scholarly journals Proteostasis collapse halts G1 progression and delimits replicative lifespan

2019 ◽  
Author(s):  
David F. Moreno ◽  
Kirsten Jenkins ◽  
Sandrine Morlot ◽  
Gilles Charvin ◽  
Attila Csikász-Nagy ◽  
...  

AbstractLoss of proteostasis and cellular senescence are key hallmarks of cell aging, but whether they are subject to direct cause-effect relationships is not known. We show that most yeast cells arrest in G1 before death with low nuclear levels of cyclin Cln3, a key activator of Start extremely sensitive to chaperone status. Chaperone availability is seriously compromised in aged cells, and the G1 arrest coincides with massive aggregation of a metastable chaperone-activity reporter. A mathematical model integrating autocatalytic protein aggregation and a minimal Start network recapitulates empirical observations. As key predictions, G1-cyclin overexpression increases lifespan in a chaperone-dependent manner, and lifespan reduction by enforced protein aggregation is greatly alleviated by increased expression of specific chaperones or cyclin Cln3. Overall, our data indicate the crucial role of chaperone malfunction in setting lifespan in yeast cells, and configure a molecular pathway whereby proteostasis breakdown acts as a direct effector of cell senescence.

eLife ◽  
2019 ◽  
Vol 8 ◽  
Author(s):  
David F Moreno ◽  
Kirsten Jenkins ◽  
Sandrine Morlot ◽  
Gilles Charvin ◽  
Attila Csikasz-Nagy ◽  
...  

Loss of proteostasis and cellular senescence are key hallmarks of aging, but direct cause-effect relationships are not well understood. We show that most yeast cells arrest in G1 before death with low nuclear levels of Cln3, a key G1 cyclin extremely sensitive to chaperone status. Chaperone availability is seriously compromised in aged cells, and the G1 arrest coincides with massive aggregation of a metastable chaperone-activity reporter. Moreover, G1-cyclin overexpression increases lifespan in a chaperone-dependent manner. As a key prediction of a model integrating autocatalytic protein aggregation and a minimal Start network, enforced protein aggregation causes a severe reduction in lifespan, an effect that is greatly alleviated by increased expression of specific chaperones or cyclin Cln3. Overall, our data show that proteostasis breakdown, by compromising chaperone activity and G1-cyclin function, causes an irreversible arrest in G1, configuring a molecular pathway postulating proteostasis decay as a key contributing effector of cell senescence.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3857-3857
Author(s):  
Katia Beider ◽  
Michal Begin ◽  
Michal Abraham ◽  
Hanna Wald ◽  
Ido Weiss ◽  
...  

Abstract Abstract 3857 Poster Board III-793 The chemokine receptor CXCR4 and its ligand CXCL12 are involved in the development and progression of a diverse number of hematological malignancies, including leukemia, lymphoma and multiple myeloma (MM). Binding CXCL12 to CXCR4 activates a variety of intracellular signal transduction pathways and effector molecules that regulate cell chemotaxis, adhesion, survival, apoptosis and proliferation. It was previously shown that CXCR4 signaling can directly induce caspase-independent cell apoptosis through the interaction with the HIV gp120 envelope protein. In the present study we investigated the effect of CXCR4 specific antagonists 4F-benzoyl-TN14003 (T140) and AMD3100 on the survival and proliferation of different human hematological cancer cells. Here, we demonstrate that T140, but not AMD3100, exhibits preferential cytotoxicity towards malignant cells of hematopoietic origin, as compared to primary normal cells or solid prostate and breast tumor cells. The in vitro treatment with T140, but not with AMD3100, significantly decreased the number of viable chronic myeloid leukemia K562 cells, acute T cell leukemia Jurkat cells, acute promyelocytic leukemia NB4 and HL60 cells, and four different MM cell lines (U266, NCI-H929, RPMI8226 and ARH77), demonstrating the highest sensitivity to T140 (p<0.01). Notably, T140 inhibited the growth of freshly isolated leukemia and MM cells obtained from consenting patients. T140 inhibits the growth of MM and leukemic cells by inducing their apoptotic cell death. The apoptotic changes in the cells were associated with morphological changes, phosphatidylserine externalization, sub-G1 arrest, DNA double-stranded breaks, decrease in mitochondrial membrane potential, release of cytochrome c, and caspase 3 activation. The important role of CXCR4 in T140-mediated cell death was confirmed by demonstrating that CXCR4 over-expression in NB4 and K562 cells increased their sensitivity to T140. Furthermore, pretreatment of NB4 and HL60 cells with AMD3100 abolishes the effect of T140 on these cells, indicating the involvement of CXCR4 in T140-induced apoptosis. Importantly, the combination with novel anti-myeloma agent bortezomib significantly augments anti-myeloma activity of T140. The anti leukemic and MM effect of T140 was confirmed in xenograft in vivo tumor models. Subcutaneous (s.c.) or intra-peritoneal (i.p.) injections of T140 (100 or 300 mcg/mouse) significantly reduced, in a dose-dependent manner, the tumor size in immuno-deficient mice that were previously inoculated s.c. with human acute leukemia cells NB4 or MM cells RPMI8226 (p<0.01). Tumors from animals treated with T140 had smaller sizes and weights, larger necrotic areas and high apoptotic scores. Taken together, these data support the unique anti-cancer effect of T140 in hematological malignancies and indicate the potential therapeutic role of T140 in MM and leukemia patients. Disclosures: No relevant conflicts of interest to declare.


PLoS ONE ◽  
2021 ◽  
Vol 16 (1) ◽  
pp. e0246116
Author(s):  
Joseph Minicucci ◽  
Molly Alfond ◽  
Angelo Demuro ◽  
David Gerberry ◽  
Joe Latulippe

Alzheimer’s disease (AD) is a devastating illness affecting over 40 million people worldwide. Intraneuronal rise of amyloid beta in its oligomeric forms (iAβOs), has been linked to the pathogenesis of AD by disrupting cytosolic Ca2+ homeostasis. However, the specific mechanisms of action are still under debate and intense effort is ongoing to improve our understanding of the crucial steps involved in the mechanisms of AβOs toxicity. We report the development of a mathematical model describing a proposed mechanism by which stimulation of Phospholipase C (PLC) by iAβO, triggers production of IP3 with consequent abnormal release of Ca2+ from the endoplasmic reticulum (ER) through activation of IP3 receptor (IP3R) Ca2+ channels. After validating the model using experimental data, we quantify the effects of intracellular rise in iAβOs on model solutions. Our model validates a dose-dependent influence of iAβOs on IP3-mediated Ca2+ signaling. We investigate Ca2+ signaling patterns for small and large iAβOs doses and study the role of various parameters on Ca2+ signals. Uncertainty quantification and partial rank correlation coefficients are used to better understand how the model behaves under various parameter regimes. Our model predicts that iAβO alter IP3R sensitivity to IP3 for large doses. Our analysis also shows that the upstream production of IP3 can influence Aβ-driven solution patterns in a dose-dependent manner. Model results illustrate and confirm the detrimental impact of iAβOs on IP3 signaling.


2020 ◽  
Vol 88 (11) ◽  
Author(s):  
Yuki Sato ◽  
Ko Sato ◽  
Hideki Yamamoto ◽  
Jun Kasamatsu ◽  
Tomomitsu Miyasaka ◽  
...  

ABSTRACT Cryptococcus deneoformans is an opportunistic fungal pathogen that frequently causes fatal meningoencephalitis in patients with impaired cell-mediated immune responses such as AIDS. Caspase-associated recruitment domain 9 (CARD9) plays a critical role in the host defense against cryptococcal infection, suggesting the involvement of one or more C-type lectin receptors (CLRs). In the present study, we analyzed the role of macrophage-inducible C-type lectin (Mincle), one of the CLRs, in the host defense against C. deneoformans infection. Mincle expression in the lungs of wild-type (WT) mice was increased in the early stage of cryptococcal infection in a CARD9-dependent manner. In Mincle gene-disrupted (Mincle KO) mice, the clearance of this fungus, pathological findings, Th1/Th2 response, and antimicrobial peptide production in the infected lungs were nearly comparable to those in WT mice. However, the production of interleukin-22 (IL-22), tumor necrosis factor alpha (TNF-α), and IL-6 and the expression of AhR were significantly decreased in the lungs of Mincle KO mice compared to those of WT mice. In in vitro experiments, TNF-α production by bone marrow-derived dendritic cells was significantly decreased in Mincle KO mice. In addition, the disrupted lysates of C. deneoformans, but not those of whole yeast cells, activated Mincle-triggered signaling in an assay with a nuclear factor of activated T cells (NFAT)-green fluorescent protein (GFP) reporter cells expressing this receptor. These results suggest that Mincle may be involved in the production of Th22-related cytokines at the early stage of cryptococcal infection, although its role may be limited in the host defense against infection with C. deneoformans.


2017 ◽  
Vol 2017 ◽  
pp. 1-8 ◽  
Author(s):  
Ya-qin Chen ◽  
Xin-guang Liu ◽  
Wei Zhao ◽  
Hongjing Cui ◽  
Jie Ruan ◽  
...  

Yeast MET18, a subunit of the cytosolic iron-sulfur (Fe/S) protein assembly (CIA) machinery which is responsible for the maturation of Fe/S proteins, has been reported to participate in the oxidative stress response. However, the underlying molecular mechanisms remain unclear. In this study, we constructed a MET18/met18Δ heterozygous mutant yeast strain and found that MET18 deficiency in yeast cells impaired oxidative stress resistance as evidenced by increased sensitivity to hydrogen peroxide (H2O2) and cumene hydroperoxide (CHP). Mechanistically, the mRNA levels of catalase A (CTA1) and catalase T (CTT1) as well as the total catalase activity were significantly reduced in MET18-deficient cells. In contrast, overexpression of CTT1 or CTA1 in MET18-deficient cells significantly increased the intracellular catalase activity and enhanced the resistance ability against H2O2 and CHP. In addition, MET18 deficiency diminished the replicative capacity of yeast cells as evidenced by the shortened replicative lifespan, which can be restored by CTT1 overexpression, but not by CTA1, in the MET18-deficient cells. These results suggest that MET18, in a catalase-dependent manner, plays an essential role in enhancing the resistance of yeast cells to oxidative stress and increasing the replicative capacity of yeast cells.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Huan-Tian Zhang ◽  
Tao Gui ◽  
Ri-Xu Liu ◽  
Kui-Leung Tong ◽  
Chong-Jie Wu ◽  
...  

AbstractChondrosarcoma (CHS) is the second most common bone malignancy with limited therapeutic approaches. Our previous study has found that Yes associated protein 1 (YAP1) is downregulated in CHS cells treated with bromodomain and extraterminal domain (BET) inhibitor JQ1. However, the precise role of YAP1 in CHS is largely unknown. Herein, we found that YAP1 expression was upregulated in CHS tissues, and positively correlated with its grading score. Loss of YAP1 inhibited CHS proliferation and induced cellular senescence, while expression of YAP1 mutants revealed YAP1/TEA domain family member (TEAD)-dependent negative regulation of p21 and subsequent cellular senescence. These results were validated by in vivo experiments using stable shYAP1 cell lines. Mechanistically, negative regulation of p21 by YAP1 occurred post-transcriptionally via Dicer-regulated miRNA networks, specifically, the miR-17 family. Furthermore, we demonstrated that sequential targeting of YAP1 and p21 enhanced the elimination of JQ1-induced senescent cells in a Bcl-2-like 1 (Bcl-XL)/Caspase-3 dependent manner. Altogether, we unveil a novel role of YAP1 signaling in mediating CHS cell senescence and propose a one-two punch approach that sequentially targets the YAP1/p21 axis to eliminate senescent cells.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3611-3611
Author(s):  
Shan Fu ◽  
Jieping Wei ◽  
Binsheng Wang ◽  
He Huang

Abstract As immunomodulatory cytokines, Type 1 interferons (IFNs) have a long history of efficacy in treating chronic myeloid leukemia (CML). Recently, many research reported the combination of IFN-α and imatinib significantly increased the rates of molecular responses, comparing to single imatinib treatment. Related molecular mechanism may be the direct effect of IFN-alpha on stem cells. Therefore, IFN-α was renewed to be a vital candidate for CML treatment. Bone marrow mesenchymal stem cells (MSCs), which also be defined as mesenchymal stromal cells, are important to hematopoiesis. IFN-α was indicated as a potential inhibitor of MSCs; however the exact mechanism remains unclear. PML is known as a tumor suppressor, which locates at the downstream of IFN-α pathway. In our previous research, we have proved that PML stably expressed in human MSCs (hMSCs), which was important in maintaining the normal function of hMSCs. To our knowledge, although PML has been extensively studied in tumor cells, little is known about PML gene regulation in MSCs. In this study, we investigated the effect of IFN-α on hMSCs and the role of PML involved in this process. After approval by institutional review board, hMSCs were isolated from the bone marrow of volunteers and confirmed by flow cytometry. Cells were treated with different concentration of IFN-α up to 14 days. We found that IFN-α treated cells were growing slowly, and had a dramatically decreased number of colone in a dose dependent manner (Fig A). However, IFN-α did not induce significant cell apoptosis. Then a variety of senescence-associated detection was measured. hMSCs senescence induced by IFN-α had a dose and time dependent manner (Fig B). After treated with IFN-α at 1000 U/ml for 7 or 14 days, we found that up to 18% ± 1.1 or 27.56% ± 1.33 of hMSCs became SA-b-gal-positive as compared with 7.53% ± 0.55 or 6.47% ± 2.5 of untreated cells (P<0.05 for both). Real time PCR analysis proved this process by an increase in production of the senescence marker p53 and p21. Expression of PML was detected by real-time PCR and immunofluorescence in hMSCs treated with IFN-α. Consistent with other studies, mRNA expression of PML can be up-regulated by IFN-α in hMSCs. When cells were treated with IFN-α at 1000 U/ml for 7 or 14 days, PML gene expression in hMSCs was increased by more than 2 fold. At the same time, both the number and size of PML-NBs were increased markedly and had a concentration dependent manner. These results indicate that PML protein can be up regulated by IFN-α in hMSCs. Then, PML expression was inhibited using an RNAi-mediated PML knockdown system. After treated with IFN-α at 1000 U/ml for 7 days, hMSCs senescence can be rescued by the knocking down of PML. The percentage of SA-b-gal positive cells in PML knocking down hMSCs has a significant decrease as compared with cell transfected with control-RNAi (4.49% ±1.27 vs. 17.26% ± 1.44, P < 0.05) (Fig C). To further characterize the effect of PML on cellular senescence in hMSCs, PML-overexpressed hMSCs were used. 7 days post-transfection, PML overexpressing hMSCs were strongly positive for SA-b-gal activity (47.43%±3.8), as compared with normal and empty vector transfected cells (4.9%±0.7, 5.97%±0.75) (P< 0.001) (Fig D). mRNA levels of P53 and P21 were also enhanced in PML-overexpressed hMSCs. P53 pathway contributes to cell senescence and the role of PML has been proved in the regulation of P53 activity, we wondered whether upregulation of PML induced by IFN-α has relationship with P53 pathway in hMSCs. In the process of IFN-α induced hMSCs senescence, an increasing co-localization of PML and P53 was observed in IFN-α treated cells (1000U/ml, 7 days) as compared with untreated cells (Fig E). To further confirm whether or not the change of P53 location was mediated through the upregulation of PML, we knocked-down the expression of PML in hMSCs. Treated with IFN-α (1000U/ml, 7 days), we did not found significant location of P53 in PML-knocking down cells as compared with control. Taken together, our results suggested that hMSCs incurred senescence upon IFN-α stimulation, while PML levels were observed significant increase. By knocking-down and overexpressing PML, we demonstrated that PML was indispensable to IFN-α mediated hMSCs senescence. The molecular mechanisms underlying this process may be an increased co-localization of PML and p53 induced by IFN-α. These findings provided a novel insight into the role of IFN-α on hMSCs. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Author(s):  
Shoshik Amram ◽  
Tal Iram ◽  
Ekaterina Lazdon ◽  
Robert Vassar ◽  
Ittai Ben-Porath ◽  
...  

ABSTRACTAlterations in astrocyte function such as a pro-inflammatory phenotype are associated with Alzheimer’s disease (AD). We had shown impairments in the ability of aged astrocytes isolated from 5xFAD mice to clear and uptake amyloid-β (Aβ) as well as to support neuronal growth. Senescent cells accumulate with age and exhibit a senescence-associated secretory phenotype, which includes secretion of pro-inflammatory cytokines. In this study, we predicted that with age, astrocytes in 5xFAD mice would exhibit a cellular senescence phenotype that could promote neurodegeneration. We found an age-dependent increase in senescent astrocytes adjacent to Aβ plaques in 5xFAD mice. Inhibition of nuclear factor kappa-light-chain-enhancer of activated B cells reduced interelukin-6 secretion by senescent astrocytes and resulted in improved neuronal support. Moreover, senescent astrocytes exhibited an increase in the induction of the TGF-β1-SMAD2/3 pathway, and inhibition of this pathway resulted in a reduction of cellular senescence. We also discovered that soluble Aβ42 induced astrocyte senescence in young naïve mice in a SMAD2/3-dependent manner. Our results suggest an important role of astrocyte senescence in AD and its role in mediating the neurotoxicity properties of astrocytes in AD and related neurodegenerative diseases.


2019 ◽  
Vol 15 ◽  
pp. 02023
Author(s):  
B. Blondin ◽  
C. Duc ◽  
J. Noble ◽  
M. Pradal ◽  
C. Tesnière

Yeast cell death can occur during wine alcoholic fermentation and lead to sluggish or stuck fermentations. The mechanisms underlying cell death during yeast starvation in alcoholic fermentations remain unclear. In this work we addressed yeast cell death using conceptual framework from ageing studies showing that yeast resistance to starvation can be influenced by the nature of the nutrient limiting cell growth. We examined cell death occurrence considering yeast cells ability to elicit an appropriate response to a set of nutrient limitations. We show that several micronutrients limitations (oleic acid, ergosterol, pantothenic acid and nicotinic acid) trigger cell death in a nitrogen-dependent manner. We provide evidence that the nitrogen Tor/Sch9 signaling pathway is involved in triggering cell death. In such conditions, yeast cells fail to acquire stress resistance given a restriction at a post-transcriptional level. We have examined the ability of different nitrogen sources to trigger cell death and show that they impact differentially on cell death and that NH4 + had a strong death inducing capacity. Finally, the QTLs approaches allowed the mapping of a set of loci controlling cell death under oleic acid and pantothenic acid starvation consistent with a multigenic control.


Author(s):  
Jing Yang ◽  
Mengmeng Liu ◽  
Dongchun Hong ◽  
Musheng Zeng ◽  
Xing Zhang

Cellular senescence occurs in proliferating cells as a consequence of various triggers including telomere shortening, DNA damage, and inappropriate expression of oncogenes. The senescent state is accompanied by failure to reenter the cell cycle under mitotic stimulation, resistance to cell death and enhanced secretory phenotype. A growing number of studies have convincingly demonstrated a paradoxical role for spontaneous senescence and therapy-induced senescence (TIS), that senescence may involve both cancer prevention and cancer aggressiveness. Cellular senescence was initially described as a physiological suppressor mechanism of tumor cells, because cancer development requires cell proliferation. However, there is growing evidence that senescent cells may contribute to oncogenesis, partly in a senescence-associated secretory phenotype (SASP)-dependent manner. On the one hand, SASP prevents cell division and promotes immune clearance of damaged cells, thereby avoiding tumor development. On the other hand, SASP contributes to tumor progression and relapse through creating an immunosuppressive environment. In this review, we performed a review to summarize both bright and dark sides of senescence in cancer, and the strategies to handle senescence in cancer therapy were also discussed.


Sign in / Sign up

Export Citation Format

Share Document