18
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      TNF-α–driven inflammation and mitochondrial dysfunction define the platelet hyperreactivity of aging

      research-article

      Read this article at

      ScienceOpenPublisherPMC
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Publisher's Note: There is a [Related article:] Blood Commentary on this article in this issue.

          Key Points

          • Aging-associated inflammation by TNF-α plays an important role in the development of platelet hyperreactivity during aging.

          • Aging-associated platelet hyperreactivity is associated with megakaryocytic inflammatory, metabolic, and mitochondrial reprogramming.

          Abstract

          Aging and chronic inflammation are independent risk factors for the development of atherothrombosis and cardiovascular disease. We hypothesized that aging-associated inflammation promotes the development of platelet hyperreactivity and increases thrombotic risk during aging. Functional platelet studies in aged-frail adults and old mice demonstrated that their platelets are hyperreactive and form larger thrombi. We identified tumor necrosis factor α (TNF-α) as the key aging-associated proinflammatory cytokine responsible for platelet hyperreactivity. We further showed that platelet hyperreactivity is neutralized by abrogating signaling through TNF-α receptors in vivo in a mouse model of aging. Analysis of the bone marrow compartments showed significant platelet-biased hematopoiesis in old mice reflected by increased megakaryocyte-committed progenitor cells, megakaryocyte ploidy status, and thrombocytosis. Single-cell RNA-sequencing analysis of native mouse megakaryocytes showed significant reprogramming of inflammatory, metabolic, and mitochondrial gene pathways in old mice that appeared to play a significant role in determining platelet hyperreactivity. Platelets from old mice (where TNF-α was endogenously increased) and from young mice exposed to exogenous TNF-α exhibited significant mitochondrial changes characterized by elevated mitochondrial mass and increased oxygen consumption during activation. These mitochondrial changes were mitigated upon TNF-α blockade. Similar increases in platelet mitochondrial mass were seen in platelets from patients with myeloproliferative neoplasms, where TNF-α levels are also increased. Furthermore, metabolomics studies of platelets from young and old mice demonstrated age-dependent metabolic profiles that may differentially poise platelets for activation. Altogether, we present previously unrecognized evidence that TNF-α critically regulates megakaryocytes resident in the bone marrow niche and aging-associated platelet hyperreactivity and thrombosis.

          Visual Abstract

          Related collections

          Most cited references47

          • Record: found
          • Abstract: found
          • Article: not found

          Functionally Distinct Subsets of Lineage-Biased Multipotent Progenitors Control Blood Production in Normal and Regenerative Conditions.

          Despite great advances in understanding the mechanisms underlying blood production, lineage specification at the level of multipotent progenitors (MPPs) remains poorly understood. Here, we show that MPP2 and MPP3 are distinct myeloid-biased MPP subsets that work together with lymphoid-primed MPP4 cells to control blood production. We find that all MPPs are produced in parallel by hematopoietic stem cells (HSCs), but with different kinetics and at variable levels depending on hematopoietic demands. We also show that the normally rare myeloid-biased MPPs are transiently overproduced by HSCs in regenerating conditions, hence supporting myeloid amplification to rebuild the hematopoietic system. This shift is accompanied by a reduction in self-renewal activity in regenerating HSCs and reprogramming of MPP4 fate toward the myeloid lineage. Our results support a dynamic model of blood development in which HSCs convey lineage specification through independent production of distinct lineage-biased MPP subsets that, in turn, support lineage expansion and differentiation.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Platelets release mitochondria serving as substrate for bactericidal group IIA-secreted phospholipase A2 to promote inflammation.

            Mitochondrial DNA (mtDNA) is a highly potent inflammatory trigger and is reportedly found outside the cells in blood in various pathologies. Platelets are abundant in blood where they promote hemostasis. Although lacking a nucleus, platelets contain functional mitochondria. On activation, platelets produce extracellular vesicles known as microparticles. We hypothesized that activated platelets could also release their mitochondria. We show that activated platelets release respiratory-competent mitochondria, both within membrane-encapsulated microparticles and as free organelles. Extracellular mitochondria are found in platelet concentrates used for transfusion and are present at higher levels in those that induced acute reactions (febrile nonhemolytic reactions, skin manifestations, and cardiovascular events) in transfused patients. We establish that the mitochondrion is an endogenous substrate of secreted phospholipase A2 IIA (sPLA2-IIA), a phospholipase otherwise specific for bacteria, likely reflecting the ancestral proteobacteria origin of mitochondria. The hydrolysis of the mitochondrial membrane by sPLA2-IIA yields inflammatory mediators (ie, lysophospholipids, fatty acids, and mtDNA) that promote leukocyte activation. Two-photon microscopy in live transfused animals revealed that extracellular mitochondria interact with neutrophils in vivo, triggering neutrophil adhesion to the endothelial wall. Our findings identify extracellular mitochondria, produced by platelets, at the midpoint of a potent mechanism leading to inflammatory responses. © 2014 by The American Society of Hematology.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Inflamm-Aging of Hematopoiesis, Hematopoietic Stem Cells, and the Bone Marrow Microenvironment

              All hematopoietic and immune cells are continuously generated by hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) through highly organized process of stepwise lineage commitment. In the steady state, HSCs are mostly quiescent, while HPCs are actively proliferating and contributing to daily hematopoiesis. In response to hematopoietic challenges, e.g., life-threatening blood loss, infection, and inflammation, HSCs can be activated to proliferate and engage in blood formation. The HSC activation induced by hematopoietic demand is mediated by direct or indirect sensing mechanisms involving pattern recognition receptors or cytokine/chemokine receptors. In contrast to the hematopoietic challenges with obvious clinical symptoms, how the aging process, which involves low-grade chronic inflammation, impacts hematopoiesis remains undefined. Herein, we summarize recent findings pertaining to functional alternations of hematopoiesis, HSCs, and the bone marrow (BM) microenvironment during the processes of aging and inflammation and highlight some common cellular and molecular changes during the processes that influence hematopoiesis and its cells of origin, HSCs and HPCs, as well as the BM microenvironment. We also discuss how age-dependent alterations of the immune system lead to subclinical inflammatory states and how inflammatory signaling might be involved in hematopoietic aging. Our aim is to present evidence supporting the concept of “Inflamm-Aging,” or inflammation-associated aging of hematopoiesis.
                Bookmark

                Author and article information

                Journal
                Blood
                Blood
                bloodjournal
                blood
                Blood
                Blood
                American Society of Hematology (Washington, DC )
                0006-4971
                1528-0020
                29 August 2019
                16 July 2019
                29 August 2019
                : 134
                : 9
                : 727-740
                Affiliations
                [1 ]Center for Cancer and Blood Disorders, Department of Pediatrics and
                [2 ]Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO;
                [3 ]Irish Center for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland;
                [4 ]Department of Mechanical Engineering, Colorado State University, Fort Collins, CO;
                [5 ]Cardiovascular Pulmonary Research Laboratory, Department of Pediatrics and
                [6 ]Cardiovascular Pulmonary Research Laboratory, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO;
                [7 ]Molecular Medicine Program and
                [8 ]Department of Internal Medicine, University of Utah, Salt Lake City, UT;
                [9 ]Department of Biochemistry and Molecular Genetics and
                [10 ]Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO;
                [11 ]Department of Internal Medicine, University of Washington, Seattle, WA;
                [12 ]Division of Geriatrics, University of Utah, Salt Lake City, UT;
                [13 ]Geriatric Research, Education, and Clinical Center, George E. Wahlen Department of Veterans Affairs Medical Center (VAMC), Salt Lake City, UT;
                [14 ]División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México;
                [15 ]Department of Pathology and Laboratory Medicine, Children’s Hospital Colorado, Aurora, CO; and
                [16 ]Department of Pathology, University of Utah, Salt Lake City, UT
                Author information
                http://orcid.org/0000-0002-9865-6176
                http://orcid.org/0000-0001-7862-6503
                http://orcid.org/0000-0002-1178-8927
                http://orcid.org/0000-0001-9112-994X
                http://orcid.org/0000-0002-2258-6490
                http://orcid.org/0000-0002-8130-991X
                http://orcid.org/0000-0002-4406-3826
                http://orcid.org/0000-0002-7229-5528
                http://orcid.org/0000-0003-3455-3730
                http://orcid.org/0000-0002-1461-2871
                Article
                PMC6716075 PMC6716075 6716075 2019/BLD2019000200
                10.1182/blood.2019000200
                6716075
                31311815
                cc3c25f1-352d-4684-8d50-5770976ef390
                © 2019 by The American Society of Hematology
                History
                : 16 February 2019
                : 02 July 2019
                Page count
                Pages: 14
                Funding
                Funded by: National Institutes of Health;
                Categories
                2
                35
                Plenary Paper
                Platelets and Thrombopoiesis
                Custom metadata
                free

                Comments

                Comment on this article