Category: Home

Autophagy activation

Autophagy activation

Lung Cancer. J Hepatol. Autophagy activation in T acgivation from activatiom donors is Autophagy activation by spermidine and correlates with function and vaccine responses. Article CAS PubMed PubMed Central Google Scholar Jain MV, Paczulla AM, Klonisch T, et al. Oxid Med Cell Longev.

Autophagy activation -

Bioinformatics analysis of autophagy—lysosomal degradation in cardiac aging. Fang, Y. Autophagy—Sirt3 axis decelerates hematopoietic aging. PubMed PubMed Central CAS Google Scholar. Alsaleh, G. Autophagy in T cells from aged donors is maintained by spermidine and correlates with function and vaccine responses.

De Risi, M. Mechanisms by which autophagy regulates memory capacity in ageing. PubMed PubMed Central Google Scholar.

Yamamuro, T. Age-dependent loss of adipose Rubicon promotes metabolic disorders via excess autophagy. Zhou, B. Mitochondrial permeability uncouples elevated autophagy and lifespan extension.

Cell , — Ezcurra, M. elegans eats its own intestine to make yolk leading to multiple senescent pathologies. Autophagy genes are required for normal lipid levels in C. Autophagy 9 , — A role for autophagy in the extension of lifespan by dietary restriction in C.

Labbadia, J. The biology of proteostasis in aging and disease. Lopez-Otin, C. The hallmarks of aging. Ravikumar, B.

Aggregate-prone proteins with polyglutamine and polyalanine expansions are degraded by autophagy. Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease.

Boland, B. Promoting the clearance of neurotoxic proteins in neurodegenerative disorders of ageing. Drug Discov.

Nuclear DNA damage signalling to mitochondria in ageing. Rubinsztein, D. Autophagy and aging. Kumsta, C. Hormetic heat stress and HSF-1 induce autophagy to improve survival and proteostasis in C. Juhasz, G. Atg7-dependent autophagy promotes neuronal health, stress tolerance, and longevity but is dispensable for metamorphosis in Drosophila.

Hara, T. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Komatsu, M. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Friedman, L. Disrupted autophagy leads to dopaminergic axon and dendrite degeneration and promotes presynaptic accumulation of α-synuclein and LRRK2 in the brain.

Schneider, J. Loss of hepatic chaperone-mediated autophagy accelerates proteostasis failure in aging. Aging Cell 14 , — elegans by inducing autophagy.

Aparicio, R. Demontis, F. Nezis, I. Ref 2 P, the Drosophila melanogaster homologue of mammalian p62, is required for the formation of protein aggregates in adult brain. Lopez, A. AT tau allele causes neurodegeneration that can be ameliorated in a zebrafish model by autophagy induction.

Brain , — Rocchi, A. Fu, H. A tau homeostasis signature is linked with the cellular and regional vulnerability of excitatory neurons to tau pathology. Schinaman, J. Rapamycin modulates tissue aging and lifespan independently of the gut microbiota in Drosophila.

Berger, Z. Rapamycin alleviates toxicity of different aggregate-prone proteins. Audesse, A. FOXO3 directly regulates an autophagy network to functionally regulate proteostasis in adult neural stem cells.

Leeman, D. Lysosome activation clears aggregates and enhances quiescent neural stem cell activation during aging. Garcia-Prat, L. Autophagy maintains stemness by preventing senescence. Nature , 37—42 The role of autophagy during the early neonatal starvation period. Yang, Z.

Atg22 recycles amino acids to link the degradative and recycling functions of autophagy. Cell 17 , — Suzuki, S. Starvation induced cell death in autophagy-defective yeast mutants is caused by mitochondria dysfunction.

PLoS ONE 6 , e Ha, J. Aspects Med. Adeva-Andany, M. Glycogen metabolism in humans. BBA Clin. Yao, W. Atg11 is required for initiation of glucose starvation-induced autophagy. Jiang, S. Starch-binding domain-containing protein 1 Stbd1 and glycogen metabolism: identification of the Atg8 family interacting motif AIM in Stbd1 required for interaction with GABARAPL1.

Weber, C. β-Oxidation and autophagy are critical energy providers during acute glucose depletion in Saccharomyces cerevisiae. Kim, K. High glucose condition induces autophagy in endothelial progenitor cells contributing to angiogenic impairment.

Kishmani, P. Pompe disease diagnosis and management guideline. Article Google Scholar. Kishnani, P. Recombinant human acid α-glucosidase: major clinical benefits in infantile-onset Pompe disease. Neurology 68 , 99— Duran, J.

Glycogen accumulation underlies neurodegeneration and autophagy impairment in Lafora disease. Singh, R. Autophagy regulates lipid metabolism. Ward, C. Autophagy, lipophagy and lysosomal lipid storage disorders.

Degradation of lipid droplet-associated proteins by chaperone-mediated autophagy facilitates lipolysis. Goeritzer, M. Active autophagy but not lipophagy in macrophages with defective lipolysis.

Kiffin, R. Altered dynamics of the lysosomal receptor for chaperone-mediated autophagy with age. Cell Sci. Palikaras, K. Ectopic fat deposition contributes to age-associated pathology in Caenorhabditis elegans. Lipid Res. Stranks, A. Autophagy controls acquisition of aging features in macrophages.

Innate Immun. Autophagy links lipid metabolism to longevity in C. Autophagy 8 , — Folick, A. Lysosomal signaling molecules regulate longevity in Caenorhabditis elegans. Science , 83—86 Autophagy and lipid metabolism coordinately modulate life span in germline-less C.

Zhang, T. SIRT3 promotes lipophagy and chaperon-mediated autophagy to protect hepatocytes against lipotoxicity. Cell Death Differ. Baur, J. Resveratrol improves health and survival of mice on a high-calorie diet.

Ding, W. Selective taste of ethanol-induced autophagy for mitochondria and lipid droplets. Autophagy 7 , — Kounakis, K. Emerging roles of lipophagy in health and disease. Cell Dev.

Chao, X. Impaired TFEB-mediated lysosome biogenesis and autophagy promote chronic ethanol-induced liver injury and steatosis in mice. Gastroenterology , — Hernandez-Gea, V.

Autophagy releases lipid that promotes fibrogenesis by activated hepatic stellate cells in mice and in human tissues. Riffelmacher, T. Autophagy-dependent generation of free fatty acids is critical for normal neutrophil differentiation.

Immunity 47 , — Kawane, K. DNA degradation and its defects. Cold Spring Harb. a Houseley, J. The many pathways of RNA degradation. Buchan, J. Guo, H. Autophagy supports genomic stability by degrading retrotransposon RNA. Fujiwara, Y. Direct uptake and degradation of DNA by lysosomes.

Discovery of a novel type of autophagy targeting RNA. Aizawa, S. Lysosomal membrane protein SIDT2 mediates the direct uptake of DNA by lysosomes. Lysosomal putative RNA transporter SIDT2 mediates direct uptake of RNA by lysosomes.

Autophagy 12 , — Sliter, D. Parkin and PINK1 mitigate STING-induced inflammation. West, A. Mitochondrial DNA stress primes the antiviral innate immune response.

Dan, X. DNA damage invokes mitophagy through a pathway involving Spata Nucleic Acids Res. Hopfner, K. Molecular mechanisms and cellular functions of cGAS—STING signalling. Johansen, T.

Selective autophagy: ATG8 family proteins, LIR motifs and cargo receptors. Pickles, S. Mitophagy and quality control mechanisms in mitochondrial maintenance.

Le Guerroue, F. Autophagosomal content profiling reveals an LC3C-dependent piecemeal mitophagy pathway. Cell 68 , — McLelland, G. Melentijevic, I. elegans neurons jettison protein aggregates and mitochondria under neurotoxic stress. Nicolas-Avila, J. A network of macrophages supports mitochondrial homeostasis in the heart.

Cell , 94— Cornelissen, T. Deficiency of parkin and PINK1 impairs age-dependent mitophagy in Drosophila. Sun, N. Measuring in vivo mitophagy. Cell 60 , — Pickrell, A. Neuron 85 , — Coordination of mitophagy and mitochondrial biogenesis during ageing in C.

McWilliams, T. Basal mitophagy occurs independently of PINK1 in mouse tissues of high metabolic demand. Cell Metab. Tomatidine enhances lifespan and healthspan in C.

Du, F. Mochida, K. Receptor-mediated selective autophagy degrades the endoplasmic reticulum and the nucleus. Hubner, C. ER-phagy and human diseases.

Park, Y. Autophagic degradation of nuclear components in mammalian cells. Autophagy 5 , — Dou, Z. Autophagy mediates degradation of nuclear lamina. Papadopoulos, C. Repair or lysophagy: dealing with damaged lysosomes. Li, Y.

The lysosomal membrane protein SCAV-3 maintains lysosome integrity and adult longevity. Vesosky, B. The influence of age on immunity to infection with Mycobacterium tuberculosis.

Detection and clearance of damaged lysosomes by the endo-lysosomal damage response and lysophagy. Gomez-Sintes, R.

Lysosomal cell death mechanisms in aging. Article CAS PubMed Google Scholar. Reggio, A. Eating the unknown: xenophagy and ER-phagy are cytoprotective defenses against pathogens.

Levine, B. Eating oneself and uninvited guests: autophagy-related pathways in cell defense. PubMed CAS Google Scholar. Rikihisa, Y. Glycogen autophagosomes in polymorphonuclear leukocytes induced by rickettsiae.

Rich, K. Cytoplasmic bacteria can be targets for autophagy. Nakagawa, I. Autophagy defends cells against invading group A Streptococcus. Gutierrez, M. Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages.

Kimmey, J. Bacterial pathogens versus autophagy: implications for therapeutic interventions. Upadhyay, S. Tuberculosis and the art of macrophage manipulation. Watson, R. The cytosolic sensor cGAS detects Mycobacterium tuberculosis DNA to induce type I interferons and activate autophagy.

Cell Host Microbe 17 , — Franco, L. The ubiquitin ligase Smurf1 functions in selective autophagy of Mycobacterium tuberculosis and anti-tuberculous host defense. Cell Host Microbe 21 , 59—72 Bah, A. Macrophage autophagy and bacterial infections. Jayaswal, S. Identification of host-dependent survival factors for intracellular Mycobacterium tuberculosis through an siRNA screen.

PLoS Pathog. Kim, J. Host cell autophagy activated by antibiotics is required for their effective antimycobacterial drug action. Cell Host Microbe 11 , — Extracellular M.

tuberculosis DNA targets bacteria for autophagy by activating the host DNA-sensing pathway. Wang, J. MicroRNA promotes autophagy to eliminate intracellular mycobacteria by targeting Rheb.

Liang, X. Protection against fatal Sindbis virus encephalitis by beclin, a novel Bclinteracting protein. Orvedahl, A. Autophagy protects against Sindbis virus infection of the central nervous system.

Cell Host Microbe 7 , — HSV-1 ICP Cell Host Microbe 1 , 23—35 Mijaljica, D. Shojaei, S. Autophagy and SARS-CoV-2 infection: a possible smart targeting of the autophagy pathway. Virulence 11 , — Carmona-Gutierrez, D. Digesting the crisis: autophagy and coronaviruses.

Cell 7 , — Choi, J. The parasitophorous vacuole membrane of Toxoplasma gondii is targeted for disruption by ubiquitin-like conjugation systems of autophagy.

Immunity 40 , — Ghartey-Kwansah, G. Autophagy in the control and pathogenesis of parasitic infections. Cell Biosci. Age-specific mortality and immunity patterns of SARS-CoV Gelino, S. Intestinal autophagy improves healthspan and longevity in C. elegans during dietary restriction.

Minnerly, J. The cell non-autonomous function of ATG is essential for neuroendocrine regulation of Caenorhabditis elegans lifespan. Bai, H. Carnio, S.

Autophagy impairment in muscle induces neuromuscular junction degeneration and precocious aging. Horvath, S. DNA methylation age of human tissues and cell types. Genome Biol. Dong, S. Chaperone-mediated autophagy sustains haematopoietic stem-cell function.

Bourdenx, M. Chaperone-mediated autophagy prevents collapse of the neuronal metastable proteome. Article PubMed CAS PubMed Central Google Scholar. Lautrup, S. Franceschi, C. Inflammaging: a new immune-metabolic viewpoint for age-related diseases.

Medzhitov, R. Origin and physiological roles of inflammation. Autophagy dictates metabolism and differentiation of inflammatory immune cells. Autophagy 14 , — Swanson, K. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Sun, Q. Inflammasome and autophagy regulation—a two-way street.

Autophagy and neurodegeneration: pathogenic mechanisms and therapeutic opportunities. Neuron 93 , — Autophagy modulation as a potential therapeutic target for diverse diseases.

Wood, J. Sirtuin activators mimic caloric restriction and delay ageing in metazoans. Morselli, E. Spermidine and resveratrol induce autophagy by distinct pathways converging on the acetylproteome.

Pietrocola, F. Spermidine induces autophagy by inhibiting the acetyltransferase EP Madeo, F. Spermidine in health and disease. aan Eisenberg, T.

Cardioprotection and lifespan extension by the natural polyamine spermidine. Zhang, H. Polyamines control eIF5A hypusination, TFEB translation, and autophagy to reverse B cell senescence. Cell 76 , — Induction of autophagy by spermidine promotes longevity.

Song, H. Huang, R. Deacetylation of nuclear LC3 drives autophagy initiation under starvation. Cell 57 , — Lee, I. A role for the NAD-dependent deacetylase Sirt1 in the regulation of autophagy. Autophagy 15 , — Mouchiroud, L. Mitchell, S. Nicotinamide improves aspects of healthspan, but not lifespan, in mice.

Ryu, D. Urolithin A induces mitophagy and prolongs lifespan in C. elegans and increases muscle function in rodents. Andreux, P.

The mitophagy activator urolithin A is safe and induces a molecular signature of improved mitochondrial and cellular health in humans. Article CAS Google Scholar. Escobar, K. Autophagy and aging: maintaining the proteome through exercise and caloric restriction.

Aging Cell 18 , e de Cabo, R. Effects of intermittent fasting on health, aging, and disease. Alexander-Floyd, J. Unexpected cell type-dependent effects of autophagy on polyglutamine aggregation revealed by natural genetic variation in C.

BMC Biol. Bjedov, I. Fine-tuning autophagy maximises lifespan and is associated with changes in mitochondrial gene expression in Drosophila. Regulation of autophagy by reactive oxygen species ROS : implications for cancer progression and treatment Vol.

Mathew R, Karp CM, Beaudoin B, Vuong N, Chen G, Chen HY, et al. Autophagy Suppresses Tumorigenesis through Elimination of p Burada F. Autophagy in colorectal cancer: An important switch from physiology to pathology. World J Gastrointest Oncol. Yue C, Yang X, Li J, Chen X, Zhao X, Chen Y, et al.

Trimethylamine N-oxide prime NLRP3 inflammasome via inhibiting ATG16L1-induced autophagy in colonic epithelial cells. Biochem Biophys Res Commun. Wang Z-L, Deng Q, Chong T, Wang Z-M. Autophagy suppresses the proliferation of renal carcinoma cell.

Eur Rev Med Pharmacol Sci. PubMed Google Scholar. Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, et al.

Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell. Petrova V, Annicchiarico-Petruzzelli M, Melino G, Amelio I. The hypoxic tumour microenvironment. Chavez-Dominguez R, Perez-Medina M, Lopez-Gonzalez JS, Galicia-Velasco M, Aguilar-Cazares D.

The double-edge sword of autophagy in cancer: from tumor suppression to pro-tumor activity. White E. Deconvoluting the context-dependent role for autophagy in cancer. Nat Rev Cancer. Peng YF, Shi YH, Ding ZB, Ke AW, Gu CY, Hui B, et al. Autophagy inhibition suppresses pulmonary metastasis of HCC in mice via impairing anoikis resistance and colonization of HCC cells.

Song J, Guo X, Xie X, Zhao X, Li D, Deng W, et al. Autophagy in hypoxia protects cancer cells against apoptosis induced by nutrient deprivation through a beclin1-dependent way in hepatocellular carcinoma.

Zhu H, Wang D, Zhang L, Xie X, Wu Y, Liu Y, et al. Oncol Rep. Macintosh RL, Timpson P, Thorburn J, Anderson KI, Thorburn A, Ryan KM. Inhibition of autophagy impairs tumor cell invasion in an organotypic model. Lock R, Kenific CM, Leidal AM, Salas E, Debnath J.

Autophagy-dependent production of secreted factors facilitates oncogenic RAS-Driven invasion. Cancer Discov. Autophagy induction impairs migration and invasion by reversing EMT in glioblastoma cells. Mol Oncol.

Peng Q, Qin J, Zhang Y, Cheng X, Wang X, Lu W, et al. Autophagy maintains the stemness of ovarian cancer stem cells by FOXA2. Li LQ, Pan D, Zhang SW, Xie D, Zheng XL, Chen H. Autophagy regulates chemoresistance of gastric cancer stem cells via the Notch signaling pathway.

Maycotte P, Jones KL, Goodall ML, Thorburn J, Thorburn A. Autophagy supports breast cancer stem cell maintenance by regulating IL6 secretion.

Mol Cancer Res. Sharif T, Martell E, Dai C, Kennedy BE, Murphy P, Clements DR, et al. Autophagic homeostasis is required for the pluripotency of cancer stem cells.

Gong C, Bauvy C, Tonelli G, Yue W, Deloménie C, Nicolas V, et al. Galavotti S, Bartesaghi S, Faccenda D, Shaked-Rabi M, Sanzone S, McEvoy A, et al. The autophagy-associated factors DRAM1 and p62 regulate cell migration and invasion in glioblastoma stem cells. Hao C, Liu G, Tian G.

Autophagy inhibition of cancer stem cells promotes the efficacy of cisplatin against non-small cell lung carcinoma. Ther Adv Respir Dis. Marinković M, Šprung M, Buljubašić M, Novak I. Autophagy modulation in cancer: Current knowledge on action and therapy.

Oxid Med Cell Longev. Amaravadi RK, Yu D, Lum JJ, Bui T, Christophorou MA, Evan GI, et al. Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma.

Levy JMM, Thompson JC, Griesinger AM, Amani V, Donson AM, Birks DK, et al. Autophagy inhibition improves chemosensitivity in BRAFVE brain tumors. Feng Y, Gao Y, Wang D, Xu Z, Sun W, Ren P. Autophagy Inhibitor LY and 5-fluorouracil 5-FU Combination-Based Nanoliposome for Enhanced Efficacy Against Esophageal Squamous Cell Carcinoma.

Nanoscale Res Lett. Liu D, Yang Y, Liu Q, Wang J. Inhibition of autophagy by 3-MA potentiates cisplatin-induced apoptosis in esophageal squamous cell carcinoma cells.

Med Oncol. Sasaki K, Tsuno NH, Sunami E, Tsurita G, Kawai K, Okaji Y, et al. Chloroquine potentiates the anti-cancer effect of 5-fluorouracil on colon cancer cells. BMC Cancer.

Yang HZ, Ma Y, Zhou Y, Xu LM, Chen XJ, Ding WB, et al. Autophagy contributes to the enrichment and survival of colorectal cancer stem cells under oxaliplatin treatment.

Cancer Lett. Kanzawa T, Germano IM, Komata T, Ito H, Kondo Y, Kondo S. Role of autophagy in temozolomide-induced cytotoxicity for malignant glioma cells. Liu F, Liu D, Yang Y, Zhao S. Effect of autophagy inhibition on chemotherapy-induced apoptosis in A lung cancer cells.

Oncol Lett. Ren JH, He WS, Nong L, Zhu QY, Hu K, Zhang RG, et al. Acquired cisplatin resistance in human lung adenocarcinoma cells is associated with enhanced autophagy. Cancer Biother Radiopharm. Han W, Sun J, Feng L, Wang KF, Li D, Pan Q, et al.

Autophagy inhibition enhances daunorubicin-induced apoptosis in K cells. Yang MC, Wang HC, Hou YC, Tung HL, Chiu TJ, Shan YS. Blockade of autophagy reduces pancreatic cancer stem cell activity and potentiates the tumoricidal effect of gemcitabine.

Hashimoto D, Bläuer M, Hirota M, Ikonen NH, Sand J, Laukkarinen J. Autophagy is needed for the growth of pancreatic adenocarcinoma and has a cytoprotective effect against anticancer drugs. Eur J Cancer. Zhang Q, Si S, Schoen S, Chen J, Jin XB, Wu G. Suppression of autophagy enhances preferential toxicity of paclitaxel to folliculin-deficient renal cancer cells.

Quan Y, Lei H, Wahafu W, Liu Y, Ping H, Zhang X. Inhibition of autophagy enhances the anticancer effect of enzalutamide on bladder cancer. Biomed Pharm. Wang F, Tang J, Li P, Si S, Yu H, Yang X, et al. Chloroquine enhances the radiosensitivity of bladder cancer cells by inhibiting autophagy and activating apoptosis.

Cell Physiol Biochem. Domagala A, Stachura J, Gabrysiak M, Muchowicz A, Zagozdzon R, Golab J, et al. Inhibition of autophagy sensitizes cancer cells to Photofrin-based photodynamic therapy. Yang X, Niu B, Wang L, Chen M, Kang X, Wang L, et al.

Han W, Pan H, Chen Y, Sun J, Wang Y, Li J, et al. EGFR tyrosine kinase inhibitors activate autophagy as a cytoprotective response in human lung cancer cells.

Li J, Yang D, Wang W, Piao S, Zhou J, Saiyin W, et al. Inhibition of autophagy by 3-MA enhances ILinduced apoptosis in human oral squamous cell carcinoma cells.

Bray K, Mathew R, Lau A, Kamphorst JJ, Fan J, Chen J, et al. Autophagy suppresses RIP kinase-dependent necrosis enabling survival to mTOR inhibition. Tomic T, Botton T, Cerezo M, Robert G, Luciano F, Puissant A, et al. Metformin inhibits melanoma development through autophagy and apoptosis mechanisms.

Cell Death Dis. Xiao X, He Q, Lu C, Werle KD, Zhao RX, Chen J, et al. Metformin impairs the growth of liver kinase B1-intact cervical cancer cells. Gynecol Oncol. Villanueva Paz M, Cotán D, Garrido-Maraver J, Cordero MD, Oropesa-Ávila M, de La Mata M, et al. Targeting autophagy and mitophagy for mitochondrial diseases treatment.

Expert Opin Ther Targets. Egan DF, Shackelford DB, Mihaylova MM, Gelino S, Kohnz RA, Mair W, et al. Phosphorylation of ULK1 hATG1 by AMP-activated protein kinase connects energy sensing to mitophagy.

Roach PJ. Kim DH, Sarbassov DD, Ali SM, King JE, Latek RR, Erdjument-Bromage H, et al. mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery.

Lin X, Han L, Weng J, Wang K, Chen T. Rapamycin inhibits proliferation and induces autophagy in human neuroblastoma cells. Biosci Rep. Shi H, Zhang L, Zhang C, Hao Y, Zhao X. Rapamycin may inhibit murine S sarcoma growth by regulating the pathways associated with autophagy and cancer stem cells.

J Cancer Res Ther. Hartford CM, Ratain MJ. Rapamycin: Something old, something new, sometimes borrowed and now renewed. Clin Pharmacol Therapeutics. Sarkar S, Floto RA, Berger Z, Imarisio S, Cordenier A, Pasco M, et al.

Lithium induces autophagy by inhibiting inositol monophosphatase. J Cell Biol. Yang YP, Hu LF, Zheng HF, Mao CJ, Hu WD, Xiong KP, et al. Application and interpretation of current autophagy inhibitors and activators. Acta Pharmacol Sin. Zhou C, Gu J, Zhang G, Dong D, Yang Q, Chen MB, et al.

AMPK-autophagy inhibition sensitizes icaritin-induced anticolorectal cancer cell activity. Wu YT, Tan HL, Shui G, Bauvy C, Huang Q, Wenk MR, et al.

Dual role of 3-methyladenine in modulation of autophagy via different temporal patterns of inhibition on class I and III phosphoinositide 3-kinase. Liu Q, Shi X, Zhou X, Wang DA, Wang LI, Li C. Effect of autophagy inhibition on cell viability and cell cycle progression in MDA-MB human breast cancer cells.

Mol Med Rep. Wolpin BM, Ng K, Zhu AX, Abrams T, Enzinger PC, McCleary NJ, et al. Multicenter phase II study of Tivozanib AV and everolimus RAD for patients with refractory Metastatic Colorectal Cancer.

Chi KH, Wang YS, Huang YC, Chiang HC, Chi MS, Chi CH, et al. Simultaneous activation and inhibition of autophagy sensitizes cancer cells to chemotherapy.

Gewirtz DA. The switch between protective and nonprotective autophagy; implications for autophagy inhibition as a therapeutic strategy in cancer.

Article CAS PubMed Central Google Scholar. An autophagic switch in the response of tumor cells to radiation and chemotherapy. Biochem Pharmacol.

Chakradeo S, Sharma K, Alhaddad A, Bakhshwin D, Le N, Harada H, et al. Yet another function of p53 - The switch that determines whether radiation-induced autophagy will be cytoprotective or nonprotective: Implications for autophagy inhibition as a therapeutic strategy.

Mol Pharmacol. Li DD, Sun T, Wu XQ, Chen SP, Deng R, Jiang S, et al. The inhibition of autophagy sensitises colon cancer cells with wild-type p53 but not mutant p53 to topotecan treatment. Sharma K, Goehe RW, Di X, Hicks MA, Torti SV, Torti FM, et al.

A novel cytostatic form of autophagy in sensitization of non-small cell lung cancer cells to radiation by vitamin D and the vitamin D analog, EB Bristol ML, Di X, Beckman MJ, Wilson EN, Henderson SC, Maiti A, et al.

Dual functions of autophagy in the response of breast tumor cells to radiation: cytoprotective autophagy with radiation alone and cytotoxic autophagy in radiosensitization by vitamin D3. Wilson EN, Bristol ML, Di X, Maltese WA, Koterba K, Beckman MJ, et al.

A switch between cytoprotective and cytotoxic autophagy in the radiosensitization of breast tumor cells by chloroquine and Vitamin D. Horm Cancer. Knockdown of autophagy-related protein 5, ATG5, decreases oxidative stress and has an opposing effect on camptothecin-induced cytotoxicity in osteosarcoma cells.

Dupéré-Richer D, Kinal M, Ménasché V, Nielsen TH, Del Rincon S, Pettersson F, et al. Vorinostat-induced autophagy switches from a death-promoting to a cytoprotective signal to drive acquired resistance.

Mariño G, Niso-Santano M, Baehrecke EH, Kroemer G. Self-consumption: The interplay of autophagy and apoptosis. Li X, Xu HL, Liu YX, An N, Zhao S, Bao JK. Autophagy modulation as a target for anticancer drug discovery. Yoshii SR, Mizushima N. Monitoring and measuring autophagy. Jiang P, Mizushima N.

LC3- and pbased biochemical methods for the analysis of autophagy progression in mammalian cells. Mizushima N, Yoshimori T, Levine B. Methods in Mammalian Autophagy Research. Download references. UKM Medical Molecular Biology Institute UMBI , Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, W.

Persekutuan, , Kuala Lumpur, Malaysia. You can also search for this author in PubMed Google Scholar. SML wrote the first draft of the manuscript.

SFC and MHEA reviewed and edited the manuscript. All authors read and approved the final manuscript. Correspondence to Siok-Fong Chin. Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Open Access This article is licensed under a Creative Commons Attribution 4. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material.

If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

Reprints and permissions. Lim, S. Is targeting autophagy mechanism in cancer a good approach? The possible double-edge sword effect. Cell Biosci 11 , 56 Download citation. Received : 26 October Accepted : 08 March Published : 20 March Anyone you share the following link with will be able to read this content:.

Sorry, a shareable link is not currently available for this article. Provided by the Springer Nature SharedIt content-sharing initiative.

Skip to main content. Search all BMC articles Search. Download PDF. Review Open access Published: 20 March Is targeting autophagy mechanism in cancer a good approach?

Abstract Autophagy is a conserved cellular process required to maintain homeostasis. Introduction Cells are naturally safeguarded by an efficient check-and-balance mechanism better known as cellular homeostasis to maintain the balance of a wide array of biochemical factors and processes.

Mechanism of autophagy Autophagy is a sequential process that involves initiation, elongation, maturation, fusion and degradation [ 4 ]. Full size image. Pathways controlling autophagy Under normal physiological conditions, autophagy occurs at a basal rate to maintain cellular viability and homeostasis [ 28 , 29 ].

Autophagy in cancer Autophagy is an evolutionarily conserved process for maintaining cellular homeostasis. Autophagy as tumour suppressor During normal conditions and early stage of cancer, autophagy serves as a shield to protect cells from harmful stimuli and malignant transformation.

Autophagy as tumour promoter As the tumour develops and progresses, autophagy, in turn, fuels and supports the growth of cancer cells. Modulation of autophagy in cancer Over the last decade, autophagy has emerged as a promising target for cancer therapy.

Table 1 Genetic and pharmacological inhibition of autophagy synergize with therapeutic agents in various malignancies Full size table. Current perspectives and future outlook Autophagy has been reported to have controversial roles in cancer progression. Conclusion Targeting autophagy in precision medicine for cancer is no doubt a very attractive strategy.

Availability of data and materials Data will be provided upon request. References Cooper GM. Google Scholar Boya P, Reggiori F, Codogno P. Article CAS Google Scholar Chen HT, Liu H, Mao MJ, Tan Y, Mo XQ, Meng XJ, et al. Article PubMed PubMed Central Google Scholar Li YJ, Lei YH, Yao N, Wang CR, Hu N, Ye WC, et al.

Article CAS Google Scholar Yun CW, Lee SH. Article PubMed Central CAS Google Scholar Parzych KR, Klionsky DJ. Article CAS Google Scholar Mizushima N. Article CAS PubMed Google Scholar Kishi-Itakura C, Koyama-Honda I, Itakura E, Mizushima N.

CAS PubMed Google Scholar Zachari M, Ganley IG. Article PubMed PubMed Central Google Scholar Shibutani ST, Yoshimori T. Article CAS PubMed Google Scholar Wei Y, Liu M, Li X, Liu J, Li H. Article PubMed PubMed Central Google Scholar Hanada T, Noda NN, Satomi Y, Ichimura Y, Fujioka Y, Takao T, et al.

Article CAS PubMed Google Scholar Mizushima N, Noda T, Yoshimori T, Tanaka Y, Ishii T, George MD, et al. Article CAS PubMed Google Scholar Fujioka Y, Noda NN, Nakatogawa H, Ohsumi Y, Inagaki F. Article CAS PubMed Google Scholar Shpilka T, Weidberg H, Pietrokovski S, Elazar Z.

Article CAS Google Scholar Weidberg H, Shvets E, Shpilka T, Shimron F, Shinder V, Elazar Z. Article CAS PubMed PubMed Central Google Scholar Xie Z, Nair U, Klionsky DJ.

Article CAS PubMed PubMed Central Google Scholar Lee YK, Lee JA. Article CAS PubMed PubMed Central Google Scholar Huang R, Xu Y, Wan W, Shou X, Qian J, You Z, et al.

Article CAS PubMed Google Scholar Ichimura Y, Kirisako T, Takao T, Satomi Y, Shimonishi Y, Ishihara N, et al. Article CAS PubMed Google Scholar Kabeya Y. Article CAS PubMed PubMed Central Google Scholar Rawet Slobodkin M, Elazar Z. Article Google Scholar Kabeya Y, Mizushima N, Yamamoto A, Oshitani-Okamoto S, Ohsumi Y, Yoshimori T.

Article CAS PubMed Google Scholar Schaaf MBE, Keulers TG, Vooijs MA, Rouschop KMA. Article CAS PubMed Google Scholar Birgisdottir ÅB, Lamark T, Johansen T. Article CAS PubMed Google Scholar Yang Z, Klionsky DJ. Article CAS PubMed Google Scholar Yorimitsu T, Klionsky DJ. Article CAS PubMed Google Scholar Musiwaro P, Smith M, Manifava M, Walker SA, Ktistakis NT.

Article CAS PubMed Google Scholar Shang L, Chen S, Du F, Li S, Zhao L, Wang X. Article CAS PubMed PubMed Central Google Scholar Kroemer G, Mariño G, Levine B. Article CAS PubMed PubMed Central Google Scholar Tan VP, Miyamoto S.

Article CAS PubMed PubMed Central Google Scholar Neufeld TP. Article CAS PubMed Google Scholar Noda T, Ohsumi Y. Article CAS PubMed Google Scholar Jhanwar-Uniyal M, Wainwright JV, Mohan AL, Tobias ME, Murali R, Gandhi CD, et al.

Article CAS Google Scholar Howell JJ, Manning BD. Article CAS Google Scholar Tato I, Bartrons R, Ventura F, Rosa JL.

Article CAS PubMed Google Scholar Alers S, Loffler AS, Wesselborg S, Stork B. Article CAS PubMed PubMed Central Google Scholar Hara K, Yonezawa K, Weng QP, Kozlowski MT, Belham C, Avruch J. Article CAS PubMed Google Scholar Kim SG, Buel GR, Blenis J. Article CAS PubMed PubMed Central Google Scholar Barbet NC, Schneider U, Helliwell SB, Stansfield I, Tuite MF, Hall MN.

Article CAS PubMed PubMed Central Google Scholar Jung CH, Ro SH, Cao J, Otto NM, Kim DH. Article CAS PubMed PubMed Central Google Scholar Arsham AM, Howell JJ, Simon MC.

Article CAS PubMed Google Scholar Hardie DG, Ross FA, Hawley SA. Article CAS PubMed PubMed Central Google Scholar Xiao B, Sanders MJ, Underwood E, Heath R, Mayer FV, Carmena D, et al.

Article CAS PubMed PubMed Central Google Scholar Woods A, Dickerson K, Heath R, Hong SP, Momcilovic M, Johnstone SR, et al. Article CAS PubMed Google Scholar Willows R, Sanders MJ, Xiao B, Patel BR, Martin SR, Read J, et al. Article CAS PubMed Google Scholar Hawley SA, Boudeau J, Reid JL, Mustard KJ, Udd L, Mäkelä TP, et al.

Article Google Scholar Inoki K, Zhu T, Guan KL. Article CAS PubMed Google Scholar Kim J, Kundu M, Viollet B, Guan KL. Article CAS PubMed PubMed Central Google Scholar Høyer-Hansen M, Bastholm L, Szyniarowski P, Campanella M, Szabadkai G, Farkas T, et al. Article PubMed CAS Google Scholar Pérez-Plasencia C, López-Urrutia E, García-Castillo V, Trujano-Camacho S, López-Camarillo C, Campos-Parra AD.

Article Google Scholar Petherick KJ, Williams AC, Lane JD, Ordóñez-Morán P, Huelsken J, Collard TJ, et al. The supernatant protein concentration was determined using a bicinchoninic acid BCA protein assay kit Bio-Rad, Hercules, CA.

Each experiment was repeated at least 3 times. Densitometric analysis of the immunoreactive bands was performed using ImageJ software NIH. As an index of cell death, release into extracellular medium was measured using the LDH assay.

Briefly, supernatants were collected at the times indicated and intact cells were lysed using Triton Xcontaining lysis buffer. The amount of LDH release was determined spectrophotometrically at nm using the LDH-Cytotoxicity Assay Kit Sigma.

The quantitative data are presented as mean ± S. We first examined the dose effect of MDMA on cell viability. Cortical neuron cultures were treated with different concentration of MDMA 0.

MDMA produced a concentration-dependent decrease in cell viability Fig. Treatment with 0. When cells were treated with higher concentration of MMDA 1.

Cultured cortical neurons were treated with different concentration of MDMA for 48 h, and then cell viability was determined by MTT assay. The data are expressed as percentage of untreated controls mean ± S. We investigated the dose-dependent effect of MDMA on autophagy activity and neurite outgrowth in cultured rat cortical neurons.

Autophagy activity was assessed using immunoblotting and immunofluorescence to detect the essential autophagy protein LC3. Upon autophagy activation, the LC3-I protein localized in the cytoplasm is cleaved, lipidated, and inserted as LC3-II into autophagosome membranes.

In the present study, cortical neurons were treated with different concentrations 0. Treatment with MDMA at 0. Beclin-1 expression are known to be involved in the formation of preautophagosomal structures.

However, the expression level of beclin-1 was not affected by MDMA Fig. We then performed double immunofluorescence to correlate the LC3 expression and neurite outgrowth using anti-MAP2 a general neurite marker and anti-LC3B antibodies after 48 h of MDMA treatment.

As shown in Fig. At µM of MDMA treatment, anti-LC3B staining exhibited a more condensed diffuse distribution. After exposure to 1 mM MDMA, typical cytoplasmic LC3B punctate was formed notably in almost all neurons with shortening of total neurite length per neuron from ±18 µm control group to ±15 µm 1 mM MDMA treatment.

At higher concentrations of MDMA 1. Immunofluorescence demonstrated that MDMA triggers dose-dependent autophagosome formation accompanied by reduction of neurite outgrowth in cultured neuronal cells also see inset showing a magnified view of LC3B-labeled aggregation in MDMA-treated neuron in Fig.

Cultured cortical neurons were treated with the indicated concentrations of MDMA for 48 h, and then western blot analysis and double immunofluorescence were performed. A Western blot analysis of LC3B and beclin β-actin was used as the internal control.

Quantitative data are expressed as intensity relative to the control mean value mean ± S. C Double immunofluorescence stained images and merged images of representative cells using anti-LC3B and anti-MAP2. Intense LC3 punctate fluorescence shown in the MDMA-treated group see insets.

D Quantitation of neurite outgrowth. The total neurite length are shown as mean ± S. Although 1 mM MDMA had no obvious effect on cell viability, pharmacological actions significantly caused a decrease in neurite outgrowth and upregulation of autophagy.

Therefore, 1 mM MDMA was applied to cultures in the following experiments. We then examined the time dependent effect of MDMA on autophagy activation. The cells were treated with 1 mM MDMA for 18 h, 24 h or 48 h and subjected to western blot analysis using LC3 antibody.

The results showed that treatment with MDMA for 18, 24 and 48 h significantly induced a time-dependent increase in ration of LC3B-II to LC3B-I to 1. Double immunofluorescence staining with anti-MAP2 and anti-LC3B antibodies appeared that LC3B punctate staining was not observed within 13 h of MDMA treatment data not shown , whereas positive aggregations increased rapidly within 18 h and even more punctate staining was observed after 24 h and 48 h of MDMA treatment see inset showing a magnified view of LC3B-labeled neuronal cells in Fig.

MDMA removal for 48 h reduced autophagosome accumulation almost to the baseline, suggesting that this process is reversible data not shown. Cultured cortical neurons were treated with MDMA for different times as indicated, and then subjected to western blot analysis and double immunofluorescence staining with anti-LC3B and MAP2 antibodies.

A Western blot analysis of LC3B. β-actin was used as the internal control upper panel. B Representative double immunofluorescence and merged images using anti-LC3B and anti-MAP2 antibodies.

Intense LC3 punctate staining pattern shown in the MDMA-treated group see insets. C Quantitation of neurite outgrowth. Monodansylcadaverine MDC staining can be used to detect autophagic vacuoles AVs [29].

The autofluorescent drug MDC accumulates in AVs, but not in the early endosome compartment. Under a fluorescence microscope, AVs stained by MDC appear as distinct punctuate structures throughout the cytoplasm or within the perinuclear region.

The autophagy inhibitor 3-MA is commonly used to define the role of autophagy under various physiological conditions [30]. Pretreatment with 1 mM 3-MA effectively decreased the number 1. To further address the role of MDMA in autophagy induction, cells were pre-incubated with 3-MA for 30 min prior to induction of autophagy by MDMA for 48 h.

Cells were treated as the same above, and then double immunostaining were performed with anti-MAP2 and anti-LC3B. Interestingly, treatment with 1 mM 3-MA alone significantly promote neurite outgrowth Fig. We advance to prove 3-MA protection against MDMA-induced neuronal cell death.

Cells were pre-incubated with 3-MA for 30 min prior to different concentration of MDMA treatment for 48 h, then the neuronal death was determined by LDH assay. We further investigated the apoptotic effect of inhibition of autophagy by 3-MA after MDMA exposure.

The appearance of cleaved caspase-3 immunoreactivity is used as a marker of apoptosis. Cortical neuron cultures were treated with 2 mM MDMA in the presence or absence of 1 mM 3-MA for 24 h, and then western blot analysis and immunofluorescence were performed using antibodies against cleaved-caspase 3 and caspase 3.

Therefore, MDMA-induced neuronal death are at least partly autophagy-dependent. However, other autophagy inhibitors, wortmannin and LY, had no effect on MDMA-elicited autophagy and neurite damage data not shown.

Cultured cortical neurons were exposed to 1 mM MDMA for 48 h in the presence or absence of 1 mM 3-MA for 48 h. Autophagy activity was assay by MDC incorporation, western blot analysis and double immunofluorescence staining with anti-LC3B and MAP2 antibodies. A The autophagic vacuoles were visualized by monodansylcadaverine MDC staining and the mean number of MDC-labeled vacuoles per cortical neurons arrows was quantitation.

Quantitative data are expressed as mean ± S. B Western blot analysis of LC3B expression. Immunoblot signal ratio of LC3B-II-to-LC3B-I was quantitative by densitometric assay. C Representative double immunofluorescence and merged images using anti-LC3B and anti-MAP2 antibodies. A Cortical neuron cultures were treated with different concentrations of MDMA in the presence or absence of 1 mM 3-MA.

After 48 h, the neuronal death was determined by LDH cytotoxicity assay. The data are presented as percentage of dead cells. B Cortical neuron cultures were treated with 2 mM MDMA in the presence or absence of 1 mM 3-MA. After 48 h, the cells were immunostained with anti-NeuN antibody.

C Quantitative data of the number of NeuN-positive cells. The data are presented as percentage of the control value mean ± S. D 3-MA attenuated MDMA-induced activation of caspase 3. Cortical neuron cultures were treated with 2 mM MDMA in the presence or absence of 1 mM 3-MA for 24 h, and then western blot analysis was performed using antibodies against cleaved-caspase 3 and caspase 3.

E Representative images from cortical neuron cultures treated with 2 mM MDMA in the presence or absence of 1 mM 3-MA. After 24 h, the apoptotic cells were determined by immunostaining with anti-cleaved caspase 3.

F Quantitative data of the number of cleaved-caspase 3 positive neurons. To further clarify the detrimental role of autophagosome accumulation in MDMA-induced neurotoxicity, we employed rapamycin and Bafilomycin A1.

Cells were treated with MDMA in the presence or absence of rapamycin or Bafilomycin for 48 h and double immunostained with anti-MAP2 and anti-LC3B antibodies. However, rapamycin and Bafilomycin A1 in the presence of MDMA acted synergistically with MDMA to enhance autophagosome accumulation and neurite degeneration Fig.

Another mTOR inhibitor Torin-1 directly inhibits both mTORC1 and mTORC2. Cotreatment of MDMA with Torin-1 nM also synergistically resulted in LC3B-labed autophagosome accumulation and aggravated neurite degeneration, whereas Torin-1 alone increased LC3B immunostaining intensity slightly without affecting neurite outgrowth S1 Fig.

This result was similar to that observed in treatment with rapamycin. After treatment, the cells were performed western blot analysis or double immunofluorescence with anti-LC3B and anti-MAP2 antibodies.

A Western blot analysis of LC3B expression. The data are presented as intensity relative to the control mean value mean ± S. MDMA treatment. AMP-activated protein kinase AMPK is the key signal triggering autophagy through inactivation of mTOR and phosphorylation of ULK1 [32] , [33].

Therefore, the deactivation of mTOR triggers autophagy. To further investigate the kinase involved in MDMA-induced autophagy, levels of phosphorylated AMPK, mTOR, and ULK1 were measured. MDMA also caused a dose-dependent increase in the phosphorylation of ULK1 in Ser Fig.

Cultured cortical neurons were treated with the indicated concentrations of MDMA for 48 h, and then western blot analysis was performed using antibodies against phospho-AMPK and AMPK A , phospho-mTOR and mTOR B , phospho-ULK1 and ULK1 C.

MDMA is a neurotoxin. Autophagy activation involves in brain ischemia, trauma and neurodegeneration. In the present study, we examined the role of autophagy in MDMA-induced neurotoxicity.

Our data provide the first evidence that MDMA elicits a robust autophagic response in primary rat cortical neurons, as judged by an increase in autophagosome formation and protein expression of LC3B-II. The autophagy inhibitor 3-MA significantly reduced MDMA-induced autophagy activation and partially protected neurons against MDMA-induced neurite shortening and neuronal death.

Either enhancement of autophagosome generation by rapamycin or impairment of autophagosome clearance by Bafilomycin A1 resulted in augmented MDMA-induced autophagosome accumulation and neurite degeneration, indicating that excessive autophagy may play a detrimental role in MDMA-triggered neurotoxicity.

Furthermore, MDMA altered the phosphorylation status of AMPK and ULK1, which plays an important role in initiating autophagosome generation. Autophagy can be classified into basal and induced modes [35]. Under basal conditions in our culture system, autophagosomes and LC3-II expression were hardly detectable in healthy neurons, although the basal level of Atg proteins, such as beclin-1 and LC3-I were highly expressed.

These observations are consistent with previous observations [5]. Instead of a low level of basal autophagy, constitutive autophagy occurs in healthy neurons, and autophagic degradation is so efficient that autophagosomes cannot accumulate at detectable levels [36].

It is now well accepted that basal or low-level autophagy is cell-protective for maintenance of homeostasis and survival, while excessive or sustained autophagy can lead to self destruction or autophagic cell death, either directly or indirectly [37]. Autophagy induction and subsequent neuronal death occur in the CNS under several pathological conditions, including ischemia, trauma, glutamate-mediated excitotoxicity, and neurodegenerative protein aggregation diseases.

Autophagic flux implies a balance between autophagosome formation and autophagic degradation. Either increased autophagosome formation in the early stage or defective degradation in the late stage can lead to accumulation of autophagic vacuoles.

MDMA-induced LC3B-II accumulation could be a result of either LC3B-I to LC3B-II conversion or defects in LC3B-II degradation. In this study, 3-MA, an autophagy inhibitor at the early stage of autophagosome biogenesis, prevented MDMA-induced autophagy vacuole accumulation, suggesting MDMA may activate signaling cascades that initiate autophagosome generation.

However, it cannot be completely excluded that autophagosome accumulation might be because of organelle turnover inhibition. It is well known that MDMA induced neuron loss through apoptosis or necrosis mechanism.

In the present study, we further confirm that MDMA caused neuron loss through autophagy upregulation. Extensive or dysregulated autophagy may lead to autophagic cell death, called type II programmed cell death.

Suppression of autophagy by 3-MA can block neuronal death in response to various stress conditions in the CNS such as kainate-induced excitotoxicity in rats [40] , and rodent traumatic or ischemic brain injury [39] , [41].

Similarly, we found that 3-MA alleviated MDMA-induced neuronal death and apoptosis, suggesting autophagic cell death involves in MDMA-induced neuron loss. In addition, autophagy is a double-edged sword in neuron cell survival. There is extensive cross-talk between autophagy and other forms of cell death, such as apoptosis and necrosis.

Depending on cell types, environment and stimulation manners, autophagy can precede, inhibit or enhance apoptotic cell death and both autophagic and necrotic morphologies are observed in the same cell [43].

The discrepancies may be duo to the complex and diverse interactions among autophagy, apoptosis and cell death.

Macroautophagy, often referred to as autophagy, is a Functional strength exercises process that Augophagy in the autophagosomic-lysosomal degradation of Ajtophagy cytoplasmic actlvation, abnormal protein aggregates, Autpohagy Autophagy activation or damaged organelles. Autophagy is generally activated by conditions Autophagy activation activagion deprivation Autophagy activation has RMR and weight loss been Autophagy activation activvation physiological as well as pathological processes Autophayg as development, differentiation, neurodegenerative Autophagy activation, stress, infection, Autophagy activation cancer. The kinase mTOR is a critical regulator of autophagy induction, with activated mTOR Akt and MAPK signaling suppressing autophagy, and negative regulation of mTOR AMPK and p53 signaling promoting it. ULK1 and ULK2 form a large complex with the mammalian homolog of an autophagy-related Atg gene product mAtg13 and the scaffold protein FIP an ortholog of yeast Atg Class III PI3K complex, containing hVps34, Beclin-1 a mammalian homolog of yeast Atg6p a mammalian homolog of yeast Vps15and Atglike protein Atg14L or Barkor or ultraviolet irradiation resistance-associated gene UVRAGis required for the induction of autophagy. The Atg genes control autophagosome formation through AtgAtg5 and LC3-II Atg8-II complexes. The term Autophagy was introduced acgivation Christian Autlphagy Duve during the Autophagy activation Foundation Symposium on Lysosomes — which was Autophagy activation in Activatipn in February Autophagy activation Metabolic health solutions he was Autophagy activation with the Nobel price activatjon Autophagy activation or Medicine for Autophaagy pioneering research about peroxisomes and lysosomes. Autophagy Autophagocytosis describes the fundamental catabolic mechanism during which cells degrade dysfunctional and unnecessary cellular components. This process is driven by the action of lysosomes and promotes survival during starvation periods as the cellular energy level can thus be maintained. During classical autophagy, autophagosomes are generates, organelles which are surrounded by double membranes see Fig. The autophagosome formation is induced by class 3 phosphoinositidekinase, Atg 6 autophagy-related gene 6 and ubiquitin or ubiquitin-like modifications of the target proteins. Autophagy activation

Video

Autophagy: Exercise vs. Fasting

Author: Nicage

0 thoughts on “Autophagy activation

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com