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

P Davizon-Castillo, B McMahon… - Blood, The Journal …, 2019 - ashpublications.org
Blood, The Journal of the American Society of Hematology, 2019ashpublications.org
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 …
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.
ashpublications.org