Skip to main content
Erschienen in: Cancer and Metastasis Reviews 2/2010

Open Access 01.06.2010

Senescent cells as a source of inflammatory factors for tumor progression

verfasst von: Albert R. Davalos, Jean-Philippe Coppe, Judith Campisi, Pierre-Yves Desprez

Erschienen in: Cancer and Metastasis Reviews | Ausgabe 2/2010

Abstract

Cellular senescence, which is associated with aging, is a process by which cells enter a state of permanent cell cycle arrest, therefore constituting a potent tumor suppressive mechanism. Recent studies show that, despite the beneficial effects of cellular senescence, senescent cells can also exert harmful effects on the tissue microenvironment. The most significant of these effects is the acquisition of a senescent-associated secretory phenotype (SASP), which entails a striking increase in the secretion of pro-inflammatory cytokines. Here, we summarize our knowledge of the SASP and the impact it has on tissue microenvironments and ability to stimulate tumor progression.

1 Introduction

Organisms with renewable tissues are at risk for developing hyperproliferative diseases, the most deadly of which is cancer. This risk is mitigated by tumor suppressor mechanisms, which suppress cancer for much of the life span. However, because these mechanisms are imperfect, cancer still poses a major challenge to the longevity of such organisms [1, 2]. Some tumor suppressor mechanisms act by curtailing the acquisition of mutations, either by preventing DNA damage or optimizing DNA repair. Others act by preventing damaged or mutant cells from developing into a tumor by eliminating them entirely (apoptosis) or permanently arresting their proliferation (cellular senescence). Several lines of evidence suggest that these tumor suppressor mechanisms can be doubled-edged swords: whereas they undoubtedly suppress the development of cancer early in life, they can also result in altered tissue structure, organization, and homeostasis. These tissue changes can drive phenotypes and pathologies associated with aging, including, ironically, late-life cancer.
In support of a role in tumor suppression, cellular senescence is induced by many potentially oncogenic stimuli [35]. In addition, the senescence response depends on two potent tumor suppressor pathways: that governed by the p53 protein and that governed by the pRB and p16INK4a proteins. Indeed, germline mutations in p53 or p16INK4A allow cells to ignore certain senescence-inducing signals and greatly increase cancer susceptibility [6, 7]. Further, virtually all cancers harbor mutations in the p53 and/or p16/pRB pathways, which, among other activities, are crucial for the senescence response [8, 9]. Finally, senescent cells are found in pre-malignant lesions in mice and humans, and in mouse models, the senescence response prevents malignant progression [10]. Interestingly, in culture and in vivo, some tumor cells retain the ability to senesce and do so in response to DNA damaging chemotherapy; in mouse models, this response is associated with arrested tumor progression and eventual regression [1113].
Consistent with a role in aging, senescent cells accumulate with age in many rodent, non-human primate, and human tissues [14, 15]. Moreover, they are found at sites of age-related pathology, including degenerative disorders such as osteoarthritis and atherosclerosis [14] and hyperproliferative lesions such as benign prostatic hyperplasia [16] and melanocytic naevi [17]. A limited number of cell culture and mouse xenograft studies support the idea that senescent cells secrete factors that can disrupt tissue structure and function and promote cancer progression [1821]. Recent studies on the senescence-associated secretory phenotype (SASP) of human and mouse fibroblasts show it is conserved across cell types and species and that specific secreted factors are strong candidates for stimulating malignant phenotypes in neighboring cells [22, 23].
The idea that a biological process such as cellular senescence can be both beneficial (tumor suppressive) and deleterious (pro-tumorigenic) is consistent with a major evolutionary theory of aging termed antagonistic pleiotropy [24]. The SASP may be the major reason for the deleterious side of the senescence response and is the focus of this review. We particularly emphasize the potential effects of the SASP [25] on cell behavior in the context of tumor progression.

2 Cellular senescence as a stress response

Cellular senescence was first identified as a process that limits the proliferation of human cells in culture [26]. These early experiments showed that cultured human fibroblasts gradually lose proliferative capacity until all cells in the culture cease division. Much of this growth arrest is known to occur because most human cells do not express telomerase. Consequently, with each cell cycle, telomeres shorten and eventually fail, generating a persistent DNA damage signal that permanently arrests growth [27]. It is now known, however, that cellular senescence occurs in culture and in vivo as a response to excessive intracellular or extracellular stress (see Fig. 1). The senescence program locks the cell into a cell cycle arrest that prevents the damage from affecting the next cell generation, thereby preventing potential malignant transformation [24]. Senescent cells have been shown to accumulate over the life span of rodents, non-human primates, and humans and are found primarily in renewable tissues [15, 28].
Many types of stresses can provoke cellular senescence [5, 29] (see Fig. 1). These stresses include dysfunctional telomeres resulting from repeated cell division (replicative senescence) or other telomeric damage. They also include oxidative stress resulting from mitochondrial deterioration or other causes, severe or irreparable DNA damage from external sources and disrupted chromatin organization due to DNA replication or damage (genotoxic stress), and the expression of certain oncogenes (oncogene-induced senescence) [10, 14, 3035]. Stresses that cause cellular senescence can be induced by external or internal chemical or physical insults encountered during the course of the life span, during therapeutic interventions (for example, X-irradiation or chemotherapy), or as a consequence of endogenous processes such as oxidative respiration or mitogenic signals. External mitogenic signals, for example growth-related oncogene alpha (GROα) secretion by tumor cells in close to normal cells [36] or circulating angiotensin II [37, 38], have also been shown to induce cellular senescence. All somatic cells that have the ability to divide can potentially undergo senescence. Regardless of the disparate mechanisms of senescent-inducing stresses, the senescence program is activated once a cell has sensed a critical level of damage or dysfunction. Senescent cells are detected in culture and in vivo by a variety of markers, including the senescence-associated β-galactosidase (SA-βgal) [28], p16INK4a [39], telomere-associated DNA damage foci [40], senescence-associated heterochromatin foci [41], and several other molecules; all of these markers have some limitations, and thus must be used in combination with each other, as well as proliferation markers (absent in senescent cells) [42].

3 The secretory phenotype of senescent cells

Cellular senescence is accompanied by a striking increase in the secreted levels of >40 factors involved in intercellular signaling [22, 25, 43]. This phenotype has been termed the “senescence-associated secretory phenotype”, or SASP [22]. The SASP has many of the paracrine effects one would expect from a pro-inflammatory stimulus, which can be deleterious if left unchecked (see Fig. 2). For example, senescent cells promote the proliferation and tumorigenesis of epithelial cells [18], stimulate angiogenesis [44], trigger an epithelial to mesenchymal transition, accelerate the invasion of transformed cells [22], and increase the growth of xenograft tumors in vivo [19]. Further, the SASP has been shown to occur after treatment of cancer patients with DNA damaging chemotherapy [22].
The SASP includes several families of soluble and insoluble factors. These factors can affect surrounding cells by activating various cell surface receptors and corresponding signal transduction pathways that may lead to multiple pathologies, including cancer. SASP factors can globally be divided into the following major categories: soluble signaling factors (interleukins, chemokines, and growth factors), secreted proteases, and secreted insoluble components. SASP proteases can have three major effects: shedding of membrane-associated proteins resulting in soluble versions of membrane-bound receptors, cleavage/degradation of signaling molecules, and degradation or processing of the extracellular matrix. These activities provide potent mechanisms by which senescent cells can modify the tissue microenvironment. In the following sections, we discuss the SASP subsets and some of their known paracrine effects on nearby cells with an emphasis on their ability to facilitate cancer progression.

3.1 Soluble factors

The most prominent cytokine of the SASP is interleukin (IL)-6, a pleiotropic pro-inflammatory cytokine. IL-6 secretion has been shown to increase markedly after DNA damage- and oncogene-induced senescence of mouse and human keratinocytes, melanocytes, monocytes, fibroblasts, and epithelial cells [22, 4547]. Another interleukin signaling pathway that is upregulated by senescent cells is IL-1 [48, 49]. Both IL-1α and β are overexpressed and secreted by senescent endothelial cells [50], fibroblasts [51, 52], and chemotherapy-induced senescent epithelial cells [53]. As discussed below, IL-6 and IL-8 expressions depend on the expression and secretion of IL-1α, indicating a hierarchy on how SASP components are regulated.
Most senescent cells overexpress IL-8 (CXCL-8), along with GROα and GROβ (CXCL-1, -2; the murine CXCL-1 is named KC) [16, 46, 54]. Among CCL family members that are generally upregulated in senescent cells are: MCP-2, -4, -1 (CCL-8, -13, -2), HCC-4 (CCL-16), eotaxin-3 (CCL-26), MIP-3α, and -1α (CCL-20,-3). MCP-3 (CCL-7) is overexpressed by senescent liver stellate cells and by prostate and skin fibroblasts. Fibroblasts induced to senesce by oncogenic RAS secrete high levels of MCP-3, as well as I-309 (CCL-1).
The insulin-like growth factor (IGF)/IGFR network may also contribute to the effect senescent cells exert on their microenvironment. Senescent endothelial, epithelial, and fibroblast cells express high levels of almost all the IGF-binding proteins (IGFBPs) including IGFBP-2, -3, -4, -5, and -6 [22, 55, 56] and their regulators, IGFBP-rP1 and IGFBP-rP2 (connective tissue growth factor—CTGF) [57, 58]. There are additional soluble factors associated with the SASP. For example, inflammatory cytokines such as colony stimulating factors (CSFs including granulocyte-macrophage (GM)-CSF and G-CSF) are secreted at high levels by senescent fibroblasts [22].

3.2 Secreted proteases

Aside from soluble signaling cytokines and growth factors, senescent cells also secrete increased levels of some matrix metalloproteinases (MMPs). The MMP family members that are consistently upregulated in human and mouse fibroblasts undergoing replicative or stress-induced senescence are stromelysin-1 and -2 (respectively, MMP-3 and -10) and collagenase-1 (MMP-1) [19, 21, 5961]. In some instances, the MMP-1 and -3 produced by senescent cells [62] can also regulate the activity of the soluble factors present in SASP. For example, these MMPs can cleave MCP-1, -2, and -4 and IL-8 [63].
Another family of proteases involved in carcinogenesis and present in the SASP are serine proteases and regulators of the plasminogen activation pathway: urokinase- or tissue-type plasminogen activators (uPA or tPA), the uPA receptor (uPAR), and inhibitors of these serine proteases (PAI-1 and -2) [64].

3.3 Non-protein secretions

Due to senescence-induced changes in cellular metabolism, senescent cells may exert influences on tissue microenvironments due to the secretion of molecules other than proteins. These molecules include reactive oxygen species (ROS) and transported ions. For example, senescent cells have been shown to release nitric oxide and ROS due to alterations in inducible nitric oxide synthase, endothelial nitric oxide synthase, and superoxide-dismutase activities [6569]. These molecules can enhance cancer cell aggressiveness, as well as promote aging and age-related degeneration [70, 71].

4 Regulation of the inflammatory component of the SASP

Overall, the gene expression profiles (mRNA) of senescent cells determined by transcript analyses resemble the profiles of secreted proteins determined by antibody arrays [22, 23]. This finding suggests that the secretory phenotype of senescent cells is at least in part regulated at the transcriptional level. However, because the changes in gene expression that occurs at senescence are so widespread, the transcriptional control may well be at the level of chromatin organization, rather than changes in specific transcription factors. In fact, dramatic chromatin alterations are known to occur at senescence [7275]. Support for the idea that gene expression specific to the senescence program may be partially attributed to larger changes in chromatin conformation is suggested by the physical clustering of genes that comprise the SASP [23].
The expression of many SASP components depends on the transcription factors NF-κB and C/EBPβ, which have increased activity at senescence [47, 76] (A. Freund and J. Campisi, unpublished data). Knockdown of C/EBPβ diminishes the expression of both IL-6 and IL-8, which are among the most upregulated cytokines at senescence [47], and NF-κB knockdown significantly decreases the levels of numerous (75%) SASP factors (A. Freund and J. Campisi, unpublished data).
A surprising recent finding was that the DNA damage response (DDR) is required for the increased secretion of a subset of SASP factors, including IL-6 and IL-8 [77]. The DDR is a signal amplification cascade that senses DNA damage, inducing cell cycle arrest and initiating DNA damage repair. If the extent of DNA damage is irreparable, the cell undergoes cellular senescence but maintains a chronic, low level DDR [78]. It is this persistent DDR that is necessary for a robust SASP; depletion of upstream components of the DDR cascade, specifically ATM, NBS1, or CHK2, prevents an increase in secreted protein levels of IL-6, IL-8, and the GRO family, among others.
However, the DDR cannot be the sole regulator of the SASP because the DDR is activated immediately after damage, whereas the SASP, like other aspects of the senescence phenotype such as SA-βgal activity, takes several days to develop. Additionally, a transient DDR—for example, caused by a low level of ionizing radiation that does not induce senescence—does not induce a SASP [77]. Thus, while the DDR is important, it is not sufficient; there must be other, slower events that cooperate with the DDR to induce the SASP. One such event is p38MAPK activation, which increases slowly after DNA damage, reaching peak levels after several days (A. Freund and J. Campisi, unpublished data).
Like many cytokine networks, the SASP also has an important positive-feedback component. IL-1α, a cytokine that regulates its own synthesis in an autocrine, receptor-mediated, positive-feedback loop via NF-κB [79, 80], is a key positive regulator of IL-6 and IL-8 expression at senescence [81]. IL-1α depletion by RNAi in senescent cells markedly reduced the extracellular protein levels of IL-6 and IL-8. Similar results were achieved using an IL-1 receptor (IL-1R) antagonist or neutralizing antibodies, demonstrating that sustained IL-1R stimulation by surface-bound IL-1α is required to maintain senescence-associated IL-6 and IL-8 extracellular levels [81].
MicroRNAs also play a role in SASP regulation. Thus far, two microRNAs, miR-146a and miR-146b (miR-146a/b), have been demonstrated to negatively regulate the senescence-associated secretion of IL-6 and IL-8 [82]. Senescent human fibroblasts with a strong SASP upregulate these microRNAs late in senescence, tuning down the secretion of inflammatory cytokines. IRAK1 is an established target of miR-146a/b and is indeed targeted in senescent cells, suggesting that these microRNAs downregulate the SASP by reducing NF-κB activity [83]. When miR-146a/b are overexpressed in human fibroblasts, IRAK1 levels decline, along with the enhanced secretion of senescence-associated IL-6/IL-8. In addition, blockage of IL-1 receptor signaling prevented upregulation of miR-146a/b, consistent with these microRNAs being part of an NF-κB feedback loop [83]. These data once again highlight the important role of NF-κB, and place miR146a/b as central players of IL-6 and IL-8 secretion within the SASP.
Although the SASP is at least partly regulated by the activation of transcription factors, the general gene expression profile acquired at senescence may be more attributable to the larger changes in chromatin conformation that senescent cells develop [7274, 84]. Consistent with this idea is the physical clustering of SASP genes such as matrix metalloproteinases (MMP-1, MMP-3, MMP-10, and MMP-12) as well as CXCL and CCL family members [23]. Moreover, recent work indicates the key role for the High Mobility Group Box 1 protein (HMGB1) in senescent cells.

5 HMGB1 regulates senescence-associated inflammatory cytokine secretion

HMGB1 is a member of the non-histone, chromatin-binding high mobility group family of proteins [85]. Unlike histones, HMGB1 loosely binds the minor grove of DNA, stabilizing nucleosome formation, thereby influencing the expression of certain genes [8688]. HMGB1 was shown to directly interact with p53, and recombinant HMGB1 protein enhanced p53 DNA binding in vitro [89].
The discovery that necrotic cells, but not apoptotic cells, passively release HMGB1 focused recent studies on the role of extracellular HMGB1 [90]. Initial studies suggested that extracellular recombinant HMGB1 promoted secretion of inflammatory cytokines (TNFα, IL-1β, and IL-6) by macrophages [91]. However, later work revealed that recombinant HMGB1 protein alone exhibited only weak activity, but synergized with the ability of bacterial components like lipopolysaccharides to stimulate inflammatory cytokine secretion [92, 93]. Further, HMGB1 can enhance the pro-inflammatory activity of cytokines [94]. These findings appear in conflict with those showing that anti-HMGB1 antibodies or antagonist reduced inflammation in arthritis models [95]. However, the general consensus now is that HMGB1 alone may not promote inflammation, but rather augment the inflammatory response in certain biological contexts.
While it was not observed that cells cultured with recombinant HMGB1 stimulated cytokine secretion, extracellular HMGB1 may enhance of the effect of the SASP (A. Davalos and J. Campisi, unpublished data). Similar to recent data showing that apoptotic cells release “find-me signals”, which recruit innate immune to clear the dying cells, senescent cells also secrete HMGB1, known to recruit and activate cells from the innate immune system [96]. This notion is consistent with two studies that suggest senescent cells may undergo clearance in vivo [13, 97].
Despite the obvious benefit of recruiting and activating innate immune cells to clear senescence cells, extracellular HMGB1 protein may promote cancer and metastatic progression. Experiments in cell culture showed that HMGB1 stimulated the migration of neuroblastoma cells, which was attenuated by an antibody against HMGB1 [98]. Further, in an in vitro angiogenesis assay, HMGB1 stimulated sprouting of endothelial [99]. When immunocompromised mice were injected with rat glioma cells, followed by injection with anti-RAGE (a HMGB1 receptor) or anti-HMGB1 antibodies, both antibodies individually reduced tumor volume, but maximal reduction occurred with both antibodies [100]. Thus, extracellular HMGB1 is able to provoke both beneficial and deleterious biological consequences. It may act to alert and identify senescent cells that require clearance. Failure to clear all senescent cells could leave sites of persistent secretion of inflammatory molecules.

6 Senescent cells create a permissive microenvironment allowing cancer cells to thrive

Inflammation has been shown to initiate or enhance several age-related diseases, including atherosclerosis, Alzheimer’s, osteoarthritis, and cancer [101103]. Factors secreted by senescent cells can promote tumor development in vivo and malignant phenotypes such as proliferation and invasiveness in cell culture models. These effects have been observed with a number of cell types, including cells derived from breast [18, 19, 21, 22, 104], skin [105], prostate [22, 106], pancreas [107], and oro-pharyngial mucosa [108]. The effects of the complex SASP is, of course, dependent on the tissue context. In the following sections, we present the various behavioral changes cells can undergo when residing in the proximity of senescent cells and discuss how the senescent tissue microenvironment can facilitate tumor initiation and progression (see Fig. 2).

6.1 Effects on cell proliferation

One of the most direct, pro-tumorigenic effects of the SASP is to promote the proliferation of epithelial cells. In the case of breast epithelial cells, senescent human fibroblasts can stimulate the growth of pre-malignant and malignant mammary epithelial cells [18, 21, 22]. Irradiated stromal cells, which are presumed to be senescent, have been shown to perturb the mammary epithelial microenvironment and fuel inappropriate epithelial cell growth in the mammary gland [104, 109]. Furthermore, MMPs secreted by senescent fibroblasts have been shown to be responsible for the higher tumorigenicity of breast epithelial cell xenografts in mice, most likely by allowing mitogenic and chemotactic signals greater access to breast cancer cells [19, 104]. In addition to secreted soluble factors, there is evidence that the matrix laid down by senescent cells can also stimulate mammary epithelial cell growth [18].
Fibroblasts from the human prostate gland that undergo senescence in culture have been shown to create a local tissue environment that favors prostate epithelial cell hyperproliferation, in part owing to amphiregulin secretion [20]. Furthermore, senescent fibroblasts increase the expression of CTGF (or IGFBPr2) [58]. CTGF was shown to promote prostate tumor progression in xenografts and is also expressed by the cancer-associated reactive stroma [110]. Moreover, it was recently determined that senescence induced by irradiation in prostate cancer patients was associated with a significantly increased release of exosome-like microvesicles [111]. This novel secretory phenotype was dependent on the activation of p53.
In the skin, unidentified factors secreted by human fibroblasts were shown to be capable of inducing clonal expansion of keratinocytes [112]. In addition, senescent endometrial fibroblasts promoted anchorage-independent epithelial cell growth, due primarily to IL-1 oversecretion [52]. In the oro-bucal cavity, tobacco-driven senescence of supportive stromal cells was shown to stimulate the hyperplastic growth of epithelial cells and was associated with the loss of E-cadherin, ZO-1, and involucrin as well as with the loss of epithelial integrity [108].
Melanocytic naevi (moles) are often composed of senescent melanocytes, which were induced to senesce by oncogenic mutations in BRAF (V600E mutations) [17]. Only rare cell variants in naevi can evolve into melanoma. Malignant melanocytes express high levels of the CXCR2 receptor [113] and can be stimulated to grow by its ligands GROα [114] or IL-8 [115]. Given that both GROα and IL-8 are part of the core SASP, the senescent microenvironment may therefore stimulate the proliferation of rare pre-malignant cells in naevi and thus drive the development of melanoma.
Endothelial cells can undergo proliferation during angiogenesis, which is stimulated by vascular endothelial growth factor (VEGF), IL-8, I-309, and eotaxin [44, 116, 117], all of which are SASP components. Indeed, senescent cells can stimulate endothelial cell migration in culture and angiogenesis in vivo in mouse xenografts of breast cancer cells. The blood vessel density was significantly higher when tumors developed in the presence of senescent, but not pre-senescent, fibroblasts [44]. RAS-driven tumors are also known to contain significant numbers of senescent cells [118]. These tumors are also highly vascularized [119]. Many of the SASP factors can also affect leukocyte proliferation during the course of cancer development. For example, IL-7 directly promotes lymphocyte proliferation in peripheral tissues, and GM-CSF stimulates myeloid suppressor cells, which are known to have important immunosuppressive functions that affect cancer progression [120].

6.2 Effects on cell migration and invasion

Senescent cells oversecrete an array of chemokines that can create a gradient to promote cell migration and invasion. In pancreatic cancer, hepatocyte growth factor (HGF), and to a lesser degree bFGF, promote cancer cell invasion in culture and potentially can drive cancer dissemination in vivo [107, 121]. In breast cancer, high levels of IL-6 and -8 secreted by senescent fibroblasts are responsible for enhancing the invasiveness of a panel of cancer cell lines in cell culture models [22]. Moreover, the secretion of MMP-2 and -3 by senescent cells can also promote the invasion of multiple epithelial cell types [19, 21, 104, 122, 123]. Other proteases, such as uPA and its regulator (PAI1), are likewise implicated in cancer cell invasion. Senescent stromal cells may promote an epithelial-to-mesenchymal transition (EMT) [22], which is an important phenotypic switch that enables cancer cells to migrate and invade [108]. Thus, senescent cells and the SASP can induce phenotypes in nearby human epithelial cells that are common during the acquisition of an aggressive phenotype in tumors.
In cell culture models, endothelial cells are induced to migrate by factors secreted by senescent fibroblasts [44]. This is in part due to VEGF secretion and chemokine gradients set up by senescent cells [124]. Neo-angiogenesis, which is dependent on endothelial cell motility and invasion, is enhanced in xenograft models containing senescent fibroblasts [44]. Further, it is known that IL-1, which is a SASP component, activates the endothelium and consequently increases the adhesive potential of cancer cells to vessel walls [125]. Thus, senescent cells might promote extravasation of cancer cells to secondary metastatic sites. However, the effects of senescent cells on angiogenesis might be cell-type dependent. For example, senescent keratinocytes oversecrete maspin, which acts as a dominant inhibitor of endothelial cell migration and invasion [126].
Senescent fibroblasts may promote leukocyte recruitment, since they chronically release chemokines [127]. In p53-deficient RAS-driven tumors induced to senesce by reestablishing p53 function [13], innate immune cells were shown to migrate into the vicinity of the senescent tumor area. CSF-1, CXCL-1, or MCP-1 and ICAM-1 transcripts were found to be higher in these senescent tumor masses and may be responsible for the immune response. For example, neutrophils express CXCR-1, -2, and -4 to sense their microenvironment and invade tissues; eosinophils use the broad spectrum receptor CCR-3 to fulfill their function; monocytes use CCR-1, -2, -5, CXCR-4, and CX3CR1 to extravasate and enter peripheral sites where they differentiate; natural killer cells express CCR-2, -5, CXCR-4, CX3CR1, and XCR1; and immature myeloid dendritic cells display CCR-1, -2, -5, -6, and CXCR-4, which facilitate their transport, migration, and function [127130].

6.3 Effects on cell differentiation

Senescent human and mouse fibroblasts disrupt the differentiation of mammary epithelial cells and inhibit the expression of differentiation markers [21, 104]. This activity is due in large measure to the secretion of MMP-3 by the senescent cells. Furthermore, weakly tumorigenic pancreatic [107] and mammary [22] epithelial cells undergo morphologic changes in culture resembling an EMT in the presence of senescent conditioned medium. The effect on mammary epithelial cells is attributable to IL-6 and IL-8 [22], as well as HGF, uPAR, and MMPs [21], which are all capable of disrupting epithelial cell clusters and stimulating de-differentiation in culture and in vivo [131133].
Strikingly, no angiostatic factors have been reported among SASP constituents (for example, IFNγ, TSP-1, MIG, PF4, IP-10, IL-4, IL-12, and endostatin). This contrasts with the largely pro-angiogenic profile of the SASP (IL-8, MCP-1 and -2, GROs, PGE2, VEGF, EGF, CSFs, uPA/tPA, MMPs, FN, and laminin) [134]. Furthermore, there may be an amplifying activation loop since senescent stromal cells secrete MCPs, CSFs, MIPs, GROs, and CXCLs, which in turn recruit inflammatory and immune cells that also secrete pro-angiogenic factors (VEGFs, IL-8, and MMPs). Thus, senescent cells are well poised to support the differentiation of a new vasculature around and within a progressing tumor.

6.4 Effects on tumor immunology

No anti-inflammatory factors (e.g., IFNα, IFNγ, IL-3, IL-5) are significantly secreted by senescent fibroblasts, and some of these factors are even downregulated upon senescence (e.g., IL-2, IL-12). Nonetheless, some reports show that massive amounts of MCP-1 or IL-8, which are prominent components of the SASP, lead to tumor destruction [135, 136]. Senescent fibroblasts might influence the macrophage balance in the tumor environment. Molecules that are implicated in the recruitment and differentiation of circulating monocytes to tumor sites also happen to be overexpressed by senescent fibroblasts [137]. These molecules can lead to an inadequate immune response within the close proximity of senescent cells. Senescent fibroblasts might affect lymphocytic populations infiltrating the tumor. Specific T-cell populations associated with tumor progression (i.e., Th2 and T-reg) respond to inflammatory cytokines that are commonly present in the SASP of fibroblasts.

7 Conclusions

From the above discussion, the SASP may play an important role in senescence, especially in the days immediately after senescence induction, by helping the growth arrest to occur efficiently and the immune system to target senescent cells for clearance. However, senescent cells appear to accumulate in vivo despite clearance, suggesting that either the clearance is not 100% efficient, or the rate of senescence increases with age, outpacing the rate of clearance. Consequently, the SASP’s deleterious, chronic inflammatory effects may only become apparent with time. It is therefore important to understand the pathways that regulate the SASP, to determine how it can be modulated and whether the potentially deleterious effects of the SASP can be inhibited without sacrificing the beneficial effects of senescence.
Most insoluble components of the extracellular matrix (ECM) are enzymatic targets of secreted proteases. Therefore, the senescence-associated changes in proteolytic activities could affect the physical properties of the tissue structure around cells. In particular, the accumulation of senescent cells could lessen the supportive role of the ECM, globally diminishing tissue tension and elasticity. In addition, the relaxed tissue structure and higher levels of MMPs might help tumor cells migrate and invade through the ECM, thus enabling metastasis. In fact, the panel of proteases secreted by senescent cells extensively overlaps with those found in malignant tumors.
Overall, senescence is a molecular program with a unique phenotypic outcome. How its extracellular molecular signature is activated and maintained, and the extent to which it influences the tissue milieu in healthy tissues, aged tissues, and diseased tissues, are some of the many questions that remain unanswered. However, even with our current limited knowledge of the senescent secretory phenotype and its potential effects on carcinogenesis, promising new strategies for cancer therapies emerge. For example, restoring the activity of tumor suppressor genes is an attractive, potentially powerful therapeutic approach. Taking into account our current understanding of the cell’s non-autonomous effects of these tumor suppressor genes, small chemicals that can pharmacologically restore their normal function would help reestablish the proper tissue and cell signals, thereby stimulating cancer regression [138141]. Such approaches would limit inflammation and thus possibly allow proper tissue repair, and they would directly promote the immune-mediated clearance of cells that drive cancer progression.
Chronic inflammation, once established, often takes on a momentum of its own due to the feedback loops of the immune system: cytokines activate leukocytes, which produce more cytokines [142]. Therefore, even a small pro-inflammatory stimulus, such as a population of senescent cells scattered throughout organs and tissues, could be the seed for a more systemic chronic inflammatory response over time, leading to age-related diseases such as cancer.

Acknowledgments

The authors are supported by grants from the National Institutes of Health (AG09909, AG017242, CA12654, AG025708, ES015566, AG025901, and AG032117), the Larry L. Hillblom Foundation, and DOE contract AC03-76SF00098.

Open Access

This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.
Open AccessThis is an open access article distributed under the terms of the Creative Commons Attribution Noncommercial License (https://​creativecommons.​org/​licenses/​by-nc/​2.​0), which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
2.
3.
Zurück zum Zitat Campisi, J. (2001). Cellular senescence as a tumor-suppressor mechanism. Trends in Cell Biology, 11(11), 27–31.CrossRef Campisi, J. (2001). Cellular senescence as a tumor-suppressor mechanism. Trends in Cell Biology, 11(11), 27–31.CrossRef
4.
Zurück zum Zitat Wright, W. E., & Shay, J. W. (2001). Cellular senescence as a tumor-protection mechanism: The essential role of counting. Current Opinion in Genetics and Development, 11, 98–103.PubMedCrossRef Wright, W. E., & Shay, J. W. (2001). Cellular senescence as a tumor-protection mechanism: The essential role of counting. Current Opinion in Genetics and Development, 11, 98–103.PubMedCrossRef
5.
Zurück zum Zitat Ben-Porath, I., & Weinberg, R. A. (2004). When cells get stressed: An integrative view of cellular senescence. Journal of Clinical Investigation, 113, 8–13.PubMed Ben-Porath, I., & Weinberg, R. A. (2004). When cells get stressed: An integrative view of cellular senescence. Journal of Clinical Investigation, 113, 8–13.PubMed
6.
Zurück zum Zitat Collins, C. J., & Sedivy, J. M. (2003). Involvement of the INK4a/Arf gene locus in senescence. Aging Cell, 2, 145–150.PubMedCrossRef Collins, C. J., & Sedivy, J. M. (2003). Involvement of the INK4a/Arf gene locus in senescence. Aging Cell, 2, 145–150.PubMedCrossRef
7.
Zurück zum Zitat Lowe, S. W., & Sherr, C. J. (2003). Tumor suppression by Ink4a-Arf: Progress and puzzles. Current Opinion in Genetics and Development, 13, 77–83.PubMedCrossRef Lowe, S. W., & Sherr, C. J. (2003). Tumor suppression by Ink4a-Arf: Progress and puzzles. Current Opinion in Genetics and Development, 13, 77–83.PubMedCrossRef
8.
Zurück zum Zitat Ohtani, N., et al. (2004). The p16INK4a-RB pathway: Molecular link between cellular senescence and tumor suppression. Journal of Medical Investigation, 51, 146–153.PubMedCrossRef Ohtani, N., et al. (2004). The p16INK4a-RB pathway: Molecular link between cellular senescence and tumor suppression. Journal of Medical Investigation, 51, 146–153.PubMedCrossRef
9.
Zurück zum Zitat Gil, J., & Peters, G. (2006). Regulation of the INK4b–ARF–INK4a tumour suppressor locus: All for one or one for all. Nature Reviews. Molecular Cell Biology, 7, 667–677.PubMedCrossRef Gil, J., & Peters, G. (2006). Regulation of the INK4b–ARF–INK4a tumour suppressor locus: All for one or one for all. Nature Reviews. Molecular Cell Biology, 7, 667–677.PubMedCrossRef
10.
Zurück zum Zitat Braig, M., & Schmitt, C. A. (2006). Oncogene-induced senescence: Putting the brakes on tumor development. Cancer Research, 66, 2881–2884.PubMedCrossRef Braig, M., & Schmitt, C. A. (2006). Oncogene-induced senescence: Putting the brakes on tumor development. Cancer Research, 66, 2881–2884.PubMedCrossRef
11.
Zurück zum Zitat Shay, J. W., & Roninson, I. B. (2004). Hallmarks of senescence in carcinogenesis and cancer therapy. Oncogene, 23, 2919–2933.PubMedCrossRef Shay, J. W., & Roninson, I. B. (2004). Hallmarks of senescence in carcinogenesis and cancer therapy. Oncogene, 23, 2919–2933.PubMedCrossRef
12.
Zurück zum Zitat Ventura, A., et al. (2007). Restoration of p53 function leads to tumour regression in vivo. Nature, 445, 661–665.PubMedCrossRef Ventura, A., et al. (2007). Restoration of p53 function leads to tumour regression in vivo. Nature, 445, 661–665.PubMedCrossRef
13.
Zurück zum Zitat Xue, W., et al. (2007). Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature, 445, 656–660.PubMedCrossRef Xue, W., et al. (2007). Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature, 445, 656–660.PubMedCrossRef
14.
Zurück zum Zitat Campisi, J. (2005). Senescent cells, tumor suppression and organismal aging: Good citizens, bad neighbors. Cell, 120, 513–522.PubMedCrossRef Campisi, J. (2005). Senescent cells, tumor suppression and organismal aging: Good citizens, bad neighbors. Cell, 120, 513–522.PubMedCrossRef
15.
Zurück zum Zitat Jeyapalan, J. C., et al. (2007). Accumulation of senescent cells in mitotic tissue of aging primates. Mechanisms of Ageing and Development, 128, 36–44.PubMedCrossRef Jeyapalan, J. C., et al. (2007). Accumulation of senescent cells in mitotic tissue of aging primates. Mechanisms of Ageing and Development, 128, 36–44.PubMedCrossRef
16.
Zurück zum Zitat Castro, P., et al. (2003). Cellular senescence in the pathogenesis of benign prostatic hyperplasia. Prostate, 55, 30–38.PubMedCrossRef Castro, P., et al. (2003). Cellular senescence in the pathogenesis of benign prostatic hyperplasia. Prostate, 55, 30–38.PubMedCrossRef
17.
Zurück zum Zitat Michaloglou, C., et al. (2005). BRAFE600-associated senescence-like cell cycle arrest of human nevi. Nature, 436, 720–724.PubMedCrossRef Michaloglou, C., et al. (2005). BRAFE600-associated senescence-like cell cycle arrest of human nevi. Nature, 436, 720–724.PubMedCrossRef
18.
Zurück zum Zitat Krtolica, A., et al. (2001). Senescent fibroblasts promote epithelial cell growth and tumorigenesis: A link between cancer and aging. Proceedings of the National Academy of Sciences of the United States of America, 98, 12072–12077.PubMedCrossRef Krtolica, A., et al. (2001). Senescent fibroblasts promote epithelial cell growth and tumorigenesis: A link between cancer and aging. Proceedings of the National Academy of Sciences of the United States of America, 98, 12072–12077.PubMedCrossRef
19.
Zurück zum Zitat Liu, D., & Hornsby, P. J. (2007). Senescent human fibroblasts increase the early growth of xenograft tumors via matrix metalloproteinase secretion. Cancer Research, 67, 3117–3126.PubMedCrossRef Liu, D., & Hornsby, P. J. (2007). Senescent human fibroblasts increase the early growth of xenograft tumors via matrix metalloproteinase secretion. Cancer Research, 67, 3117–3126.PubMedCrossRef
20.
Zurück zum Zitat Bavik, C., et al. (2006). The gene expression program of prostate fibroblast senescence modulates neoplastic epithelial cell proliferation through paracrine mechanisms. Cancer Research, 66, 794–802.PubMedCrossRef Bavik, C., et al. (2006). The gene expression program of prostate fibroblast senescence modulates neoplastic epithelial cell proliferation through paracrine mechanisms. Cancer Research, 66, 794–802.PubMedCrossRef
21.
Zurück zum Zitat Parrinello, S., et al. (2005). Stromal–epithelial interactions in aging and cancer: Senescent fibroblasts alter epithelial cell differentiation. Journal of Cell Science, 118(Pt 3), 485–496.PubMedCrossRef Parrinello, S., et al. (2005). Stromal–epithelial interactions in aging and cancer: Senescent fibroblasts alter epithelial cell differentiation. Journal of Cell Science, 118(Pt 3), 485–496.PubMedCrossRef
22.
Zurück zum Zitat Coppe, J. P., et al. (2008). Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biology, 6(12), 2853–2868.PubMedCrossRef Coppe, J. P., et al. (2008). Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biology, 6(12), 2853–2868.PubMedCrossRef
23.
Zurück zum Zitat Coppe, J. P., et al. (2010). A human-like senescence-associated secretory phenotype is conserved in mouse cells dependent on physiological oxygen. PLoS ONE, 5(2), e9188.PubMedCrossRef Coppe, J. P., et al. (2010). A human-like senescence-associated secretory phenotype is conserved in mouse cells dependent on physiological oxygen. PLoS ONE, 5(2), e9188.PubMedCrossRef
24.
Zurück zum Zitat Campisi, J., & d'Adda di Fagagna, F. (2007). Cellular senescence: When bad things happen to good cells. Nature Reviews. Molecular Cell Biology, 8, 729–740.PubMedCrossRef Campisi, J., & d'Adda di Fagagna, F. (2007). Cellular senescence: When bad things happen to good cells. Nature Reviews. Molecular Cell Biology, 8, 729–740.PubMedCrossRef
25.
26.
Zurück zum Zitat Hayflick, L. (1965). The limited in vitro lifetime of human diploid cell strains. Experimental Cell Research, 37, 614–636.PubMedCrossRef Hayflick, L. (1965). The limited in vitro lifetime of human diploid cell strains. Experimental Cell Research, 37, 614–636.PubMedCrossRef
27.
Zurück zum Zitat d'Adda di Fagagna, F., et al. (2003). A DNA damage checkpoint response in telomere-initiated senescence. Nature, 426, 194–198.PubMedCrossRef d'Adda di Fagagna, F., et al. (2003). A DNA damage checkpoint response in telomere-initiated senescence. Nature, 426, 194–198.PubMedCrossRef
28.
Zurück zum Zitat Dimri, G. P., et al. (1995). A novel biomarker identifies senescent human cells in culture and in aging skin in vivo. Proceedings of the National Academy of Sciences of the United States of America, 92, 9363–9367.PubMedCrossRef Dimri, G. P., et al. (1995). A novel biomarker identifies senescent human cells in culture and in aging skin in vivo. Proceedings of the National Academy of Sciences of the United States of America, 92, 9363–9367.PubMedCrossRef
29.
Zurück zum Zitat Ben-Porath, I., & Weinberg, R. A. (2005). The signals and pathways activating cellular senescence. International Journal of Biochemistry and Cell Biology, 37(5), 961–976.PubMedCrossRef Ben-Porath, I., & Weinberg, R. A. (2005). The signals and pathways activating cellular senescence. International Journal of Biochemistry and Cell Biology, 37(5), 961–976.PubMedCrossRef
30.
Zurück zum Zitat Schmitt, C. A. (2003). Senescence, apoptosis and therapy—Cutting the lifelines of cancer. \Nature Reviews. Cancer, 3(4), 286–295.PubMedCrossRef Schmitt, C. A. (2003). Senescence, apoptosis and therapy—Cutting the lifelines of cancer. \Nature Reviews. Cancer, 3(4), 286–295.PubMedCrossRef
31.
Zurück zum Zitat Martin, G. M. (2005). Genetic modulation of senescent phenotypes in Homo sapiens. Cell, 120, 523–532.PubMedCrossRef Martin, G. M. (2005). Genetic modulation of senescent phenotypes in Homo sapiens. Cell, 120, 523–532.PubMedCrossRef
32.
Zurück zum Zitat Chien, K. R., & Karsenty, G. (2005). Longevity and lineages: Toward the integrative biology of degenerative diseases in heart, muscle, and bone. Cell, 120(4), 533–544.PubMedCrossRef Chien, K. R., & Karsenty, G. (2005). Longevity and lineages: Toward the integrative biology of degenerative diseases in heart, muscle, and bone. Cell, 120(4), 533–544.PubMedCrossRef
33.
Zurück zum Zitat Lombard, D. B., et al. (2005). DNA repair, genome stability, and aging. Cell, 120, 497–512.PubMedCrossRef Lombard, D. B., et al. (2005). DNA repair, genome stability, and aging. Cell, 120, 497–512.PubMedCrossRef
34.
Zurück zum Zitat Balaban, R. S., Nemoto, S., & Finkel, T. (2005). Mitochondria, oxidants, and aging. Cell, 120, 483–495.PubMedCrossRef Balaban, R. S., Nemoto, S., & Finkel, T. (2005). Mitochondria, oxidants, and aging. Cell, 120, 483–495.PubMedCrossRef
35.
Zurück zum Zitat Collado, M., & Serrano, M. (2006). The power and the promise of oncogene-induced senescence markers. Nature Reviews. Cancer, 6, 472–476.PubMedCrossRef Collado, M., & Serrano, M. (2006). The power and the promise of oncogene-induced senescence markers. Nature Reviews. Cancer, 6, 472–476.PubMedCrossRef
36.
Zurück zum Zitat Hill, R., et al. (2005). Selective evolution of stromal mesenchyme with p53 loss in response to epithelial tumorigenesis. Cell, 123, 1001–1011.PubMedCrossRef Hill, R., et al. (2005). Selective evolution of stromal mesenchyme with p53 loss in response to epithelial tumorigenesis. Cell, 123, 1001–1011.PubMedCrossRef
37.
Zurück zum Zitat Najjar, S. S., Scuteri, A., & Lakatta, E. G. (2005). Arterial aging: Is it an immutable cardiovascular risk factor? Hypertension, 46(3), 454–462.PubMedCrossRef Najjar, S. S., Scuteri, A., & Lakatta, E. G. (2005). Arterial aging: Is it an immutable cardiovascular risk factor? Hypertension, 46(3), 454–462.PubMedCrossRef
38.
Zurück zum Zitat Kunieda, T., et al. (2006). Angiotensin II induces premature senescence of vascular smooth muscle cells and accelerates the development of atherosclerosis via a p21-dependent pathway. Circulation, 114(9), 953–960.PubMedCrossRef Kunieda, T., et al. (2006). Angiotensin II induces premature senescence of vascular smooth muscle cells and accelerates the development of atherosclerosis via a p21-dependent pathway. Circulation, 114(9), 953–960.PubMedCrossRef
39.
Zurück zum Zitat Krishnamurthy, J., et al. (2004). Ink4a/Arf expression is a biomarker of aging. Journal of Clinical Investigation, 114, 1299–1307.PubMed Krishnamurthy, J., et al. (2004). Ink4a/Arf expression is a biomarker of aging. Journal of Clinical Investigation, 114, 1299–1307.PubMed
40.
Zurück zum Zitat Herbig, U., et al. (2004). Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21(CIP1), but not p16(INK4a). Molecular Cell, 14(4), 501–513.PubMedCrossRef Herbig, U., et al. (2004). Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21(CIP1), but not p16(INK4a). Molecular Cell, 14(4), 501–513.PubMedCrossRef
41.
Zurück zum Zitat Narita, M., et al. (2003). Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell, 113, 703–716.PubMedCrossRef Narita, M., et al. (2003). Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell, 113, 703–716.PubMedCrossRef
42.
Zurück zum Zitat Collado, M., & Serrano, M. (2010). Senescence in tumours: Evidence from mice and humans. Nature Reviews. Cancer, 10(1), 51–57.PubMedCrossRef Collado, M., & Serrano, M. (2010). Senescence in tumours: Evidence from mice and humans. Nature Reviews. Cancer, 10(1), 51–57.PubMedCrossRef
43.
Zurück zum Zitat Coppe, J. P., et al. (2010). The senescence-associated secretory phenotype: The dark side of tumor suppression. Annual Review of Pathology: Mechanisms of Disease, 5, 99–118.CrossRef Coppe, J. P., et al. (2010). The senescence-associated secretory phenotype: The dark side of tumor suppression. Annual Review of Pathology: Mechanisms of Disease, 5, 99–118.CrossRef
44.
Zurück zum Zitat Coppe, J. P., et al. (2006). Secretion of vascular endothelial growth factor by primary human fibroblasts at senescence. Journal of Biological Chemistry, 281(40), 29568–29574.PubMedCrossRef Coppe, J. P., et al. (2006). Secretion of vascular endothelial growth factor by primary human fibroblasts at senescence. Journal of Biological Chemistry, 281(40), 29568–29574.PubMedCrossRef
45.
Zurück zum Zitat Lu, S. Y., et al. (2006). Ripe areca nut extract induces G1 phase arrests and senescence-associated phenotypes in normal human oral keratinocyte. Carcinogenesis, 27(6), 1273–1284.PubMedCrossRef Lu, S. Y., et al. (2006). Ripe areca nut extract induces G1 phase arrests and senescence-associated phenotypes in normal human oral keratinocyte. Carcinogenesis, 27(6), 1273–1284.PubMedCrossRef
46.
Zurück zum Zitat Sarkar, D., et al. (2004). Human polynucleotide phosphorylase (hPNPaseold-35): A potential link between aging and inflammation. Cancer Research, 64(20), 7473–7478.PubMedCrossRef Sarkar, D., et al. (2004). Human polynucleotide phosphorylase (hPNPaseold-35): A potential link between aging and inflammation. Cancer Research, 64(20), 7473–7478.PubMedCrossRef
47.
Zurück zum Zitat Kuilman, T., et al. (2008). Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell, 133, 1019–1031.PubMedCrossRef Kuilman, T., et al. (2008). Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell, 133, 1019–1031.PubMedCrossRef
48.
Zurück zum Zitat Garfinkel, S., et al. (1994). Post-transcriptional regulation of interleukin 1 alpha in various strains of young and senescent human umbilical vein endothelial cells. Proceedings of the National Academy of Sciences of the United States of America, 91(4), 1559–1563.PubMedCrossRef Garfinkel, S., et al. (1994). Post-transcriptional regulation of interleukin 1 alpha in various strains of young and senescent human umbilical vein endothelial cells. Proceedings of the National Academy of Sciences of the United States of America, 91(4), 1559–1563.PubMedCrossRef
49.
Zurück zum Zitat McLachlan, J. A., et al. (1995). Immunological functions of aged human monocytes. Pathobiology, 63(3), 148–159.PubMedCrossRef McLachlan, J. A., et al. (1995). Immunological functions of aged human monocytes. Pathobiology, 63(3), 148–159.PubMedCrossRef
50.
Zurück zum Zitat Maier, J. A. M., et al. (1990). Extension of the life-span of human endothelial cells by an interleukin-1a antisense oligomer. Science, 249, 1570–1574.PubMedCrossRef Maier, J. A. M., et al. (1990). Extension of the life-span of human endothelial cells by an interleukin-1a antisense oligomer. Science, 249, 1570–1574.PubMedCrossRef
51.
Zurück zum Zitat Kumar, S., Millis, A. J., & Baglioni, C. (1992). Expression of interleukin 1-inducible genes and production of interleukin 1 by aging human fibroblasts. Proceedings of the National Academy of Sciences of the United States America, 89(10), 4683–4687.CrossRef Kumar, S., Millis, A. J., & Baglioni, C. (1992). Expression of interleukin 1-inducible genes and production of interleukin 1 by aging human fibroblasts. Proceedings of the National Academy of Sciences of the United States America, 89(10), 4683–4687.CrossRef
52.
Zurück zum Zitat Palmieri, D., Watson, J. M., & Rinehart, C. A. (1999). Age-related expression of PEDF/EPC-1 in human endometrial stromal fibroblasts: Implications for interactive senescence. Experimental Cell Research, 247(1), 142–147.PubMedCrossRef Palmieri, D., Watson, J. M., & Rinehart, C. A. (1999). Age-related expression of PEDF/EPC-1 in human endometrial stromal fibroblasts: Implications for interactive senescence. Experimental Cell Research, 247(1), 142–147.PubMedCrossRef
53.
Zurück zum Zitat Chang, B. D., et al. (2002). Molecular determinants of terminal growth arrest induced in tumor cells by a chemotherapeutic agent. Proceedings of the National Academy of Sciences of the United States of America, 99, 389–394.PubMedCrossRef Chang, B. D., et al. (2002). Molecular determinants of terminal growth arrest induced in tumor cells by a chemotherapeutic agent. Proceedings of the National Academy of Sciences of the United States of America, 99, 389–394.PubMedCrossRef
54.
Zurück zum Zitat Bode-Boger, S. M., Scalera, F., & Martens-Lobenhoffer, J. (2005). Asymmetric dimethylarginine (ADMA) accelerates cell senescence. Vascular Medicine, 10(Suppl 1), S65–S71.PubMedCrossRef Bode-Boger, S. M., Scalera, F., & Martens-Lobenhoffer, J. (2005). Asymmetric dimethylarginine (ADMA) accelerates cell senescence. Vascular Medicine, 10(Suppl 1), S65–S71.PubMedCrossRef
55.
Zurück zum Zitat Wang, S., et al. (1996). Characterization of IGFBP-3, PAI-1 and SPARC mRNA expression in senescent fibroblasts. Mechanisms of Ageing and Development, 92(2–3), 121–132.PubMedCrossRef Wang, S., et al. (1996). Characterization of IGFBP-3, PAI-1 and SPARC mRNA expression in senescent fibroblasts. Mechanisms of Ageing and Development, 92(2–3), 121–132.PubMedCrossRef
56.
Zurück zum Zitat Grillari, J., et al. (2000). Subtractive hybridization of mRNA from early passage and senescent endothelial cells. Experimental Gerontology, 35(2), 187–197.PubMedCrossRef Grillari, J., et al. (2000). Subtractive hybridization of mRNA from early passage and senescent endothelial cells. Experimental Gerontology, 35(2), 187–197.PubMedCrossRef
57.
Zurück zum Zitat Lopez-Bermejo, A., et al. (2000). Characterization of insulin-like growth factor-binding protein-related proteins (IGFBP-rPs) 1, 2, and 3 in human prostate epithelial cells: Potential roles for IGFBP-rP1 and 2 in senescence of the prostatic epithelium. Endocrinology, 141(11), 4072–4080.PubMedCrossRef Lopez-Bermejo, A., et al. (2000). Characterization of insulin-like growth factor-binding protein-related proteins (IGFBP-rPs) 1, 2, and 3 in human prostate epithelial cells: Potential roles for IGFBP-rP1 and 2 in senescence of the prostatic epithelium. Endocrinology, 141(11), 4072–4080.PubMedCrossRef
58.
Zurück zum Zitat Kim, K. H., et al. (2004). Expression of connective tissue growth factor, a biomarker in senescence of human diploid fibroblasts, is up-regulated by a transforming growth factor-beta-mediated signaling pathway. Biochemical and Biophysical Research Communications, 318(4), 819–825.PubMedCrossRef Kim, K. H., et al. (2004). Expression of connective tissue growth factor, a biomarker in senescence of human diploid fibroblasts, is up-regulated by a transforming growth factor-beta-mediated signaling pathway. Biochemical and Biophysical Research Communications, 318(4), 819–825.PubMedCrossRef
59.
Zurück zum Zitat West, M. D., Pereira-Smith, O. M., & Smith, J. R. (1989). Replicative senescence of human skin fibroblasts correlates with a loss of regulation and overexpression of collagenase activity. Experimental Cell Research, 184, 138–147.PubMedCrossRef West, M. D., Pereira-Smith, O. M., & Smith, J. R. (1989). Replicative senescence of human skin fibroblasts correlates with a loss of regulation and overexpression of collagenase activity. Experimental Cell Research, 184, 138–147.PubMedCrossRef
60.
Zurück zum Zitat Millis, A. J. T., et al. (1992). Differential expression of metalloproteinase and tissue inhibitor of metalloproteinase genes in diploid human fibroblasts. Experimental Cell Research, 201, 373–379.PubMedCrossRef Millis, A. J. T., et al. (1992). Differential expression of metalloproteinase and tissue inhibitor of metalloproteinase genes in diploid human fibroblasts. Experimental Cell Research, 201, 373–379.PubMedCrossRef
61.
Zurück zum Zitat Zeng, G., & Millis, A. J. (1996). Differential regulation of collagenase and stromelysin mRNA in late passage cultures of human fibroblasts. Experimental Cell Research, 222(1), 150–156.PubMedCrossRef Zeng, G., & Millis, A. J. (1996). Differential regulation of collagenase and stromelysin mRNA in late passage cultures of human fibroblasts. Experimental Cell Research, 222(1), 150–156.PubMedCrossRef
62.
Zurück zum Zitat Hornebeck, W., & Maquart, F. X. (2003). Proteolyzed matrix as a template for the regulation of tumor progression. Biomedicine and Pharmacotherapy, 57(5–6), 223–230.CrossRef Hornebeck, W., & Maquart, F. X. (2003). Proteolyzed matrix as a template for the regulation of tumor progression. Biomedicine and Pharmacotherapy, 57(5–6), 223–230.CrossRef
63.
Zurück zum Zitat McQuibban, G. A., et al. (2002). Matrix metalloproteinase processing of monocyte chemoattractant proteins generates CC chemokine receptor antagonists with anti-inflammatory properties in vivo. Blood, 100(4), 1160–1167.PubMed McQuibban, G. A., et al. (2002). Matrix metalloproteinase processing of monocyte chemoattractant proteins generates CC chemokine receptor antagonists with anti-inflammatory properties in vivo. Blood, 100(4), 1160–1167.PubMed
64.
Zurück zum Zitat Blasi, F., & Carmeliet, P. (2002). uPAR: A versatile signalling orchestrator. Nature Reviews. Molecular Cell Biology, 3(12), 932–943.PubMedCrossRef Blasi, F., & Carmeliet, P. (2002). uPAR: A versatile signalling orchestrator. Nature Reviews. Molecular Cell Biology, 3(12), 932–943.PubMedCrossRef
65.
Zurück zum Zitat Sato, I., et al. (1993). Reduction of nitric oxide producing activity associated with in vitro aging in cultured human umbilical vein endothelial cell. Biochemical and Biophysical Research Communications, 195(2), 1070–1076.PubMedCrossRef Sato, I., et al. (1993). Reduction of nitric oxide producing activity associated with in vitro aging in cultured human umbilical vein endothelial cell. Biochemical and Biophysical Research Communications, 195(2), 1070–1076.PubMedCrossRef
66.
Zurück zum Zitat Lee, A. C., et al. (1999). Ras proteins induce senescence by altering the intracellular levels of reactive oxygen species. Journal of Biological Chemistry, 274(12), 7936–7940.PubMedCrossRef Lee, A. C., et al. (1999). Ras proteins induce senescence by altering the intracellular levels of reactive oxygen species. Journal of Biological Chemistry, 274(12), 7936–7940.PubMedCrossRef
67.
Zurück zum Zitat van der Loo, B., et al. (2000). Enhanced peroxynitrite formation is associated with vascular aging. Journal of Experimental Medicine, 192(12), 1731–1744.PubMedCrossRef van der Loo, B., et al. (2000). Enhanced peroxynitrite formation is associated with vascular aging. Journal of Experimental Medicine, 192(12), 1731–1744.PubMedCrossRef
68.
Zurück zum Zitat Macip, S., et al. (2002). Inhibition of p21-mediated ROS accumulation can rescue p21-induced senescence. EMBO Journal, 21, 2180–2188.PubMedCrossRef Macip, S., et al. (2002). Inhibition of p21-mediated ROS accumulation can rescue p21-induced senescence. EMBO Journal, 21, 2180–2188.PubMedCrossRef
69.
Zurück zum Zitat Xin, M. G., et al. (2003). Senescence-enhanced oxidative stress is associated with deficiency of mitochondrial cytochrome c oxidase in vascular endothelial cells. Mechanisms of Ageing and Development, 124(8–9), 911–919.PubMedCrossRef Xin, M. G., et al. (2003). Senescence-enhanced oxidative stress is associated with deficiency of mitochondrial cytochrome c oxidase in vascular endothelial cells. Mechanisms of Ageing and Development, 124(8–9), 911–919.PubMedCrossRef
70.
Zurück zum Zitat Finkel, T., & Holbrook, N. J. (2000). Oxidants, oxidative stress and the biology of ageing. Nature, 408, 239–247.PubMedCrossRef Finkel, T., & Holbrook, N. J. (2000). Oxidants, oxidative stress and the biology of ageing. Nature, 408, 239–247.PubMedCrossRef
71.
Zurück zum Zitat Finkel, T., Serrano, M., & Blasco, M. A. (2007). The common biology of cancer and ageing. Nature, 448, 767–774.PubMedCrossRef Finkel, T., Serrano, M., & Blasco, M. A. (2007). The common biology of cancer and ageing. Nature, 448, 767–774.PubMedCrossRef
72.
Zurück zum Zitat Funayama, R., & Ishikawa, F. (2007). Cellular senescence and chromatin structure. Chromosoma, 116(5), 431–440.PubMedCrossRef Funayama, R., & Ishikawa, F. (2007). Cellular senescence and chromatin structure. Chromosoma, 116(5), 431–440.PubMedCrossRef
73.
Zurück zum Zitat Mehta, I. S., et al. (2007). Alterations to nuclear architecture and genome behavior in senescent cells. Annals of the New York Academy of Sciences, 1100, 250–263.PubMedCrossRef Mehta, I. S., et al. (2007). Alterations to nuclear architecture and genome behavior in senescent cells. Annals of the New York Academy of Sciences, 1100, 250–263.PubMedCrossRef
74.
Zurück zum Zitat Narita, M. (2007). Cellular senescence and chromatin organisation. British Journal of Cancer, 96(5), 686–691.PubMedCrossRef Narita, M. (2007). Cellular senescence and chromatin organisation. British Journal of Cancer, 96(5), 686–691.PubMedCrossRef
75.
76.
Zurück zum Zitat Acosta, J. C., et al. (2008). Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell, 133(6), 1006–1018.PubMedCrossRef Acosta, J. C., et al. (2008). Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell, 133(6), 1006–1018.PubMedCrossRef
77.
Zurück zum Zitat Rodier, F., et al. (2009). Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nature Cell Biology, 11(8), 973–979.PubMedCrossRef Rodier, F., et al. (2009). Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nature Cell Biology, 11(8), 973–979.PubMedCrossRef
78.
Zurück zum Zitat d'Adda di Fagagna, F. (2008). Living on a break: Cellular senescence as a DNA-damage response. Nature Reviews. Cancer, 8(7), 512–22.PubMedCrossRef d'Adda di Fagagna, F. (2008). Living on a break: Cellular senescence as a DNA-damage response. Nature Reviews. Cancer, 8(7), 512–22.PubMedCrossRef
79.
Zurück zum Zitat Hiscott, J., et al. (1993). Characterization of a functional NF-kappa B site in the human interleukin 1 beta promoter: Evidence for a positive autoregulatory loop. Molecular and Cellular Biology, 13(10), 6231–6240.PubMed Hiscott, J., et al. (1993). Characterization of a functional NF-kappa B site in the human interleukin 1 beta promoter: Evidence for a positive autoregulatory loop. Molecular and Cellular Biology, 13(10), 6231–6240.PubMed
80.
Zurück zum Zitat Niu, J., et al. (2004). Identification of an autoregulatory feedback pathway involving interleukin-1alpha in induction of constitutive NF-kappaB activation in pancreatic cancer cells. Journal of Biological Chemistry, 279(16), 16452–16462.PubMedCrossRef Niu, J., et al. (2004). Identification of an autoregulatory feedback pathway involving interleukin-1alpha in induction of constitutive NF-kappaB activation in pancreatic cancer cells. Journal of Biological Chemistry, 279(16), 16452–16462.PubMedCrossRef
81.
Zurück zum Zitat Orjalo, A. V., et al. (2009). Cell surface-bound IL-1alpha is an upstream regulator of the senescence-associated IL-6/IL-8 cytokine network. Proceedings of the National Academy of Sciences of the United States of America, 106(40), 17031–17036.PubMedCrossRef Orjalo, A. V., et al. (2009). Cell surface-bound IL-1alpha is an upstream regulator of the senescence-associated IL-6/IL-8 cytokine network. Proceedings of the National Academy of Sciences of the United States of America, 106(40), 17031–17036.PubMedCrossRef
82.
Zurück zum Zitat Bhaumik, D., et al. (2009). MicroRNAs miR-146a/b negatively modulate the senescence-associated inflammatory mediators IL-6 and IL-8. Aging, 1(4), 402–411.PubMed Bhaumik, D., et al. (2009). MicroRNAs miR-146a/b negatively modulate the senescence-associated inflammatory mediators IL-6 and IL-8. Aging, 1(4), 402–411.PubMed
83.
Zurück zum Zitat Taganov, K. D., et al. (2006). NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proceedings of the National Academy of Sciences of the United States of America, 103(33), 12481–12486.PubMedCrossRef Taganov, K. D., et al. (2006). NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proceedings of the National Academy of Sciences of the United States of America, 103(33), 12481–12486.PubMedCrossRef
84.
Zurück zum Zitat Adams, P. D. (2007). Remodeling of chromatin structure in senescent cells and its potential impact on tumor suppression and aging. Gene, 397(1–2), 84–93.PubMedCrossRef Adams, P. D. (2007). Remodeling of chromatin structure in senescent cells and its potential impact on tumor suppression and aging. Gene, 397(1–2), 84–93.PubMedCrossRef
85.
Zurück zum Zitat Bianchi, M. E., & Agresti, A. (2005). HMG proteins: Dynamic players in gene regulation and differentiation. Current Opinion in Genetics and Development, 15(5), 496–506.PubMedCrossRef Bianchi, M. E., & Agresti, A. (2005). HMG proteins: Dynamic players in gene regulation and differentiation. Current Opinion in Genetics and Development, 15(5), 496–506.PubMedCrossRef
86.
Zurück zum Zitat Lotze, M. T., & Tracey, K. J. (2005). High-mobility group box 1 protein (HMGB1): Nuclear weapon in the immune arsenal. Nature Reviews. Immunology, 5(4), 331–342.PubMedCrossRef Lotze, M. T., & Tracey, K. J. (2005). High-mobility group box 1 protein (HMGB1): Nuclear weapon in the immune arsenal. Nature Reviews. Immunology, 5(4), 331–342.PubMedCrossRef
87.
Zurück zum Zitat Park, J. S., et al. (2003). Activation of gene expression in human neutrophils by high mobility group box 1 protein. American Journal of Physiology Cell Physiology, 284(4), C870–C879.PubMed Park, J. S., et al. (2003). Activation of gene expression in human neutrophils by high mobility group box 1 protein. American Journal of Physiology Cell Physiology, 284(4), C870–C879.PubMed
88.
Zurück zum Zitat Stros, M., et al. (2004). High-affinity binding of tumor-suppressor protein p53 and HMGB1 to hemicatenated DNA loops. Biochemistry, 43(22), 7215–7225.PubMedCrossRef Stros, M., et al. (2004). High-affinity binding of tumor-suppressor protein p53 and HMGB1 to hemicatenated DNA loops. Biochemistry, 43(22), 7215–7225.PubMedCrossRef
89.
Zurück zum Zitat Jayaraman, L., et al. (1998). High mobility group protein-1 (HMG-1) is a unique activator of p53. Genes and Development, 12(4), 462–472.PubMedCrossRef Jayaraman, L., et al. (1998). High mobility group protein-1 (HMG-1) is a unique activator of p53. Genes and Development, 12(4), 462–472.PubMedCrossRef
90.
Zurück zum Zitat Scaffidi, P., Misteli, T., & Bianchi, M. E. (2002). Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature, 418(6894), 191–195.PubMedCrossRef Scaffidi, P., Misteli, T., & Bianchi, M. E. (2002). Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature, 418(6894), 191–195.PubMedCrossRef
91.
Zurück zum Zitat Kokkola, R., et al. (2005). RAGE is the major receptor for the proinflammatory activity of HMGB1 in rodent macrophages. Scandinavian Journal of Immunology, 61(1), 1–9.PubMedCrossRef Kokkola, R., et al. (2005). RAGE is the major receptor for the proinflammatory activity of HMGB1 in rodent macrophages. Scandinavian Journal of Immunology, 61(1), 1–9.PubMedCrossRef
92.
Zurück zum Zitat Rouhiainen, A., et al. (2007). Pivotal advance: Analysis of proinflammatory activity of highly purified eukaryotic recombinant HMGB1 (amphoterin). Journal of Leukocyte Biology, 81(1), 49–58.PubMedCrossRef Rouhiainen, A., et al. (2007). Pivotal advance: Analysis of proinflammatory activity of highly purified eukaryotic recombinant HMGB1 (amphoterin). Journal of Leukocyte Biology, 81(1), 49–58.PubMedCrossRef
93.
Zurück zum Zitat Bianchi, M.E., (2009). HMGB1 loves company. J Leukoc Biol. Bianchi, M.E., (2009). HMGB1 loves company. J Leukoc Biol.
94.
Zurück zum Zitat Sha, Y., et al. (2008). HMGB1 develops enhanced proinflammatory activity by binding to cytokines. Journal of Immunology, 180(4), 2531–2537. Sha, Y., et al. (2008). HMGB1 develops enhanced proinflammatory activity by binding to cytokines. Journal of Immunology, 180(4), 2531–2537.
95.
Zurück zum Zitat Kokkola, R., et al. (2003). Successful treatment of collagen-induced arthritis in mice and rats by targeting extracellular high mobility group box chromosomal protein 1 activity. Arthritis and Rheumatism, 48(7), 2052–2058.PubMedCrossRef Kokkola, R., et al. (2003). Successful treatment of collagen-induced arthritis in mice and rats by targeting extracellular high mobility group box chromosomal protein 1 activity. Arthritis and Rheumatism, 48(7), 2052–2058.PubMedCrossRef
96.
Zurück zum Zitat Elliott, M. R., et al. (2009). Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature, 461(7261), 282–286.PubMedCrossRef Elliott, M. R., et al. (2009). Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature, 461(7261), 282–286.PubMedCrossRef
97.
Zurück zum Zitat Krizhanovsky, V., et al. (2008). Senescence of activated stellate cells limits liver fibrosis. Cell, 134(4), 657–667.PubMedCrossRef Krizhanovsky, V., et al. (2008). Senescence of activated stellate cells limits liver fibrosis. Cell, 134(4), 657–667.PubMedCrossRef
98.
Zurück zum Zitat Fages, C., et al. (2000). Regulation of cell migration by amphoterin. Journal of Cell Science, 113(Pt 4), 611–620.PubMed Fages, C., et al. (2000). Regulation of cell migration by amphoterin. Journal of Cell Science, 113(Pt 4), 611–620.PubMed
99.
Zurück zum Zitat Schlueter, C., et al. (2005). Angiogenetic signaling through hypoxia: HMGB1: An angiogenetic switch molecule. American Journal of Pathology, 166(4), 1259–1263.PubMed Schlueter, C., et al. (2005). Angiogenetic signaling through hypoxia: HMGB1: An angiogenetic switch molecule. American Journal of Pathology, 166(4), 1259–1263.PubMed
100.
Zurück zum Zitat Taguchi, A., et al. (2000). Blockade of RAGE-amphoterin signalling suppresses tumour growth and metastases. Nature, 405(6784), 354–360.PubMedCrossRef Taguchi, A., et al. (2000). Blockade of RAGE-amphoterin signalling suppresses tumour growth and metastases. Nature, 405(6784), 354–360.PubMedCrossRef
101.
Zurück zum Zitat Brennan, F. M., Maini, R. N., & Feldmann, M. (1995). Cytokine expression in chronic inflammatory disease. British Medical Bulletin, 51(2), 368–384.PubMed Brennan, F. M., Maini, R. N., & Feldmann, M. (1995). Cytokine expression in chronic inflammatory disease. British Medical Bulletin, 51(2), 368–384.PubMed
102.
Zurück zum Zitat Brod, S. A. (2000). Unregulated inflammation shortens human functional longevity. Inflammation Research, 49(11), 561–570.PubMedCrossRef Brod, S. A. (2000). Unregulated inflammation shortens human functional longevity. Inflammation Research, 49(11), 561–570.PubMedCrossRef
103.
Zurück zum Zitat Caruso, C., et al. (2004). Aging, longevity, inflammation, and cancer. Annals of the New York Academy of Sciences, 1028, 1–13.PubMedCrossRef Caruso, C., et al. (2004). Aging, longevity, inflammation, and cancer. Annals of the New York Academy of Sciences, 1028, 1–13.PubMedCrossRef
104.
Zurück zum Zitat Tsai, K. K., et al. (2005). Cellular mechanisms for low-dose ionizing radiation-induced perturbation of the breast tissue microenvironment. Cancer Research, 65, 6734–6744.PubMedCrossRef Tsai, K. K., et al. (2005). Cellular mechanisms for low-dose ionizing radiation-induced perturbation of the breast tissue microenvironment. Cancer Research, 65, 6734–6744.PubMedCrossRef
105.
Zurück zum Zitat Sun, P., et al. (2007). PRAK is essential for ras-induced senescence and tumor suppression. Cell, 128, 295–308.PubMedCrossRef Sun, P., et al. (2007). PRAK is essential for ras-induced senescence and tumor suppression. Cell, 128, 295–308.PubMedCrossRef
106.
Zurück zum Zitat Choi, J., et al. (2000). Expression of senescence-associated beta-galactosidase in enlarged prostates from men with benign prostatic hyperplasia. Urology, 56, 160–166.PubMedCrossRef Choi, J., et al. (2000). Expression of senescence-associated beta-galactosidase in enlarged prostates from men with benign prostatic hyperplasia. Urology, 56, 160–166.PubMedCrossRef
107.
Zurück zum Zitat Ohuchida, K., et al. (2004). Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor–stromal interactions. Cancer Research, 64(9), 3215–3222.PubMedCrossRef Ohuchida, K., et al. (2004). Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor–stromal interactions. Cancer Research, 64(9), 3215–3222.PubMedCrossRef
108.
Zurück zum Zitat Coppe, J. P., et al. (2008). A role for fibroblasts in mediating the effects of tobacco-induced epithelial cell growth and invasion. Molecular Cancer Research, 6(7), 1085–1098.PubMedCrossRef Coppe, J. P., et al. (2008). A role for fibroblasts in mediating the effects of tobacco-induced epithelial cell growth and invasion. Molecular Cancer Research, 6(7), 1085–1098.PubMedCrossRef
109.
Zurück zum Zitat Barcellos-Hoff, M. H., & Ravani, S. A. (2000). Irradiated mammary gland stroma promotes the expression of tumorigenic potential by unirradiated epithelial cells. Cancer Research, 60, 1254–1260.PubMed Barcellos-Hoff, M. H., & Ravani, S. A. (2000). Irradiated mammary gland stroma promotes the expression of tumorigenic potential by unirradiated epithelial cells. Cancer Research, 60, 1254–1260.PubMed
110.
Zurück zum Zitat Yang, F., et al. (2005). Stromal expression of connective tissue growth factor promotes angiogenesis and prostate cancer tumorigenesis. Cancer Research, 65(19), 8887–8895.PubMedCrossRef Yang, F., et al. (2005). Stromal expression of connective tissue growth factor promotes angiogenesis and prostate cancer tumorigenesis. Cancer Research, 65(19), 8887–8895.PubMedCrossRef
111.
Zurück zum Zitat Lehmann, B. D., et al. (2008). Senescence-associated exosome release from human prostate cancer cells. Cancer Research, 68, 7864–7871.PubMedCrossRef Lehmann, B. D., et al. (2008). Senescence-associated exosome release from human prostate cancer cells. Cancer Research, 68, 7864–7871.PubMedCrossRef
112.
Zurück zum Zitat Dilley, T. K., Bowden, G. T., & Chen, Q. M. (2003). Novel mechanisms of sublethal oxidant toxicity: Induction of premature senescence in human fibroblasts confers tumor promoter activity. Experimental Cell Research, 290, 38–48.PubMedCrossRef Dilley, T. K., Bowden, G. T., & Chen, Q. M. (2003). Novel mechanisms of sublethal oxidant toxicity: Induction of premature senescence in human fibroblasts confers tumor promoter activity. Experimental Cell Research, 290, 38–48.PubMedCrossRef
113.
Zurück zum Zitat Dhawan, P., & Richmond, A. (2002). Role of CXCL1 in tumorigenesis of melanoma. Journal of Leukocyte Biology, 72(1), 9–18.PubMed Dhawan, P., & Richmond, A. (2002). Role of CXCL1 in tumorigenesis of melanoma. Journal of Leukocyte Biology, 72(1), 9–18.PubMed
114.
Zurück zum Zitat Balentien, E., et al. (1991). Effects of MGSA/GRO alpha on melanocyte transformation. Oncogene, 6(7), 1115–1124.PubMed Balentien, E., et al. (1991). Effects of MGSA/GRO alpha on melanocyte transformation. Oncogene, 6(7), 1115–1124.PubMed
115.
Zurück zum Zitat Schadendorf, D., et al. (1993). IL-8 produced by human malignant melanoma cells in vitro is an essential autocrine growth factor. Journal of Immunology, 151(5), 2667–2675. Schadendorf, D., et al. (1993). IL-8 produced by human malignant melanoma cells in vitro is an essential autocrine growth factor. Journal of Immunology, 151(5), 2667–2675.
116.
Zurück zum Zitat Bernardini, G., et al. (2000). I-309 binds to and activates endothelial cell functions and acts as an angiogenic molecule in vivo. Blood, 96(13), 4039–4045.PubMed Bernardini, G., et al. (2000). I-309 binds to and activates endothelial cell functions and acts as an angiogenic molecule in vivo. Blood, 96(13), 4039–4045.PubMed
117.
Zurück zum Zitat Salcedo, R., et al. (2001). Eotaxin (CCL11) induces in vivo angiogenic responses by human CCR3+ endothelial cells. Journal of Immunology, 166(12), 7571–7578. Salcedo, R., et al. (2001). Eotaxin (CCL11) induces in vivo angiogenic responses by human CCR3+ endothelial cells. Journal of Immunology, 166(12), 7571–7578.
118.
Zurück zum Zitat Collado, M., et al. (2005). Tumor biology: Senescent in premalignant tumours. Nature, 436, 642.PubMedCrossRef Collado, M., et al. (2005). Tumor biology: Senescent in premalignant tumours. Nature, 436, 642.PubMedCrossRef
119.
Zurück zum Zitat Sparmann, A., & Bar-Sagi, D. (2004). Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis. Cancer Cell, 6(5), 447–458.PubMedCrossRef Sparmann, A., & Bar-Sagi, D. (2004). Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis. Cancer Cell, 6(5), 447–458.PubMedCrossRef
120.
Zurück zum Zitat Frey, A. B. (2006). Myeloid suppressor cells regulate the adaptive immune response to cancer. Journal of Clinical Investigation, 116(10), 2587–2590.PubMedCrossRef Frey, A. B. (2006). Myeloid suppressor cells regulate the adaptive immune response to cancer. Journal of Clinical Investigation, 116(10), 2587–2590.PubMedCrossRef
121.
Zurück zum Zitat Birchmeier, C., et al. (2003). Met, metastasis, motility and more. Nature Reviews. Molecular Cell Biology, 4(12), 915–925.PubMedCrossRef Birchmeier, C., et al. (2003). Met, metastasis, motility and more. Nature Reviews. Molecular Cell Biology, 4(12), 915–925.PubMedCrossRef
122.
Zurück zum Zitat Camphausen, K., et al. (2001). Radiation therapy to a primary tumor accelerates metastatic growth in mice. Cancer Research, 61(5), 2207–2211.PubMed Camphausen, K., et al. (2001). Radiation therapy to a primary tumor accelerates metastatic growth in mice. Cancer Research, 61(5), 2207–2211.PubMed
123.
Zurück zum Zitat Qian, L. W., et al. (2002). Radiation-induced increase in invasive potential of human pancreatic cancer cells and its blockade by a matrix metalloproteinase inhibitor, CGS27023. Clinical Cancer Research, 8(4), 1223–1227.PubMed Qian, L. W., et al. (2002). Radiation-induced increase in invasive potential of human pancreatic cancer cells and its blockade by a matrix metalloproteinase inhibitor, CGS27023. Clinical Cancer Research, 8(4), 1223–1227.PubMed
124.
Zurück zum Zitat Strieter, R. M., et al. (2006). Cancer CXC chemokine networks and tumour angiogenesis. European Journal of Cancer, 42(6), 768–778.PubMedCrossRef Strieter, R. M., et al. (2006). Cancer CXC chemokine networks and tumour angiogenesis. European Journal of Cancer, 42(6), 768–778.PubMedCrossRef
125.
Zurück zum Zitat Orr, F. W., & Wang, H. H. (2001). Tumor cell interactions with the microvasculature: A rate-limiting step in metastasis. Surgical Oncology Clinics of North America, 10(2), 357–81. ix−x.PubMed Orr, F. W., & Wang, H. H. (2001). Tumor cell interactions with the microvasculature: A rate-limiting step in metastasis. Surgical Oncology Clinics of North America, 10(2), 357–81. ix−x.PubMed
126.
Zurück zum Zitat Nickoloff, B. J., et al. (2004). Tumor suppressor maspin is up-regulated during keratinocyte senescence, exerting a paracrine antiangiogenic activity. Cancer Research, 64(9), 2956–2961.PubMedCrossRef Nickoloff, B. J., et al. (2004). Tumor suppressor maspin is up-regulated during keratinocyte senescence, exerting a paracrine antiangiogenic activity. Cancer Research, 64(9), 2956–2961.PubMedCrossRef
127.
Zurück zum Zitat Mantovani, A. (2004). Chemokines in neoplastic progression. Seminars in Cancer Biology, 14(3), 147–148.PubMedCrossRef Mantovani, A. (2004). Chemokines in neoplastic progression. Seminars in Cancer Biology, 14(3), 147–148.PubMedCrossRef
128.
Zurück zum Zitat Homey, B., Muller, A., & Zlotnik, A. (2002). Chemokines: Agents for the immunotherapy of cancer? Nature Reviews. Immunology, 2(3), 175–184.PubMedCrossRef Homey, B., Muller, A., & Zlotnik, A. (2002). Chemokines: Agents for the immunotherapy of cancer? Nature Reviews. Immunology, 2(3), 175–184.PubMedCrossRef
129.
Zurück zum Zitat Balkwill, F. (2004). Cancer and the chemokine network. Nature Reviews. Cancer, 4(7), 540–550.PubMedCrossRef Balkwill, F. (2004). Cancer and the chemokine network. Nature Reviews. Cancer, 4(7), 540–550.PubMedCrossRef
130.
Zurück zum Zitat Ben-Baruch, A. (2006). Inflammation-associated immune suppression in cancer: The roles played by cytokines, chemokines and additional mediators. Seminars in Cancer Biology, 16(1), 38–52.PubMedCrossRef Ben-Baruch, A. (2006). Inflammation-associated immune suppression in cancer: The roles played by cytokines, chemokines and additional mediators. Seminars in Cancer Biology, 16(1), 38–52.PubMedCrossRef
131.
Zurück zum Zitat Potempa, S., & Ridley, A. J. (1998). Activation of both MAP kinase and phosphatidylinositide 3-kinase by Ras is required for hepatocyte growth factor/scatter factor-induced adherens junction disassembly. Molecular Biology of the Cell, 9(8), 2185–2200.PubMed Potempa, S., & Ridley, A. J. (1998). Activation of both MAP kinase and phosphatidylinositide 3-kinase by Ras is required for hepatocyte growth factor/scatter factor-induced adherens junction disassembly. Molecular Biology of the Cell, 9(8), 2185–2200.PubMed
132.
Zurück zum Zitat Paumelle, R., et al. (2002). Hepatocyte growth factor/scatter factor activates the ETS1 transcription factor by a RAS–RAF–MEK–ERK signaling pathway. Oncogene, 21(15), 2309–2319.PubMedCrossRef Paumelle, R., et al. (2002). Hepatocyte growth factor/scatter factor activates the ETS1 transcription factor by a RAS–RAF–MEK–ERK signaling pathway. Oncogene, 21(15), 2309–2319.PubMedCrossRef
133.
Zurück zum Zitat Thiery, J. P. (2002). Epithelial–mesenchymal transitions in tumour progression. Nature Reviews. Cancer, 2(6), 442–454.PubMedCrossRef Thiery, J. P. (2002). Epithelial–mesenchymal transitions in tumour progression. Nature Reviews. Cancer, 2(6), 442–454.PubMedCrossRef
134.
Zurück zum Zitat Tonini, T., Rossi, F., & Claudio, P. P. (2003). Molecular basis of angiogenesis and cancer. Oncogene, 22(42), 6549–6556.PubMedCrossRef Tonini, T., Rossi, F., & Claudio, P. P. (2003). Molecular basis of angiogenesis and cancer. Oncogene, 22(42), 6549–6556.PubMedCrossRef
135.
Zurück zum Zitat Nesbit, M., et al. (2001). Low-level monocyte chemoattractant protein-1 stimulation of monocytes leads to tumor formation in nontumorigenic melanoma cells. Journal of Immunology, 166(11), 6483–6490. Nesbit, M., et al. (2001). Low-level monocyte chemoattractant protein-1 stimulation of monocytes leads to tumor formation in nontumorigenic melanoma cells. Journal of Immunology, 166(11), 6483–6490.
136.
Zurück zum Zitat Schaider, H., et al. (2003). Differential response of primary and metastatic melanomas to neutrophils attracted by IL-8. International Journal of Cancer, 103(3), 335–343.CrossRef Schaider, H., et al. (2003). Differential response of primary and metastatic melanomas to neutrophils attracted by IL-8. International Journal of Cancer, 103(3), 335–343.CrossRef
137.
Zurück zum Zitat Sica, A., et al. (2006). Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: Potential targets of anti-cancer therapy. European Journal of Cancer, 42(6), 717–727.PubMedCrossRef Sica, A., et al. (2006). Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: Potential targets of anti-cancer therapy. European Journal of Cancer, 42(6), 717–727.PubMedCrossRef
138.
Zurück zum Zitat Selivanova, G., et al. (1997). Restoration of the growth suppression function of mutant p53 by a synthetic peptide derived from the p53 C-terminal domain. Nature Medicine, 3(6), 632–638.PubMedCrossRef Selivanova, G., et al. (1997). Restoration of the growth suppression function of mutant p53 by a synthetic peptide derived from the p53 C-terminal domain. Nature Medicine, 3(6), 632–638.PubMedCrossRef
139.
Zurück zum Zitat Foster, B. A., et al. (1999). Pharmacological rescue of mutant p53 conformation and function. Science, 286(5449), 2507–2510.PubMedCrossRef Foster, B. A., et al. (1999). Pharmacological rescue of mutant p53 conformation and function. Science, 286(5449), 2507–2510.PubMedCrossRef
140.
Zurück zum Zitat Chene, P. (2003). Inhibiting the p53–MDM2 interaction: An important target for cancer therapy. Nature Reviews. Cancer, 3(2), 102–109.PubMedCrossRef Chene, P. (2003). Inhibiting the p53–MDM2 interaction: An important target for cancer therapy. Nature Reviews. Cancer, 3(2), 102–109.PubMedCrossRef
141.
Zurück zum Zitat Selivanova, G., & Wiman, K. G. (2007). Reactivation of mutant p53: Molecular mechanisms and therapeutic potential. Oncogene, 26(15), 2243–2254.PubMedCrossRef Selivanova, G., & Wiman, K. G. (2007). Reactivation of mutant p53: Molecular mechanisms and therapeutic potential. Oncogene, 26(15), 2243–2254.PubMedCrossRef
142.
Zurück zum Zitat Barnes, P. J., & Karin, M. (1997). Nuclear factor-kappaB: A pivotal transcription factor in chronic inflammatory diseases. New England Journal of Medicine, 336(15), 1066–1071.PubMedCrossRef Barnes, P. J., & Karin, M. (1997). Nuclear factor-kappaB: A pivotal transcription factor in chronic inflammatory diseases. New England Journal of Medicine, 336(15), 1066–1071.PubMedCrossRef
Metadaten
Titel
Senescent cells as a source of inflammatory factors for tumor progression
verfasst von
Albert R. Davalos
Jean-Philippe Coppe
Judith Campisi
Pierre-Yves Desprez
Publikationsdatum
01.06.2010
Verlag
Springer US
Erschienen in
Cancer and Metastasis Reviews / Ausgabe 2/2010
Print ISSN: 0167-7659
Elektronische ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-010-9220-9

Weitere Artikel der Ausgabe 2/2010

Cancer and Metastasis Reviews 2/2010 Zur Ausgabe

EditorialNotes

Preface

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Perioperative Checkpointhemmer-Therapie verbessert NSCLC-Prognose

28.05.2024 NSCLC Nachrichten

Eine perioperative Therapie mit Nivolumab reduziert das Risiko für Rezidive und Todesfälle bei operablem NSCLC im Vergleich zu einer alleinigen neoadjuvanten Chemotherapie um über 40%. Darauf deuten die Resultate der Phase-3-Studie CheckMate 77T.

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.