2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Troublemaking mutations: Clonal hematopoiesis for the prediction of prognosis in ST-segment elevation myocardial infarction

      discussion
      a , a , b , *
      EBioMedicine
      Elsevier

      Read this article at

      Bookmark
          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

          In recent years, a new risk factor has emerged in cardiovascular disease (CVD): clonal hematopoiesis of indeterminate potential (CHIP). This condition occurs when acquired somatic mutations in cancer-related genes provide a selective advantage to hematopoietic stem and progenitor cells (HSPCs), leading to the expansion of mutant hematopoietic cells over the years. 1 The cellular progeny of HSPCs inherits these mutations and, therefore, CHIP can be detected in peripheral blood by DNA sequencing approaches. While CHIP confers an increased relative risk of developing hematological neoplasia, most CHIP mutation carriers will never transition to malignancy, as these conditions are relatively rare and typically require the acquisition of several mutations. However, CHIP is associated with higher all-cause mortality rates, mainly due to an increased risk of atherosclerotic CVD and related conditions. 1 , 2 Experimental studies in mice suggest that this connection may be accounted for by an exacerbated inflammatory response driven by mutant immune cells.2, 3, 4 While research on CHIP has soared recently, there is still much to be learned about this new cardiovascular risk factor and its potential impact on the clinical management of CVD. In this issue of EBioMedicine, Wang et al. 5 shed light on the clinical implications of CHIP in patients affected by a myocardial infarction with ST-segment elevation (STEMI), a common acute clinical manifestation of coronary artery disease that is associated with increased risk of death and ischemic heart failure (HF) development. The authors performed deep targeted sequencing in a cohort of 485 patients with STEMI (median age of 62 years) to assess the prevalence and prognostic value of CHIP in this setting. While the sequencing panel included 51 different genes, the study was primarily focused on mutations in the epigenetic regulatory genes DNMT3A and TET2, the two most common CHIP driver genes. Most analyses considered exclusively mutations with a variant allele fraction (VAF) > 2% (i.e. 4% mutant blood cells, assuming a monoallelic mutation), which is the threshold of mutant clone size most frequently used to define CHIP, although an exploratory analysis of the effect of smaller mutant clones was also executed. This undertaking yielded 3 major findings. First, the authors found that CHIP mutations were present in 16.5% of STEMI patients considering VAF ≥ 2%, with 70% of these mutations occurring in DNMT3A or TET2. Second, DNMT3A or TET2 mutations with a VAF ≥ 2% were associated with >2-fold higher incidence of death or major adverse cardiac events (a composite of all-cause death, recurrent nonfatal MI, nonfatal stroke or HF-related hospitalization) during a median follow up of 3 years, independently of traditional CVD risk factors. Third, DNMT3A and TET2 mutation carriers showed a modest, but statistically significant increase in circulating levels of the inflammatory cytokines IL-1β and IL-6, consistent with findings in previous CHIP studies in mice and humans.2, 3, 4 , 6 This increase in inflammatory cytokines was not accompanied by significant differences in high-sensitivity C-reactive levels, suggesting that this common indicator of systemic inflammation does not capture the exacerbated inflammation that accompanies CHIP. The interesting findings reported by Wang and co-workers need to be interpreted in the context of the fast-evolving literature on CHIP. While some controversies have arisen about the intricacies of the connection between CHIP and CVD, a number of independent lines of evidence suggest that CHIP mutations, particularly in DNMT3A and TET2, are potent predictors of adverse outcomes in CVD patients. 1 Chronic HF patients who carry CHIP mutations display a substantially higher risk of adverse clinical progression, defined as higher risk of death or composites of all-cause or HF-specific death and HF-related hospitalizations. 1 , 6, 7, 8 The current work extends the prognostic value of CHIP to STEMI patients and fuels the growing interest in using anti-inflammatory approaches to prevent recurrent CVD events. 1 , 9 Based on the findings in the current study and prior work,2, 3, 4 , 10 it is logical to wonder whether the heightened IL-6/IL-1β-driven inflammation associated with some CHIP mutations may be a precision target for the prevention of recurrent CVD events post-MI in CHIP mutation carriers. Testing this possibility will require the development of carefully- designed clinical trials. A major strength of the work by Wang et al. 5 is the use of a deep sequencing strategy, which allows for greater sensitivity in the detection of CHIP with relatively small mutant clones (e.g. VAF < 5%) than the whole exome/genome approaches used in previous larger studies. 2 , 10 The findings reported herein suggest that CHIP with VAF as low as 1% may be clinically relevant in the context of STEMI, at least for DNMT3A and TET2 mutations, similar to previous reports in the setting of ischemic HF. 8 Although it is not currently recommended to screen unselected patients with STEMI or HF for CHIP, these findings indicate that sensitive sequencing technologies, such as the one used in this study, may be required in the future to capture clinically relevant CHIP clones in CVD patients. An important caveat that must be considered is the limited percentage of women in the study cohort (∼27%), which does not allow to evaluate potential sex-differences in the clinical impact of CHIP. Similarly, the potential prognostic value of CHIP mutations beyond those affecting DNMT3A or TET2 remains unexplored and requires further investigation. In summary, the novel results reported by Wang et al. 5 have important implications for our understanding of the prognostic value of CHIP mutations. This work expands the mounting body of evidence linking CHIP to CVD, and reinforces the notion that the detection of CHIP may in the future aid decision-making in the clinical management of the growing population of patients who survive a MI. Declaration of interests JJF is a consultant for FL86. MD-D has no relationships relevant to the contents of this article to disclose.

          Related collections

          Most cited references10

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

          Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease

          Background Clonal hematopoiesis of indeterminate potential (CHIP), defined by the presence of an expanded somatic blood cell clone in those without other hematologic abnormalities, is common in older individuals and associates with an increased risk of developing hematologic cancer. We previously found preliminary evidence for an association of CHIP with human atherosclerotic cardiovascular disease, but the nature of this association was unclear. Methods We used whole exome sequencing to detect the presence of CHIP in peripheral blood cells and associated this with coronary heart disease in four case-control studies together comprising 4,794 cases and 3,537 controls. To assess causality, we perturbed the function of Tet2, the second most commonly mutated gene linked to clonal hematopoiesis, in the hematopoietic cells of atherosclerosis-prone mice. Results In nested case-control analyses from two prospective cohorts, carriers of CHIP had a 1.9-fold (95% confidence interval 1.4–2.7) increased risk of coronary heart disease compared to non-carriers. In two retrospective case-control cohorts for early-onset myocardial infarction, those with CHIP had a 4.0-fold greater risk (95% confidence interval 2.4–6.7) of having myocardial infarction. Mutations in DNMT3A, TET2, ASXL1, and JAK2 were each individually associated with coronary heart disease. Those with clonal hematopoiesis also had increased coronary artery calcification, a marker of coronary atherosclerosis burden. Hyperlipidemic mice engrafted with Tet2−/− or Tet2+/− bone marrow developed larger atherosclerotic lesions in the aortic root and aorta than mice receiving control marrow. Analyses of Tet2−/− macrophages demonstrated elevated expression of several chemokine and cytokine genes that contribute to atherosclerosis. Conclusions Clonal hematopoiesis robustly associates with coronary heart disease in humans and causes accelerated atherosclerosis in mice.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice

            Human aging is associated with an increased frequency of somatic mutations in hematopoietic cells. Several of these recurrent mutations, including those in the gene encoding the epigenetic modifier enzyme TET2, promote expansion of the mutant blood cells. This clonal hematopoiesis correlates with an increased risk of atherosclerotic cardiovascular disease. We studied the effects of the expansion of Tet2 -mutant cells in atherosclerosis-prone, low-density lipoprotein receptor–deficient ( Ldlr −/− ) mice. We found that partial bone marrow reconstitution with TET2-deficient cells was sufficient for their clonal expansion and led to a marked increase in atherosclerotic plaque size. TET2-deficient macrophages exhibited an increase in NLRP3 inflammasome–mediated interleukin-1β secretion. An NLRP3 inhibitor showed greater atheroprotective activity in chimeric mice reconstituted with TET2-deficient cells than in nonchimeric mice. These results support the hypothesis that somatic TET2 mutations in blood cells play a causal role in atherosclerosis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Targeting inflammation in atherosclerosis — from experimental insights to the clinic

              Atherosclerosis, a dominant and growing cause of death and disability worldwide, involves inflammation from its inception to the emergence of complications. Targeting inflammatory pathways could therefore provide a promising new avenue to prevent and treat atherosclerosis. Indeed, clinical studies have now demonstrated unequivocally that modulation of inflammation can forestall the clinical complications of atherosclerosis. This progress pinpoints the need for preclinical investigations to refine strategies for combatting inflammation in the human disease. In this Review, we consider a gamut of attractive possibilities for modifying inflammation in atherosclerosis, including targeting pivotal inflammatory pathways such as the inflammasomes, inhibiting cytokines, manipulating adaptive immunity and promoting pro-resolution mechanisms. Along with lifestyle measures, pharmacological interventions to mute inflammation could complement traditional targets, such as lipids and hypertension, to make new inroads into the management of atherosclerotic risk. The contribution of inflammation to atherosclerosis is substantial, and is just beginning to be understood. In this Review, Soehnlein and Libby discuss how inflammation promotes atherosclerosis and its consequences, and how such processes could be targeted therapeutically. The potential pitfalls of targeting immune processes — namely the increased potential for infections — are also discussed, along with ways to modulate cardiovascular therapies in time and space to make them more effective.
                Bookmark

                Author and article information

                Contributors
                Journal
                EBioMedicine
                EBioMedicine
                EBioMedicine
                Elsevier
                2352-3964
                28 April 2022
                May 2022
                28 April 2022
                : 79
                : 104015
                Affiliations
                [a ]Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
                [b ]CIBER en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
                Author notes
                [* ]Corresponding author at: Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain. jjfuster@ 123456cnic.es
                Article
                S2352-3964(22)00199-2 104015
                10.1016/j.ebiom.2022.104015
                9062663
                35490554
                9779b031-397e-44e5-b086-14b2b59942d9
                © 2022 The Authors

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 5 April 2022
                : 5 April 2022
                Categories
                Comment

                Comments

                Comment on this article