets despite the lack of a nucleus, and the key role played by mitochondria in platelet function and survival both in health and disease.
Collapse
Affiliation(s)
- Hannah Melchinger
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - Kanika Jain
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - Tarun Tyagi
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - John Hwa
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
24
|
Basak I, Bhatlekar S, Manne B, Stoller M, Hugo S, Kong X, Ma L, Rondina MT, Weyrich AS, Edelstein LC, Bray PF. miR-15a-5p regulates expression of multiple proteins in the megakaryocyte GPVI signaling pathway. J Thromb Haemost 2019; 17:511-524. [PMID: 30632265 PMCID: PMC6397079 DOI: 10.1111/jth.14382] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Indexed: 12/22/2022]
Abstract
Essentials The action of microRNAs (miRs) in human megakaryocyte signaling is largely unknown. Cord blood-derived human megakaryocytes (MKs) were used to test the function of candidate miRs. miR-15a-5p negatively regulated MK GPVI-mediated αIIbβ3 activation and α-granule release. miR-15a-5p acts as a potential "master-miR" regulating genes in the MK GPVI signaling pathway. SUMMARY: Background Megakaryocytes (MKs) invest their progeny platelets with proteins and RNAs. MicroRNAs (miRs), which inhibit mRNA translation into protein, are abundantly expressed in MKs and platelets. Although platelet miRs have been associated with platelet reactivity and disease, there is a paucity of information on the function of miRs in human MKs. Objective To identify MK miRs that regulate the GPVI signaling pathway in the MK-platelet lineage. Methods Candidate miRs associated with GPVI-mediated platelet aggregation were tested for functionality in cultured MKs derived from cord blood. Results An unbiased, transcriptome-wide screen in 154 healthy donors identified platelet miR-15a-5p as significantly negatively associated with CRP-induced platelet aggregation. Platelet agonist dose-response curves demonstrated activation of αIIbβ3 in suspensions of cord blood-derived cultured MKs. Overexpression and knockdown of miR-15a-5p in these MKs reduced and enhanced, respectively, CRP-induced αIIbβ3 activation but did not alter thrombin or ADP stimulation. FYN, SRGN, FCER1G, MYLK. and PRKCQ, genes involved in GPVI signaling, were identified as miR-15a-5p targets and were inhibited or de-repressed in MKs with miR-15a-5p overexpression or inhibition, respectively. Lentiviral overexpression of miR-15a-5p also inhibited GPVI-FcRγ-mediated phosphorylation of Syk and PLCγ2, GPVI downstream signaling molecules, but effects of miR-15a-5p on αIIbβ3 activation did not extend to other ITAM-signaling receptors (FcγRIIa and CLEC-2). Conclusion Cord blood-derived MKs are a useful human system for studying the functional effects of candidate platelet genes. miR-15a-5p is a potential "master-miR" for specifically regulating GPVI-mediated MK-platelet signaling. Targeting miR-15a-5p may have therapeutic potential in hemostasis and thrombosis.
Collapse
Affiliation(s)
- I. Basak
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
| | - S. Bhatlekar
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
| | - B.K. Manne
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
| | - M. Stoller
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
| | - S. Hugo
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
| | - X. Kong
- The Cardeza Foundation for Hematologic Research and the Department of Medicine, Thomas Jefferson University, Jefferson Medical College, Philadelphia, PA 19107
| | - L. Ma
- The Cardeza Foundation for Hematologic Research and the Department of Medicine, Thomas Jefferson University, Jefferson Medical College, Philadelphia, PA 19107
| | - M. T. Rondina
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
| | - A. S. Weyrich
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
| | - L. C. Edelstein
- The Cardeza Foundation for Hematologic Research and the Department of Medicine, Thomas Jefferson University, Jefferson Medical College, Philadelphia, PA 19107
| | - P. F. Bray
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
- Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|