1
|
Zhang J, Xiang H, Jiang L, Wang M, Yang G. Construction of a novel platelet‑related gene risk model to predict the prognosis and drug response in virus‑related hepatocellular carcinoma. Oncol Lett 2024; 28:592. [PMID: 39417040 PMCID: PMC11481168 DOI: 10.3892/ol.2024.14725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
Platelet activity in the tumor microenvironment (TME) is crucial for the development of tumors. However, the roles and clinical potential of platelet activity in the TME for virus-related hepatocellular carcinoma (HCC) remain unclear. The present study aimed to identify a novel signature based on platelet activity for prognostic prediction and treatment decisions in virus-related HCC. First, a novel platelet signature score (PSS) for each patient with virus-related HCC from The Cancer Genome Atlas was calculated using gene set variation analysis, and the patients were divided into two subgroups (high and low PSS). It was demonstrated that the patients with a high PSS had a worse prognosis, higher platelet activity, stronger inflammation and immunosuppression in TME than patients with a low PSS. Furthermore, 137 differentially expressed genes (DEGs; fold change >2; P<0.05) were identified using 'DESeq2' and 'edgeR' software. Subsequently, 3 genes (cyclin-J-Like protein, nuclear receptor subfamily 0 group B member 1 and tripartite motif containing 54) were identified from DEGs using univariate Cox and least absolute shrinkage and selection operator (LASSO) analyses. Risk score (RS) was calculated based on gene expression and coefficients from LASSO. Patients were divided into high and low RS groups according to the median value, and the 3-gene model was used to predict prognoses and drug responses. Notably, it was demonstrated that patients with a low RS may be better candidates for immune therapy due to lower levels of tumor immune dysfunction and exclusion scores. Moreover, patients with a high RS may be better candidates for nonimmune therapy due to lower half-maximal inhibitory concentration values of drugs (such as AKT inhibitors and gemcitabine). Finally, it was demonstrated that patients with a high PSS and RS had a higher platelet activity, inflammation status, tumor hallmarks and the worst prognosis than patients with a low PSS and RS. This helped to better find patients with these characteristics and suitable treatments using this method. Collectively, the findings of the present study indicate that PSS combined with RS has great potential to evaluate the prognosis of patients with virus -related HCC and assist in deciding treatment strategies.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Honglin Xiang
- Department of Orthopaedics, Laboratory of Biological Tissue Engineering and Digital Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Ling Jiang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Mei Wang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Guodong Yang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
2
|
Dominguez JH, Xie D, Kelly KJ. Renal, but not platelet or skin, extracellular vesicles decrease oxidative stress, enhance nascent peptide synthesis, and protect from ischemic renal injury. Am J Physiol Renal Physiol 2023; 325:F164-F176. [PMID: 37318988 PMCID: PMC10393335 DOI: 10.1152/ajprenal.00321.2022] [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: 01/03/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/17/2023] Open
Abstract
Acute kidney injury (AKI) is deadly and expensive, and specific, effective therapy remains a large unmet need. We have demonstrated the beneficial effects of transplanted adult tubular cells and extracellular vesicles (EVs; exosomes) derived from those renal cells on experimental ischemic AKI, even when administered after renal failure is established. To further examine the mechanisms of benefit with renal EVs, we tested the hypothesis that EVs from other epithelia or platelets (a rich source of EVs) might be protective, using a well-characterized ischemia-reperfusion model. When given after renal failure was present, renal EVs, but not those from skin or platelets, markedly improved renal function and histology. The differential effects allowed us to examine the mechanisms of benefit with renal EVs. We found significant decreases in oxidative stress postischemia in the renal EV-treated group with preservation of renal superoxide dismutase and catalase as well as increases in anti-inflammatory interleukin-10. In addition, we propose a novel mechanism of benefit: renal EVs enhanced nascent peptide synthesis following hypoxia in cells and in postischemic kidneys. Although EVs have been used therapeutically, these results serve as "proof of principle" to examine the mechanisms of injury and protection.NEW & NOTEWORTHY Acute kidney injury is common and deadly, yet the only approved treatment is dialysis. Thus, a better understanding of injury mechanisms and potential therapies is needed. We found that organ-specific, but not extrarenal, extracellular vesicles improved renal function and structure postischemia when given after renal failure occurred. Oxidative stress was decreased and anti-inflammatory interleukin-10 increased with renal, but not skin or platelet, exosomes. We also propose enhanced nascent peptide synthesis as a novel protective mechanism.
Collapse
Affiliation(s)
- Jesus H. Dominguez
- Division of Nephrology and Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, United States
| | - Danhui Xie
- Division of Nephrology and Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - K. J. Kelly
- Division of Nephrology and Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, United States
| |
Collapse
|
3
|
Xi C, Pang J, Zhi W, Chang CSS, Siddaramappa U, Shi H, Horuzsko A, Pace BS, Zhu X. Nrf2 sensitizes ferroptosis through l-2-hydroxyglutarate-mediated chromatin modifications in sickle cell disease. Blood 2023; 142:382-396. [PMID: 37267508 PMCID: PMC10485372 DOI: 10.1182/blood.2022018159] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 06/04/2023] Open
Abstract
Sickle cell disease (SCD) is a chronic hemolytic and systemic hypoxia condition with constant oxidative stress and significant metabolic alterations. However, little is known about the correlation between metabolic alterations and the pathophysiological symptoms. Here, we report that Nrf2, a master regulator of cellular antioxidant responses, regulates the production of the metabolite l-2-hydroxyglutarate (L2HG) to mediate epigenetic histone hypermethylation for gene expression involved in metabolic, oxidative, and ferroptotic stress responses in SCD. Mechanistically, Nrf2 was found to regulate the expression of L2HG dehydrogenase (L2hgdh) to mediate L2HG production under hypoxia. Gene expression profile analysis indicated that reactive oxygen species (ROS) and ferroptosis responses were the most significantly affected signaling pathways after Nrf2 ablation in SCD. Nrf2 silencing and L2HG supplementation sensitize human sickle erythroid cells to ROS and ferroptosis stress. The absence of Nrf2 and accumulation of L2HG significantly affect histone methylation for chromatin structure modification and reduce the assembly of transcription complexes on downstream target genes to regulate ROS and ferroptosis responses. Furthermore, pharmacological activation of Nrf2 was found to have protective effects against ROS and ferroptosis stress in SCD mice. Our data suggest a novel mechanism by which Nrf2 regulates L2HG levels to mediate SCD severity through ROS and ferroptosis stress responses, suggesting that targeting Nrf2 is a viable therapeutic strategy for ameliorating SCD symptoms.
Collapse
Affiliation(s)
- Caixia Xi
- Georgia Cancer Center, Augusta University, Augusta, GA
| | - Junfeng Pang
- Georgia Cancer Center, Augusta University, Augusta, GA
| | - Wenbo Zhi
- Center for Biotechnology and Genomic Medicine, Consolidated Proteomics and Mass Spectrometry Core Lab, Augusta University, Augusta, GA
| | - Chang-Sheng S. Chang
- Bioinformatics Shared Resource and Integrated Genomics, Georgia Cancer Center, Augusta University, Augusta, GA
| | - Umapathy Siddaramappa
- Division of Hematology/Oncology, Department of Pediatrics, Augusta University, Augusta, GA
| | - Huidong Shi
- Georgia Cancer Center, Augusta University, Augusta, GA
| | | | - Betty S. Pace
- Division of Hematology/Oncology, Department of Pediatrics, Augusta University, Augusta, GA
| | - Xingguo Zhu
- Georgia Cancer Center, Augusta University, Augusta, GA
- Division of Hematology/Oncology, Department of Pediatrics, Augusta University, Augusta, GA
| |
Collapse
|
4
|
Momma TY, Ottaviani JI. There is no direct competition between arginase and nitric oxide synthase for the common substrate l-arginine. Nitric Oxide 2022; 129:16-24. [PMID: 36126859 DOI: 10.1016/j.niox.2022.09.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/08/2022] [Accepted: 09/10/2022] [Indexed: 11/19/2022]
Abstract
AIMS Extrahepatic arginases are postulated to be involved in cardiovascular-related pathologies by competing with nitric oxide synthase (NOS) for the common substrate l-arginine, subsequently decreasing nitric oxide production. However, previous models used to study arginase and NOS competition did not account for steady state level of l-arginine pool, which is dependent on conditions of l-arginine supply and utilization pathways. This work aimed at revisiting the concept of NOS and arginase competition while considering different conditions of l-arginine supply and l-arginine utilization pathways. METHODS AND RESULTS Mouse macrophage-like RAW cells and human vascular endothelial cells co-expressing NOS and arginase were used to reevaluate the concept of substrate competition between arginase and NOS under conditions of l-arginine supply that mimicked either a continuous (similar to in vivo conditions) or a limited supply (similar to previous in vitro models). Enzyme kinetics simulation models were used to gain mechanistic insight and to evaluate the tenability of a substrate competition between the two enzymes. In addition to arginase and NOS, other l-arginine pathways such as transporters and utilization towards protein synthesis were considered to understand the intricacies of l-arginine metabolism. Our results indicate that when there is a continuous supply of l-arginine, as is the case for most cells in vivo, arginase does not affect NOS activity by a substrate competition. Furthermore, we demonstrate that l-arginine pathways such as transporters and protein synthesis are more likely to affect NOS activity than arginase. CONCLUSIONS Arginase does not outcompete NOS for the common substrate l-arginine. Findings from this study should be considered to better understand the role of arginase in certain pathologies and for the interpretation of in vivo studies with arginase inhibitors.
Collapse
Affiliation(s)
- Tony Y Momma
- College of Agricultural and Environmental Sciences, University of California, Davis, CA, 95616, USA.
| | - Javier I Ottaviani
- College of Agricultural and Environmental Sciences, University of California, Davis, CA, 95616, USA; Mars Inc., McLean, VA, 22101, USA
| |
Collapse
|
5
|
Sagar RC, Ajjan RA, Naseem KM. Non-Traditional Pathways for Platelet Pathophysiology in Diabetes: Implications for Future Therapeutic Targets. Int J Mol Sci 2022; 23:ijms23094973. [PMID: 35563363 PMCID: PMC9104718 DOI: 10.3390/ijms23094973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 11/23/2022] Open
Abstract
Cardiovascular complications remain the leading cause of morbidity and mortality in individuals with diabetes, driven by interlinked metabolic, inflammatory, and thrombotic changes. Hyperglycaemia, insulin resistance/deficiency, dyslipidaemia, and associated oxidative stress have been linked to abnormal platelet function leading to hyperactivity, and thus increasing vascular thrombotic risk. However, emerging evidence suggests platelets also contribute to low-grade inflammation and additionally possess the ability to interact with circulating immune cells, further driving vascular thrombo-inflammatory pathways. This narrative review highlights the role of platelets in inflammatory and immune processes beyond typical thrombotic effects and the impact these mechanisms have on cardiovascular disease in diabetes. We discuss pathways for platelet-induced inflammation and how platelet reprogramming in diabetes contributes to the high cardiovascular risk that characterises this population. Fully understanding the mechanistic pathways for platelet-induced vascular pathology will allow for the development of more effective management strategies that deal with the causes rather than the consequences of platelet function abnormalities in diabetes.
Collapse
|
6
|
Platelets Purification Is a Crucial Step for Transcriptomic Analysis. Int J Mol Sci 2022; 23:ijms23063100. [PMID: 35328521 PMCID: PMC8953733 DOI: 10.3390/ijms23063100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 01/24/2023] Open
Abstract
Platelets are small anucleate cells derived from the fragmentation of megakaryocytes and are involved in different biological processes especially hemostasis, thrombosis, and immune response. Despite their lack of nucleus, platelets contain a reservoir of megakaryocyte-derived RNAs and all the machinery useful for mRNA translation. Interestingly, platelet transcriptome was analyzed in health and diseases and led to the identification of disease-specific molecular signatures. Platelet contamination by leukocytes and erythrocytes during platelet purification is a major problem in transcriptomic analysis and the presence of few contaminants in platelet preparation could strongly alter transcriptome results. Since contaminant impacts on platelet transcriptome remains theoretical, we aimed to determine whether low leukocyte and erythrocyte contamination could cause great or only minor changes in platelet transcriptome. Using microarray technique, we compared the transcriptome of platelets from the same donor, purified by common centrifugation method or using magnetic microbeads to eliminate contaminating cells. We found that platelet transcriptome was greatly altered by contaminants, as the relative amount of 8274 transcripts was different between compared samples. We observed an increase of transcripts related to leukocytes and erythrocytes in platelet purified without microbeads, while platelet specific transcripts were falsely reduced. In conclusion, serious precautions should be taken during platelet purification process for transcriptomic analysis, in order to avoid platelets contamination and result alteration.
Collapse
|
7
|
Lecot P, Ardin M, Dussurgey S, Alcazer V, Moudombi L, Pereira Abrantes M, Hubert M, Swalduz A, Hernandez‐Vargas H, Viari A, Caux C, Michallet M. Gene signature of circulating platelet‐bound neutrophils is associated with poor prognosis in cancer patients. Int J Cancer 2022; 151:138-152. [PMID: 35253899 PMCID: PMC9311065 DOI: 10.1002/ijc.33991] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 02/08/2022] [Accepted: 02/17/2022] [Indexed: 11/15/2022]
Abstract
Beyond their critical role in hemostasis, platelets physically interact with neutrophils to form neutrophil‐platelet aggregates (NPAs), enhancing neutrophil effector functions during inflammation. NPAs may also promote disease worsening in various inflammatory diseases. However, characterization of NPAs in cancer remains totally unexplored. Using ImageStreamX (ISX) imaging flow cytometer, we were not only allowed able to detect CD15+ CD14− CD36+ ITGA2B+ NPAs in both healthy donors' (HDs) and cancer patients' bloods, but we also showed that NPAs result from the binding of platelets preferentially to low‐density neutrophils (LDNs) as opposed to normal‐density neutrophils (NDNs). By reanalyzing two independent public scRNAseq data of whole blood leukocytes from cancer patients and HDs, we could identify a subset of neutrophils with high platelet gene expression that may correspond to NPAs. Moreover, we showed that cancer patients' derived NPAs possessed a distinct molecular signature compared to the other neutrophil subsets, independently of platelet genes. Gene ontology (GO) term enrichment analysis of this NPAs‐associated neutrophil transcriptomic signature revealed a significant enrichment of neutrophil degranulation, chemotaxis and trans‐endothelial migration GO terms. Lastly, using The Cancer Genome Atlas (TCGA), we could show by multivariate Cox analysis that the NPAs‐associated neutrophil transcriptomic signature was associated with a worse patient prognosis in several cancer types. These results suggest that neutrophils from NPAs are systemically primed by platelets empowering them with cancer progression capacities once at tumor site. NPAs may therefore hold clinical utility as novel noninvasive blood prognostic biomarker in cancer patients with solid tumors.
Collapse
Affiliation(s)
- Pacôme Lecot
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Maude Ardin
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Sébastien Dussurgey
- Université de Lyon, SFR Biosciences, ENS de Lyon, Inserm US8, CNRS UMS3444, UCBL ‐ 50 Avenue Tony Garnier Lyon France
| | - Vincent Alcazer
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Lyvia Moudombi
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Manuela Pereira Abrantes
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Margaux Hubert
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Aurélie Swalduz
- Department of Lung and Thoracic Medical Oncology Centre Léon Bérard Lyon France
| | - Hector Hernandez‐Vargas
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Alain Viari
- Synergie Lyon Cancer, Plateforme de Bio‐informatique ‘Gilles Thomas’ Lyon France
| | - Christophe Caux
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Marie‐Cécile Michallet
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| |
Collapse
|
8
|
Gee BE, Pearson A, Buchanan-Perry I, Simon RP, Archer DR, Meller R. Whole Blood Transcriptome Analysis in Children with Sickle Cell Anemia. Front Genet 2022; 12:737741. [PMID: 35095995 PMCID: PMC8793691 DOI: 10.3389/fgene.2021.737741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/20/2021] [Indexed: 12/27/2022] Open
Abstract
Whole transcriptome RNA-sequencing was performed to quantify RNA expression changes in whole blood samples collected from steady state sickle cell anemia (SCA) and control subjects. Pediatric SCA and control subjects were recruited from Atlanta (GA)-based hospital(s) systems and consented for RNA sequencing. RNA sequencing was performed on an Ion Torrent S5 sequencer, using the Ion Total RNA-seq v2 protocol. Data were aligned to the hg19 reference genome and analyzed in the Partek Genomics studio package (v7.0). 223 genes were differentially expressed between SCA and controls (± 1.5 fold change FDR p < 0.001) and 441 genes show differential transcript expression (± 1.5 fold FDR p < 0.001). Differentially expressed RNA are enriched for hemoglobin associated genes and ubiquitin-proteasome pathway genes. Further analysis shows higher gamma globin gene expression in SCA (33-fold HBG1 and 49-fold HBG2, both FDR p < 0.05), which did not correlate with hemoglobin F protein levels. eQTL analysis identified SNPs in novel non-coding RNA RYR2 gene as having a potential regulatory role in HBG1 and HBG2 expression levels. Gene expression correlation identified JHDM1D-AS1(KDM7A-DT), a non-coding RNA associated with angiogenesis, enhanced GATA1 and decreased JAK-STAT signaling to correlate with HBG1 and HBG2 mRNA levels. These data suggest novel regulatory mechanisms for fetal hemoglobin regulation, which may offer innovative therapeutic approaches for SCA.
Collapse
Affiliation(s)
- Beatrice E. Gee
- Department of Pediatrics, Morehouse School of Medicine, Atlanta, GA, United States
- Morehouse School of Medicine, Cardiovascular Research Institute, Atlanta, GA, United States
- Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Andrea Pearson
- Translational Stroke Program, Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, United States
| | - Iris Buchanan-Perry
- Department of Pediatrics, Morehouse School of Medicine, Atlanta, GA, United States
- Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Roger P. Simon
- Translational Stroke Program, Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, United States
- Grady Memorial Hospital, Atlanta, GA, United States
- Department of Neurology, Morehouse School of Medicine, Atlanta, GA, United States
| | - David R. Archer
- Aflac Cancer and Blood Disorders Center of Emory University and Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Robert Meller
- Translational Stroke Program, Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
9
|
Sloan AR, Lee-Poturalski C, Hoffman HC, Harris PL, Elder TE, Richardson B, Kerstetter-Fogle A, Cioffi G, Schroer J, Desai A, Cameron M, Barnholtz-Sloan J, Rich J, Jankowsky E, Sen Gupta A, Sloan AE. Glioma stem cells activate platelets by plasma-independent thrombin production to promote glioblastoma tumorigenesis. Neurooncol Adv 2022; 4:vdac172. [PMID: 36452274 PMCID: PMC9700385 DOI: 10.1093/noajnl/vdac172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background The interaction between platelets and cancer cells has been underexplored in solid tumor models that do not metastasize, for example, glioblastoma (GBM) where metastasis is rare. Histologically, it is known that glioma stem cells (GSCs) are found in perivascular and pseudsopalisading regions of GBM, which are also areas of platelet localization. High platelet counts have been associated with poor clinical outcomes in many cancers. While platelets are known to promote the progression of other tumors, mechanisms by which platelets influence GBM oncogenesis are unknown. Here, we aimed to understand how the bidirectional interaction between platelets and GSCs drives GBM oncogenesis. Methods Male and female NSG mice were transplanted with GSC lines and treated with antiplatelet and anti-thrombin inhibitors. Immunofluorescence, qPCR, and Western blots were used to determine expression of coagulation mechanism in GBM tissue and subsequent GSC lines. Results We show that GSCs activate platelets by endogenous production of all the factors of the intrinsic and extrinsic coagulation cascades in a plasma-independent manner. Therefore, GSCs produce thrombin resulting in platelet activation. We further demonstrate that the endogenous coagulation cascades of these cancer stem cells are tumorigenic: they activate platelets to promote stemness and proliferation in vitro and pharmacological inhibition delays tumor growth in vivo. Conclusions Our findings uncover a specific preferential relationship between platelets and GSCs that drive GBM malignancies and identify a therapeutically targetable novel interaction.
Collapse
Affiliation(s)
- Anthony R Sloan
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Neurological Surgery, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Christine Lee-Poturalski
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
- Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Harry C Hoffman
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, Ohio, USA
| | - Peggy L Harris
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Neurological Surgery, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Theresa E Elder
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Neurological Surgery, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Brian Richardson
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Population and Quantitative Health Science, Case Western Reserve University, Cleveland, Ohio, USA
| | - Amber Kerstetter-Fogle
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Gino Cioffi
- Division of Cancer Epidemiology and Genetics, Trans-Divisional Research Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Julia Schroer
- Geisinger Commonwealth School of Medicine, Scranton, Pennsylvania, USA
| | - Ansh Desai
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, Ohio, USA
| | - Mark Cameron
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Population and Quantitative Health Science, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jill Barnholtz-Sloan
- Division of Cancer Epidemiology and Genetics, Trans-Divisional Research Program, National Cancer Institute, Bethesda, Maryland, USA
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Bethesda, Maryland, USA
| | - Jeremy Rich
- Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Eckhard Jankowsky
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
- Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Anirban Sen Gupta
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering, Cleveland, Ohio, USA
| | - Andrew E Sloan
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Neurological Surgery, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Neurosciences, Piedmont Health, Atlanta Georgia, USA
| |
Collapse
|
10
|
Shen Z, Du W, Perkins C, Fechter L, Natu V, Maecker H, Rowley J, Gotlib J, Zehnder J, Krishnan A. Platelet transcriptome identifies progressive markers and potential therapeutic targets in chronic myeloproliferative neoplasms. Cell Rep Med 2021; 2:100425. [PMID: 34755136 PMCID: PMC8561315 DOI: 10.1016/j.xcrm.2021.100425] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/08/2021] [Accepted: 09/23/2021] [Indexed: 12/11/2022]
Abstract
Predicting disease progression remains a particularly challenging endeavor in chronic degenerative disorders and cancer, thus limiting early detection, risk stratification, and preventive interventions. Here, profiling the three chronic subtypes of myeloproliferative neoplasms (MPNs), we identify the blood platelet transcriptome as a proxy strategy for highly sensitive progression biomarkers that also enables prediction of advanced disease via machine-learning algorithms. The MPN platelet transcriptome reveals an incremental molecular reprogramming that is independent of patient driver mutation status or therapy. Subtype-specific markers offer mechanistic and therapeutic insights, and highlight impaired proteostasis and a persistent integrated stress response. Using a LASSO model with validation in two independent cohorts, we identify the advanced subtype MF at high accuracy and offer a robust progression signature toward clinical translation. Our platelet transcriptome snapshot of chronic MPNs demonstrates a proof-of-principle for disease risk stratification and progression beyond genetic data alone, with potential utility in other progressive disorders.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blood Platelets/metabolism
- Blood Platelets/pathology
- Cellular Reprogramming
- Child
- Child, Preschool
- Cohort Studies
- Diagnosis, Differential
- Disease Progression
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Middle Aged
- Polycythemia Vera/diagnosis
- Polycythemia Vera/genetics
- Polycythemia Vera/metabolism
- Polycythemia Vera/pathology
- Primary Myelofibrosis/diagnosis
- Primary Myelofibrosis/genetics
- Primary Myelofibrosis/metabolism
- Primary Myelofibrosis/pathology
- Proteostasis/genetics
- Risk Assessment
- Thrombocythemia, Essential/diagnosis
- Thrombocythemia, Essential/genetics
- Thrombocythemia, Essential/metabolism
- Thrombocythemia, Essential/pathology
- Transcriptome
Collapse
Affiliation(s)
- Zhu Shen
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Wenfei Du
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Cecelia Perkins
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Lenn Fechter
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Vanita Natu
- Stanford Functional Genomics Facility, Stanford University School of Medicine, Stanford, CA, USA
| | - Holden Maecker
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jesse Rowley
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Jason Gotlib
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - James Zehnder
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Anandi Krishnan
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| |
Collapse
|
11
|
Rolfes V, Ribeiro LS, Hawwari I, Böttcher L, Rosero N, Maasewerd S, Santos MLS, Próchnicki T, Silva CMDS, Wanderley CWDS, Rothe M, Schmidt SV, Stunden HJ, Bertheloot D, Rivas MN, Fontes CJ, Carvalho LH, Cunha FQ, Latz E, Arditi M, Franklin BS. Platelets Fuel the Inflammasome Activation of Innate Immune Cells. Cell Rep 2021; 31:107615. [PMID: 32402278 PMCID: PMC7225754 DOI: 10.1016/j.celrep.2020.107615] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 03/12/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
The inflammasomes control the bioactivity of pro-inflammatory cytokines of the interleukin (IL)-1 family. The inflammasome assembled by NLRP3 has been predominantly studied in homogeneous cell populations in vitro, neglecting the influence of cellular interactions that occur in vivo. Here, we show that platelets boost the inflammasome capacity of human macrophages and neutrophils and are critical for IL-1 production by monocytes. Platelets license NLRP3 transcription, thereby enhancing ASC oligomerization, caspase-1 activity, and IL-1β secretion. Platelets influence IL-1β production in vivo, and blood platelet counts correlate with plasmatic IL-1β levels in malaria. Furthermore, we reveal an enriched platelet gene signature among the highest-expressed transcripts in IL-1β-driven autoinflammatory diseases. The platelet effect is independent of cell-to-cell contact, platelet-derived lipid mediators, purines, nucleic acids, and a host of platelet cytokines, and it involves the triggering of calcium-sensing receptors on macrophages. Hence, platelets provide an additional layer of regulation of inflammasomes and IL-1-driven inflammation. Platelets license NLRP3 for inflammasome activattion in innate immune cells Platelets are required for optimal monocyte inflammasome activation Platelets shape IL-1β in vivo, and platelet counts correlate with IL-1β in plasma A constitutive, heat-sensitive soluble platelet-factor boost IL-1β in macrophages
Collapse
Affiliation(s)
- Verena Rolfes
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Lucas Secchim Ribeiro
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany.
| | - Ibrahim Hawwari
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Lisa Böttcher
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Nathalia Rosero
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Salie Maasewerd
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Marina Lima Silva Santos
- Laboratório de Malária, Instituto René Rachou, Fundação Oswaldo Cruz, 30190-002 Belo Horizonte, MG, Brazil
| | - Tomasz Próchnicki
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Camila Meirelles de Souza Silva
- Center for Research in Inflammatory Diseases, School of Medicine of Ribeirão Preto, University of Sao Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Carlos Wagner de Souza Wanderley
- Center for Research in Inflammatory Diseases, School of Medicine of Ribeirão Preto, University of Sao Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Maximilian Rothe
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Susanne V Schmidt
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - H James Stunden
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Damien Bertheloot
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Magali Noval Rivas
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Cor Jesus Fontes
- Departamento de Clínica Médica, Universidade Federal de Mato Grosso, 78060-900 Cuiabá, MT, Brazil
| | - Luzia Helena Carvalho
- Laboratório de Malária, Instituto René Rachou, Fundação Oswaldo Cruz, 30190-002 Belo Horizonte, MG, Brazil
| | - Fernando Queiroz Cunha
- Center for Research in Inflammatory Diseases, School of Medicine of Ribeirão Preto, University of Sao Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Eicke Latz
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01655, USA; German Center for Neurodegenerative Diseases, 53127 Bonn, NRW, Germany
| | - Moshe Arditi
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA.
| | | |
Collapse
|
12
|
Abstract
Platelets have long been known to play important roles beyond hemostasis and thrombosis. Now recognized as a bona fide mediator of malignant disease, platelets influence various aspects of cancer progression, most notably tumor cell metastasis. Interestingly, platelets isolated from cancer patients often display distinct RNA and protein profiles, with no clear alterations in hemostatic activity. This phenotypically distinct population, termed tumor-educated platelets, now receive significant attention for their potential use as a readily available liquid biopsy for early cancer detection. Although the mechanisms underpinning platelet education are still being defined, direct uptake and storage of tumor-derived factors, signal-dependent changes in platelet RNA processing, and differential platelet production by tumor-educated megakaryocytes are the most prominent scenarios. This article aims to cover the various modalities of platelet education by tumors, in addition to assessing their diagnostic potential.
Collapse
|
13
|
Multifaceted Functions of Platelets in Cancer: From Tumorigenesis to Liquid Biopsy Tool and Drug Delivery System. Int J Mol Sci 2020; 21:ijms21249585. [PMID: 33339204 PMCID: PMC7765591 DOI: 10.3390/ijms21249585] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
Platelets contribute to several types of cancer through plenty of mechanisms. Upon activation, platelets release many molecules, including growth and angiogenic factors, lipids, and extracellular vesicles, and activate numerous cell types, including vascular and immune cells, fibroblasts, and cancer cells. Hence, platelets are a crucial component of cell-cell communication. In particular, their interaction with cancer cells can enhance their malignancy and facilitate the invasion and colonization of distant organs. These findings suggest the use of antiplatelet agents to restrain cancer development and progression. Another peculiarity of platelets is their capability to uptake proteins and transcripts from the circulation. Thus, cancer-patient platelets show specific proteomic and transcriptomic expression patterns, a phenomenon called tumor-educated platelets (TEP). The transcriptomic/proteomic profile of platelets can provide information for the early detection of cancer and disease monitoring. Platelet ability to interact with tumor cells and transfer their molecular cargo has been exploited to design platelet-mediated drug delivery systems to enhance the efficacy and reduce toxicity often associated with traditional chemotherapy. Platelets are extraordinary cells with many functions whose exploitation will improve cancer diagnosis and treatment.
Collapse
|
14
|
Gutmann C, Joshi A, Mayr M. Platelet "-omics" in health and cardiovascular disease. Atherosclerosis 2020; 307:87-96. [PMID: 32646580 DOI: 10.1016/j.atherosclerosis.2020.05.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/28/2020] [Accepted: 05/27/2020] [Indexed: 12/21/2022]
Abstract
The importance of platelets for cardiovascular disease was established as early as the 19th century. Their therapeutic inhibition stands alongside the biggest achievements in medicine. Still, certain aspects of platelet pathophysiology remain unclear. This includes platelet resistance to antiplatelet therapy and the contribution of platelets to vascular remodelling and extends beyond cardiovascular disease to haematological disorders and cancer. To address these gaps in our knowledge, a better understanding of the underlying molecular processes is needed. This will be enabled by technologies that capture dysregulated molecular processes and can integrate them into a broader network of biological systems. The advent of -omics technologies, such as mass spectrometry proteomics, metabolomics and lipidomics; highly multiplexed affinity-based proteomics; microarray- or RNA-sequencing-(RNA-seq)-based transcriptomics, and most recently ribosome footprint-based translatomics, has enabled a more holistic understanding of platelet biology. Most of these methods have already been applied to platelets, and this review will summarise this information and discuss future developments in this area of research.
Collapse
Affiliation(s)
- Clemens Gutmann
- King's British Heart Foundation Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, United Kingdom
| | - Abhishek Joshi
- King's British Heart Foundation Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, United Kingdom
| | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, United Kingdom.
| |
Collapse
|
15
|
Momma TY, Ottaviani JI. Arginase inhibitor, N ω-hydroxy-L-norarginine, spontaneously releases biologically active NO-like molecule: Limitations for research applications. Free Radic Biol Med 2020; 152:74-82. [PMID: 32131024 DOI: 10.1016/j.freeradbiomed.2020.02.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/25/2019] [Accepted: 02/28/2020] [Indexed: 12/21/2022]
Abstract
There has been a renewed interest in the enzyme arginase for its role in various physiological and pathological processes that go beyond the urea cycle. One such role ascribed to arginase has been that of regulating nitric oxide (NO) production by a substrate (l-arginine) competition between arginase and nitric oxide synthase (NOS). Several arginase inhibitors have been developed to investigate the biological roles of arginase, of which Nω-hydroxy-l-norarginine (nor-NOHA) is commercially available and is used widely from cell culture models to clinical investigations in humans. Despite the prevalence of nor-NOHA to investigate the substrate competition between arginase and NOS, little is known regarding interferences that nor-NOHA could have on common methods to assess NO production. Therefore, we investigated if nor-NOHA has unintended consequences on common NO assessment methods. We show that nor-NOHA spontaneously releases biologically active NO-like molecule in cell culture media by reacting with riboflavin. This NO-like molecule is indistinguishable from an NO donor (NOR-3) using common methods to assess NO. Besides riboflavin, nor-NOHA spontaneously reacts with H2O2 to diminish H2O2 content and produce NO-like molecule in the process. Our investigation provides detailed evidence on unintended artefacts related to nor-NOHA that can limit its use in cell culture, as well as some ex vivo and in vivo models. Future studies on arginase should take into consideration the limitations presented here when using nor-NOHA as a research tool, not only in investigations related to arginase and NOS competition, but also for investigating other biological roles of arginase.
Collapse
Affiliation(s)
- Tony Y Momma
- Department of Nutrition, University of California, Davis, CA, 95616, USA.
| | - Javier I Ottaviani
- Department of Nutrition, University of California, Davis, CA, 95616, USA; Mars, Inc., McLean, VA, 22101, USA
| |
Collapse
|
16
|
Smiljanovic B, Grützkau A, Sörensen T, Grün JR, Vogl T, Bonin M, Schendel P, Stuhlmüller B, Claussnitzer A, Hermann S, Ohrndorf S, Aupperle K, Backhaus M, Radbruch A, Burmester GR, Häupl T. Synovial tissue transcriptomes of long-standing rheumatoid arthritis are dominated by activated macrophages that reflect microbial stimulation. Sci Rep 2020; 10:7907. [PMID: 32404914 PMCID: PMC7220941 DOI: 10.1038/s41598-020-64431-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/15/2020] [Indexed: 12/30/2022] Open
Abstract
Advances in microbiome research suggest involvement in chronic inflammatory diseases such as rheumatoid arthritis (RA). Searching for initial trigger(s) in RA, we compared transcriptome profiles of highly inflamed RA synovial tissue (RA-ST) and osteoarthritis (OA)-ST with 182 selected reference transcriptomes of defined cell types and their activation by exogenous (microbial) and endogenous inflammatory stimuli. Screening for dominant changes in RA-ST demonstrated activation of monocytes/macrophages with gene-patterns induced by bacterial and fungal triggers. Gene-patterns of activated B- or T-cells in RA-ST reflected a response to activated monocytes/macrophages rather than inducing their activation. In contrast, OA-ST was dominated by gene-patterns of non-activated macrophages and fibroblasts. The difference between RA and OA was more prominent in transcripts of secreted proteins and was confirmed by protein quantification in synovial fluid (SF) and serum. In total, 24 proteins of activated cells were confirmed in RA-SF compared to OA-SF and some like CXCL13, CCL18, S100A8/A9, sCD14, LBP reflected this increase even in RA serum. Consequently, pathogen-like response patterns in RA suggest that direct microbial influences exist. This challenges the current concept of autoimmunity and immunosuppressive treatment and advocates new diagnostic and therapeutic strategies that consider microbial persistence as important trigger(s) in the etiopathogenesis of RA.
Collapse
Affiliation(s)
- Biljana Smiljanovic
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin, Berlin, Germany
| | - Andreas Grützkau
- Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Till Sörensen
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin, Berlin, Germany
| | - Joachim R Grün
- Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Thomas Vogl
- Institute of Immunology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Marc Bonin
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin, Berlin, Germany
| | - Pascal Schendel
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin, Berlin, Germany
| | - Bruno Stuhlmüller
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin, Berlin, Germany
| | - Anne Claussnitzer
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin, Berlin, Germany
| | - Sandra Hermann
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin, Berlin, Germany
| | - Sarah Ohrndorf
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin, Berlin, Germany
| | - Karlfried Aupperle
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin, Berlin, Germany
| | - Marina Backhaus
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin, Berlin, Germany
| | - Andreas Radbruch
- Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Gerd R Burmester
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin, Berlin, Germany
| | - Thomas Häupl
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
17
|
Davizon-Castillo P, Rowley JW, Rondina MT. Megakaryocyte and Platelet Transcriptomics for Discoveries in Human Health and Disease. Arterioscler Thromb Vasc Biol 2020; 40:1432-1440. [PMID: 32295424 PMCID: PMC7253186 DOI: 10.1161/atvbaha.119.313280] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Anucleate platelets, long viewed as merely cell fragments with a limited repertoire of rapid-acting hemostatic functions, are now recognized to have a complex and dynamic transcriptome mirroring that of many nucleated cells. The field of megakaryocyte and platelet transcriptomics has been rapidly growing, particularly with the advent of newer technologies such as next-generation RNA-sequencing. Studies interrogating the megakaryocyte and platelet transcriptome have led to a number of key insights into human health and disease. In this brief focused review, we will discuss some of the recent discoveries made through transcriptome analysis of megakaryocytes and platelets. We will also highlight the utility of integrating ribosome footprint analysis to augment discoveries. Both bulk and single-cell sequencing approaches will be reviewed, along with comparative studies between human and murine platelets under basal healthy settings and during acute systemic inflammatory diseases.
Collapse
Affiliation(s)
- Pavel Davizon-Castillo
- From the Section of Pediatric Hematology, Oncology, and Bone Marrow Transplant, University of Colorado, Aurora (P.D.-C)
| | - Jesse W Rowley
- University of Utah Molecular Medicine Program, University of Utah, Salt Lake City (J.W.R., M.T.R.).,Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City (J.W.R., M.T.R.)
| | - Matthew T Rondina
- From the Section of Pediatric Hematology, Oncology, and Bone Marrow Transplant, University of Colorado, Aurora (P.D.-C).,University of Utah Molecular Medicine Program, University of Utah, Salt Lake City (J.W.R., M.T.R.).,Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City (J.W.R., M.T.R.).,Department of Pathology, University of Utah, Salt Lake City (M.T.R.).,George E. Wahlen VAMC, Salt Lake City, UT (M.T.R.)
| |
Collapse
|
18
|
Zhang J, Ding Y, Jiang D, Xie J, Liu Y, Ma J, Mu Y, Zhang X, Yu C, Zhang Y, Yi X, Zhou Z, Fang L, Shen S, Yang Y, Cheng K, Zhuang R, Zhang Y. Deficiency of platelet adhesion molecule CD226 causes megakaryocyte development and platelet hyperactivity. FASEB J 2020; 34:6871-6887. [PMID: 32248623 DOI: 10.1096/fj.201902142r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/27/2019] [Accepted: 03/16/2020] [Indexed: 12/17/2022]
Abstract
This study used constitutive CD226 gene knockout (KO) mice as a model to investigate the functions and mechanisms of CD226 in megakaryocyte (MK) maturation and platelet activation. Although CD226 deficiency did not cause MK polyploidization or platelet granule abnormalities, increased MK counts were detected in the femora bone marrow (BM) and spleen of CD226 KO mice. Particularly, CD226 KO mice have a more extensive membrane system in MKs and platelets than wild-type (WT) mice. We also demonstrated that CD226 KO mice displayed increased platelet counts, shortened bleeding time, and enhanced platelet aggregation. CD226 KO platelets had an increased mature platelet ratio compared to the control platelets. In addition, the observed reduction in bleeding time may be due to decreased nitric oxide (NO) production in the platelets. Platelet-specific CD226-deficient mice showed similar increased MK counts, shortened bleeding time, enhanced platelet aggregation, and decreased NO production in platelets. Furthermore, we performed middle cerebral artery occlusion-reperfusion surgery on WT and CD226 KO mice to explore the potential effect of CD226 on acute ischemia-reperfusion injury; the results revealed that CD226 deficiency led to significantly increased infarct area. Thus, CD226 is a promising candidate for the treatment of thrombotic disorders.
Collapse
Affiliation(s)
- Jinxue Zhang
- Orthopedic Department of Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yong Ding
- Orthopedic Department of Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Dongxu Jiang
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Jiangang Xie
- Department of Emergency, Fourth Military Medical University, Xi'an, China
| | - Yongming Liu
- Orthopedic Department of Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jingchang Ma
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Yang Mu
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Xuexin Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Chaoping Yu
- Department of Emergency, Fourth Military Medical University, Xi'an, China
| | - Yun Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Xin Yi
- Orthopedic Department of Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Ziqing Zhou
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Liang Fang
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Shen Shen
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Yixin Yang
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Kun Cheng
- Transplant Immunology Laboratory, Fourth Military Medical University, Xi'an, China
| | - Ran Zhuang
- Department of Immunology, Fourth Military Medical University, Xi'an, China.,Transplant Immunology Laboratory, Fourth Military Medical University, Xi'an, China
| | - Yuan Zhang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
19
|
Sighting acute myocardial infarction through platelet gene expression. Sci Rep 2019; 9:19574. [PMID: 31863085 PMCID: PMC6925116 DOI: 10.1038/s41598-019-56047-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 12/06/2019] [Indexed: 11/20/2022] Open
Abstract
Acute myocardial infarction is primarily due to coronary atherosclerotic plaque rupture and subsequent thrombus formation. Platelets play a key role in the genesis and progression of both atherosclerosis and thrombosis. Since platelets are anuclear cells that inherit their mRNA from megakaryocyte precursors and maintain it unchanged during their life span, gene expression profiling at the time of an acute myocardial infarction provides information concerning the platelet gene expression preceding the coronary event. In ST-segment elevation myocardial infarction (STEMI), a gene-by-gene analysis of the platelet gene expression identified five differentially expressed genes: FKBP5, S100P, SAMSN1, CLEC4E and S100A12. The logistic regression model used to combine the gene expression in a STEMI vs healthy donors score showed an AUC of 0.95. The same five differentially expressed genes were externally validated using platelet gene expression data from patients with coronary atherosclerosis but without thrombosis. Platelet gene expression profile highlights five genes able to identify STEMI patients and to discriminate them in the background of atherosclerosis. Consequently, early signals of an imminent acute myocardial infarction are likely to be found by platelet gene expression profiling before the infarction occurs.
Collapse
|
20
|
Carpentier S, Romagné F, Vivier E. A comprehensive approach to gene expression profiling in immune cells. Methods Enzymol 2019; 636:1-47. [PMID: 32178815 DOI: 10.1016/bs.mie.2019.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
With the advent of whole-transcriptome studies and the growing need for public repositories, it has become essential to combine multiple heterogeneous datasets for immune cells. In this chapter, we describe the implementation of a compendium of 10,833 genes for 975 samples, corresponding to 52 resting immune cell types. We begin by describing the datasets, and their selection, in particular. We then explain the methodology implemented to create a qualified compendium: the processing of each array (quality control, normalization and bias correction), integration (merging rules, global normalization and batch removal) and validation. Finally some examples of use will be detailed. The utility and limitations of the compendium are also discussed, as an introduction to the next version.
Collapse
Affiliation(s)
| | | | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France; Aix-Marseille Univ, APHM, CNRS, INSERM, CIML, Hôpital de la Timone, Marseille-Immunopole, Marseille, France.
| |
Collapse
|
21
|
Couldwell G, Machlus KR. Modulation of megakaryopoiesis and platelet production during inflammation. Thromb Res 2019; 179:114-120. [PMID: 31128560 DOI: 10.1016/j.thromres.2019.05.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/19/2019] [Accepted: 05/13/2019] [Indexed: 12/24/2022]
Abstract
Megakaryocytes (MKs) are widely known as the progenitor cells of platelets. These large, polyploid cells are a derivative of the hematopoietic stem cell (HSC), and reside in the bone marrow, lining blood vessel walls where they release their platelet progeny into circulation. Although little is known about how MKs differ under various environmental stressors, both chronic and acute inflammation alter the differentiation and molecular content of MKs. Furthermore, evidence suggests that the release of inflammatory cytokines may induce MK rupture and rapid release of platelets as a mechanism to quickly replenish diminished platelet counts in response to inflammation. Similarities between MKs and their close relatives, white blood cells, have introduced the notion that MKs may play a role in combating infection by engulfing and presenting antigens, and passing this information to circulating platelets. In addition, MKs exposed to varying bone marrow environments produce different platelets which enter circulation primed to respond to and combat inflammation, infection, or injury. This review focuses on how inflammation alters MK production, maturation, and platelet production. In addition, it introduces the idea that inflammation reprograms MKs to create different, more pathogenic platelets and leads them to take on different roles as responders to deleterious conditions. In the future, studies determining how platelets are altered in disease states may lead to novel MK- and platelet-based therapeutic targets to mitigate inflammation-related morbidity and mortality.
Collapse
Affiliation(s)
| | - Kellie R Machlus
- Division of Hematology, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
22
|
Wang W, Wang L, Gulko PS, Zhu J. Computational deconvolution of synovial tissue cellular composition: presence of adipocytes in synovial tissue decreased during arthritis pathogenesis and progression. Physiol Genomics 2019; 51:241-253. [PMID: 31100034 PMCID: PMC6620645 DOI: 10.1152/physiolgenomics.00009.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/18/2019] [Accepted: 05/13/2019] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) and rheumatoid arthritis (RA) are the most common forms of arthritis. The synovial tissue is the major site of inflammation of OA and RA and consists of diverse cells. Synovial tissue cell composition changes during arthritis pathogenesis and progression have not been systematically characterized and may provide critical insights into disease processes. In this study we aimed at systematically examining cellular changes in synovial tissue. Publicly available synovial tissue transcriptomic data sets were used. We computationally estimated cell compositions in synovial tissue based on transcriptomic data and compared cell compositions in different diseases or at different disease stages. Synovial fibroblasts, macrophages, adipocytes, and immune cells were the major cell types in all synovial tissue. Both OA and RA patients had a significantly lower adipocyte fraction compared with healthy controls. The decrease trend was also observed during OA and RA progression. The fraction of monocytes was also increased in both OA and RA arthritis patients, consistent with the observations that inflammation involved in both OA and RA. But the monocyte fraction in RAs was much higher than the ones in healthy controls and OAs. The M2 macrophage fraction was reduced in RA compared with OA, the reduction trend continued during RA progression from the early- to the late-stage. There were consistent cell composition differences between different types or stages of arthritis. Both in RA and OA, the new discovery of changes in the adipocyte and M2 macrophage fractions has potential leading to novel therapeutic development.
Collapse
Affiliation(s)
- Wenhui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai , New York, New York
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Li Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai , New York, New York
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai , New York, New York
- Sema4, a Mount Sinai venture, Stamford, Connecticut
| | - Percio S Gulko
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai , New York
| | - Jun Zhu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai , New York, New York
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai , New York, New York
- Sema4, a Mount Sinai venture, Stamford, Connecticut
| |
Collapse
|
23
|
Toledo SLDO, Guedes JVM, Alpoim PN, Rios DRA, Pinheiro MDB. Sickle cell disease: Hemostatic and inflammatory changes, and their interrelation. Clin Chim Acta 2019; 493:129-137. [PMID: 30825426 DOI: 10.1016/j.cca.2019.02.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/26/2019] [Accepted: 02/26/2019] [Indexed: 12/23/2022]
Abstract
Sickle cell disease, the most common genetic blood disorder in the world, has high clinical variability, negatively impacts quality of life and contributes to early mortality. Sickled erythrocytes cause blood flow obstruction, hemolysis, and several hemostatic changes that promote coagulation. These events, in turn, induce chronic inflammation, characterized by elevated plasma levels of pro-inflammatory markers, which aggravates the already unfavorable state of the circulatory system. Empirical evidence indicates that the hemostatic and inflammatory systems continuously interact with each other and thereby further propagate the hypercoagulability and inflammatory conditions. In this review article, we discuss the pathophysiological aspects of sickle cell disease and the hemostatic and inflammatory changes that underlie its pathogenesis.
Collapse
Affiliation(s)
- Sílvia L de O Toledo
- Federal University of São João del-Rei (UFSJ), Dona Lindu Center-West Campus, Sebastião Gonçalves Coelho Street, 400, Chanadour, 35501-296 Divinópolis, MG, Brazil
| | - João V M Guedes
- Federal University of São João del-Rei (UFSJ), Dona Lindu Center-West Campus, Sebastião Gonçalves Coelho Street, 400, Chanadour, 35501-296 Divinópolis, MG, Brazil
| | - Patrícia N Alpoim
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais (MG), Brazil
| | - Danyelle R A Rios
- Federal University of São João del-Rei (UFSJ), Dona Lindu Center-West Campus, Sebastião Gonçalves Coelho Street, 400, Chanadour, 35501-296 Divinópolis, MG, Brazil
| | - Melina de B Pinheiro
- Federal University of São João del-Rei (UFSJ), Dona Lindu Center-West Campus, Sebastião Gonçalves Coelho Street, 400, Chanadour, 35501-296 Divinópolis, MG, Brazil.
| |
Collapse
|
24
|
Shang C, Wuren T, Ga Q, Bai Z, Guo L, Eustes AS, McComas KN, Rondina MT, Ge R. The human platelet transcriptome and proteome is altered and pro-thrombotic functional responses are increased during prolonged hypoxia exposure at high altitude. Platelets 2019; 31:33-42. [PMID: 30721642 DOI: 10.1080/09537104.2019.1572876] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Exposure to hypoxia, through ascension to high altitudes (HAs), air travel, or human disease, is associated with an increased incidence of thrombosis in some settings. Mechanisms underpinning this increased thrombosis risk remain incompletely understood, and the effects of more sustained hypoxia on the human platelet molecular signature and associated functional responses have never been examined. We examined the effects of prolonged (≥2 months continuously) hypobaric hypoxia on platelets isolated from subjects residing at HA (3,700 meters) and, for comparison, matched subjects residing under normoxia conditions at sea level (50 meters). Using complementary transcriptomic, proteomic, and functional methods, we identified that the human platelet transcriptome is markedly altered under prolonged exposure to hypobaric hypoxia at HA. Among the significantly, differentially expressed genes (mRNA and protein), were those having canonical roles in platelet activation and thrombosis, including membrane glycoproteins (e.g. GP4, GP6, GP9), integrin subunits (e.g. ITGA2B), and alpha-granule chemokines (e.g. SELP, PF4V1). Platelets from subjects residing at HA were hyperactive, as demonstrated by increased engagement and adhesion to fibrinogen, fewer alpha granules by transmission electron microscopy, increased circulating PF4 and ADP, and significantly enhanced clot retraction. In conclusion, we identify that prolonged hypobaric hypoxia exposure due to HA alters the platelet transcriptome and proteome, triggering increased functional activation responses that may contribute to thrombosis. Our findings may also have relevance across a range of human diseases where chronic hypoxia, platelet activation, and thrombosis are increased.
Collapse
Affiliation(s)
- Chunxiang Shang
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai, China.,Oncology Department, The Fifth Hospital of Qinghai Provinces, Xining, Qinghai, China
| | - Tana Wuren
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai, China.,Departments of Internal Medicine and Pathology, University of Utah, Salt Lake City, UT, USA
| | - Qing Ga
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai, China
| | - Zhenzhong Bai
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai, China
| | - Li Guo
- The University of Utah Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
| | - Alicia S Eustes
- Departments of Internal Medicine and Pathology, University of Utah, Salt Lake City, UT, USA.,The University of Utah Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
| | - Kyra N McComas
- Departments of Internal Medicine and Pathology, University of Utah, Salt Lake City, UT, USA
| | - Matthew T Rondina
- Departments of Internal Medicine and Pathology, University of Utah, Salt Lake City, UT, USA.,The University of Utah Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA.,Hematological Department, George E. Wahlen VAMC GRECC, Salt Lake City, UT, USA
| | - Rili Ge
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai, China
| |
Collapse
|
25
|
Liu FF, Tu TT, Zhang HF, Hu F, Huang L, Deng LF, Guo M, Wei Q, Li K. Coexpression network analysis of platelet genes in sickle cell disease. Platelets 2019; 30:1022-1029. [PMID: 30663491 DOI: 10.1080/09537104.2018.1562170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 12/04/2018] [Accepted: 12/18/2018] [Indexed: 10/27/2022]
Abstract
Platelets play important roles in vascular health. Activation of platelet may contribute to coagulation and inflammation. Evidence suggests circulating platelets are chronically activated in sickle cell disease (SCD) patients with steady state and further activated in vaso-occlusive crisis. However, the molecular basis of sickle platelet dysfunction remains obscure. Here, we used weighted gene coexpression network analysis combined with differentially expressed genes (DEGs) analysis to further investigate this basis. We found 57 DEGs were closely related to platelet dysfunction in SCD. Enrichment analysis showed that these 57 genes were mostly related to protein synthesis, adenosine triphosphate (ATP) synthase activity and inflammation, suggesting a hyperactivation status of platelets in SCD. We identified six hub genes from the 57 DEGs according to their Gene Significance value ranking, including CRYM, CCT6P1, SUCNR1, PRKAB2, GSTM3 and FCGR2C. Altogether, our results offered some new insight into platelet activation and identified novel potential targets for antiplatelet therapy in SCD.
Collapse
Affiliation(s)
- Fang-Fang Liu
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , P. R. China
| | - Tong-Tao Tu
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , P. R. China
| | - Hong-Feng Zhang
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , P. R. China
| | - Fan Hu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , P. R. China
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology , Wuhan , P. R.China
| | - Liang Huang
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , P. R. China
| | - Lin-Feng Deng
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , P. R. China
| | - Mao Guo
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , P. R. China
| | - Qing Wei
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , P. R. China
| | - Ke Li
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , P. R. China
| |
Collapse
|
26
|
|
27
|
Ben Hamda C, Sangeda R, Mwita L, Meintjes A, Nkya S, Panji S, Mulder N, Guizani-Tabbane L, Benkahla A, Makani J, Ghedira K. A common molecular signature of patients with sickle cell disease revealed by microarray meta-analysis and a genome-wide association study. PLoS One 2018; 13:e0199461. [PMID: 29979707 PMCID: PMC6034806 DOI: 10.1371/journal.pone.0199461] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 06/07/2018] [Indexed: 12/16/2022] Open
Abstract
A chronic inflammatory state to a large extent explains sickle cell disease (SCD) pathophysiology. Nonetheless, the principal dysregulated factors affecting this major pathway and their mechanisms of action still have to be fully identified and elucidated. Integrating gene expression and genome-wide association study (GWAS) data analysis represents a novel approach to refining the identification of key mediators and functions in complex diseases. Here, we performed gene expression meta-analysis of five independent publicly available microarray datasets related to homozygous SS patients with SCD to identify a consensus SCD transcriptomic profile. The meta-analysis conducted using the MetaDE R package based on combining p values (maxP approach) identified 335 differentially expressed genes (DEGs; 224 upregulated and 111 downregulated). Functional gene set enrichment revealed the importance of several metabolic pathways, of innate immune responses, erythrocyte development, and hemostasis pathways. Advanced analyses of GWAS data generated within the framework of this study by means of the atSNP R package and SIFT tool identified 60 regulatory single-nucleotide polymorphisms (rSNPs) occurring in the promoter of 20 DEGs and a deleterious SNP, affecting CAMKK2 protein function. This novel database of candidate genes, transcription factors, and rSNPs associated with SCD provides new markers that may help to identify new therapeutic targets.
Collapse
Affiliation(s)
- Cherif Ben Hamda
- Laboratory of Bioinformatics, Biomathematics and Biostatistics, Institute Pasteur of Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
- Faculty of Science of Bizerte, Jarzouna, University of Carthage, Tunisia
- * E-mail: (KG); (CBH)
| | - Raphael Sangeda
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Liberata Mwita
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | | | - Siana Nkya
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Sumir Panji
- University of Cape Town, Cape Town, South Africa
| | | | - Lamia Guizani-Tabbane
- University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institute Pasteur of Tunis, Tunis, Tunisia
| | - Alia Benkahla
- Laboratory of Bioinformatics, Biomathematics and Biostatistics, Institute Pasteur of Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
| | - Julie Makani
- Faculty of Science of Bizerte, Jarzouna, University of Carthage, Tunisia
| | - Kais Ghedira
- Laboratory of Bioinformatics, Biomathematics and Biostatistics, Institute Pasteur of Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
- * E-mail: (KG); (CBH)
| | | |
Collapse
|
28
|
Fleischer LM, Somaiya RD, Miller GM. Review and Meta-Analyses of TAAR1 Expression in the Immune System and Cancers. Front Pharmacol 2018; 9:683. [PMID: 29997511 PMCID: PMC6029583 DOI: 10.3389/fphar.2018.00683] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 06/06/2018] [Indexed: 12/29/2022] Open
Abstract
Since its discovery in 2001, the major focus of TAAR1 research has been on its role in monoaminergic regulation, drug-induced reward and psychiatric conditions. More recently, TAAR1 expression and functionality in immune system regulation and immune cell activation has become a topic of emerging interest. Here, we review the immunologically-relevant TAAR1 literature and incorporate open-source expression and cancer survival data meta-analyses. We provide strong evidence for TAAR1 expression in the immune system and cancers revealed through NCBI GEO datamining and discuss its regulation in a spectrum of immune cell types as well as in numerous cancers. We discuss connections and logical directions for further study of TAAR1 in immunological function, and its potential role as a mediator or modulator of immune dysregulation, immunological effects of psychostimulant drugs of abuse, and cancer progression.
Collapse
Affiliation(s)
- Lisa M Fleischer
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Rachana D Somaiya
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Gregory M Miller
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States.,Department of Chemical Engineering, Northeastern University, Boston, MA, United States.,Center for Drug Discovery, Northeastern University, Boston, MA, United States
| |
Collapse
|
29
|
Xia L, Zeng Z, Tang WH. The Role of Platelet Microparticle Associated microRNAs in Cellular Crosstalk. Front Cardiovasc Med 2018; 5:29. [PMID: 29670887 PMCID: PMC5893844 DOI: 10.3389/fcvm.2018.00029] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/15/2018] [Indexed: 01/11/2023] Open
Abstract
Platelet is an anucleate cell containing abundant messenger RNAs and microRNAs (miRNAs), and their functional roles in hemostasis and inflammation remain elusive. Accumulating evidence has suggested that platelets can actively transfer RNAs to hepatocytes, vascular cells, macrophages, and tumor cells. The incorporated mRNAs are translated into proteins, and miRNAs were found to regulate the gene expression, resulting in the functional change of the recipient cells. This novel intercellular communication opens up a new avenue for the pathophysiological role of platelet in platelet-associated vascular diseases. Therefore, understanding the underlying mechanism and identification of the platelet miRNAs involved in this biological process would provide novel diagnostic and therapeutic targets for cardiovascular diseases.
Collapse
Affiliation(s)
- Luoxing Xia
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Zhi Zeng
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Wai Ho Tang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
30
|
Abstract
Platelets are equipped with RNA processing machineries, such as pre-mRNA splicing, pre-miRNA processing, and mRNA translation. Since platelets are devoid of a nucleus, most RNA transcripts in platelets are derived from megakaryocytes during thrombocytogenesis. However, platelets can also ingest RNA molecules during circulation and/or interaction with other cell types. Since platelets were first described by Bizzozero in 1881, their well-established role in hemostasis and thrombosis has been intensively studied. However, in the past decades, the list of biological processes in which platelets play an important role keeps expanding. In this review, we discuss how platelet RNA biomarker signatures can be altered in the presence of cancer.
Collapse
Affiliation(s)
- Nik Sol
- Department of Neurology, VU University Medical Center, Amsterdam, The Netherlands. .,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.
| | - Thomas Wurdinger
- Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.,Department of Neurosurgery, VU University Medical Center, Amsterdam, The Netherlands.,Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
31
|
Cox SE, Ellins EA, Marealle AI, Newton CR, Soka D, Sasi P, Luca Di Tanna G, Johnson W, Makani J, Prentice AM, Halcox JP, Kirkham FJ. Ready-to-use food supplement, with or without arginine and citrulline, with daily chloroquine in Tanzanian children with sickle-cell disease: a double-blind, random order crossover trial. Lancet Haematol 2018; 5:e147-e160. [PMID: 29548623 PMCID: PMC5871644 DOI: 10.1016/s2352-3026(18)30020-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 01/15/2018] [Accepted: 01/15/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Sickle-cell disease increases the risk of malnutrition. Low arginine and nitric oxide bioavailability are implicated in morbidity related to sickle-cell disease. Simple interventions are required, especially in low-income settings. We aimed to test the hypotheses that: (1) supplementary arginine, citrulline, and daily chloroquine increase bioavailable arginine and flow-mediated dilatation (FMD; maximal diameter change; FMDmax%), a measure of nitric oxide-dependent endothelial function; and (2) protein energy supplementation in the form of ready-to-use supplementary food (RUSF) improves the height-for-age and body-mass index-for-age Z-scores in children with sickle-cell disease. METHODS We performed a double-blind, random order crossover trial with two 4-month intervention periods (each followed by 4-month washout periods) in Muhimbili National Hospital in Dar-es-Salaam, Tanzania. We enrolled 119 children from the Muhimbili Sickle Cohort who were aged 8-12 years, naive to hydroxyurea, and had documented HbSS phenotype. Two formulations of RUSF (providing 500 kcal/day) were tested: basic (RUSF-b), with which children also received weekly chloroquine (150 mg or 225 mg chloroquine base, dependent on bodyweight); and vascular (RUSF-v), which was fortified with arginine and citrulline (designed to achieve mean intakes of 0·2 g/kg per day of arginine and 0·1 g/kg per day of citrulline), and with which children received daily chloroquine (maximum 3 mg chloroquine base/kg per day). Children were randomly allocated to receive either RUSF-b first or RUSF-v first and, after a washout period, were then given the other treatment. The primary outcomes in comparing the two RUSF formulations were mean plasma arginine, arginine to ornithine ratio, and arginine to asymmetric dimethylarginine ratio, and mean FMDmax%. The primary outcomes of the combined effect of both RUSF interventions were mean height-for-age Z-score and body-mass index-for-age Z-score. Analyses were done on the eligible intention-to-treat population. This trial is registered with ClinicalTrials.gov, number NCT01718054; and with ISRCTN74331412. FINDINGS Between Aug 9, 2012, and Feb 26, 2014, 145 children were randomised (71 children to RUSF-v first and 74 children to RUSF-b first) and 119 children were treated, of whom 114 children yielded complete data for all reported endpoints. The ratio of arginine to ornithine (mean of individual differences -8·67%, 95% CI -19·55 to 2·20; p=0·12) and the mean FMDmax% (1·00, -0·47 to 2·47; p=0·18) did not significantly differ between the RUSF-b and RUSF-v treatments. However, the arginine to asymmetric dimethylarginine ratio was significantly increased by RUSF-v compared with RUSF-b (56·26%, 31·13 to 81·38; p<0·0001). In planned analyses that used mixed effects models to estimate the effect of each intervention compared with the participants at baseline or during washout periods, the arginine to asymmetric dimethylarginine ratio increased following both RUSF-v treatment (86%; p<0·0001) and RUSF-b treatment (40%; p<0·0001). However, FMDmax% was higher after treatment with RUSF-v (0·92; p<0·0001) but not RUSF-b (0·39; p=0·22). Following either intervention (RUSF-b and RUSF-v, pooled) body-mass index-for-age Z-score (0·091; p=0·001) and height-for-age Z-score (0·013; p=0·081) increased compared with baseline and washout timepoints. In 83 participants in the treated population, there were 71 adverse events during the intervention, of which 21 (30%) were serious, and 81 adverse events during the washout periods, of which 26 (32%) were serious (p=0·31), including one patient who died in the second washout period. INTERPRETATION RUSF providing 500 kcal/day results in small weight gains in children with sickle-cell disease. However, even without arginine and citrulline fortification, RUSF seems to ameliorate arginine dysregulation and might improve endothelial function. Long-term studies are required to assess whether these physiological effects translate to improved clinical outcomes and better growth and development in patients with sickle-cell disease. FUNDING Wellcome Trust.
Collapse
Affiliation(s)
- Sharon E Cox
- Department of Population Health, London School of Hygiene & Tropical Medicine, London, UK; School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan; Muhimbili Wellcome Programme, School of Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania.
| | - Elizabeth A Ellins
- Institute of Life Sciences, Swansea University Medical School, Swansea University, Swansea, UK
| | - Alphonce I Marealle
- Muhimbili Wellcome Programme, School of Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania; Department of Clinical Pharmacy and Pharmacology, School of Pharmacy, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Charles R Newton
- Muhimbili Wellcome Programme, School of Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania; Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - Deogratias Soka
- Muhimbili Wellcome Programme, School of Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania; Department of Haematology, School of Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Philip Sasi
- Department of Clinical Pharmacology, School of Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Gian Luca Di Tanna
- Tropical Epidemiology Group, London School of Hygiene & Tropical Medicine, London, UK; Centre for Primary Care and Public Health, Queen Mary University of London, London, UK
| | - William Johnson
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, Leicestershire, UK
| | - Julie Makani
- Muhimbili Wellcome Programme, School of Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania; Department of Haematology, School of Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Andrew M Prentice
- Medical Research Council Unit The Gambia, London School of Hygiene & Tropical Medicine, London, UK
| | - Julian P Halcox
- Institute of Life Sciences, Swansea University Medical School, Swansea University, Swansea, UK
| | - Fenella J Kirkham
- Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, UK; Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK; University Child Health, University Hospital Southampton, Southampton, UK
| |
Collapse
|
32
|
Li J, Eberwine J. The successes and future prospects of the linear antisense RNA amplification methodology. Nat Protoc 2018; 13:811-818. [PMID: 29599441 PMCID: PMC7086549 DOI: 10.1038/nprot.2018.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/04/2018] [Indexed: 12/03/2022]
Abstract
This Perspective discusses the development of the linear amplified RNA amplification technique over the last 25 years, and future applications of this important and versatile methodology. It has been over a quarter of a century since the introduction of the linear RNA amplification methodology known as antisense RNA (aRNA) amplification. Whereas most molecular biology techniques are rapidly replaced owing to the fast-moving nature of development in the field, the aRNA procedure has become a base that can be built upon through varied uses of the technology. The technique was originally developed to assess RNA populations from small amounts of starting material, including single cells, but over time its use has evolved to include the detection of various cellular entities such as proteins, RNA-binding-protein-associated cargoes, and genomic DNA. In this Perspective we detail the linear aRNA amplification procedure and its use in assessing various components of a cell's chemical phenotype. This procedure is particularly useful in efforts to multiplex the simultaneous detection of various cellular processes. These efforts are necessary to identify the quantitative chemical phenotype of cells that underlies cellular function.
Collapse
Affiliation(s)
- Jifen Li
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James Eberwine
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
33
|
de Bruin RG, Rabelink TJ, van Zonneveld AJ, van der Veer EP. Emerging roles for RNA-binding proteins as effectors and regulators of cardiovascular disease. Eur Heart J 2018; 38:1380-1388. [PMID: 28064149 DOI: 10.1093/eurheartj/ehw567] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 11/02/2016] [Indexed: 12/18/2022] Open
Abstract
The cardiovascular system comprises multiple cell types that possess the capacity to modulate their phenotype in response to acute or chronic injury. Transcriptional and post-transcriptional mechanisms play a key role in the regulation of remodelling and regenerative responses to damaged cardiovascular tissues. Simultaneously, insufficient regulation of cellular phenotype is tightly coupled with the persistence and exacerbation of cardiovascular disease. Recently, RNA-binding proteins such as Quaking, HuR, Muscleblind, and SRSF1 have emerged as pivotal regulators of these functional adaptations in the cardiovascular system by guiding a wide-ranging number of post-transcriptional events that dramatically impact RNA fate, including alternative splicing, stability, localization and translation. Moreover, homozygous disruption of RNA-binding protein genes is commonly associated with cardiac- and/or vascular complications. Here, we summarize the current knowledge on the versatile role of RNA-binding proteins in regulating the transcriptome during phenotype switching in cardiovascular health and disease. We also detail existing and potential DNA- and RNA-based therapeutic approaches that could impact the treatment of cardiovascular disease in the future.
Collapse
Affiliation(s)
- Ruben G de Bruin
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| | - Ton J Rabelink
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| | - Anton Jan van Zonneveld
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| | - Eric P van der Veer
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| |
Collapse
|
34
|
Marcantoni E, Allen N, Cambria MR, Dann R, Cammer M, Lhakhang T, O’Brien MP, Kim B, Worgall T, Heguy A, Tsirigos A, Berger JS. Platelet Transcriptome Profiling in HIV and ATP-Binding Cassette Subfamily C Member 4 (ABCC4) as a Mediator of Platelet Activity. JACC Basic Transl Sci 2018; 3:9-22. [PMID: 30062189 PMCID: PMC6058944 DOI: 10.1016/j.jacbts.2017.10.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/20/2017] [Accepted: 10/23/2017] [Indexed: 11/30/2022]
Abstract
An unbiased platelet transcriptome profile identified ATP binding cassette subfamily C member 4 (ABCC4) as a novel mediator of platelet activity in virologically suppressed human immunodeficiency virus (HIV)-infected subjects on antiretroviral therapy. Using ex vivo and in vitro cellular and molecular assays we demonstrated that ABCC4 regulated platelet activation by altering granule release and cyclic nucleotide homeostasis through a cAMP-protein kinase A (PKA)-mediated mechanism. Platelet ABCC4 inhibition attenuated platelet activation and effector cell function by reducing the release of inflammatory mediators, such as sphingosine-1-phosphate. ABCC4 inhibition may represent a novel antithrombotic strategy in HIV-infected subjects on antiretroviral therapy.
Collapse
Key Words
- ABCC4
- ABCC4, ATP binding cassette subfamily C member 4
- ART, antiretroviral therapy
- BSA, bovine serum albumin
- CVD, cardiovascular disease
- HIV
- HIV, human immunodeficiency virus
- HUVEC, human umbilical vein endothelial cell(s)
- IL, interleukin
- NSAID, nonsteroidal anti-inflammatory drug
- PAH, pulmonary artery hypertension
- PBS, phosphate-buffered saline
- RNA-Seq, RNA sequencing
- RT, room temperature
- S1P, sphingosine-1-phosphate
- VASP, vasodilator-stimulated phosphoprotein
- cAMP, cyclic adenosine monophosphate
- cardiovascular disease
- platelet activity
- qPCR, quantitative polymerase chain reaction
Collapse
Affiliation(s)
- Emanuela Marcantoni
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Nicole Allen
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Matthew R. Cambria
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Rebecca Dann
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Michael Cammer
- DART Microscopy Laboratory, NYU Langone Medical Center, New York, New York
| | - Tenzin Lhakhang
- Applied Bioinformatics Laboratories, New York University School of Medicine, New York, New York
| | - Meagan P. O’Brien
- Divisions of Infectious Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Benjamin Kim
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - Tilla Worgall
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - Adriana Heguy
- Department of Pathology, New York University School of Medicine, New York, New York
- Genome Technology Center, Division of Advanced Research Technologies, NYU Langone Medical Center, New York, New York
| | - Aristotelis Tsirigos
- Applied Bioinformatics Laboratories, New York University School of Medicine, New York, New York
| | - Jeffrey S. Berger
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York
- Division of Vascular Surgery, Department of Surgery, New York University School of Medicine, New York, New York
| |
Collapse
|
35
|
Abstract
Sickle cell disease (SCD) is a monogenetic disorder caused by a mutation in the β-globin gene HBB leading to polymerization of red blood cells causing damage to cell membranes, increasing its rigidity and intravascular hemolysis. Multiple lines of evidence suggest that SCD can be viewed as pan-vasculopathy associated with multiple mechanisms but driven by hemoglobin S polymerization. Here we review the pathophysiology, clinical manifestations and management strategies for cerebrovascular disease, pulmonary hypertension and renal disease associated with SCD. These "vascular phenotypes" reflect the systemic nature of the complications of SCD and are a major threat to the well-being of patients with the disorder.
Collapse
Affiliation(s)
- Ashar Usmani
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, USA
| | - Roberto F. Machado
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
36
|
Lu CJ, Wang Y, Huang YL, Li XH. Individualized identification of disturbed pathways in sickle cell disease. Open Life Sci 2017. [DOI: 10.1515/biol-2017-0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
AbstractBackgroundSickle cell disease (SCD) is one of the most common genetic blood disorders. Identifying pathway aberrance in an individual SCD contributes to the understanding of disease pathogenesis and the promotion of personalized therapy. Here we proposed an individualized pathway aberrance method to identify the disturbed pathways in SCD.MethodsBased on the transcriptome data and pathway data, an individualized pathway aberrance method was implemented to identify the altered pathways in SCD, which contained four steps: data preprocessing, gene-level statistics, pathway-level statistics, and significant analysis. The changed percentage of altered pathways in SCD individuals was calculated, and a differentially expressed gene (DEG)-based pathway enrichment analysis was performed to validate the results.ResultsWe identified 618 disturbed pathways between normal and SCD conditions. Among them, 6 pathways were altered in > 80% SCD individuals. Meanwhile, forty-six DEGs were identified between normal and SCD conditions, and were enriched in heme biosynthesis. Relative to DEG-based pathway analysis, the new method presented richer results and more extensive application.ConclusionThis study predicted several disturbed pathways via detecting pathway aberrance on a personalized basis. The results might provide new sights into the pathogenesis of SCD and facilitate the application of custom treatment for SCD.
Collapse
Affiliation(s)
- Chun-Juan Lu
- Department of Blood Transfusion, Heilongjiang Provincial Hospital, Haerbin150036, Heilongjiang, China
| | - Yan Wang
- Medical Laboratory Diagnosis Center, Jinan Central Hospital, Jinan250013, Shandong, China
| | - Ya-Li Huang
- Nuclear Medicine Department, Qilu Hospital of Shandong University, Jinan, 250012, Shandong PR, China
| | - Xin-Hua Li
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan250012, Shandong, China
| |
Collapse
|
37
|
Grippi C, Izzi B, Gianfagna F, Noro F, Falcinelli E, Di Pardo A, Amico E, Donati M, de Gaetano G, Iacoviello L, Hoylaerts M, Cerletti C. Neuromedin U potentiates ADP- and epinephrine-induced human platelet activation. Thromb Res 2017; 159:100-108. [DOI: 10.1016/j.thromres.2017.09.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/12/2017] [Accepted: 09/27/2017] [Indexed: 10/18/2022]
|
38
|
Best MG, Vancura A, Wurdinger T. Platelet RNA as a circulating biomarker trove for cancer diagnostics. J Thromb Haemost 2017; 15:1295-1306. [PMID: 28671345 DOI: 10.1111/jth.13720] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Platelets are multifunctional cell fragments, circulating in blood in high abundance. Platelets assist in thrombus formation, sensing of pathogens entering the blood stream, signaling to immune cells, releasing vascular remodeling factors, and, negatively, enhancing cancer metastasis. Platelets are 'educated' by their environment, including in patients with cancer. Cancer cells appear to initiate intraplatelet signaling, resulting in splicing of platelet pre-mRNAs, and enhance secretion of cytokines. Platelets can induce leukocyte and endothelial cell modeling factors, for example, through adenine nucleotides (ATP), thereby facilitating extravasation of cancer cells. Besides releasing factors, platelets can also sequester RNAs and proteins released by cancer cells. Thus, platelets actively respond to queues from local and systemic conditions, thereby altering their transcriptome and molecular content. Platelets contain a rich repertoire of RNA species, including mRNAs, small non-coding RNAs and circular RNAs; although studies regarding the functionality of the various platelet RNA species require more attention. Recent advances in high-throughput characterization of platelet mRNAs revealed 10 to > 1000 altered mRNAs in platelets in the presence of disease. Hence, platelet RNA appears to be dynamically affected by pathological conditions, thus possibly providing opportunities to use platelet RNA as diagnostic, prognostic, predictive, or monitoring biomarkers. In this review, we cover the literature regarding the platelet RNA families, processing of platelet RNAs, and the potential application of platelet RNA as disease biomarkers.
Collapse
Affiliation(s)
- M G Best
- Department of Neurosurgery, VU University Medical Center, Amsterdam, the Netherlands
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
- Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - A Vancura
- Department of Neurosurgery, VU University Medical Center, Amsterdam, the Netherlands
- Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - T Wurdinger
- Department of Neurosurgery, VU University Medical Center, Amsterdam, the Netherlands
- Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
- Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
39
|
Wysokinski WE, Tafur A, Ammash N, Asirvatham SJ, Wu Y, Gosk-Bierska I, Grill DE, Slusser JP, Mruk J, McBane RD. Impact of atrial fibrillation on platelet gene expression. Eur J Haematol 2017; 98:615-621. [PMID: 28306170 DOI: 10.1111/ejh.12879] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2017] [Indexed: 02/01/2023]
Abstract
AIMS Platelets retain cytoplasmic messenger RNA and are capable of protein biosynthesis. Several diseases are known to impact the platelet transcriptome but the effect of non-valvular atrial fibrillation (NVAF) on platelet RNA transcript is essentially unknown. The aim of this study was to evaluate the impact of NVAF on platelet RNA transcript by measuring platelet genes expression in consecutive NVAF patients before and 3-4 months after pulmonary vein isolation (PVI) and compared to normal sinus rhythm controls (NSR). METHODS AND RESULTS RNA from isolated platelets were reverse transcribed, assayed against 15 genes using real-time qPCR, and expressed as mean cycle threshold (ΔCt) using beta-2-microglobulin as endogenous control. Expression of all evaluated genes, except cathepsin A gene, was significantly lower (higher ΔCt) in 103 NVAF patients compared to 55 NSR controls. Insulin-like growth factor binding protein acid labile subunit gene (IGFALS) had expression more than 16 fold-lower (17.0±2.8 vs 12.5±3.8, P<.001), follow by genes encoding for prostacyclin receptor, and for von Willebrand factor which had fourfold lower expression compared to NSR controls. Gender, type of atrial fibrillation, heart failure, hypertension, prior stroke, diabetes mellitus, and atherosclerosis were associated with different gene expression. Following PVI, expression of four genes significantly increased, particularly IGFALS gene (increased 256-fold) and ADAMT gene increased 16-fold); expression of three genes significantly decreased, and expression of eight genes has not changed. CONCLUSIONS Platelets are capable to respond to the circulatory environment of NVAF by altering transcript and changing prothrombotic status. This shows platelet potential for molecular "reprogramming" possibly induced by flow disturbances of NVAF.
Collapse
Affiliation(s)
| | - Alfonso Tafur
- Department of Cardiovascular Medicine, Vascular Surgery and Medicine Section, NorthShore University HealthSystem, Skokie, USA
| | - Naser Ammash
- Mayo Clinic and Foundation for Education and Research, Rochester, MN, USA
| | | | - Yanhong Wu
- Mayo Clinic and Foundation for Education and Research, Rochester, MN, USA
| | | | - Diane E Grill
- Mayo Clinic and Foundation for Education and Research, Rochester, MN, USA
| | - Joshua P Slusser
- Mayo Clinic and Foundation for Education and Research, Rochester, MN, USA
| | - Jozef Mruk
- Department of Internal Medicine, University of Kansas, School of Medicine-Wichita, Wichita, KS, USA
| | - Robert D McBane
- Mayo Clinic and Foundation for Education and Research, Rochester, MN, USA
| |
Collapse
|
40
|
Association of circulating transcriptomic profiles with mortality in sickle cell disease. Blood 2017; 129:3009-3016. [PMID: 28373264 DOI: 10.1182/blood-2016-11-752279] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/27/2017] [Indexed: 12/27/2022] Open
Abstract
Sickle cell disease (SCD) complications are associated with increased morbidity and risk of mortality. We sought to identify a circulating transcriptomic profile predictive of these poor outcomes in SCD. Training and testing cohorts consisting of adult patients with SCD were recruited and prospectively followed. A pathway-based signature derived from grouping peripheral blood mononuclear cell transcriptomes distinguished 2 patient clusters with differences in survival in the training cohort. These findings were validated in a testing cohort in which the association between cluster 1 molecular profiling and mortality remained significant in a fully adjusted model. In a third cohort of West African children with SCD, cluster 1 differentiated SCD severity using a published scoring index. Finally, a risk score composed of assigning weights to cluster 1 profiling, along with established clinical risk factors using tricuspid regurgitation velocity, white blood cell count, history of acute chest syndrome, and hemoglobin levels, demonstrated a higher hazard ratio for mortality in both the training and testing cohorts compared with clinical risk factors or cluster 1 data alone. Circulating transcriptomic profiles are a powerful method to risk-stratify severity of disease and poor outcomes in both children and adults, respectively, with SCD and highlight potential associated molecular pathways.
Collapse
|
41
|
Gresele P, Falcinelli E, Sebastiano M, Momi S. Matrix Metalloproteinases and Platelet Function. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 147:133-165. [PMID: 28413027 DOI: 10.1016/bs.pmbts.2017.01.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Platelets contain and release several matrix metalloproteinases (MMPs) and their tissue inhibitors of matrix metalloproteinases (TIMPs), including MMP-1, -2, -3, -9, and -14 and TIMP-1, -2, and -4. Although devoid of a nucleus, platelets also synthesize TIMP-2 upon activation. Platelet-released MMPs/TIMPs, as well as MMPs generated by other cells within the cardiovascular system, modulate platelet function in health and disease. In particular, a normal hemostatic platelet response to vessel wall injury may be transformed into pathologic thrombus formation by the release from platelets and/or by the local generation of some MMPs. Moreover, platelets may localize the production of leukocyte-derived MMPs to sites of vascular damage, contributing to atherosclerosis development and complications and to arterial aneurysm formation. Finally, the interaction between platelets and tumor cells is strongly influenced by MMPs/TIMPs. All these mechanisms are emerging as important in atherothrombosis, inflammatory disease, and cancer growth and dissemination. Increasing knowledge of these mechanisms may open the way to novel therapeutic approaches.
Collapse
Affiliation(s)
- Paolo Gresele
- Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy.
| | - Emanuela Falcinelli
- Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Manuela Sebastiano
- Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Stefania Momi
- Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
42
|
Joehanes R, Zhang X, Huan T, Yao C, Ying SX, Nguyen QT, Demirkale CY, Feolo ML, Sharopova NR, Sturcke A, Schäffer AA, Heard-Costa N, Chen H, Liu PC, Wang R, Woodhouse KA, Tanriverdi K, Freedman JE, Raghavachari N, Dupuis J, Johnson AD, O'Donnell CJ, Levy D, Munson PJ. Integrated genome-wide analysis of expression quantitative trait loci aids interpretation of genomic association studies. Genome Biol 2017; 18:16. [PMID: 28122634 PMCID: PMC5264466 DOI: 10.1186/s13059-016-1142-6] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/20/2016] [Indexed: 12/21/2022] Open
Abstract
Background Identification of single nucleotide polymorphisms (SNPs) associated with gene expression levels, known as expression quantitative trait loci (eQTLs), may improve understanding of the functional role of phenotype-associated SNPs in genome-wide association studies (GWAS). The small sample sizes of some previous eQTL studies have limited their statistical power. We conducted an eQTL investigation of microarray-based gene and exon expression levels in whole blood in a cohort of 5257 individuals, exceeding the single cohort size of previous studies by more than a factor of 2. Results We detected over 19,000 independent lead cis-eQTLs and over 6000 independent lead trans-eQTLs, targeting over 10,000 gene targets (eGenes), with a false discovery rate (FDR) < 5%. Of previously published significant GWAS SNPs, 48% are identified to be significant eQTLs in our study. Some trans-eQTLs point toward novel mechanistic explanations for the association of the SNP with the GWAS-related phenotype. We also identify 59 distinct blocks or clusters of trans-eQTLs, each targeting the expression of sets of six to 229 distinct trans-eGenes. Ten of these sets of target genes are significantly enriched for microRNA targets (FDR < 5%). Many of these clusters are associated in GWAS with multiple phenotypes. Conclusions These findings provide insights into the molecular regulatory patterns involved in human physiology and pathophysiology. We illustrate the value of our eQTL database in the context of a recent GWAS meta-analysis of coronary artery disease and provide a list of targeted eGenes for 21 of 58 GWAS loci. Electronic supplementary material The online version of this article (doi:10.1186/s13059-016-1142-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Roby Joehanes
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA.,Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD, USA.,Institute for Aging Research, Hebrew SeniorLife, Harvard Medical School, Boston, MA, USA
| | - Xiaoling Zhang
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA.,Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA.,Section of Biomedical Genetics, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Tianxiao Huan
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA
| | - Chen Yao
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA
| | - Sai-Xia Ying
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD, USA
| | - Quang Tri Nguyen
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD, USA
| | - Cumhur Yusuf Demirkale
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD, USA
| | - Michael L Feolo
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Nataliya R Sharopova
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Anne Sturcke
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Alejandro A Schäffer
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Nancy Heard-Costa
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Han Chen
- School of Public Health, Harvard University, Boston, MA, USA.,Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Po-Ching Liu
- DNA Sequencing and Genomics Core, National Institutes of Health, Bethesda, MD, USA
| | - Richard Wang
- DNA Sequencing and Genomics Core, National Institutes of Health, Bethesda, MD, USA
| | - Kimberly A Woodhouse
- DNA Sequencing and Genomics Core, National Institutes of Health, Bethesda, MD, USA
| | - Kahraman Tanriverdi
- Department of Medicine, University of Massachusetts Medical School, Worchester, MA, USA
| | - Jane E Freedman
- Department of Medicine, University of Massachusetts Medical School, Worchester, MA, USA
| | - Nalini Raghavachari
- National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Josée Dupuis
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA.,Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Andrew D Johnson
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA
| | - Christopher J O'Donnell
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA.,Cardiology Section, Department of Medicine, Boston VA Healthcare, Boston, MA, USA
| | - Daniel Levy
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA.
| | - Peter J Munson
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD, USA. .,National Institutes of Health, Bldg 12A, Room 2003, Bethesda, MD, 20892-5626, USA.
| |
Collapse
|
43
|
Pienimaeki‐Roemer A, Konovalova T, Musri MM, Sigruener A, Boettcher A, Meister G, Schmitz G. Transcriptomic profiling of platelet senescence and platelet extracellular vesicles. Transfusion 2016; 57:144-156. [DOI: 10.1111/trf.13896] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 07/09/2016] [Accepted: 07/17/2016] [Indexed: 12/12/2022]
Affiliation(s)
| | - Tatiana Konovalova
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic of Regensburg
| | - Melina M. Musri
- Institute for Biochemistry I, Faculty of Biology and Preclinical Medicine, University of RegensburgRegensburg Germany
| | - Alexander Sigruener
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic of Regensburg
| | - Alfred Boettcher
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic of Regensburg
| | - Gunter Meister
- Institute for Biochemistry I, Faculty of Biology and Preclinical Medicine, University of RegensburgRegensburg Germany
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic of Regensburg
| |
Collapse
|
44
|
Abstract
Publisher's Note: This article has a companion Point by Brass et al. Publisher's Note: Join in the discussion of these articles at Blood Advances Community Conversations.
Collapse
|
45
|
Steppan J, Tran HT, Bead VR, Oh YJ, Sikka G, Bivalacqua TJ, Burnett AL, Berkowitz DE, Santhanam L. Arginase Inhibition Reverses Endothelial Dysfunction, Pulmonary Hypertension, and Vascular Stiffness in Transgenic Sickle Cell Mice. Anesth Analg 2016; 123:652-8. [PMID: 27537757 PMCID: PMC5032625 DOI: 10.1213/ane.0000000000001378] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND In sickle cell disease (SCD), hemolysis results in the release and activation of arginase, an enzyme that reciprocally regulates nitric oxide (NO) synthase activity and thus, NO production. Simply supplementing the common substrate L-arginine, however, fails to improve NO bioavailability. In this study, we tested the hypothesis that arginase inhibition would improve NO bioavailability and thereby attenuate systemic and pulmonary vascular endothelial dysfunction in transgenic mice with SCD. METHODS We studied 5-month-old transgenic sickle cell (SC) mice and age matched wild-type (WT) controls. SC mice were treated with the arginase inhibitor, 2(S)-amino-6-boronohexanoic acid (ABH; approximately 400 μg/d) for 4 weeks or left untreated. RESULTS Vascular arginase activity was significantly higher at baseline in untreated SC mice compared to WT controls (SC versus WT, 346 ± 69.3 vs 69 ± 17.3 pmol urea/mg protein/minute; P = 0.0043; n = 4-5 animals per group). Treatment with ABH may significantly decrease arginase activity to levels near WT controls (SC + ABH 125.2 ± 17.3 pmol urea/mg protein/minute; P = 0.0213). Aortic strips from untreated SC mice showed decreased NO and increased reactive oxygen species (ROS) production (NO: fluorescence rate 0.76 ± 0.14 vs 1.34 ± 0.17 RFU/s; P = 0.0005 and ROS: fluorescence rate 3.96 ± 1.70 vs 1.63 ± 1.20 RFU/s, P = 0.0039; n = 3- animals per group). SC animals treated with ABH for 4 weeks demonstrated NO (fluorescence rate: 1.16 ± 0.16) and ROS (fluorescence rate: 2.02 ± 0.45) levels comparable with age-matched WT controls (n = 3- animals per group). The maximal endothelial-dependent vasorelaxation response to acetylcholine was impaired in aortic rings from SC mice compared with WT (57.7% ± 8.4% vs 80.3% ± 11.0%; P = 0.02; n = 6 animals per group). The endothelial-independent response was not different between groups. In SC mice, the right ventricular cardiac output index and end-systolic elastance were similar (4.60 ± 0.51 vs 2.9 ± 0.85 mL/min/100 g and 0.89 ± 0.48 vs 0.58 ± 0.11 mm Hg/μL), whereas the pulmonary vascular resistance index and right ventricular end-systolic pressure were greater (2.9 ± 0.28 vs 5.5 ± 2.0 mm Hg × min/μL/100 g and 18.9 ± 1.1 vs 23.1 ± 4.0 mm Hg; n = 8 animals per group). Pulse wave velocity (a measure of arterial stiffness) was greater in SC mice compared with WT (3.74 ± 0.54 vs 3.25 ± 0.21 m/s; n = 20 animals per group), arginase inhibition for 4 weeks significantly reduced the vascular SC phenotype to one similar to WT animals (P = 0.0009). CONCLUSIONS Arginase inhibition improves NO bioavailability and thereby attenuates systemic and pulmonary vascular endothelial dysfunction in transgenic mice with SCD. Therefore, arginase is a potential therapeutic target in the treatment of cardiovascular dysfunction in SCD.
Collapse
MESH Headings
- Anemia, Sickle Cell/drug therapy
- Anemia, Sickle Cell/enzymology
- Anemia, Sickle Cell/physiopathology
- Animals
- Arginase/antagonists & inhibitors
- Arginase/metabolism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/physiopathology
- Enzyme Inhibitors/pharmacology
- Enzyme Inhibitors/therapeutic use
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/enzymology
- Hypertension, Pulmonary/physiopathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Pulse Wave Analysis/methods
- Vascular Stiffness/drug effects
- Vascular Stiffness/physiology
- Vasodilation/drug effects
- Vasodilation/physiology
- Vasodilator Agents/pharmacology
- Vasodilator Agents/therapeutic use
Collapse
Affiliation(s)
- Jochen Steppan
- From the *Department of Anesthesiology and Critical Care Medicine and †Department of Medicine, Division of Cardiology, Johns Hopkins Medical Institution, Baltimore, Maryland; ‡Department of Anesthesiology and Pain Medicine, Yonsei University, College of Medicine, Seoul, South Korea; and §Department of Urology, Johns Hopkins Medical Institution, Baltimore, Maryland
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Platelets are megakaryocyte-derived cellular fragments, which lack a nucleus and are the smallest circulating cells and are classically known to have a major role in supporting hemostasis. Apart from this well-established role, it is now becoming evident that platelets are also capable of conveying other important functions, such as during infection and inflammation. This paper will outline these nonhemostatic functions in two major sections termed "Platelets versus pathogens" and "Platelet-target cell communication". Platelets actively contribute to protection against invading pathogens and are capable of regulating immune functions in various target cells, all through sophisticated and efficient mechanisms. These relatively novel features will be highlighted, illustrating the multifunctional role of platelets in inflammation.
Collapse
Affiliation(s)
- Rick Kapur
- Toronto Platelet Immunobiology Group, Keenan Research Centre for Biomedical Science, St. Michael׳s Hospital, Canadian Blood Services, Toronto, Ontario, Canada
| | - John W Semple
- Toronto Platelet Immunobiology Group, Keenan Research Centre for Biomedical Science, St. Michael׳s Hospital, Canadian Blood Services, Toronto, Ontario, Canada; Departments of Pharmacology, Medicine, and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
47
|
Platelets constitutively express IL-33 protein and modulate eosinophilic airway inflammation. J Allergy Clin Immunol 2016; 138:1395-1403.e6. [PMID: 27056266 DOI: 10.1016/j.jaci.2016.01.032] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/29/2015] [Accepted: 01/07/2016] [Indexed: 11/22/2022]
Abstract
BACKGROUND Although platelets play a key role in allergic inflammation in addition to their well-established role in hemostasis, the precise mechanisms of how platelets modulate allergic inflammation are not fully understood. IL-33 is an essential regulator of innate immune responses and allergic inflammation. OBJECTIVE We sought to determine the expression of IL-33 protein by platelets and its functional significance in airway inflammation. METHODS IL-33 protein in human platelets, the human megakaryocyte cell line MEG-01, and bone marrow-derived mouse megakaryocytes was detected by using Western blot analysis and fluorescent immunostaining. We examined the functional relevance of IL-33 protein in platelets by comparing platelet-intact and platelet-depleted groups in a murine model of IL-33-dependent airway eosinophilia elicited by intranasal administration of papain. We further compared the additive effect of administration of platelets derived from wild-type versus IL-33-deficient mice on the papain-induced eosinophilia. RESULTS Platelets and their progenitor cells, megakaryocytes, constitutively expressed IL-33 protein (31 kDa). Papain-induced IL-33-dependent airway eosinophilia in mice was significantly attenuated by platelet depletion. Conversely, concomitant administration of platelets derived from wild-type mice but not IL-33-deficient mice enhanced the papain-induced airway eosinophilia. CONCLUSIONS Our novel findings suggest that platelets might be important cellular sources of IL-33 protein in vivo and that platelet-derived IL-33 might play a role in airway inflammation. Therefore platelets might become an attractive novel therapeutic target for asthma and probably allergic inflammation.
Collapse
|
48
|
New molecular insights into modulation of platelet reactivity in aspirin-treated patients using a network-based approach. Hum Genet 2016; 135:403-414. [DOI: 10.1007/s00439-016-1642-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/23/2016] [Indexed: 10/22/2022]
|
49
|
Best MG, Sol N, Tannous BA, Wesseling P, Wurdinger T. Re: a Word of Caution on New and Revolutionary Diagnostic Tests. Cancer Cell 2016; 29:143-4. [PMID: 26859454 DOI: 10.1016/j.ccell.2016.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/11/2016] [Accepted: 01/11/2016] [Indexed: 11/24/2022]
Affiliation(s)
- Myron G Best
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Nik Sol
- Department of Neurology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Bakhos A Tannous
- Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Pathology, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Thomas Wurdinger
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA; thromboDx B.V., 1098 EA Amsterdam, the Netherlands.
| |
Collapse
|
50
|
Best MG, Sol N, Kooi I, Tannous J, Westerman BA, Rustenburg F, Schellen P, Verschueren H, Post E, Koster J, Ylstra B, Ameziane N, Dorsman J, Smit EF, Verheul HM, Noske DP, Reijneveld JC, Nilsson RJA, Tannous BA, Wesseling P, Wurdinger T. RNA-Seq of Tumor-Educated Platelets Enables Blood-Based Pan-Cancer, Multiclass, and Molecular Pathway Cancer Diagnostics. Cancer Cell 2015; 28:666-676. [PMID: 26525104 PMCID: PMC4644263 DOI: 10.1016/j.ccell.2015.09.018] [Citation(s) in RCA: 607] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 07/02/2015] [Accepted: 09/25/2015] [Indexed: 12/12/2022]
Abstract
Tumor-educated blood platelets (TEPs) are implicated as central players in the systemic and local responses to tumor growth, thereby altering their RNA profile. We determined the diagnostic potential of TEPs by mRNA sequencing of 283 platelet samples. We distinguished 228 patients with localized and metastasized tumors from 55 healthy individuals with 96% accuracy. Across six different tumor types, the location of the primary tumor was correctly identified with 71% accuracy. Also, MET or HER2-positive, and mutant KRAS, EGFR, or PIK3CA tumors were accurately distinguished using surrogate TEP mRNA profiles. Our results indicate that blood platelets provide a valuable platform for pan-cancer, multiclass cancer, and companion diagnostics, possibly enabling clinical advances in blood-based "liquid biopsies".
Collapse
Affiliation(s)
- Myron G Best
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Nik Sol
- Department of Neurology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Irsan Kooi
- Department of Clinical Genetics, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Jihane Tannous
- Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA
| | - Bart A Westerman
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - François Rustenburg
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Pepijn Schellen
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; thromboDx B.V., 1098 EA Amsterdam, the Netherlands
| | - Heleen Verschueren
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; thromboDx B.V., 1098 EA Amsterdam, the Netherlands
| | - Edward Post
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; thromboDx B.V., 1098 EA Amsterdam, the Netherlands
| | - Jan Koster
- Department of Oncogenomics, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Bauke Ylstra
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Najim Ameziane
- Department of Clinical Genetics, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Josephine Dorsman
- Department of Clinical Genetics, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Egbert F Smit
- Department of Pulmonary Diseases, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Henk M Verheul
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - David P Noske
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Jaap C Reijneveld
- Department of Neurology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - R Jonas A Nilsson
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; thromboDx B.V., 1098 EA Amsterdam, the Netherlands; Department of Radiation Sciences, Oncology, Umeå University, 90185 Umeå, Sweden
| | - Bakhos A Tannous
- Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Pathology, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Thomas Wurdinger
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA; thromboDx B.V., 1098 EA Amsterdam, the Netherlands.
| |
Collapse
|