Influence of FOX genes on aging and aging-associated diseases

Text
Read preview
Mark as finished
How to read the book after purchase
Influence of FOX genes on aging and aging-associated diseases
Font:Smaller АаLarger Aa

Elena Tschumak

INFLUENCE OF FOX GENES ON AGING AND AGING-ASSOCIATED DISEASES

Inhaltsverzeichnis

FOX GENES AFFECT AGING AND RELATED DISEASES

FOXP2 genes have a direct and indirect effect on aging

Influence of other members of Fox-family on aging processes

FOX genes and anti-aging agents

FOXP2 and neuroplasticity

FOXP2 and neurodegeneration

FOXP2 and Alzheimer

Further aging and neurodegeneration relevant FOXP2 targets

Structure and function of FoxP genes is responsible for his function

Natural products and FOXP Targets

Post-stroke FOXP2 expression and discussion on appropriate animal models to study FOXP2 language function

Further literature:

FOX GENES AFFECT AGING AND RELATED DISEASES

FOXP2 genes have a direct and indirect effect on aging

FOX genes include 19 highly conserved, structurally related families from FoxA to FoxS (Sridhar Hannenhalli and Klaus H. Kaestner, 2009). Different publications mentioned the role of FOXgenes in aging processes.

One of the main factors of aging is telomere attrition. Telomeres modulate Rb and p53, that decrease PGC-1α and PGC-1β expression (Sahin and Depinho, 2012) and activates the cyclin-dependent kinase inhibitor p21CIP1.16 H. Shelterin (consists of Tin2, Rap1, TRF1, TRF2, Pot1-TPP1 heterodimer) protects telomers and play a role in mtDNA damage. Telomeric heterochromatin is influenced by lncRNA TERRA via binding to TRF1, TRF2,ORC, HP1 and H3K9me3(Scaffidi et al., 2005; Han and Brunet, 2011).

Tanabe et al. described 2011 in „FOXP2 Promotes the Nuclear Translocation of POT1, but FOXP2 (R553H), Mutation Related to Speech-Language Disorder, Partially Prevents It“, how

FOXP2 influences on POT1 (protection of telomeres 1) as a FOXP2-associated protein. (Tanabe et al. reported protection of nuclear-colocated telomerase.) This gene POT1 as member of the telombin family encodes a nuclear protein involved in telomere maintenance. The loss of its POT1 function induces the cell arrest. The researchers identified that POT1 is alone localized in the cytoplasm but co-localized with FOXP2 and the forkhead domain of FOXP2 in nuclei. FOXP2 with mutated nuclear localization signals as well as R553H mutated forkhead, which is associated with speech-language disorder, prevented the nuclear translocation of POT1. The authors propose FOXP2 as a binding partner for the nuclear translocation of POT1.

According to Multanii and Chang, 2007 „WRN at telomeres: implications for aging and cancer“

POT1 is critically important for telomere extension by telomerase, POT1 functions to negatively regulate telomere length by competing with telomerase for access to the telomeric substrate.

Review Wohlgemuth et al, 2014 showed that FOXP2 also provided the NMDAR-mediated neuronal plasticity affecting MAPKK. It is known that Aging also influences free radicals via Fc-gamma receptor and phagocytosis via p42/p44 MAPK signalling pathways and aging relevant ROS activate inflammatory cascade reaction via JAK/STAT, NF-κB/MAPK.

Molina-Serrano et al. showed 2019 in „Histone Modifications as an Intersection Between Diet and Longevity“ that Lysine 4 on histone H3 can be found in one of three possible methylated forms: mono, di or trimethylated (H3K4me1, -2, and -3).The trimethylated form usually localizes at the promoter of actively transcribed genes (Santos-Rosa et al., 2002), and has been shown to have a strong implication with aging in several model organisms. S. cerevisiae H3K4 methyltransferase complex COMPASS mutants had significantly reduced lifespan (Smith et al., 2008; Ryu et al., 2014; Cruz et al., 2018). “H3K9me3 was only associated with antioxidant genes in females (Strakovsky et al., 2014). Paternal HF diet changed the epigenome in spermatozoa and offspring liver. Specifically, under HF diet, H3K4me was enriched in paternal sperm around the transcriptional start site (TSS) of genes involved in development regulatory processes such as Hoxd11, Hoxd13, Bai3, Foxp2, and Foxa2. In contrast, in the offspring liver, H3K4me was enriched in genes controlling lipid biosynthesis, fatty acid synthesis and the oxidation-reduction process (Terashima et al., 2015).Aging relevant H3K27ac and H3K14ac are acetylated via p300/CBP and its co-activator CREB. cAMP responsive CREB expression is responsible to fasting. So CBP, CREB, CRTC2 and TAF-4 activate together gluconeogenesis genes (Altarejos and Montminy, 2011)” (Molina-Serrano et al., 2019, p.11)

FOXP2 indirectly regulates the FEZF2, the RELN, the FOXO1 and the DYRK1A genes via theRUNX-AUTS2-TBR1cascade In a feedback loop the FEZF2 regulates FOXP2 expression. (Molyneaux et al., 2005) TBR1 also regulates RELN expression (Chen et al., 2002). The RELN in turn regulates FOXO1 expression, which is also indirectly regulated by RUNX2 (Kuhlwilm et al., 2013). It would be of great scientific interest to investigate further whether and to what extent FOXP2 expression via FOXO interaction can influence tumorigenesis and antiaging, as FOXO genes are generally known for their importance in cell cycle and apoptosis. (Kops et al., 2002)

Gascoyne et al. suggested 2015 in „The Forkhead Transcription Factor FOXP2 Is Required for Regulation of p21WAF1/CIP1 in 143B Osteosarcoma Cell Growth Arrest“ that “FOXP2 expression could be induced by MAPK pathway inhibition in growth-arrested 143B cells, but not in traditional cell line models of osteoblast differentiation (MG-63, C2C12, MC3T3-E1)” (Gascoyne et al., 2015, p.1) They studied a model in which transient upregulation of Foxp2 in pre-osteoblast mesenchymal cells regulates a p21-dependent growth arrest checkpoint and identified that Foxp2 expression is not neuronally restricted and is linked to regulation of the cell cycle via various mechanisms: p53 target gene cyclin-dependent kinase inhibitor p21, is as a primary controller of multiple diverse cellular pathways, e.g. neuronal differentiation, growth arrest of pre-osteoblast type 143B osteosarcoma cells etc. They reported that regarding the RUNX-2-dependent pathway for FOXP the bone-specific isoform of RUNX2, which these cells lack, is not a requirement for FOXP2 function and in “developing murine mesenchymal cells the loss of growth factor signalling upon entry to the periosteum might up-regulate Foxp2. Expression of Foxp2 during both osteoblast and some neuronal development appears to be transient and this is also observed for other family members”(Gascoyne et al., 2015, p.13), considered potential connections between FOXP2 and p53 pathways. “Normal osteoblast development is compromised in bone metastases of solid tumours and in bone malignancies such as multiple myeloma, and this characterisation of FOXP2 growth arrest function in a disease context may identify novel malignant pathways. Furthermore, FOXP2 status in osteosarcoma may provide information regarding the stage of developmental block, with potential clinical significance. Direct connection of FOXP2 to the mutated p53 pathway in osteosarcoma, via a common target p21/CDKN1A, is also likely to have implications for understanding osteosarcoma biology.” (Gascoyne et al., 2015, p.14)

Also the review of Wohlgemuth et al., 2014 showed that FOXP2 also provided the NMDAR-mediated neuronal plasticity affecting MAPKK and tyrosine phosphatase. The MARK is a RAS Downstream Effector. RAS is a GTPase that indirectly interacts with oncological relevant p21 and p53 indirectly. So FOXP2 can act alone or in combination with other members of its or other gene families. Its own expression can be influenced both by the cooperation partners and by the feedback loops or by other interactions. These innumerable tissue-specific regulation possibilities allow a fine-tuning and a rapid adaptation to ever-changing environmental conditions which need to be studied more closely.

Influence of other members of Fox-family on aging processes

According to Ni et al. 2012, Ribarič 2012 Foxo plays an important epigenetic role in aging and Hansen et al, 2013 describe DAF/FOXO effect on longevity. Steroid hormone dehydrogenases, cytochrome P450s and others aging relevant steroid hormone signalling pathways are influenced not only by LIPL-4 and TOR regulated autophagy but also by daf-16/FOXO (Lapierre et al., 2011).In C. Elegans daf-16/FOXO affects aging and tumor growth. (Pinkston-Gosse and Kenyon, 2007) Experiments with germline-less C. Elegans showed that FOXO3A effects NAD-dependent protein deacetylases and ADP-ribosyl transferases activity of aging relevant sirtuin (Oberdoerffer et al., 2008; Wang et al., 2008) e.g. with the help of histone H3K9 deacetylation, glucose homeostasis, genomic stability and via subunit RelA NF-κB regulating SIRT6 (Kanfi et al., 2010; Kawahara et al., 2009; Zhong et al., 2010). SIRT6 also effects telomers. (Michishita et al., 2008; Mostoslavsky et al., 2006; Tennen & Chua, 2011) AMPK and SIRT1 also directly influence PGC-1α via deacetylation and phosphorylation). Aging is associated with upregulation of chaperones and proteases level via misfolded proteins also appeared by Alzheimer’s and Parkinson’s disease,(Bernales et al., 2012) and UPRmt. UPRmt as well as FOXO signalling can be activated by NAD+ and increase longevity (Mouchiroud et al., 2013) Lon protease is another important for degradation of oxidized proteins within the mitochondrial matrix aging factor (Ngo et al., 2013, 2009; Bota et al, 2002)

 

FOXO3 (like mTOR and AMPK) effects aging relevant mitophagy (Rodriguez-Hernandez et al., 2009). Kim et al.,2014 describe how phosphorylation, acetylation, or ubiquitination of FoxO-genes effect histones and chromatin and change this way ROS-level. Therefore FoxO4 shows negativ effect on PI3K/Akt pathways (Karger et al. 2009) as well as on MAFbx and MURF1 in muscle aging (Clavel et al. 2006). Oxidative stress leads to FoxO phosphorylation via Akt and its transport from the nucleus to the cytoplasm. FoxO3 and FoxO4 deacetylation by Sirt1 (Jian et al. 2011) and its upregulation via catalase (Fukuoka et al. 2003; Brunet et al. 1999) play an important role in aging. FoxO1 also regulates apoptosis, DNA repair and cell cycle arrest (Yamaza et al.2010; McLoughlin, 2009; Ma et al., 2016), but also plays a key role in stem cell pluripotency. (Zhang et al., 2011). FoxO6 effects gastric carcinoma (Kim et al. 2011) and together with PGC-1a influences oxidative metabolism in skeletal muscle. Chung et al.,2013 reported that FoxO6 and PGC-1a form a regulatory loop to regulate oxidative metabolism in skeletal muscle. In addition, Foxoa2 plays a potential role in sepsis (Berg et al., 2006) and in asthmatic mucus secretion (Park et al., 2009).

An interesting aging approach (connected with adjacent to Paneth cells and crypts) are stem cells and their regulation e.g. via cancer relevant Lrig1, Wnt, Olfm4, Hopx, p57, Sox9, Tert, Bmi1, β-catenin, Ascl2, Lgr5, Myc, Ephb2, CD44, PPAR and SMAD signalling (Nalapareddy et al., 2017; Akunuru, and Geiger, 2016) Deficiencies in DNA damage repair limits the function of haematopoietic stem cells with age. (Rossi et al., 2007) Low ISC and increased ROS levels relate to Foxo1, Foxo3a and Foxo4 activity. (Tothova et al., 2007; Miyamoto et al., 2007)

But FOXO like CoQ and PGC-1α also influences electron transport changes contribute to aging effects such as cardiac failure. (Rosca et al., 2008; Frenzel et al., 2010; López-Lluch et al.,2010; Houtkooper et al., 2010) Shijin et al. mentioned 2016 in „An Update on inflammaging: Mechanisms, Prevention, and Treatment“ the influence of inflammatory cytokines on lymphocytes. Type I and type II cytokines effect CD4+ T lymphocytes (Alberti et al., 2006) and CD8+ and CD4+ T lymphocytes (Franceschi et al., 2000). Especially role on imflamaging play IL-1, IL-6, TNF-α and PGE2 (Bruunsgaard et al., 2003; Cesari et al., 2004). Aging is associated with IGF-1 down- and IL-6- and TNF-α- upregulation (Lio et al., 2003) IL-1, IL-6, and IL-8 are activated via NF-κB signalling pathway (Bartek et al., 2008).

Different publications mentioned that FOX genes are an important factor in the development of tumors (Nishimura et al.,1998; Wang et al., 2015; Tian et al., 2015; Lo et al., 2016,2018; Lu et al., 2017; Nik et al., 2013; Shi et al., 2016; Yu et al., 2017; Dou et al., 2017; Milewski et al., 2017; Cai et al., 2015; Xian et al., 2018; Herrero and Gitton, 2018; Toma et al., 2011; Zhang et al., 2012; Rousso et al., 2012; Howarth et al., 2008; Teufel et al., 2003; Li and Tucker, 1993; Ji et al., 2016; Myatt et al., 2007) The FOXO family and the transcription factor NF-kB influence the upstream protein kinase B e.g. in connection with the receptors of the Trk family (e.g. the Neurotrophin). Inhibition of NF-κB signalling, NLRP3 inflammasome and other pro-inflammatory pathways or hormonal treatments eventually can restore aging relevant Progerin level, elevated by telomeres disfunction. (Osorio et al., 2010; Cao et al., 2011) In the same time senescence is accompanied by increased IL-1ß level and activated tumor necrosis factor and interferons production. (Review López-Otín, 2013; Green et al., 2011; Salminen et al., 2012, 2019; Adler et al.,2007). The Hsp90 and the IκB-Kinase (IKK) inactivate the NF-κB pathway and inhibit autophagy via induction a cell signalling switch from autophagy to apoptosis in tumor cells downregulation and of Beclin 1 expression. (Jiang et al, 2011) NF-κB also reduces GnRH production (Zhang et al., 2013) and affects this way muscle weakness, reduced neurogenesis, bone fragility and skin atrophy. Further NF-κB decreases H3K27me3 level via H3K27me3 demethylase (Lauren et al., 2016; De Santa et al., 2007). BCL2, transcriptionally regulated by nuclear factor-kappa B, affects cell shrinkage (Chakraborty et al., 2015; Lambie1 and Conradt, 2016) and acts directly on apoptosis-activating BH3. Reqmi et al. discovered that BCL2-related proteins also regulate mitochondrial dynamics using dynamin-related GTPases. (Lüpertz, 2008; Hornstein et al., 2013)

Hsp90 protects 20S proteasome from oxidative damage inactivation (Höhn et al., 2017; Conconi et al., 1998) and helps to fold oncogenic proteins e.g. of p53 (Saibi et al., 2013). Aging relevant Hsp70 is activated with the help of HSF-1deacetylation by SIRT1 (Westerheide et al., 2009). Aging dependent changes in the amount of sensitive to diet and exercise NAD+ (Cantó et al., 2015) can affect the activity of sirtuins. SIRT1 activity can be decreased via NAD+ level, which also increases activity of the HIF-1α transcription factor resulting in changed in oxidative phosphorylation and mitochondrial dysfunction. (Gomes et al., 2012, 2013; Greer et al., 2007) SIRT1, p53 and HIF-1α in turn effect NAD+ level. Also, AMPK, which inhibits insulin/IGF-1/mTOR, cooperate with SIRT1 to create new mitochondria with the help of PGC-1 and p53. In the same time mitochondria influences TCA cycle. (Salminen et al., 2014) Tollefsbol described 2014 the connection between caloric restriction and longevity. Schultz and Sinclair described 2016 that intestinal stem cells can renewal via BST1 (converts NAD+ cADPR) (Yilmaz et al., 2012 These cells express Notch ligand delta Dl and ESG.

Low protein-high carbohydrate diet influences energy level via enhancing FGF21 expression and in the same time reduction of mTOR activity. This increases Brain-derived neurotrophic factor expression (Zaptan et al., 2015) and influences neural precursor cells (Marosi and Mattson, 2014; Vivar and van Praag, 2013) Neural progenitor cells are also positively influenced by nutrient-sensing protein FOXO3 ( Renault et al., 2009; Devin et al., 2016; Renault et al., 2009) Also grey matter volume of subcortical regions is positively affected this way. (Colman et al., 2009)

Low caloric intake has also positive effect on SIRT1 activity which leads to dendritic outgrowth and plasticity and low inflammatory cytokine activity. (Maalouf et al., 2009) Ohkura et al., 2010 described how FOXO interacts with the sirtuin, which is relevant to aging and cancer. Sirtuin in turn decreases apoptosis via FOXO or BAX. (Dang et al., 2009; Dang et al., 2009; Martins et al., 2015) According to „CYB5R3: a key player in aerobic metabolism and aging?“ (de Cabo et al. 2010) coenzyme Q (CoQ) helps to reduce NADH-level in mitochondria and regulate this way NAD+-dependent enzymes e.g., sirtuins. So, cytochrome b5 coding reductase CYB5R needs NADH and CoQ for its function.(Villalba et al., 1995) Also low calories intake enhanced aerobic metabolism. This needs cytosolic cooperation of N-myristoylated CYB5R3 (a component of P-450 mediated hydroxylation of drugs and steroid hormones (Passon and Hultquist, 1972), fatty acid elongation (Keyes and Cinti ,1980) and cholesterol biosynthesis (Reddy et al, 1977)) and SIRT1. SIRT1 in turn needs lysine acetylation for this cooperation. It modifies CYB5R3 activity and regulates this way the cytosolic NAD+/NADH level.

The activity of FOXO/DAF-16 is positively and negatively regulated by different molecular players (e.g., the insulin/IGF signalling pathway and the nutrient sensor AMPK) and stresses (e.g., oxidative and heat stress). (Eijkelenboom and Burgering, 2013; Salih and Brunet, 2008; Dervis and al., 2008) Sirtuins and H2S induced AMPK and IIS (influences FOXO and mTOR (effected by GH)) have opposite actions. (Barzilai et al., 2012; Fontana et al., 2010; Kenyon, 2010, 2005; Blagosklonny, 2006; Kapahi et al., 2010; Stanfelet al., 2009; Polak and Hall, 2015; Moskalevet al., 2014) FOXO also effects detoxification enzymes MnSOD and GADD45. (Kops et al., 2002; Nemoto and Finkel, 2002)

Apolipoprotein E4 (apoE4) and FOXO are not only associated with quick cell aging (Aksenov et al., 2001; Martins, et al., 2015) but also with Alzheimer’s disease (Sando et al., 2008; DiLoreto & Murphy, 2015).

Further aging factors are rapamycin target S6K activator and 4EBP1 inhibiter mTORC1 and cytoskeletal relevant TORC2 protein kinases (Zoncu et al., 2011),

AMP/ATP ratio and metformin activated AMPK, insulin and IGF1 influenced (FOXO) , which in turn activates MnSOD.

FOX genes and anti-aging agents

Aging relevant SIRT2 deacetylase can be regulated via Resveratrol. Resveratrol is known for his positive affect on Alzheimer disease, on neuroendocrine tumors, on multiple myeloma, on follicular lymphoma, on colon-ca etc. Like EGCG and alpha-M it inhibits PI3K/PTEN/Akt/mTORC1 and WNT/beta-catenin pathway. Resveratrol effects c-Myc, MMP-7 and SIRT1activity and increases SLUG-, vimentin- and NF-kappa-B level. NF-kappa-B level in turn influences caspase-3, MMP-9- and CXCR4- level as well as EMT-level via TGF-beta/SMAD. TGF-beta/SMAD regulates SNAIL/E-cadherin level. ( Ji et al., 2015)

Resveratrol also increases PARP-1 and AMPK level (which in turn decreases MDR1 and CREB phosphorylation level (Wang et al., 2015), E-cadherin-level and apoptosis. It decreases cell migration, invasion and proliferation. Resveratrol interacts with type II topoisomerase and causes double strand DNA breaks and also influences TP53 via ATM. (Demoulin et al., 2015) Together with mitomycin it effects C p21it level and shows an anti-proliferative effect. (Ali et al., 2014) Resveratrol increases the expression of miR-34 which resulted in the decreased expression of E2F3 and SIRT1 (Kumazaki et al., 2013) Tumor suppressor Klotho gene have also aging relevant effect. It increases SIRT1 level (Melatonin shows similar effects) and activates FOXO3 phosphorylation via PI3K/PTEN/Akt pathway. Resveratrol has positive effect on liver cancer and macular degeneration (via VEGF-A and VEGF-C negative regulation), on cardiovascular diseases, on endometriosis, on PCOS, on dyslipidaemia, on diabetes, on chronic renal insufficiency, on Friedreich Ataxia, on Huntington disease and on brain ca via TP53 and p21, 1A/1B light chain 3B (LC3-II), Atg5, beclin-1 and NANOG Cip1 etc. Resveratrol inhibits cyclooxygenase activity, CYP A1 metabolism and influences aging relevant TNFRSF6,TNF-alpha, HIF-1alpha, VEGF, NF-kappaB activity, FAS/FAS-ligand, TP53, FAS-L = CD95, IL-17 but also apoptosis of activated T cells interleukin 17. In head and neck cancer Resveratrol decreases ALDH1, CD44 , HNC-TICs, EMT, NANOG, NESTIN and OCT4 level (Hu et al., 2012) In leukaemia it decreases pLKB1level via SIRT1 and STK11 (Peng et al., 2015) Antiaging phytochemicals are Stilbenoid Resveratrol from Veratrum, which among others influences caspase-1 (Yang et al., 2014; Pietrocola et al., 2012) as well as Catechins and EGCG from Green, which also influence Beclin 1 (Yang et al., 2013; Yang, 2008; Fan et al, 2014) and Camellia sinensis. which also positively influences Helicobacter pylori-triggered caspase-1 signalling pathway.

Not only Resveratrol but also Andrographolide and Parthenolide effect NF-kappaB (Gunn et al., 2011) Further antiaging products are e.g. Iberis amara, which effects apoptosis and ROS ( Weidner et al., 2016) and Beta-carotene, which interact with FOXP2 and piperine, which like Vitamin A kill CSCs. ( Scarpa et al., 2015) Omega-3 polyunsaturated fatty acids negatively influences PI3K/PTEN/Akt/mTORC1 signalling and effects apoptosis (Vasudevan et al., 2014) According to (Rafael de Cabo et al., 2014) caloric restriction influences FGF21, TOR/S6K pathways, insulin, SIRT3, mitochondrial acetyl proteome (Hebert et al., 2013) and metformin - ATP Level in mitochondria, SKN-1/Nrf (Berstein, 2012) (in the same time in worms it lowers ATP levels (Dillin et al., 2002; Lee et al., 2003) possible via mutations in the mitochondrial leucyl-tRNA synthetase gene (Lee et al., 2003),Atg5 (Pyo et al., 2013), TOR signalling, p53 (Jia and Levine, 2007; Tavernarakis et al., 2008).Also acetylproteome, spermidine and sirtuin, which influences autophagy, histone acetyltransferase like resveratrol effects histone deacetylase (sirtuin) (Morselli et al., 2010) and Atg5 (Morselli et al., 2011).SIRT3 is positively affected by dietary restriction via deacetylation of mitochondrial proteins (Someya et al., 2010) and also can be upregulated by Resveratrol. ROS activates hypoxia-inducible factor 1 (HIF-1) and (AMP)-activated protein kinase (AMPK) and affects transcription factors NRF2/SKN-1 and p53/CEP-1 (Chang et al., 2015; Ventura et al., 2009 ). AMPK activates PGC1α , which regulates mitochondrial respiration and detoxification( Liang and Ward, 2006 ). Low PGC1α level effects antioxidant NO (Borniquel et al., 2006 ) which also contribute to proper memory functions. CR also positively influences Hormesis (Calabrese et al., 1987).and anti-stress transcription factors e.g. Gis1, Msn2/Msn4 and Rim15 in yeast and FOXO in mammals.(Wang et al., 2011) In flies it needs help of Complex I and IV (Zid et al., 2009), d4E-BP (Zid et al., 2009) and catalase (Schriner et al., 2005).

 

Several reports illustrated that Metformin activates AMP-activated protein kinase (Zhou et al., 2001) and inhibits tyrosine phosphates activity (Holland et al., 2004) . and mTORC1 signalling (Kalender et al., 2010)

Campisi showed in her review „Senescent Cells, Tumor Suppression, and Organismal Aging: Good Citizens, Bad Neighbors25 February 2005“ p53 and RB tumor suppressor proteins as aging relevant agents with anticancer effect. Senescence is associated with RAS-RAF-MEK signalling cascade. Different senescence signals can converge (Bringold and Serrano, 2000; Lundberg et al., 2000; Narita and Lowe, 2004), so p53 can be influenced by telomeres (d’Adda di Fagagna et al., 2003, Takai et al., 2003) and RAS (Serrano et al., 1997; Ferbeyre et al., 200;, Pearson et al., 2000), RAS also upregulates pRB (responsible for repressive heterochromatin at loci containing transcription factor E2F) and effects p16 (Beausejour et al., 2003;Wright and Shay, 200;, Collins and Sedivy, 2003; Ben-Porath and Weinberg, 2004; Harvey et al., 1993; Smogorzewska and de Lange, 2002) and plays a role in oxygen toxicity during replicative senescence (Parrinello et al., 2003), p16, p53 pathway, which overlap with oncogenic RAS (Beausejour et al., 2003; Brookes et al., 2002; Huot et al., 2002), but also with ROS (Irani et al., 1997), IGF1 signalling pathway, ARF, MDM2 (Krishnamurthy et al., 2004), p16 transcriptional activator Ets-1 (Ohtani et al., 2001), correlated with p16 expression (Krishnamurthy et al., 2004). It is possibly due to increased sensitivity of the INK4a/ARF locus to transcriptional activation.

According to Blagosklonny, 2012 sirtuins negatively regulate rDNA recombination (Kaeberlein et al., 1999) and number of extrachromosomal rDNA circles. (Stinclair and Guarente, 1997) Sir.2 improves FOXO ortholog, DAF-16 function in worms possibly via IIS (Tissenbaum et al., 2001) and in flies via Rpd3 deacetylation. (Rogina and Helfand, 2004)

Rapamycin inhibits mTOR and rejuvenates cardiac and skeletal muscles via increasing NAD+ level (Cantó et al., 2012)and improves aged immune system (Mannick et al., 2014). It also activates Sirtuin via STACs. STACs are impaired by TGF-β, the levels of which increases during aging in mouse and human sera. Another possibly aging relevant effect of Rapamycin is inhibition of S6K1 (Selman et al., 2009 ). Similar effect on increases longevity has nutrient supplementation with spermidine and polyamine-production of gut flora in mice (Tofalo et al., 2019; Matsumoto et al., 2011, 2007)

According to „Six plant extracts delay yeast chronological aging through different signalling pathways“ Lutchman et al., 2016 Cimicifuga via SNF1 (TORC1 inhibition), Ginkgo biloba via PKA inhibition, Valeriana officinalis L. via PKA pathway, Apium graveolens L. and Salix alba via PKH1/2-sensitive form of Sch9 inhibition as well as Passiflora incarnata L, caffeine, myriocin, spermidine, cryptotanshinone, rapamycin, lithocholic acid, resveratrol and methionine sulfoxide positively effects Saccharomyces cerevisiae aging . (de Cabo et al., 2014; Eisenberget al., 2009 Fontanaet al., 2010; Goldberget al., 2010, 2009; Hubbard and Sinclair, 2014; Kaeberlein, 2010; Leonov et al., 2015; Minois et al., 2011; Huang et al., 2014; Morselli et al., 2009; Arlia-Ciommo et al., 2014; Burstein et al., 2012; Lutchman et al., 2016)

SNF5 component SWI/SNF acts in opposite to polycomb-mediated PcG silencing in Drosophila and to p16INK4a silencing in mammals, leads to increased p16 level and is decreased in cancer cells. (Oruetxebarria et al., 2004)

Further natural aging relevant compounds describe McCubrey et al. 2017 in “Effects of resveratrol, curcumin, berberine and other nutraceuticals on aging, cancer development, cancer stem cells and microRNAs”

Rapamycin also protects again Alzheimer's disease, Parkinson and cardiomyopathy via mTOR (Cai Z and Yan LJ, 2013; Bitto et al., 2015). TOR overlaps with metformin, which increases insulin sensitivity, activates AMPK and inhibits IGF-1 signalling65. H2Ss is also a goal of metformin. (Anisimov, 2010; Awartani, 2002). Nestler et al., 2002 describe that Biguanide have positive effect on ovulation and gynaecological diseases. It also improves immune response (Dilman ,1994), has a positive effect on N-nitrosobis-(2-oxopropyl) induced pancreatic cancer malignancies (Schneider, 2001), inhibits lung carcinogenesis induced by tobacco carcinogen (Memmott et al., 2010) and benzo(a)pyrene-induced skin and cervico-vaginal carcinogenesis in mice as well as on melatonin level (Deriabina et al., 2010) .Phenformin improves immunity and inhibits carcinogenesis in mice (Vinnitski and Iakumenko, 1981) and can inhibit carcinogenesis induced by X-rays in rats. (Anisimov et al., 1982) Diabenol® showed anticancerogenic effects, eg. in colon cancer, improved oestrous function, decreased the size of mammary adenocarcinoma metastases into the lung, multiplicity of all colon tumors in mice. (Popovich et al., 2005)

Metformin as buformin positively effect aging relevant glycation end products (Kiho et al., 2005; Diamanti-Kandarakis et al., 2007) Therefore pentosidine is an aging-marker (Ulrich, 2001)

According to Anisimov et al., 2013; Perridon et al., 2016 ; Wu et al., 2012 various H2S upregulates SIRT, which can inhibit human colon adenocarcinoma. H2S also improves mitochondrial function and antioxidants level.( Kimura et al., 2010, 2004) It happens via ROS-scavengers (Sun et al., 2012) and S-sulfhydration of p66Shc (Xie et al., 2014) glutathione peroxidase, superoxide dismutase. H2S stabilize and activates p21 (van Deursen et al., 2014) It also effects anti-oxidative transcription factor Nrf2 activation via S-sulfhydration of Keap1 (Yang et al., 2013), induced the S-sulfhydration of MEK1 and protects against human platelet aggregation in vitro ( Gao et al., 2015) According to Review „The role of hydro gen sulphide in aging and age-related pathologies“ Bernard et al., 2016, H2S has positive effects of on genome stability via increasing MEK1 S-sulfhydration, ERK1/2 and PARP-1 activity leading to the activation of DNA damage repair mechanisms and protection from cellular senescence.(Zhao et al., 2014)

The CSE/H2S pathway is important in genome stability and cell proliferation due to downregulation of ERK1/2 activity: its inhibition in hepatoma cells decreases their proliferation and increases ROS production, mitochondrial disruption, DNA damage and apoptosis. H2S inhibition activates p53, p21, Bax and other pro-apoptotic genes (Pan et al., 2014) and protects against oxidative damage of Alzheimer's patients . H2S also effects telomere maintenance and influences expression and activity of SIRT1 (López‐Otín et al., 2013; Zhang et al., 2014; Li et al., 2014), IL-6, TNF-α (Rios et al., 2015; Zhang et al., 2013) and methylation via CSE/H2S. (Li et al., 2015; Kamatet all., 2015) NaHS inhibits glycation in humans. (Houtkooper et al., 2010)