15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Paradoxes of senolytics

      article-commentary
      1 ,
      Aging (Albany NY)
      Impact Journals
      senescence, cancer, diseases, senolytic, mTOR, gerosuppressants

      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

          Senolytics are drugs that extend lifespan and delay some age-related diseases by killing senescent cells [1–4]. In fact, drug screens have identified a diverse group of drugs that are preferentially toxic to at least some senescent cells in some cellular models [2–9]. So far, however, their selectivity against senescent cells is modest and cell-type-specific [8–11]. Nevertheless, targeting senescent cells has been shown in animal models to prevent such age-related pathologies as emphysema [12], lung fibrosis [13–15], atherosclerosis [16,17], osteoporosis [18], osteoarthritis [19,20], renal disease [21], intervertebral disk pathology [2], hepatic steatosis [22] and other age-related conditions [4,7,18,23,24]. In this editorial commentary, I want to draw your attention to the paradoxes associated with senolytics, which argue against the dogma that says aging is a functional decline caused by molecular damage. This dogma predicts that senolytics should accelerate aging. If aging is caused by loss of function, then killing senescent cells would be expected to accelerate aging, given that dead cells have no functionality at all. Instead, however, senolytics slow aging, which highlights a contradiction in the prevailing dogma. The theory of hyperfunctional aging [25–32] addresses this paradox. Killing senescent cells is beneficial because senescent cells are hyperfunctional [33]. The hypersecretory phenotype or Senescence-Associated Secretory Phenotype (SASP) is the best-known example of universal hyperfunction [34–36]. Most such hyperfunctions are tissue-specific. For example, senescent beta cells overproduce insulin [37] and thus activate mTOR in hepatocytes, adipocytes and other cells, causing their hyperfunction, which in turn leads to metabolic syndrome (obesity, hypertension, hyperlipidemia and hyperglycemia) and is also a risk factor for cancer [38–40]. SASP, hyperinsulinemia and obesity, hypertension, hyperlipidemia and hyperglycemia are all examples of absolute hyperfunction (an increase in functionality). In comparison, relative hyperfunction is an insufficient decrease of unneeded function. For example, protein synthesis decreases with aging, but that decrease is not sufficient [30]. In analogy, a car moving on the highway at 65 mph is not “hyperfunctional.” But if the car were to exit the highway and enter a residential driveway at only 60 mph it would be “hyperfunctional,” and stopping that car would likely prevent damage to other objects. Similarly, killing hyperfunctional cells can prevent organismal damage. Senolytics eliminate hyperfunctional cells, which otherwise damage organs (Figure 1). Figure 1 Target of senolytics in the aging quasi-program. In post-mitotic quiescent cells in an organism, growth-promoting effectors such as mTOR drive conversion to senescence. Hyperfunctional senescent cells activate other cells (including cells in distant organs), rendering them also hyperfunctional, which eventually leads to organ damage. This process manifests as functional decline, a terminal event secondary to initial hyperfunction. Senolytics such as ABT263 or 737 kill hyperfunctional senescent cells, preventing damage to organs. Gerosuppressants such as rapamycin suppress geroconversion and may decrease hyperfunction of already senescent cells, thereby slowing disease progression (not shown here in scheme). Senolytics should not be confused with gerosuppressants (Figure 1). Gerosuppressants, such as rapamycin, do not kill cells; they instead prevent cellular conversion to senescence (geroconversion) [33]. Rapamycin also slows disease progression by limiting the hyperfunction of senescent cells. Notably, some senolytics are also gerosuppressants. For example, inhibitors of MEK [41–43] or PI3K [2,41] are both gerosuppressants [41] and senolytics [2,42,43]. It may seem paradoxical that senolytics are anticancer drugs [44] because standard anticancer agents cause molecular damage. According to the hyperfunction theory [45], molecular damage does not cause aging. Although accumulation of molecular damage does happen and would destroy the organism eventually, no organism lives long enough for that to occur because TOR-driven (hyperfunctional) aging kills it first. If TOR-driven aging (i.e., aging as we currently know it) were abolished, then organisms would die from “post-aging syndrome” due to molecular damage (see Figure 8 in ref. [25].). Molecular damage contributes to some age-related diseases. But these diseases would arise even without molecular damage [45]. Molecular damage is essential for most types of cancer, but a senescent microenvironment [46] and overall organism aging (and associated diseases such as diabetes) also play roles [47], as does clonal selection for mTOR activation in cancer cells [48]. Importantly, molecular damage renders cancer cells robust and hyperfunctional. Cancer cells kill an organism not because molecular damage makes them weak; it is because the molecular damage makes them robust and hyperfunctional. If accumulation of molecular damage leads to immortalization and robustness, then aging cannot represent functional decline caused by molecular damage [48]. All senolytics, without exception, were initially investigated or specifically developed as anticancer drugs. But not all anticancer drugs are senolytics. Both senolytics and gerosuppressants belong to a very special subgroup of oncotargeted drugs [49]. Various pathways involving IGF-1, Ras, MEK, AMPK, TSC1/2, FOXO, PI3K, mTOR, S6K, and NFκB comprise a mTOR-related network and are involved in aging [49]. Oncoproteins promote aging, while tumor suppressors are gerosuppressors, which inhibit aging [48,50]. As depicted a decade ago (see Figure 3 in ref. [51]. and Figures 4 and 9 in ref. [25].), oncotargets are gerotargets that are also mTOR activators, while tumor and aging suppressors are mTOR inhibitors. In brief, geroconversion and oncogenic transformation are two sides of the same process [50]. Gerogenic oncogenes activate the mTOR pathway, driving geroconversion of cell cycle-arrested cells. When cell cycle control is disabled, they drive oncogenic transformation [48,50]. Many puzzles remain. For example, killing senescent adipocytes, macrophages or foam cells will slow diseases such as atherosclerosis and metabolic diseases, and killing senescent glial cells can prevent cognitive decline [23]. On the other hand, killing some senescent cell types may be counterproductive. For example, killing senescent beta cells may lead to diabetes [37], and killing of senescent hyperfunctional neurons in Alzheimer’s disease may have unpredictable consequences. Fortunately, senolytics are tissue-specific and only kill some types of senescent cells [8–11], which may make them safer. To add further complication to the paradoxes associated with senolytics, it was shown that many detected p16/β-gal-positive cells are not senescent cells, but are instead hyperfunctional macrophages, which contribute to aging [52–54]. Notably, β-gal staining is a marker of hyperfunctional lysosomes [55]. A combination of markers, including mTOR targets, is needed to define senescence [33]. Some senolytics that target Bcl2 family proteins may theoretically kill leukemia/lymphoma cells. I hope to discuss these and other issues in a scheduled review “Senolytics, gerosuppressants and conventional life-extending drugs.”

          Related collections

          Most cited references40

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs

          The healthspan of mice is enhanced by killing senescent cells using a transgenic suicide gene. Achieving the same using small molecules would have a tremendous impact on quality of life and the burden of age-related chronic diseases. Here, we describe the rationale for identification and validation of a new class of drugs termed senolytics, which selectively kill senescent cells. By transcript analysis, we discovered increased expression of pro-survival networks in senescent cells, consistent with their established resistance to apoptosis. Using siRNA to silence expression of key nodes of this network, including ephrins (EFNB1 or 3), PI3Kδ, p21, BCL-xL, or plasminogen-activated inhibitor-2, killed senescent cells, but not proliferating or quiescent, differentiated cells. Drugs targeting these same factors selectively killed senescent cells. Dasatinib eliminated senescent human fat cell progenitors, while quercetin was more effective against senescent human endothelial cells and mouse BM-MSCs. The combination of dasatinib and quercetin was effective in eliminating senescent MEFs. In vivo, this combination reduced senescent cell burden in chronologically aged, radiation-exposed, and progeroid Ercc1 −/Δ mice. In old mice, cardiac function and carotid vascular reactivity were improved 5 days after a single dose. Following irradiation of one limb in mice, a single dose led to improved exercise capacity for at least 7 months following drug treatment. Periodic drug administration extended healthspan in Ercc1 −/Δ mice, delaying age-related symptoms and pathology, osteoporosis, and loss of intervertebral disk proteoglycans. These results demonstrate the feasibility of selectively ablating senescent cells and the efficacy of senolytics for alleviating symptoms of frailty and extending healthspan.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation.

            The TOR (target of rapamycin) kinase limits longevity by poorly understood mechanisms. Rapamycin suppresses the mammalian TORC1 complex, which regulates translation, and extends lifespan in diverse species, including mice. We show that rapamycin selectively blunts the pro-inflammatory phenotype of senescent cells. Cellular senescence suppresses cancer by preventing cell proliferation. However, as senescent cells accumulate with age, the senescence-associated secretory phenotype (SASP) can disrupt tissues and contribute to age-related pathologies, including cancer. MTOR inhibition suppressed the secretion of inflammatory cytokines by senescent cells. Rapamycin reduced IL6 and other cytokine mRNA levels, but selectively suppressed translation of the membrane-bound cytokine IL1A. Reduced IL1A diminished NF-κB transcriptional activity, which controls much of the SASP; exogenous IL1A restored IL6 secretion to rapamycin-treated cells. Importantly, rapamycin suppressed the ability of senescent fibroblasts to stimulate prostate tumour growth in mice. Thus, rapamycin might ameliorate age-related pathologies, including late-life cancer, by suppressing senescence-associated inflammation.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Identification of a novel senolytic agent, navitoclax, targeting the Bcl‐2 family of anti‐apoptotic factors

              Summary Clearing senescent cells extends healthspan in mice. Using a hypothesis‐driven bioinformatics‐based approach, we recently identified pro‐survival pathways in human senescent cells that contribute to their resistance to apoptosis. This led to identification of dasatinib (D) and quercetin (Q) as senolytics, agents that target some of these pathways and induce apoptosis preferentially in senescent cells. Among other pro‐survival regulators identified was Bcl‐xl. Here, we tested whether the Bcl‐2 family inhibitors, navitoclax (N) and TW‐37 (T), are senolytic. Like D and Q, N is senolytic in some, but not all types of senescent cells: N reduced viability of senescent human umbilical vein epithelial cells (HUVECs), IMR90 human lung fibroblasts, and murine embryonic fibroblasts (MEFs), but not human primary preadipocytes, consistent with our previous finding that Bcl‐xl siRNA is senolytic in HUVECs, but not preadipocytes. In contrast, T had little senolytic activity. N targets Bcl‐2, Bcl‐xl, and Bcl‐w, while T targets Bcl‐2, Bcl‐xl, and Mcl‐1. The combination of Bcl‐2, Bcl‐xl, and Bcl‐w siRNAs was senolytic in HUVECs and IMR90 cells, while combination of Bcl‐2, Bcl‐xl, and Mcl‐1 siRNAs was not. Susceptibility to N correlated with patterns of Bcl‐2 family member proteins in different types of human senescent cells, as has been found in predicting response of cancers to N. Thus, N is senolytic and acts in a potentially predictable cell type‐restricted manner. The hypothesis‐driven, bioinformatics‐based approach we used to discover that dasatinib (D) and quercetin (Q) are senolytic can be extended to increase the repertoire of senolytic drugs, including additional cell type‐specific senolytic agents.
                Bookmark

                Author and article information

                Journal
                Aging (Albany NY)
                Aging (Albany NY)
                Aging
                Aging (Albany NY)
                Impact Journals
                1945-4589
                December 2018
                28 December 2018
                : 10
                : 12
                : 4289-4293
                Affiliations
                [1 ]Cell Stress Biology, Roswell Park Cancer Institute , Buffalo, , NY, 14263, USA
                Author notes
                Correspondence to: Mikhail V. Blagosklonny; email: Blagosklonny@ 123456rapalogs.com
                Article
                101750
                10.18632/aging.101750
                6326665
                30594910
                1f9461d9-3d07-44d1-95ea-b676fb84abe3
                Copyright © 2018 Blagosklonny

                This is an open-access article distributed under the terms of the Creative Commons Attribution (CC BY) 3.0 License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                : 24 December 2018
                : 27 December 2018
                Categories
                Commentary

                Cell biology
                senescence,cancer,diseases,senolytic,mtor,gerosuppressants
                Cell biology
                senescence, cancer, diseases, senolytic, mtor, gerosuppressants

                Comments

                Comment on this article