Category: Family

Autophagy and therapeutic targeting

Autophagy and therapeutic targeting

Karsli-Uzunbas, G. Efficacy and therapsutic of sirolimus in lymphangioleiomyomatosis. Power sports nutrition tips Therapeutoc, Moreau K, Autophagy and therapeutic targeting L, Puri C, Rubinsztein DC. Ann Surg Oncol ;16 3 — Similarly, lipoteichoic acid, the major toxin from Gram-positive bacteria, also provokes the activation of neutrophils, accompanied by massive cytokine release Autophagy and therapeutic targeting

Autophagy and therapeutic targeting -

Recent data suggest that these proteins work together to mediate the clearance of dysfunctional mitochondria via mitophagy As disease-associated mutations have impaired mitophagy-related activity, this supports previous assertions that mitochondrial dysfunction may contribute to these forms of Parkinson's disease.

However, the relevance of mitophagy to the common sporadic forms of Parkinson's disease is still unclear. Furthermore, impaired autophagy is implicated in a rare degenerative form of epilepsy caused by mutations in laforin, called Lafora epilepsy 60 , as well as in forms of motor neuron disease caused by mutations in dynactin 61 , It is thus likely that neurodegenerative diseases associated with mutations that impair autophagy will have an increased propensity towards aggregate formation and cellular toxicity, and that some of the neuronal stress may be due to defective mitophagy.

Therapeutic strategies and challenges. Autophagy defects can occur at different stages of the pathway in different diseases, and this may influence treatment strategies.

Defects in autophagosome formation may be amenable to drugs that enhance autophagosome biogenesis. For example, laforin mutations causing Lafora epilepsy impair autophagosome formation by enhancing mTOR activity; mTOR inhibitors for example, rapamycin may therefore be beneficial in this context Similarly, nitric oxide induction — a frequent occurrence in neurodegenerative diseases — blocks autophagosome formation and this effect can be reversed in Huntington's disease models by N - L -arginine methyl ester L -NAME , which inhibits nitric oxide generation Indeed, L -NAME induces autophagy, enhances the degradation of mutant huntingtin and alleviates toxicity in in vivo disease models However, it may not be beneficial to induce autophagosome formation in certain disease settings: if the mutation or disease prevents the delivery of autophagosomes to lysosomes which occurs if there are mutations in the dynein apparatus 62 ; if the mutation results in impaired lysosome activity, which is observed in various lysosomal storage diseases 64 , 65 ; or in familial Alzheimer's disease caused by presenilin 1 mutations In such cases, increasing autophagosome formation will not necessarily enhance autophagic substrate degradation, and may result in cellular membrane build-up, as the newly formed autophagosome would not be efficiently delivered to lysosomes.

When the initial studies were performed to examine the possibility of upregulating autophagy to enable the clearance of intracytoplasmic aggregation-prone proteins, the only known pharmacological method of inducing autophagy chronically was using rapamycin.

However, the side effects of rapamycin which are unrelated to autophagy may make it unattractive for use in pre-symptomatic patients who may require long-term therapy. Various screens have identified pathways and compounds that regulate autophagy independently of mTOR.

For instance, imidazoline receptor agonists such as clonidine and rilmenidine induce autophagy and have protective effects in cell culture, D. melanogaster and zebrafish models of Huntington's disease Rilmenidine also has protective effects in a mouse model of Huntington's disease Rilmenidine is a safe, centrally acting antihypertensive drug that lowers blood pressure by activating imidazoline receptors in the brain which are widely distributed — the same receptors it acts on to induce autophagy.

To date, there have been no reports of deleterious effects associated with specific autophagy upregulation in vivo. Indeed, rapamycin prolongs lifespan in D. melanogaster and in rodents 67 , 68 , 69 and, at least in D. melanogaster , these effects are largely autophagy-dependent Although it is still not known whether specific upregulation of autophagy is beneficial in mice, deletion of the polyglutamine tract in the wild-type huntingtin protein induces autophagy in mice.

This zero glutamine allele is associated with enhanced lifespan in otherwise wild-type mice; it increases lifespan and decreases motor symptoms in a knock-in mouse model of Huntington's disease From a therapeutic perspective, however, constitutive autophagy induction may not be necessary.

The therapeutic regimes with rapamycin and rilmenidine in mice have used dosing protocols that are likely to result in pulsatile upregulation of autophagosome formation, with periods of normality between dosing 51 , Thus, intermittent upregulation of autophagy may be effective and associated with fewer side effects in patients.

One major challenge when considering clinical trials for neurodegenerative diseases will be the feasibility of monitoring autophagic flux not simply autophagosome numbers in the correct tissue at the correct time.

This will be a major challenge in the brain, but it will also be complex in tissues that are likely to be used for obtaining biopsy samples, such as tumours.

Simply assaying autophagy in white blood cells or other easily accessible tissues may not be informative as there may be issues associated with tissue access for drugs for example, the blood—brain barrier or tumour cores as well as different receptors on different cells; therefore, certain compounds that act in the brain may have no effects in white blood cells.

Cancer is both the first disease that was linked to a deficiency in autophagy 70 and, somewhat paradoxically, the first disease for which clinical trials are being carried out in patients to inhibit autophagy This apparent paradox can perhaps be reconciled by the differential effects of autophagy in different stages of tumorigenesis.

The prevailing current view is that autophagy functions both as a tumour suppressor pathway that prevents tumour initiation and as a pro-survival pathway that helps tumour cells endure metabolic stress and resist death triggered by chemotherapeutic agents The notion of autophagy as a potential target in cancer has been recently discussed in the literature reviewed in Refs 71 , 72 , 73 , Here, we briefly highlight key discoveries and central controversies.

Tumour initiation. Although the role of autophagy in tumour progression is complex, there is a general consensus that autophagy suppresses tumour initiation. Genetic deletion of the autophagy gene BECN1 is associated with enhanced susceptibility to breast, ovarian and prostate cancer in humans 70 , 75 , and increased spontaneous malignancies in mice 76 , Mice deficient in ATG4C show increased susceptibility to chemically-induced fibrosarcomas 78 , and mice with deletion of Atg5 or Atg7 develop benign liver tumours Besides this direct evidence that autophagy gene mutations promote tumorigenesis, there is also a strong overlap between oncogenic signalling activation and suppression of autophagy reviewed in Ref.

Conversely, mTOR-activating oncogenic signals — such as oncogenic receptor tyrosine kinases, class I PI3Ks and AKT — inhibit autophagy. Other tumour suppressors such as death-associated protein kinase 1 and cyclin-dependent kinase inhibitor 2A also known as p19 ARF and oncogenes such as BCL-2 positively or negatively regulate autophagy, respectively, through their effects on BECN1 Refs 81 , However, the most commonly mutated tumour suppressor gene in human cancer, cellular tumour antigen p53, has both positive effects on autophagy as a nuclear transcription factor and negative effects through its cytoplasmic actions, including an interaction with the human orthologue of yeast Atg17, RB1-inducible coiled-coil 1 RB1CC1; also known as FIP 83 , Mutant forms of p53 that accumulate in the cytoplasm in human cancers suppress autophagy Three recent studies have shown that autophagy is essential for RAS-induced malignant cell transformation 90 , 91 , By contrast, another study showed that autophagy gene knockdown enhanced clonogenic survival in cells expressing oncogenic RAS It is not yet known whether the requirement for autophagy in the RAS-driven transformation observed in most studies is a unique feature of cells with activated RAS or whether it will also be observed in the context of other activated oncogenes.

One proposed hypothesis is that loss of mitochondrial function mediated by oncogenic RAS-induced mitophagy overcomes the cellular energy deficit resulting from glucose insufficiency Even though autophagy may be required for cellular transformation by RAS and potentially other oncogenes , there is little doubt that basal autophagy is a bona fide tumour suppressor pathway.

Although the precise mechanisms by which autophagy mediates tumour suppression are not completely understood, several pathways are likely to have a contributing role reviewed in Ref.

Many of these relate to the ability of autophagy to remove damaged organelles — especially mitochondria — that generate ROS, which in turn promote genotoxic stress as well as pro-inflammatory and pro-tumorigenic signalling. Tumours formed by autophagy-deficient cells with bi-allelic loss of Atg5 or monoallelic loss of Becn1 display genomic instability and DNA damage, which is in part mediated by ROS 93 , In addition, PARK2 — which encodes an E3 ubiquitin protein ligase that is involved in mitophagy — is a tumour suppressor gene that is frequently mutated in colon cancer, lung cancer and glioblastoma 95 , However, it is not yet known whether its tumour suppressor effects are due to its role in mitophagy or due to mitophagy-independent E3 ligase functions.

The accumulation of p62 in autophagy-deficient cells promotes tumorigenesis through a mechanism that is postulated to involve the role of p62 as a scaffold protein that functions in the activation of the transcription factors nuclear factor-κB and NFE2-related factor 2 Refs 97 , In addition, p62 interacts with and activates the mitogenic signalling and autophagy-suppressive molecule mTOR Interestingly, SMAD ubiquitylation regulatory factor 1 SMURF1 — a pinteracting protein that also has a role in mitophagy — is amplified in pancreatic carcinomas , , and its genetic knockdown in human pancreatic carcinoma cells leads to reduced tumour cell invasion and anchorage-independent growth Tumour progression.

There is increasing evidence that autophagy may be necessary for tumour progression. The retention of a wild-type Becn1 allele in tumours arising in mice with heterozygous deletion of Becn1 Ref.

This concept has been further underscored by the recent observation that hepatic deletion of Atg7 or Atg5 results in benign but not malignant hepatic tumours However, an alternative explanation is that the background mouse strain may not develop the additional mutations needed for malignant transformation.

For example, heterozygous Becn1 -deficient mice develop pre-neoplastic mammary lesions but not mammary carcinomas, even though Becn1 allelic loss is associated with breast cancer in humans and other malignancies in mice 70 , 76 , Nonetheless, several other studies also support a role for autophagy in promoting the growth of established tumours in vivo.

Bi-allelic deletion of Atg5 or Atg7 impaired tumour growth of RAS-transformed immortalized infant mouse kidney epithelial cells in nude mice 90 , and Atg5 short hairpin RNA shRNA impaired tumour growth of human pancreatic ductal adenocarcinoma cells in a mouse xenograft model The concept underlying these phenotypes is that autophagy provides a survival advantage to tumour cells by enabling them to overcome the metabolic stress that is inherently present in the tumour microenvironment.

Consistent with this notion, autophagy is induced by cellular stress — including nutrient, growth factor and oxygen deprivation — and functions to maintain the survival of normal cells, organisms as well as tumour cells in such settings reviewed in Ref. The pro-oncogenic function of autophagy in established cancer may be context-dependent; not all data are consistent with such a role for autophagy in established tumours.

Although autophagy upregulation, in part mediated by the effects of hypoxia-inducible factor 1α HIF1α on BNIP3 Ref. In a mouse melanoma xenograft model, heterozygous Becn1 -deficient mice display a more aggressive tumour phenotype with increased angiogenesis under hypoxic conditions through a mechanism that is postulated to involve the upregulation of HIF2α but not HIF1α.

Furthermore, several clinicopathological studies have shown a correlation between levels of BECN1 expression and cancer prognosis; low levels of BECN1 expression are associated with worse cancer prognosis in gastric cancer , colorectal cancer , pancreatic cancer , oesophageal cancer , chondrosarcoma and breast cancer , whereas high levels of BECN1 expression are associated with improved survival in high-grade gliomas , hepatocellular carcinomas and B cell lymphomas Although it is not known whether low levels of BECN1 expression directly correlate with low levels of autophagy in these tumours, such studies point to the need for further careful analyses of the relationship between levels of autophagy and tumour progression in different types of tumours.

To address this question, there is an urgent need for in vivo models in which autophagy can be selectively regulated at defined stages of tumorigenesis. One caveat in the use of such models is that complete inhibition of autophagy which results in cell death may not accurately reflect those phenotypes that are associated with partial inhibition of autophagy.

Autophagy inhibition in cancer therapy. The evolutionarily conserved role of autophagy in promoting cell survival during metabolic stress has stimulated research to determine whether autophagy may promote therapeutic resistance to cytotoxic therapy.

The first in vivo data that affirmed this concept involved the demonstration that treatment with the lysosomotropic inhibitor chloroquine which inhibits autophagosome degradation enhanced the ability of p53 activation or alkylating agents to induce tumour regression in a mouse model of MYC-induced lymphoma However, chloroquine has activities on lysosomal processes that are distinct from autophagy as well as lysosome-independent processes such as DNA intercalation.

There have now been several studies indicating that autophagy inhibition — with 3-methyladenine treatment, genetic knockdown of autophagy genes or chloroquine or hydroxychloroquine treatment — sensitizes tumour cells to cell death induced by diverse cytotoxic agents reviewed in Refs 71 , 72 , 74 , Although many chemotherapeutic agents induce autophagy at least indirectly by inducing cellular stress , most efforts have focused on using autophagy inhibitors in tumour cell lines with high levels of basal autophagy such as those with oncogenic RAS mutations or on using autophagy inhibitors in conjunction with agents that directly stimulate autophagy signalling pathways such as mTOR inhibitors, dual PI3K and mTOR inhibitors, epidermal growth factor receptor EGFR inhibitors and proteasome inhibitors.

The poor prognosis of certain tumours such as pancreatic carcinoma with oncogenic RAS mutations, coupled with the preclinical data suggesting a role for autophagy in RAS-mediated transformation and tumour growth 86 , , has led to the initiation of early-phase trials of lysosomal inhibitors in patients with RAS-driven tumours reviewed in Ref.

In addition, more specific inhibitors of the autophagic machinery for example, PIK3C3 inhibitors, ATG4B inhibitors and ATG7 inhibitors are in preclinical development for potential use in cancer clinical trials.

Pharmacological inhibition of autophagy adaptor proteins such as p62 that have a role in pro-tumorigenic signalling, and perhaps of other components of the p62 autophagy adaptor complex, may represent novel strategies for cancer therapy.

Although the rationale for such studies is supported by strong preclinical data, many open questions and controversies remain regarding autophagy as a target in cancer therapy. First, although the available data support the concept that cancer cells exhibiting increased autophagy die in response to lysosomal inhibitors, no study has unequivocally demonstrated that the cytotoxic effects of these agents in cancer cells arise specifically from autophagy inhibition rather than from another effect on lysosomal function or even from lysosome-independent effects.

Alternatively, lysosomal inhibitors may potentiate cytotoxicity in cancer cells via autophagy-independent mechanisms. Indeed, some recent studies in glioma and breast cancer cells illustrate a mechanistic dissociation between the actions of lysosomotropic agents and autophagy inhibition in governing sensitivity to cytotoxic therapy.

For example, bafilomycin A1 enhanced the cytotoxicity of temozolomide a DNA alkylating agent in glioma cells , whereas shRNA-mediated knockdown of BECN1 and ATG5 protected the same cells against temozolomide-induced death Chloroquine sensitized breast cancer cells to death induced by a DNA-damaging agent, a PI3K inhibitor or an mTOR inhibitor, but similar effects were not observed in these cells with Atg12 or Becn1 knockdown or following treatment with a different lysosomal inhibitor, bafilomycin A1 Ref.

Second, there is still controversy as to whether autophagy may represent a mechanism of cell death during chemotherapy. Yet, there are numerous studies demonstrating that genetic knockdown of autophagy genes blocks tumour cell death induced by oncogenic RAS or by various chemotherapeutic agents reviewed in Refs 72 , , ; one common theme is that cell death via autophagy is more likely to occur in tumour cells that are deficient in apoptosis or in tumour cells treated with a combination of pan-BCL-2 inhibitors such as gossypol or GX and other agents.

Further studies are required to delineate more precisely the chemotherapeutic contexts in which autophagy functions as a pro-survival or pro-death mechanism. Some other potential caveats associated with autophagy inhibition in cancer therapy also warrant consideration. Given the tumour suppressor effects of autophagy discussed above and the protective effects of autophagy in other diseases such as neurodegeneration and infectious diseases, as well as in ageing , there are concerns about whether autophagy inhibition treatment may increase the incidence of secondary tumours or other diseases in patients.

Thus, even if short-term benefits on tumour progression are observed, a long duration of patient follow-up may be required before increased secondary malignancies and other adverse effects with a long latency period , if they occur, are detected.

The drugs chloroquine and hydroxychloroquine have been used extensively in the treatment of malaria and systemic lupus erythematosus, and are fairly well tolerated; however, it is not known whether more proximal inhibitors of autophagy will have similar safety profiles.

Nonetheless, a recent study raises the concern that acute inhibition of autophagy may limit chemotherapy responses by preventing autophagy-dependent anticancer immune responses Autophagy-competent but not autophagy-deficient cells have been shown to release cellular ATP and recruit immune cells into the tumour bed, leading to effective immunogenic cell death and chemotherapeutic responses in mice with intact immune systems.

Autophagy activation in cancer therapy. Certain agents that are used as a preventive form of cancer therapy such as the use of tamoxifen in patients who are at risk of developing familial breast cancer induce autophagy Certain epidemiological factors associated with increased cancer incidence reduce autophagy.

For example, vitamin D is a potent inducer of autophagy , , , and patients with low vitamin D levels exhibit an increased risk of developing breast, colon, prostate and other cancers Conversely, certain epidemiological factors associated with decreased cancer risk can increase autophagy.

For example, exercise induces autophagy 38 , and patients who regularly exercise more than minutes per week of moderate-intensity exercise have a decreased risk of breast, prostate, endometrial and colon cancer , , Furthermore, the use of metformin an AMPK activator and autophagy-inducing agent; Fig.

At present, the association between interventions that reduce cancer incidence and those that induce autophagy is only correlative. Further studies are required to determine whether autophagy upregulation has a mechanistic role in the efficacy of such cancer prevention strategies.

If so, the use of more direct activators of autophagy may be a feasible, new and alternative cancer prevention strategy. Future directions. Ultimately, the question of whether autophagy represents a useful target in cancer prevention or cancer treatment will need to be addressed by conducting clinical trials in patients.

Preclinical data have been useful in formulating testable hypotheses; however, there are several conflicting reports regarding the role of autophagy in tumour initiation, tumour progression and cell death decisions during chemotherapy.

Some of these may ultimately be reconciled by a better understanding of the diverse set of roles that autophagy has in different oncogenic contexts and in different stages of tumorigenesis. However, there are also limitations to studies in tumour cell lines, mouse xenograft models and targeted mutant mouse models, as well as problems with respect to the specificity of approaches used to modulate and measure autophagy, which may account for some of the apparent discrepancies.

If the results of early trials are promising, it will be important to determine the antitumour mechanisms of action of lysosomal inhibitors. Further preclinical and clinical studies are also warranted to explore the role of autophagy upregulation in cancer prevention, the feasibility of blocking p62 in autophagy-deficient tumours and the possibility of exploiting autophagy as a death pathway in tumour cells.

Another important area of future investigation will be to determine whether tumour cell autophagy-dependent survival can be selectively inhibited in tumour cells while bypassing the potential adverse effects of systemic autophagy inhibition by targeting tumour cell-specific autophagy activation, such as the platelet-derived growth factor receptor PDGFR -induced promotion of HIF1α-dependent hypoxia-selective autophagy Moreover, it is likely that emerging research regarding the role of autophagy in cancer stem cells, in epithelial-to-mesencyhmal transition, in DNA damage and cell cycle control, in the regulation of inflammatory signalling and in other aspects of cancer biology will further shift the existing paradigms that dictate our current understanding of autophagy as a target in cancer therapy.

Autophagic machinery is used in a multipronged defence against microorganisms, including via the selective delivery of microorganisms to degradative lysosomes a process referred to as xenophagy and via the delivery of microbial nucleic acids and antigens to endolysosomal compartments for the activation of innate and adaptive immunity , , In , enforced neuronal expression of the autophagy gene Becn1 was shown to protect mice against lethal alphavirus encephalitis , providing the first clue that autophagy upregulation may be beneficial in the treatment of infectious diseases It is now known that numerous medically important pathogens are degraded in vitro by xenophagy, including: bacteria such as group A Streptococcus pyogenes , Mycobacterium tuberculosis , Shigella flexneri , Salmonella enterica , Listeria monocytogenes and Francisella tularensis ; viruses such as herpes simplex virus type 1 HSV-1 and chikungunya virus; and parasites such as Toxoplasma gondii , There are also in vivo data indicating that autophagy genes have a protective role against numerous pathogens, including L.

monocytogenes , M. tuberculosis , S. enterica , T. gondii , HSV-1, Sindbis virus and chikungunya virus , , , , , Moreover, there are emerging links between host genes that regulate autophagy and host susceptibility to M. tuberculosis infection , Based on these studies, there is a strong possibility that pharmacological agents that increase autophagy may be effective therapeutic agents for treating certain intracellular bacterial infections, parasitic infections and viral infections.

In support of this concept, vitamin D treatment has been shown to inhibit both HIV and M. tuberculosis replication in human macrophages through an autophagy-dependent mechanism , In addition, the antimycobacterial action of standard antituberculous agents is associated with autophagy induction , raising the possibility that some drugs that are already in clinical use for the treatment of certain infections may be acting, at least in part, via autophagy.

Innate immunity. In addition to enhancing pathogen degradation, the upregulation of autophagy may facilitate optimal regulation of innate immune signalling and enhancement of antigen presentation , , , The interrelationship between autophagy and innate immune signalling is complex; in some contexts, autophagy can enhance type I interferon IFN production and innate immune responses by delivering viral nucleic acids to endosomal Toll-like receptors, whereas in other contexts autophagy proteins prevent the innate immune response from being excessive and detrimental, either through direct protein—protein interactions with innate immune signalling molecules or indirectly by controlling cellular levels of ROS production.

Despite this complexity, a consensus is emerging that autophagy has a central role in titrating the innate immune response so that it is adaptive rather than maladaptive Thus, upregulation of autophagy may be useful in increasing antimicrobial innate immunity while preventing excessive inflammatory responses that can be destructive to the host during infection.

Adaptive immunity. With respect to adaptive immunity, autophagy is involved in the delivery of endogenously synthesized microbial antigens to MHC class II antigen-presenting molecules, leading to the activation of CD4 T lymphocytes , In addition, the autophagy gene ATG5 is required for dendritic cells to process and present extracellular microbial antigens for MHC class II presentation It has been reported that autophagy also enhances the presentation of endogenous viral antigens on MHC class I molecules , and that autophagy in tumour cells is essential for antigen cross-presentation by dendritic cells The link between autophagy and CD4 T cell responses suggests that pharmacological induction of autophagy may be beneficial not only in the treatment of acute infection by enhancing xenophagy, innate immunity and adaptive immunity but also in enhancing vaccine efficacy.

In support of this principle, the targeting of the influenza virus matrix protein 1 MP1 to autophagosomes via fusion with LC3 enhanced anti-MP1 CD4 T cell responses , and mice immunized with rapamycin-treated dendritic cells infected with bacille Calmette—Guérin BCG or candidate mycobacterial vaccine strains showed enhanced CD4 T cell-mediated protection when they were challenged with virulent M.

tuberculosis Further studies are warranted to determine whether strategies to augment autophagy-dependent adaptive immune responses could be beneficial in vaccine development. In principle, this may be accomplished in various ways: by enhancing autophagy in cells infected with live attenuated vaccines; by targeting microbial antigens to the autophagosome via fusion with LC3 or an autophagy adaptor or receptor molecule that binds to LC3; or by administering antigens that are processed by autophagy-dependent dendritic cells for MHC class II presentation.

Autophagy-independent functions of autophagy proteins in immunity. There is increasing evidence that autophagy proteins may exhibit diverse functions in innate and adaptive immunity, independently of the autophagy pathway reviewed in Refs , , Abnormalities in some of these functions, such as aberrant regulation of the inflammasome and defects in granule cell exocytosis in Paneth cells , are thought to be relevant to the pathogenesis of subtypes of Crohn's disease a type of inflammatory bowel disease that are associated with a polymorphism in the autophagy gene ATG16L1.

At present, it is not known whether general stimulation of autophagy will increase the autophagy-independent functions of autophagy proteins in immunity.

A more likely long-term strategy may be to devise new agents that mimic the beneficial autophagy-independent functions of autophagy proteins in the control of infectious diseases and autoinflammatory diseases. Microorganisms co-opt autophagy. In parallel with increasing evidence that autophagy has a role in host defence, there is a growing list of viruses and intracellular bacteria that exploit the host autophagic machinery to enhance their own replication reviewed in Refs , These include medically important viruses such as HIV, hepatitis C virus HCV , hepatitis B virus HBV , picornaviruses and Dengue virus, as well as intracellular bacteria such as F.

tularensis , Coxiella burnetii and Brucella abortus , , , , Of note, for some viruses such as HIV there is evidence for both an anti- and proviral function of autophagy , , Until recently, all evidence indicating a promicrobial function of autophagy stemmed from studies in cultured mammalian cell lines.

However, recent studies have demonstrated reduced HBV DNA replication in mice with liver-specific knockout of Atg5 Ref. For the list of pathogens that exploit the host autophagic machinery for enhanced replication, the potential dangers of autophagy upregulation or the potential benefits of autophagy inhibition will probably depend on whether the specific pathogen utilizes the complete autophagic pathway or just certain components of the autophagic machinery to enhance its intracellular replication or survival, as well as whether pharmacological targeting manipulates the whole autophagic pathway or only specific components of the autophagic machinery required for pathogen replication.

For example, intracellular Brucella abortus survives by promoting the formation of B. abortus -containing vacuoles, which requires the activity of the autophagy-initiating proteins ULK1, BECN1, ATG14L and PIK3C3, but not the activity of the autophagy elongation proteins ATG5, ATG16L1, ATG4B, ATG7 and LC3B In this scenario, one might predict that inhibitors of PIK3C3 or signals upstream of ULK1 may exert protective functions, whereas such effects would not be observed with inhibitors of the autophagy protein conjugation systems or inhibitors of autophagosome maturation.

By contrast, Dengue virus replication is thought to be enhanced by autophagy-dependent lipid metabolism, which increases cellular β-oxidation and generates ATP In this scenario, blocking autophagy at any stage in the pathway might suppress viral replication.

It is prudent to exercise caution in considering the use of autophagy-inducing agents for the treatment of patients with infections that may otherwise be alleviated by autophagy upregulation such as tuberculosis if these patients are also co-infected with pathogens that may exploit the autophagy pathway such as chronic concurrent infections with HCV, HBV and possibly HIV.

Another potential concern is the possibility that excess levels of autophagy may exacerbate certain infectious diseases through alternative mechanisms that are independent of pathogen replication. In a recent report, the H5N1 pandemic strain of influenza virus induced autophagic alveolar epithelial cell death, and pharmacological and genetic inhibition of autophagy ameliorated acute lung injury and decreased mortality in H5N1-infected mice without affecting viral replication Promising future therapeutic strategies.

A promising future direction in the treatment of infectious diseases is the development of agents that block the activity of specific microbial gene products that antagonize the functions of autophagy or autophagy proteins in antimicrobial host defence.

Several viral virulence gene products have been shown to block either autophagy initiation or autophagolysosome maturation through their interaction with BECN1 reviewed in Refs , These include: the HSV-1 neurovirulence protein ICP The interaction of some of these viral proteins with BECN1 has been shown to be important in viral pathogenesis in mice; a mutant HSV-1 lacking the BECN1-binding domain of ICP These studies suggest that inhibitors of the interactions between viral proteins that antagonize autophagy and their host autophagy protein targets may be useful for treating diseases such as HSV-1 encephalitis, in which viral antagonism of autophagy is essential for viral virulence.

A related concept emerges from the identification of specific bacterial virulence factors — such as L. monocytogenes actin assembly-inducing protein ActA and Shigella flexneri IscB — that enable intracellular bacteria to escape recognition by the autophagic pathway reviewed in Ref.

Presumably, inhibition of these bacterial virulence factors would enhance xenophagic degradation of intracellular bacteria and thereby protect the host against diseases caused by such pathogens.

The strategy of developing pharmacological inhibitors of microbial antagonists of autophagy is attractive not only because of its potential efficacy in controlling certain viral and intracellular bacterial infections but also because of its potential safety and specificity in comparison with general strategies to modulate autophagy.

By targeting specific microbial virulence factors, such approaches might avoid the potential pitfalls associated with manipulation of systemic autophagy. Another approach towards harnessing the autophagic pathway for the treatment of infectious diseases is emerging from our rapidly expanding understanding of the machinery involved in the recognition and targeting of viruses and intracellular bacteria to the autophagosome.

In addition, SMURF1 binds to alphavirus nucleocapsid and targets it to autophagosomes through a mechanism that may involve its C2 phospholipid-binding domain Moreover, the function of certain microbial adaptor proteins can be regulated by phosphorylation; for example, TANK-binding kinase 1 phosphorylates optineurin, thus enhancing its LC3-binding affinity and the autophagic clearance of cytosolic S.

enterica This suggests that strategies to augment the activity of optineurin or other autophagy adaptors may be effective in enhancing the xenophagic degradation of intracellular pathogens. Future research to further delineate the mechanisms of regulating the function of autophagy adaptors should lay the groundwork for the preclinical development of agents that selectively enhance microbial autophagy for the treatment of certain infectious diseases.

One potential caveat of this approach is that many of the autophagy adaptors are not specific for pathogens and they also function in the selective autophagy of damaged mitochondria and other host cell components; thus, such strategies may have additional unwanted effects on host cell function.

Considerations for clinical trials. Given the central role of the autophagic machinery in controlling infection and immunity either through the classical autophagy pathway or through autophagy-independent functions of autophagy proteins , it will be important for all clinical trials using autophagy inhibitors to carefully monitor the incidence, prevalence and severity of infectious diseases, autoimmune diseases and inflammatory diseases.

Conversely, patients enrolled in clinical trials with autophagy inducers, such as patients who are at risk of developing neurodegenerative diseases, may experience protection against mycobacterial infections and other infectious diseases in which autophagy has a central role in host defence.

For medical purposes, it may be valuable to identify drugs that induce or inhibit autophagy some examples from above are summarized in Table 2 , their sites of action illustrated in Figs 1 , 2 and possible uses listed in Table 3. Notably, many of the compounds being considered are still under investigation or are tool compounds and may not therefore be suitable for clinical use, although they do illustrate potentially druggable points in the autophagic pathway.

Inducers may have particular value in certain neurodegenerative diseases, some infectious diseases and in cytoprotection. As discussed above, it has been suggested that autophagy inhibition may be valuable in cancers.

However, this is largely based on studies using the lysosomotropic drug chloroquine or its derivatives, which affect all acidic compartments in cells and have many autophagy-independent effects Some of the agents discussed are in clinical trials for example, lysosomotropic drugs for certain cancers and rilmenidine for Huntington's disease ; however, a deeper understanding of the roles of autophagy and autophagy modulators is still required for many of the conditions in which there may be therapeutic possibilities.

Furthermore, as many of these drugs have autophagy-independent effects, one may need to be cautious before inferring that all of their effects are autophagy-dependent although this may not necessarily preclude the consideration of these drugs for therapeutic purposes.

mTOR complex 1. Drugs or signals that modulate autophagy can be considered in two categories, depending on whether or not they act via mTOR Fig. mTORC1 is inhibited by rapamycin and its analogues CCI, RAD and AP , which induce autophagy in yeast, mammalian cell lines, primary cultures and in vivo Studies have revealed that rapamycin does not inhibit mTORC1 completely, leading to the concept that certain functions of mTORC1 are rapamycin-resistant such as cap-dependent translation and the phosphorylation of eukaryotic translation initiation factor 4E-binding protein 1 EIF4EBP1; also known as 4EBP1.

These studies have identified two selective ATP-competitive small-molecule mTOR inhibitors, PP and Torin 1, that directly inhibit both mTORC1 and mTORC2 Ref.

The maximal autophagy induction mediated by ATP-competitive inhibitors appears to be higher than that seen with rapamycin, although rapamycin can induce autophagy at lower concentrations than ATP-competitive inhibitors in certain cell types The activity of mTOR is regulated by the class I PI3K—AKT pathway.

PI is a molecule within the new class of dual mTOR and class I PI3K inhibitors. This combinatorial inhibition appears to be very effective as it addresses the negative feedback mechanism between mTORC1 and the PI3K—AKT pathway: rapamycin inhibits mTORC1 but this releases the negative feedback mechanism such that PI3K—AKT signalling is enhanced , As expected, PI has been shown to be a strong inducer of autophagy Although PI itself does not appear to have suitable structural properties for clinical development, it could stimulate the development of this new class of dual inhibitors It is important to note that mTOR affects many processes that are distinct from autophagy, and AKT inhibition may result in increased susceptibility to cell death.

Although this may be attractive in the context of treating cancers, AKT inhibition may prove to be a liability if it is used for treating neurodegenerative diseases. Targeting AMPK activity.

As discussed above, AMPK activates autophagy via at least two mechanisms: mTOR inhibition and direct ULK1 activation Fig. Metformin, a widely used antidiabetic agent, activates AMPK and induces autophagy As discussed above, it is possible that some of the effects of this drug in diabetes may be mediated via autophagy, and the use of metformin may therefore be worth considering for conditions in which autophagy upregulation is beneficial.

Phosphatidylinositol signalling pathway. Intracellular inositol and inositol-1,4,5-trisphosphate Ins 1,4,5 P 3 levels negatively regulate autophagy via an mTOR-independent mechanism Fig. Autophagy can be induced via this pathway using various mood-stabilizing drugs such as lithium, carbamazepine and valproic acid However, one should bear in mind that these drugs have additional targets; for example, valproic acid is a histone deactylase inhibitor.

Interestingly, carbamazepine has been shown to have beneficial effects in a mouse model of α1 antitrypsin deficiency, and these effects may be at least partly mediated via autophagy induction As many of the drugs acting on this pathway are used in the clinic for example, clonidine and rilmenidine have been used to treat hypertension, and verapamil is used to treat hypertension, cluster headaches, cardiac arrhythmias and angina and have favourable safety profiles, they may represent a tractable set of compounds for therapeutic applications in diseases that may benefit from autophagy induction.

Other autophagy-modulating drugs. In addition to the compounds described above, numerous other compounds have been described that induce autophagy, including resveratrol , spermidine , EGFR antagonists , BH3 mimetics which decrease the inhibitory interactions of BCL-2 and BCL-X L with BECN1 29 , and L -NAME 63 which blocks nitric oxide formation.

The precise mechanisms by which some of these compounds for example, L -NAME induce autophagy are still unclear, and many of these drugs and their targets affect processes that are distinct from autophagy.

Conversely, chloroquine is a lysosomotropic agent that impairs autophagosome degradation as well as other lysosomal degradation pathways. Furthermore, lysosomotropic agents have effects on diverse cellular compartments that require an acidic pH, and this may cause complex perturbations in cells in addition to autophagy arrest; however, the possible enhanced toxicity mediated by such drugs that have both autophagy-blocking and other effects may be beneficial in certain cancers.

Drugs with unforeseen autophagy-blocking effects. In addition to studies that have revealed autophagy-modulating drugs of potential therapeutic benefit in various diseases, similar studies have identified drugs that have unforeseen and possibly toxic autophagy-inhibiting effects.

In many neurodegenerative diseases, such as Huntington's disease, oxidative stress is increased, and antioxidants have therefore been proposed as a rational therapeutic strategy. Indeed, there are at least two major trials of antioxidants underway for Huntington's disease.

However, a diverse range of antioxidant drugs inhibit autophagy in vivo but exacerbate mutant protein aggregation and toxicity in animal models Thus, the potential benefits of ROS scavengers in neurodegenerative diseases may be compromised by their autophagy-blocking properties.

An alternative strategy that has been considered for neurodegenerative diseases associated with intracellular aggregate formation is to boost the chaperone activity of cells to decrease protein aggregation.

This can be achieved using heat shock protein 90 HSP90 inhibitors, such as geldanamycin, which induce HSP chaperones via the heat shock response Recent data suggest that HSP90 inhibition may impair autophagy , which may partly explain the minimal efficacy of this strategy in a mouse model of Huntington's disease Another drug that has been identified as an autophagy inhibitor is the macrolide antibiotic azithromycin, which has been widely used for its anti-inflammatory properties in patients with cystic fibrosis and, similarly to bafilomycin A1, can block autophagosome maturation 69 , Recent epidemiological evidence indicates that the use of azithromycin in patients with cystic fibrosis may be accompanied by an increasing incidence of drug-resistant non-tuberculous mycobacterial infection 69 , , Moreover, azithromycin inhibited intracellular killing of drug-resistant mycobacteria in macrophages and enhanced pulmonary disease in drug-resistant mycobacteria-infected mice through a mechanism that is postulated to involve azithromycin-mediated blockade of lysosomal acidification, autophagic maturation and phagosomal degradation Although further studies are warranted to confirm a causal relationship among chronic azithromycin use, blockade of lysosomal acidification and patient susceptibility to drug-resistant mycobacterial infections, these data highlight the need for cautious monitoring of the prevalence of infectious diseases in patient populations that receive chronic treatment with lysosomotropic agents such as patients in oncology trials with hydroxychloroquine.

Autophagy drug screens. In recent years there has been a rapid increase in reports in the literature on drugs that affect autophagy. Although this has revealed many drugs of potential clinical utility, it is important to review studies carefully before any conclusions are made.

The tools we use to measure autophagy are imperfect and frequently require multiple orthogonal assays to allow robust conclusions to be made about their effects. For instance, many screens have relied on LC3 vesicle count as a primary read-out, as this correlates with autophagosome numbers. This count can increase when autophagy is induced if autophagosome formation exceeds degradation Conversely, LC3 vesicle numbers also increase if autophagosome degradation is impaired.

Another commonly used read-out for autophagy studies involves measurements of the levels of p62, an endogenous autophagy substrate. Analyses of p62 levels may be confounded by agents that regulate its transcription This caveat may be addressed by using stable inducible cell lines overexpressing p62 and by measuring p62 clearance after switching off transgene expression Comparisons between different reported screens have yielded both concordant and discordant findings reviewed in Ref.

In some cases, apparently discrepant results can be explained by LC3-based assays that have not accounted for the effects of flux and have misinterpreted LC3 vesicle counts or increases in LC3-II as being simply due to increased autophagosome formation. Other reasons for discrepant results include the fact that drugs can have different effects on autophagy if different concentrations are used; for instance, a drug that induces autophagy at a low concentration at which it has relatively good target specificity could block autophagy at a high concentration as a result of additional targets being engaged.

Recent compound screens have revealed important insights into the biology of autophagy. For instance, 'spautin-1' specific and potent autophagy inhibitor 1 has been identified and shown to inhibit autophagy by blocking two proteins, ubiquitin-specific peptidase 10 USP10 and USP13, that target the BECN1-containing PIK3C3 complex, thereby enhancing the degradation of PIK3C3 complexes BECN1 also regulates the stability of the proteins USP10 and USP13, which might explain the novel observation that BECN1 regulates p53 levels, as USP10 mediates p53 deubiquitylation.

These data suggest that BECN1 haploinsufficiency may contribute to tumorigenesis by reducing the levels of the tumour suppressor gene p For many compounds that have been reported to modulate autophagy, the targets are still unclear.

It is therefore desirable to confirm whether the proposed targets of autophagy-modifying drugs act as one would predict when they are modulated genetically for example, by knockdown or overexpression ; it is also desirable to ascertain whether the drugs modulate target activity at physiological concentrations, and whether the drugs act primarily via a particular target for example, by showing that the drugs do not modulate autophagy when the target is knocked down or knocked out.

The development of more specific autophagy modulators is vital, both for therapeutic applications and for their use as chemical probes to allow the acute modulation of this process for cell biology and physiological studies. Such reagents will be crucial for inferring the direct effects of autophagy on biological and disease processes by limiting the influences of autophagy-independent effects.

Various strategies have been proposed. The first involves inhibitors of the class III PI3K PIK3C3 , the crystal structure of which exhibits a smaller ATP-binding pocket than that of class I PI3K isoforms , indicating that structure-based design may be used to develop compounds that have considerably higher selectivity for class III PI3Ks than for class I PI3Ks.

The two ubiquitin-like conjugation systems that are crucial for the structural formation of autophagosomes may also be amenable to the development of specific inhibitors. ATG7 an E1-activating enzyme , ATG3 and ATG10 E2-conjugating enzymes , and the ATG12—ATG5 conjugate an E3-like ligase may be suitable targets for structure-based drug design , Such approaches may also be used to exploit related strategies that have been developed for targeting ubiquitin and ubiquitin-like E1 enzymes using semi-synthetic protein inhibitors.

ATG4B, the cysteine protease that cleaves ATG8 at its carboxy-terminal to allow phosphatidylethanolamine conjugation, may also be a suitable candidate for structure-based drug design. Although the active site cleft of ATG4B is masked in the free form of the protein, thus eliminating this region as a target for inhibitors, possible targets for the design of specific inhibitors have been identified and include the inhibitory loop of ATG4B or the substrate ubiquitin core binding site , However, even the design of specific inhibitors of ATG4 proteins may not guarantee the absence of autophagy-independent off-target effects, as recent data suggest that autophagic machinery may have roles in seemingly distinct processes that do not appear to require conventional autophagosomes, such as osteoclast function , phagocytosis , entosis and mitochondrial cell death Conversely, it also seems as though inhibition of certain autophagy core proteins, including PIK3C3, BECN1 and ULK1, does not necessarily ablate autophagy completely In principle, one may be able to influence the clearance of autophagy substrates by acting not only on the autophagy pathway itself but also on processes influencing substrate recruitment as well as on lysosomal activity Substrate recruitment may be influenced by altered activities of autophagy adaptor molecules that help to recruit substrates to autophagosomes This may allow enhanced clearance of a repertoire of selected substrates.

Although this may be achievable, in principle, by altering the levels of such adaptors, it is possible that post-translational modifications of these adaptors may be a more effective means of influencing substrate recognition, and this approach may also be more amenable to targeting with drugs.

An elegant example of this phenomenon discussed above is the observation that phosphorylation of optineurin, which can act as an autophagy receptor, promoted the selective autophagy of ubiquitin-coated cytosolic S. In a similar mode, the PINK1—PARK2 machinery is thought to regulate selective autophagic degradation of mitochondria with disrupted membrane potentials.

A recent genome-wide screen identified many genes involved in the selective clearance of the Sindbis virus capsid protein; some of these genes also regulated mitochondrial degradation after depolarization This suggests that there may be many different types of adaptors and regulatory mechanisms that are amenable to perturbation, and also that this type of mechanism may ultimately affect a range of selective autophagic substrates.

One other step that may enhance the degradation of autophagic substrates is at the level of the lysosome. This may be achieved by modulating the activity of transcription factor EB TFEB , a master transcriptional modulator that influences both lysosomal biogenesis and autophagy This may be a tractable target, as TFEB activity is modulated by phosphorylation.

Another way to enhance autophagic substrate degradation could be via the depletion of the endogenous lysosomal cathepsin inhibitor cystatin B. Indeed, this strategy appears to enhance autophagic substrate clearance and reduce amyloid-β pathology in a mouse model of Alzheimer's disease Although this article has focused on modulating autophagy using drugs, it should be noted that there may be other ways of inducing autophagy, some of which could also be beneficial to health.

These include dietary restriction, exercise and possibly gene therapy routes potentially transducing tissues with vectors that enhance or block autophagy in some instances. As we know that various hormones such as insulin affect autophagy, we also need to consider non-cell-autonomous modes of autophagy regulation, particularly in the whole-body context.

The etymology of autophagy — 'self-eating' — indicates that the final goal of autophagosomes is to deliver cargo to the lysosome for degradation. Recent studies in yeast have demonstrated that autophagosomes can fuse with the plasma membrane to release their cargo into the extracellular medium , This mechanism sheds some light on the unconventional secretion of proteins.

A mechanism of this type in mammalian cells is also involved in exporting the pro-inflammatory cytokine interleukin-1 Ref. This process is dependent on autophagy-related protein 5 ATG5 , the inflammasome, the peripheral Golgi protein Golgi reassembly stacking protein 2 55kDa GRASP55; also known as GORASP2 and on the small GTPase RAB8A.

This autophagy-based secretion mechanism can probably be extended to other proteins and small molecules that modulate the immune response Interestingly, the mechanism that leads to the formation of the secretory autophagosomes can be modulated either via the classical biogenesis pathway or via a non-classical pathway such as that described in yeast that emerges from a novel compartment for unconventional protein secretion CUPS Like the conventional site for autophagosome formation, the CUPS contains high levels of the lipid phosphatidylinositolphosphate PtdIns3P as well as ATG8 and ATG9.

However, although these two proteins are required for the formation of the phagophore assembly site also known as the pre-autophagosomal structure , they are not necessary for the formation of the CUPS. It appears that the formation of autophagosomes that are engaged in the degradative pathway is also a matter of plasticity in mammalian cells What we already know about the formation of the autophagosome relies on the seminal discovery of Atg genes in yeast Most of these genes are conserved and act in an apparent hierarchical manner to form an autophagosome.

This classical or canonical pathway can be triggered by amino acids or by serum starvation. However, in other settings — as reviewed in Refs , — only a subset of ATG proteins are used to form an autophagosome.

For example, autophagy may not require some of the components of complex I of phosphoinositide 3-kinase PI3K , such as beclin 1 and class III PI3K also known as PIK3C3. However, it is possible that in this setting the lipid PtdIns3P may still have a role. This lipid can be produced by other sources, either from the degradation of phosphatidylinositol polyphosphates such as phosphatidylinositol-3,4-bisphosphate PtdIns 3,4 P 2 , phosphatidylinositol-3,4,5-trisphosphate PtdIns 3,4,5 P 3 and phosphatidylinositol-3,5-bisphosphate PtdIns 3,5 P 2 , or via the activity of class II PI3Ks on phosphatidylinositol phosphates.

Deviation from the canonical pathway of autophagosome biosynthesis and the final destination of autophagosomes are emerging concepts that should be taken into consideration in the attempts to develop drugs to target the autophagic pathway.

Ravikumar, B. et al. Regulation of mammalian autophagy in physiology and pathophysiology. CAS PubMed Google Scholar. Yang, Z. Eaten alive: a history of macroautophagy.

Nature Cell Biol. Kuma, A. The role of autophagy during the early neonatal starvation period. Nature , — This study illustrates the importance of autophagy in newborn mammals as a process that protects against starvation in the period before breast feeding is established.

Boya, P. Inhibition of macroautophagy triggers apoptosis. CAS PubMed PubMed Central Google Scholar. Mizushima, N. Autophagy in mammalian development and differentiation. Tsukamoto, S. Autophagy is essential for preimplantation development of mouse embryos.

Science , — Al Rawi, S. Postfertilization autophagy of sperm organelles prevents paternal mitochondrial DNA transmission. Sato, M. Degradation of paternal mitochondria by fertilization-triggered autophagy in C. elegans embryos. Rubinsztein, D. Autophagy and aging.

Cell , — The role of Atg proteins in autophagosome formation. Cell Dev. Axe, E. Autophagosome formation from membrane compartments enriched in phosphatidylinositol 3-phosphate and dynamically connected to the endoplasmic reticulum.

Cell Biol. Hayashi-Nishino, M. A subdomain of the endoplasmic reticulum forms a cradle for autophagosome formation. Ylä-Anttila, P. Autophagy 5 , — References 11, 12 and 13 provide strong support for a role of the ER in autophagosome biogenesis.

PubMed Google Scholar. Hailey, D. Mitochondria supply membranes for autophagosome biogenesis during starvation. This study suggests that mitochondria contribute membrane to autophagosomes during starvation.

Plasma membrane contributes to the formation of pre-autophagosomal structures. This study provides evidence that the plasma membrane is involved in the formation of autophagosome precursor structures. Mari, M. An Atg9-containing compartment that functions in the early steps of autophagosome biogenesis.

Ohashi, Y. Membrane delivery to the yeast autophagosome from the Golgi—endosomal system. Cell 21 , — Nair, U. SNARE proteins are required for macroautophagy. This study shows how SNARE proteins are involved in autophagosome biogenesis in yeast.

Moreau, K. Autophagosome precursor maturation requires homotypic fusion. This paper describes how SNAREs regulate mammalian autophagy by aiding in the expansion of the membranes of autophagosome precursors. Noda, N. Genes Cells 13 , — Weidberg, H. Biogenesis and cargo selectivity of autophagosomes.

Kraft, C. Selective types of autophagy in yeast. Acta , — Kim, D. mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Blommaart, E. Phosphorylation of ribosomal protein S6 is inhibitory for autophagy in isolated rat hepatocytes.

Noda, T. Tor, a phosphatidylinositol kinase homologue, controls autophagy in yeast. Mammucari, C. FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab. Pattingre, S. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. This study describes BCL-2 proteins as key negative regulators of autophagy that act via interactions with BECN1.

Erlich, S. Differential interactions between Beclin 1 and Bcl-2 family members. Autophagy 3 , — Maiuri, M. Functional and physical interaction between Bcl-XL and a BH3-like domain in Beclin EMBO J. Wei, Y. JNK1-mediated phosphorylation of Bcl-2 regulates starvation-induced autophagy.

Cell 30 , — This paper describes how JNK1-mediated phosphorylation of BCL-2 during starvation results in dissociation of the interaction between BCL-2 and BECN1, and mediates autophagy induction.

Role of JNK1-dependent Bcl-2 phosphorylation in ceramide-induced macroautophagy. Rabinowitz, J. Autophagy and metabolism. Singh, R. Autophagy regulates lipid metabolism. Yang, L.

Defective hepatic autophagy in obesity promotes ER stress and causes insulin resistance. Codogno, P. Autophagy: a potential link between obesity and insulin resistance.

Autophagy regulates adipose mass and differentiation in mice. Zhang, Y. Adipose-specific deletion of autophagy-related gene 7 atg7 in mice reveals a role in adipogenesis.

Natl Acad. USA , — He, C. Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis. This paper describes exercise as a novel physiological inducer of autophagy and demonstrates a role for autophagy in the exercise-induced beneficial effects on metabolism.

Kaushik, S. Autophagy in hypothalamic AgRP neurons regulates food intake and energy balance. Meng, Q. Autophagy — alias self-eating — appetite and ageing. EMBO Rep. The roles of intracellular protein-degradation pathways in neurodegeneration. Berger, Z. Rapamycin alleviates toxicity of different aggregate-prone proteins.

Aggregate-prone proteins with polyglutamine and polyalanine expansions are degraded by autophagy. This is the first description of autophagy as a process that clears intracytoplasmic aggregation-prone proteins, and provides data in cell culture suggesting that autophagy induction may be a therapeutic strategy for certain neurodegenerative diseases.

Webb, J. α-synuclein is degraded by both autophagy and the proteasome. Shibata, M. Regulation of intracellular accumulation of mutant Huntingtin by Beclin 1. Menzies, F. Autophagy induction reduces mutant ataxin-3 levels and toxicity in a mouse model of spinocerebellar ataxia type 3.

Brain , 93— Spencer, B. Beclin 1 gene transfer activates autophagy and ameliorates the neurodegenerative pathology in α-synuclein models of Parkinson's and Lewy body diseases.

Williams, A. Novel targets for Huntington's disease in an mTOR-independent autophagy pathway. Nature Chem. This paper describes a large-scale drug screen that identified a series of mTOR-independent autophagy inducers that have protective effects in cell, D.

melanogaster and zebrafish models of Huntington's disease. CAS Google Scholar. Zheng, S. Deletion of the huntingtin polyglutamine stretch enhances neuronal autophagy and longevity in mice.

PLoS Genet. PubMed PubMed Central Google Scholar. Rapamycin pre-treatment protects against apoptosis. 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.

References 52 and 53 describe how conditional loss of autophagy in the mammalian nervous system leads to cell death and aggregation of endogenous proteins.

Youle, R. Mechanisms of mitophagy. Nature Rev. Lee, J. Lysosomal proteolysis and autophagy require presenilin 1 and are disrupted by Alzheimer-related PS1 mutations.

Pickford, F. The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid β accumulation in mice. Winslow, A. The Parkinson disease protein α-synuclein inhibits autophagy.

Autophagy 7 , — Google Scholar. Corrochano, S. α-synuclein levels affect autophagosome numbers in vivo and modulate Huntington disease pathology. Autophagy 8 , — Jin, S. PINK1- and Parkin-mediated mitophagy at a glance. Cell Sci.

Aguado, C. Laforin, the most common protein mutated in Lafora disease, regulates autophagy. Puls, I. Mutant dynactin in motor neuron disease.

Nature Genet. Dynein mutations impair autophagic clearance of aggregate-prone proteins. Sarkar, S. Complex inhibitory effects of nitric oxide on autophagy. Cell 43 , 19—32 Raben, N.

Monitoring autophagy in lysosomal storage disorders. Settembre, C. A block of autophagy in lysosomal storage disorders. Rose, C. Rilmenidine attenuates toxicity of polyglutamine expansions in a mouse model of Huntington's disease. Bjedov, I. Mechanisms of life span extension by rapamycin in the fruit fly Drosophila melanogaster.

Harrison, D. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Renna, M. Azithromycin blocks autophagy and may predispose cystic fibrosis patients to mycobacterial infection.

Liang, X. Induction of autophagy and inhibition of tumorigenesis by beclin 1. This paper describes BECN1 as a mammalian autophagy gene that is associated with tumour suppression.

doi: Weinberg R, Fisher DE, Rich J. Dynamic and transient cancer stem cells nurture melanoma. Nat Med. Article PubMed CAS Google Scholar. Shen YA, Wang CY, Hsieh YT, Chen YJ, Wei YH. Metabolic reprogramming orchestrates cancer stem cell properties in nasopharyngeal carcinoma.

Article PubMed Google Scholar. Saga I, Shibao S, Okubo J, Osuka S, Kobayashi Y, Yamada S, Fujita S, Urakami K, Kusuhara M, Yoshida K, et al. Integrated analysis identifies different metabolic signatures for tumor-initiating cells in a murine glioblastoma model. Neuro Oncol. Cell Stem Cell.

Chen D, Che G. Value of caveolin-1 in cancer progression and prognosis: emphasis on cancer-associated fibroblasts, human cancer cells and mechanism of caveolin-1 expression review. Oncol Lett. Pinilla SM, Honrado E, Hardisson D, Benitez J, Palacios J.

Caveolin-1 expression is associated with a basal-like phenotype in sporadic and hereditary breast cancer. Breast Cancer Res Treat. Wiechen K, Diatchenko L, Agoulnik A, Scharff KM, Schober H, Arlt K, Zhumabayeva B, Siebert PD, Dietel M, Schafer R, et al.

Caveolin-1 is down-regulated in human ovarian carcinoma and acts as a candidate tumor suppressor gene. Am J Pathol. Zhang ZB, Cai L, Zheng SG, Xiong Y, Dong JH. Overexpression of caveolin-1 in hepatocellular carcinoma with metastasis and worse prognosis: correlation with vascular endothelial growth factor, microvessel density and unpaired artery.

Pathol Oncol Res. Sotgia F, Del Galdo F, Casimiro MC, Bonuccelli G, Mercier I, Whitaker-Menezes D, Daumer KM, Zhou J, Wang C, Katiyar S, et al. Fang WB, Yao M, Cheng N. Priming cancer cells for drug resistance: role of the fibroblast niche. Front Biol Beijing. Gorges TM, Tinhofer I, Drosch M, Rose L, Zollner TM, Krahn T, von Ahsen O.

Circulating tumour cells escape from EpCAM-based detection due to epithelial-to-mesenchymal transition. BMC Cancer. Grover PK, Cummins AG, Price TJ, Roberts-Thomson IC, Hardingham JE.

Circulating tumour cells: the evolving concept and the inadequacy of their enrichment by EpCAM-based methodology for basic and clinical cancer research. Ann Oncol. Xu D, Hemler ME. Metabolic activation-related CDCD98 complex. Mol Cell Proteomics. Wang Q, Holst J. L-type amino acid transport and cancer: targeting the mTORC1 pathway to inhibit neoplasia.

Am J Cancer Res. Hensley CT, Faubert B, Yuan Q, Lev-Cohain N, Jin E, Kim J, Jiang L, Ko B, Skelton R, Loudat L, et al. Metabolic heterogeneity in human lung tumors. Robertson-Tessi M, Gillies RJ, Gatenby RA, Anderson AR.

Impact of metabolic heterogeneity on tumor growth, invasion, and treatment outcomes. Cancer Res. Sengupta D, Pratx G. Imaging metabolic heterogeneity in cancer. Mol Cancer. Inversed relationship between CD44 variant and c-Myc due to oxidative stress-induced canonical Wnt activation.

Therapeutic strategies targeting cancer stem cells. Hatakeyama M. The role of Helicobacter pylori CagA in gastric carcinogenesis. Int J Hematol. Yong X, Tang B, Li BS, Xie R, Hu CJ, Luo G, Qin Y, Dong H, Yang SM. Helicobacter pylori virulence factor CagA promotes tumorigenesis of gastric cancer via multiple signaling pathways.

Cell Commun Signal. Langedijk J, Mantel-Teeuwisse AK, Slijkerman DS, Schutjens MH. Drug repositioning and repurposing: terminology and definitions in literature. Drug Discov Today. Tommasino C, Gambardella L, Buoncervello M, Griffin RJ, Golding BT, Alberton M, Macchia D, Spada M, Cerbelli B, d'Amati G, et al.

New derivatives of the antimalarial drug Pyrimethamine in the control of melanoma tumor growth: an in vitro and in vivo study. Kimura T, Takabatake Y, Takahashi A, Isaka Y.

Chloroquine in cancer therapy: a double-edged sword of autophagy. Li J, Lee AS. Curr Mol Med. Lee AS. GRP78 induction in cancer: therapeutic and prognostic implications. Patel TA, Dave B, Rodriguez AA, Chang JC, Perez EA, Colon-Otero G.

Dual HER2 blockade: preclinical and clinical data. Breast Cancer Res. Puri N, Salgia R. Synergism of EGFR and c-Met pathways, cross-talk and inhibition, in non-small cell lung cancer. J Carcinog. Wen W, Wu J, Liu L, Tian Y, Buettner R, Hsieh MY, Horne D, Dellinger TH, Han ES, Jove R, et al.

Pasquier E, Andre N, Street J, Chougule A, Rekhi B, Ghosh J, Philip DS, Meurer M, MacKenzie KL, Kavallaris M, et al. Effective management of advanced angiosarcoma by the synergistic combination of propranolol and vinblastine-based metronomic chemotherapy: a bench to bedside study.

Pessetto ZY, Ma Y, Hirst JJ, von Mehren M, Weir SJ, Godwin AK. Drug repurposing identifies a synergistic combination therapy with imatinib mesylate for gastrointestinal stromal tumor. Schweitzer BI, Dicker AP, Bertino JR. Dihydrofolate reductase as a therapeutic target.

FASEB J. Gangjee A, Kurup S, Namjoshi O. Dihydrofolate reductase as a target for chemotherapy in parasites. de Castro MA, Bunt G, Wouters FS. Cathepsin B launches an apoptotic exit effort upon cell death-associated disruption of lysosomes.

Cell Death Discov. Zhou J, Tan SH, Nicolas V, Bauvy C, Yang ND, Zhang J, Xue Y, Codogno P, Shen HM. Activation of lysosomal function in the course of autophagy via mTORC1 suppression and autophagosome-lysosome fusion.

Cell Res. Rink L, Skorobogatko Y, Kossenkov AV, Belinsky MG, Pajak T, Heinrich MC, Blanke CD, von Mehren M, Ochs MF, Eisenberg B, et al. Gene expression signatures and response to imatinib mesylate in gastrointestinal stromal tumor.

Tarn C, Merkel E, Canutescu AA, Shen W, Skorobogatko Y, Heslin MJ, Eisenberg B, Birbe R, Patchefsky A, Dunbrack R, et al. Analysis of KIT mutations in sporadic and familial gastrointestinal stromal tumors: therapeutic implications through protein modeling.

Wang CM, Huang K, Zhou Y, Du CY, Ye YW, Fu H, Zhou XY, Shi YQ. Molecular mechanisms of secondary imatinib resistance in patients with gastrointestinal stromal tumors. J Cancer Res Clin Oncol. Puccio CA, Mittelman A, Lichtman SM, Silver RT, Budman DR, Ahmed T, Feldman EJ, Coleman M, Arnold PM, Arlin ZA, et al.

J Clin Oncol. Shim JS, Liu JO. Recent advances in drug repositioning for the discovery of new anticancer drugs. Int J Biol Sci. Bernstein WB, Dennis PA. Repositioning HIV protease inhibitors as cancer therapeutics. Curr Opin HIV AIDS. Dolma S, Selvadurai HJ, Lan X, Lee L, Kushida M, Voisin V, Whetstone H, So M, Aviv T, Park N, et al.

Inhibition of dopamine receptor D4 impedes autophagic flux, proliferation, and survival of glioblastoma stem cells. Heuillet E, Petitet F, Mignani S, Malleron JL, Lavayre J, Neliat G, Doble A, Blanchard JC.

The naphtosultam derivative RP fananserin has high affinity for the dopamine D4 receptor. Eur J Pharmacol. Shchors K, Massaras A, Hanahan D. Dual targeting of the autophagic regulatory circuitry in gliomas with repurposed drugs elicits cell-lethal autophagy and therapeutic benefit. Kametaka S, Okano T, Ohsumi M, Ohsumi Y.

Cao Y, Klionsky DJ. Liang XH, Kleeman LK, Jiang HH, Gordon G, Goldman JE, Berry G, Herman B, Levine B. Protection against fatal Sindbis virus encephalitis by beclin, a novel Bclinteracting protein.

J Virol. Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, Rosen J, Eskelinen EL, Mizushima N, Ohsumi Y, et al. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene.

Thorburn A. Autophagy and its effects: making sense of double-edged swords. PLoS Biol. White E, DiPaola RS. The double-edged sword of autophagy modulation in cancer. Jia G, Kong R, Ma ZB, Han B, Wang YW, Pan SH, Li YH, Sun B. The activation of c-Jun NH 2 -terminal kinase is required for dihydroartemisinin-induced autophagy in pancreatic cancer cells.

Wei Y, Sinha S, Levine B. Dual role of JNK1-mediated phosphorylation of Bcl-2 in autophagy and apoptosis regulation. Shimizu S, Yoshida T, Tsujioka M, Arakawa S.

Autophagic cell death and cancer. Int J Mol Sci. Dong LH, Cheng S, Zheng Z, Wang L, Shen Y, Shen ZX, Chen SJ, Zhao WL. Gatto F, Nielsen J. Systematic analysis of overall survival and interactions between tumor mutations and drug treatment.

Ashley EA, Dhorda M, Fairhurst RM, Amaratunga C, Lim P, Suon S, Sreng S, Anderson JM, Mao S, Sam B, et al. Spread of artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med.

Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, Walczak H, Vandenabeele P.

Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Gao M, Monian P, Pan Q, Zhang W, Xiang J, Jiang X. Ferroptosis is an autophagic cell death process. Bridges RJ, Natale NR, Patel SA. Br J Pharmacol. Lewerenz J, Hewett SJ, Huang Y, Lambros M, Gout PW, Kalivas PW, Massie A, Smolders I, Methner A, Pergande M, et al.

Dale J, Alcorn N, Capell H, Madhok R. Combination therapy for rheumatoid arthritis: methotrexate and sulfasalazine together or with other DMARDs. Nat Clin Pract Rheumatol. quiz, following Dai L, Cao Y, Chen Y, Parsons C, Qin Z.

Yoshikawa M, Tsuchihashi K, Ishimoto T, Yae T, Motohara T, Sugihara E, Onishi N, Masuko T, Yoshizawa K, Kawashiri S, et al. xCT inhibition depletes CD44v-expressing tumor cells that are resistant to EGFR-targeted therapy in head and neck squamous cell carcinoma. Buckingham SC, Campbell SL, Haas BR, Montana V, Robel S, Ogunrinu T, Sontheimer H.

Glutamate release by primary brain tumors induces epileptic activity. Sehm T, Fan Z, Ghoochani A, Rauh M, Engelhorn T, Minakaki G, Dorfler A, Klucken J, Buchfelder M, Eyupoglu IY, et al. Sulfasalazine impacts on ferroptotic cell death and alleviates the tumor microenvironment and glioma-induced brain edema.

Shitara K, Doi T, Nagano O, Imamura CK, Ozeki T, Ishii Y, Tsuchihashi K, Takahashi S, Nakajima TE, Hironaka S, et al. Gastric Cancer. Jeong HJ, Oh HA, Nam SY, Han NR, Kim YS, Kim JH, Lee SJ, Kim MH, Moon PD, Kim HM.

The critical role of mast cell-derived hypoxia-inducible factor-1alpha in human and mice melanoma growth. Int J Cancer.

Jangi SM, Ruiz-Larrea MB, Nicolau-Galmes F, Andollo N, Arroyo-Berdugo Y, Ortega-Martinez I, Diaz-Perez JL, Boyano MD. Nicolau-Galmes F, Asumendi A, Alonso-Tejerina E, Perez-Yarza G, Jangi SM, Gardeazabal J, Arroyo-Berdugo Y, Careaga JM, Diaz-Ramon JL, Apraiz A, et al.

Terfenadine induces apoptosis and autophagy in melanoma cells through ROS-dependent and -independent mechanisms.

Gammoh N, Lam D, Puente C, Ganley I, Marks PA, Jiang X. Role of autophagy in histone deacetylase inhibitor-induced apoptotic and nonapoptotic cell death. Zhang J, Ng S, Wang J, Zhou J, Tan SH, Yang N, Lin Q, Xia D, Shen HM.

Histone deacetylase inhibitors induce autophagy through FOXO1-dependent pathways. Banreti A, Sass M, Graba Y. The emerging role of acetylation in the regulation of autophagy. Westin JR. Clin Lymphoma Myeloma Leuk. Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing.

Cang S, Ma Y, Liu D. New clinical developments in histone deacetylase inhibitors for epigenetic therapy of cancer. Maggi CA, Meli A. The sensory-efferent function of capsaicin-sensitive sensory neurons. Gen Pharmacol. Pingle SC, Matta JA, Ahern GP. Capsaicin receptor: TRPV1 a promiscuous TRP channel.

Handb Exp Pharmacol. Garufi A, Pistritto G, Cirone M, D'Orazi G. Reactivation of mutant p53 by capsaicin, the major constituent of peppers. Muller PA, Vousden KH. p53 mutations in cancer. Olivier M, Hollstein M, Hainaut P.

TP53 mutations in human cancers: origins, consequences, and clinical use. Bar J, Moskovits N, Oren M. Involvement of stromal p53 in tumor-stroma interactions. Semin Cell Dev Biol. Amantini C, Ballarini P, Caprodossi S, Nabissi M, Morelli MB, Lucciarini R, Cardarelli MA, Mammana G, Santoni G.

Masumoto K, Tsukimoto M, Kojima S. Role of TRPM2 and TRPV1 cation channels in cellular responses to radiation-induced DNA damage. Abraham RT. Cell cycle checkpoint signaling through the ATM and ATR kinases. Bolderson E, Richard DJ, Zhou BB, Khanna KK. Recent advances in cancer therapy targeting proteins involved in DNA double-strand break repair.

Crighton D, Wilkinson S, O'Prey J, Syed N, Smith P, Harrison PR, Gasco M, Garrone O, Crook T, Ryan KM. DRAM, a pinduced modulator of autophagy, is critical for apoptosis. Crighton D, Wilkinson S, Ryan KM. DRAM links autophagy to p53 and programmed cell death. Nakagawa K, Umeda T, Higuchi O, Tsuzuki T, Suzuki T, Miyazawa T.

Evaporative light-scattering analysis of sulforaphane in broccoli samples: quality of broccoli products regarding sulforaphane contents. J Agric Food Chem. Myzak MC, Dashwood RH. Chemoprotection by sulforaphane: keep one eye beyond Keap1.

Cancer Lett. Kansanen E, Kuosmanen SM, Leinonen H, Levonen AL. The Keap1-Nrf2 pathway: mechanisms of activation and dysregulation in cancer. Redox Biol. Taguchi K, Motohashi H, Yamamoto M. Molecular mechanisms of the Keap1-Nrf2 pathway in stress response and cancer evolution.

Genes Cells. Kanematsu S, Uehara N, Miki H, Yoshizawa K, Kawanaka A, Yuri T, Tsubura A. Autophagy inhibition enhances sulforaphane-induced apoptosis in human breast cancer cells.

Anticancer Res. Mizushima N, Yoshimori T, Levine B. Methods in mammalian autophagy research. Pawlik A, Wiczk A, Kaczynska A, Antosiewicz J, Herman-Antosiewicz A. Sulforaphane inhibits growth of phenotypically different breast cancer cells.

Eur J Nutr. El-Khattouti A, Selimovic D, Haikel Y, Hassan M. Crosstalk between apoptosis and autophagy: molecular mechanisms and therapeutic strategies in cancer.

J Cell Death. Maiuri MC, Zalckvar E, Kimchi A, Kroemer G. Self-eating and self-killing: crosstalk between autophagy and apoptosis. Van Noorden R, Ledford H. Download references. The author thanks Professor Victor C.

Kok, MMedSc, MD, PhD, FACP Asia University, Taichung, Taiwan for the critical appraisal of the manuscript. Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, , Japan.

Japan Society for the Promotion of Science, Kojimachi, Chiyoda-ku, Tokyo, , Japan. You can also search for this author in PubMed Google Scholar. Correspondence to Go J. Open Access This article is distributed under the terms of the Creative Commons Attribution 4.

Reprints and permissions. Yoshida, G. Therapeutic strategies of drug repositioning targeting autophagy to induce cancer cell death: from pathophysiology to treatment. J Hematol Oncol 10 , 67 Download citation. Received : 09 January Accepted : 02 March Published : 09 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: 09 March Therapeutic strategies of drug repositioning targeting autophagy to induce cancer cell death: from pathophysiology to treatment Go J.

Abstract The Nobel Prize in Physiology or Medicine was awarded to the researcher that discovered autophagy, which is an evolutionally conserved catabolic process which degrades cytoplasmic constituents and organelles in the lysosome.

Background Cellular degradation processes mainly fall into two categories: macroautophagy commonly referred to as autophagy and the ubiquitin-proteasome system. Full size image. Table 1 Typical examples of drug re-positioning targeting autophagy in cancer cells Full size table.

Conclusions Conventional agents are not only pharmacologically safe but also cheaper than specialized anticancer drugs. Abbreviations AMPK: AMP-activated protein kinase ARE: Antioxidant response element Atg: Autophagy-related gene ATM: Ataxia-telangiectasia A-T mutated CAF: Cancer-associated fibroblasts CagA: Cytotoxin-associated gene A Cav Caveolin 1 CDK1: Cyclin-dependent kinase 1 CSCs: Cancer stem-like cells DHA: Dihydroartemisinin DHFR: Dihydrofolate reductase DRAM: Damage-regulated autophagy modulator DRD4: Dopamine receptor D4 EpCAM: Epithelial cell adhesion molecule ER: Endoplasmic reticulum GBM: Glioblastoma multiforme GIST: Gastrointestinal stromal tumor GNS: GBM neural stem cells GSH: Glutathione H.

pylori : Helicobacter pylori HDAC: Histone deacetylases HO Heme oxygenase 1 IL: Interleukin Iso-Pyr: Iso-pyrimethamine JNK: c-Jun NH2-terminal kinase KEAP1: Kelch-like ECH-associated protein 1 MBP: Methylbenzoprim MCT: Monocarboxylate transporter MEFs: Mouse embryonic fibroblasts MNP: m -Nitropyrimethamine MPT: Mitochondrial permeability transition mTOR: Mammalian target of rapamycin MZPES: m -Azidopyrimethamine ethanesulfonate salt NQO NAD P H-quinone oxidoreductase-1 Nrf2: Nuclear factor erythroid 2-related factor 2 PARP: Poly ADP-ribose polymerase PI3K: Phosphatidylinositol 3-kinase Pyr: Pyrimethamine ROS: Reactive oxygen species SLE: Systemic lupus erythematosus TEM: Transmission electron microscope TMZ: Temozolomide TRPV1: Transient receptor potential cation channel subfamily V member 1 UPR: Unfolded protein response VEGF: Vascular endothelial growth factor VPA: Valproic acid.

References Glick D, Barth S, Macleod KF. Article CAS PubMed PubMed Central Google Scholar Das G, Shravage BV, Baehrecke EH. Article CAS PubMed Google Scholar Korolchuk VI, Menzies FM, Rubinsztein DC. Article CAS PubMed Google Scholar Ciechanover A.

Article CAS PubMed Google Scholar Wang Y, Nartiss Y, Steipe B, McQuibban GA, Kim PK. Article CAS PubMed Google Scholar Lee J, Giordano S, Zhang J. Article CAS PubMed Google Scholar Kim J, Kundu M, Viollet B, Guan KL. Article CAS PubMed PubMed Central Google Scholar Mihaylova MM, Shaw RJ.

Article CAS PubMed PubMed Central Google Scholar Metcalf DJ, Garcia-Arencibia M, Hochfeld WE, Rubinsztein DC. Article CAS PubMed PubMed Central Google Scholar Jimenez-Sanchez M, Thomson F, Zavodszky E, Rubinsztein DC.

Article CAS PubMed PubMed Central Google Scholar Tsugawa H, Suzuki H, Saya H, Hatakeyama M, Hirayama T, Hirata K, Nagano O, Matsuzaki J, Hibi T. Article CAS PubMed Google Scholar Wileman T. Article CAS PubMed Google Scholar Chan SN, Tang BL. Article CAS PubMed Google Scholar Hamasaki M, Furuta N, Matsuda A, Nezu A, Yamamoto A, Fujita N, Oomori H, Noda T, Haraguchi T, Hiraoka Y, et al.

Article CAS PubMed Google Scholar Marchi S, Patergnani S, Pinton P. Article CAS PubMed Google Scholar Mauvezin C, Neisch AL, Ayala CI, Kim J, Beltrame A, Braden CR, Gardner MK, Hays TS, Neufeld TP.

Article CAS PubMed PubMed Central Google Scholar Yu L, McPhee CK, Zheng L, Mardones GA, Rong Y, Peng J, Mi N, Zhao Y, Liu Z, Wan F, et al. Article CAS PubMed PubMed Central Google Scholar Ding WX, Yin XM. Article CAS PubMed PubMed Central Google Scholar Saito T, Sadoshima J.

Article CAS PubMed PubMed Central Google Scholar Zhang H, Bosch-Marce M, Shimoda LA, Tan YS, Baek JH, Wesley JB, Gonzalez FJ, Semenza GL. Article CAS PubMed PubMed Central Google Scholar Lemasters JJ, Qian T, He L, Kim JS, Elmore SP, Cascio WE, Brenner DA.

Article CAS PubMed Google Scholar Kim YC, Guan KL. Article PubMed PubMed Central Google Scholar Porta C, Paglino C, Mosca A. Article PubMed PubMed Central Google Scholar Arcaro A, Guerreiro AS.

Article CAS PubMed PubMed Central Google Scholar Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, Overvatn A, Bjorkoy G, Johansen T. Article CAS PubMed Google Scholar Klionsky DJ, Abdelmohsen K, Abe A, Abedin MJ, Abeliovich H, Acevedo Arozena A, Adachi H, Adams CM, Adams PD, Adeli K, et al.

Article PubMed PubMed Central Google Scholar Loos B, du Toit A, Hofmeyr JH. Article PubMed PubMed Central CAS Google Scholar Yoshimori T, Yamamoto A, Moriyama Y, Futai M, Tashiro Y. CAS PubMed Google Scholar Hoyer-Hansen M, Jaattela M.

Article CAS PubMed Google Scholar Senft D, Ronai ZA. Article CAS PubMed PubMed Central Google Scholar Zhang J, Stevens MF, Bradshaw TD. Article CAS PubMed Google Scholar Stepanenko AA, Andreieva SV, Korets KV, Mykytenko DO, Baklaushev VP, Huleyuk NL, Kovalova OA, Kotsarenko KV, Chekhonin VP, Vassetzky YS, et al.

Article PubMed PubMed Central Google Scholar Happold C, Roth P, Wick W, Schmidt N, Florea AM, Silginer M, Reifenberger G, Weller M. Article CAS PubMed Google Scholar Seiter K, Katragadda S, Ponce D, Rasul M, Ahmed N.

Article PubMed PubMed Central CAS Google Scholar Phan LM, Yeung SC, Lee MH. CAS PubMed PubMed Central Google Scholar Ward PS, Thompson CB. Article CAS PubMed PubMed Central Google Scholar Yoshida GJ. Article PubMed PubMed Central CAS Google Scholar Kuhajda FP. Article CAS Google Scholar Hardie DG.

Article CAS PubMed Google Scholar Hardie DG, Ross FA, Hawley SA. Article CAS PubMed Google Scholar Mathew R, Karp CM, Beaudoin B, Vuong N, Chen G, Chen HY, Bray K, Reddy A, Bhanot G, Gelinas C, et al. Article CAS PubMed PubMed Central Google Scholar Takamura A, Komatsu M, Hara T, Sakamoto A, Kishi C, Waguri S, Eishi Y, Hino O, Tanaka K, Mizushima N.

Article CAS PubMed PubMed Central Google Scholar Shimizu S, Konishi A, Nishida Y, Mizuta T, Nishina H, Yamamoto A, Tsujimoto Y. Article CAS PubMed Google Scholar Sui X, Chen R, Wang Z, Huang Z, Kong N, Zhang M, Han W, Lou F, Yang J, Zhang Q, et al. Article CAS PubMed PubMed Central Google Scholar Yang ZJ, Chee CE, Huang S, Sinicrope FA.

Article CAS PubMed PubMed Central Google Scholar Jawhari S, Ratinaud MH, Verdier M. Article CAS PubMed PubMed Central Google Scholar Yan Y, Xu Z, Dai S, Qian L, Sun L, Gong Z.

Article PubMed PubMed Central CAS Google Scholar Friedman HS, Kerby T, Calvert H. CAS PubMed Google Scholar Mason WP, Cairncross JG. Article CAS PubMed Google Scholar Zhou Y, Wang HD, Zhu L, Cong ZX, Li N, Ji XJ, Pan H, Wang JW, Li WC.

CAS PubMed Google Scholar Gao AM, Ke ZP, Shi F, Sun GC, Chen H. Article CAS PubMed Google Scholar Perera RM, Stoykova S, Nicolay BN, Ross KN, Fitamant J, Boukhali M, Lengrand J, Deshpande V, Selig MK, Ferrone CR, et al. Article CAS PubMed PubMed Central Google Scholar Martina JA, Diab HI, Lishu L, Jeong AL, Patange S, Raben N, Puertollano R.

Article PubMed PubMed Central CAS Google Scholar Martina JA, Diab HI, Li H, Puertollano R. Article CAS PubMed PubMed Central Google Scholar Kauffman EC, Ricketts CJ, Rais-Bahrami S, Yang Y, Merino MJ, Bottaro DP, Srinivasan R, Linehan WM.

Article CAS PubMed PubMed Central Google Scholar Raices M, D'Angelo MA. Article CAS PubMed Google Scholar Toh PP, Luo S, Menzies FM, Rasko T, Wanker EE, Rubinsztein DC. Article CAS PubMed PubMed Central Google Scholar Granato M, Rizzello C, Romeo MA, Yadav S, Santarelli R, D'Orazi G, Faggioni A, Cirone M.

Article CAS PubMed Google Scholar Huang H, Weng H, Zhou H, Qu L. Article CAS PubMed Google Scholar Prochownik EV, Vogt PK. Article CAS PubMed PubMed Central Google Scholar Warmoes MO, Locasale JW.

Article CAS PubMed PubMed Central Google Scholar Pavlides S, Whitaker-Menezes D, Castello-Cros R, Flomenberg N, Witkiewicz AK, Frank PG, Casimiro MC, Wang C, Fortina P, Addya S, et al. Article CAS PubMed Google Scholar Zhao H, Yang L, Baddour J, Achreja A, Bernard V, Moss T, Marini JC, Tudawe T, Seviour EG, San Lucas FA, et al.

PubMed PubMed Central Google Scholar Ishimoto T, Nagano O, Yae T, Tamada M, Motohara T, Oshima H, Oshima M, Ikeda T, Asaba R, Yagi H, et al.

Article CAS PubMed Google Scholar Ishimoto T, Oshima H, Oshima M, Kai K, Torii R, Masuko T, Baba H, Saya H, Nagano O. Article CAS PubMed Google Scholar Yoshida GJ.

Article PubMed PubMed Central Google Scholar Maugeri-Sacca M, Bartucci M, De Maria R. Article CAS PubMed Google Scholar Wang QE. Article PubMed PubMed Central Google Scholar Vlashi E, Lagadec C, Vergnes L, Matsutani T, Masui K, Poulou M, Popescu R, Della Donna L, Evers P, Dekmezian C, et al.

Article CAS PubMed PubMed Central Google Scholar Yoshida GJ, Saya H. Article PubMed CAS Google Scholar Shen YA, Wang CY, Hsieh YT, Chen YJ, Wei YH. Article PubMed Google Scholar Saga I, Shibao S, Okubo J, Osuka S, Kobayashi Y, Yamada S, Fujita S, Urakami K, Kusuhara M, Yoshida K, et al.

Article CAS PubMed PubMed Central Google Scholar Wu Z, Wei D, Gao W, Xu Y, Hu Z, Ma Z, Gao C, Zhu X, Li Q. Article CAS PubMed Google Scholar Chen D, Che G.

CAS PubMed PubMed Central Google Scholar Pinilla SM, Honrado E, Hardisson D, Benitez J, Palacios J. Article CAS PubMed Google Scholar Wiechen K, Diatchenko L, Agoulnik A, Scharff KM, Schober H, Arlt K, Zhumabayeva B, Siebert PD, Dietel M, Schafer R, et al.

Article CAS PubMed PubMed Central Google Scholar Zhang ZB, Cai L, Zheng SG, Xiong Y, Dong JH. Article CAS PubMed Google Scholar Sotgia F, Del Galdo F, Casimiro MC, Bonuccelli G, Mercier I, Whitaker-Menezes D, Daumer KM, Zhou J, Wang C, Katiyar S, et al.

Article CAS PubMed PubMed Central Google Scholar Fang WB, Yao M, Cheng N. Article CAS Google Scholar Gorges TM, Tinhofer I, Drosch M, Rose L, Zollner TM, Krahn T, von Ahsen O. Article CAS PubMed PubMed Central Google Scholar Grover PK, Cummins AG, Price TJ, Roberts-Thomson IC, Hardingham JE.

Article CAS PubMed Google Scholar Xu D, Hemler ME. Article CAS PubMed PubMed Central Google Scholar Wang Q, Holst J. CAS PubMed PubMed Central Google Scholar Hensley CT, Faubert B, Yuan Q, Lev-Cohain N, Jin E, Kim J, Jiang L, Ko B, Skelton R, Loudat L, et al. Article CAS PubMed PubMed Central Google Scholar Robertson-Tessi M, Gillies RJ, Gatenby RA, Anderson AR.

Article CAS PubMed PubMed Central Google Scholar Sengupta D, Pratx G. Article PubMed PubMed Central CAS Google Scholar Yoshida GJ, Saya H.

Article CAS PubMed Google Scholar Yoshida GJ, Saya H. Article CAS PubMed Google Scholar Hatakeyama M. Article CAS PubMed Google Scholar Yong X, Tang B, Li BS, Xie R, Hu CJ, Luo G, Qin Y, Dong H, Yang SM.

Article PubMed PubMed Central CAS Google Scholar Langedijk J, Mantel-Teeuwisse AK, Slijkerman DS, Schutjens MH. Article PubMed Google Scholar Tommasino C, Gambardella L, Buoncervello M, Griffin RJ, Golding BT, Alberton M, Macchia D, Spada M, Cerbelli B, d'Amati G, et al. Article PubMed PubMed Central CAS Google Scholar Kimura T, Takabatake Y, Takahashi A, Isaka Y.

Article CAS PubMed Google Scholar Li J, Lee AS. Article CAS PubMed Google Scholar Lee AS. Article CAS PubMed Google Scholar Patel TA, Dave B, Rodriguez AA, Chang JC, Perez EA, Colon-Otero G.

Article PubMed PubMed Central CAS Google Scholar Puri N, Salgia R. Article PubMed PubMed Central Google Scholar Wen W, Wu J, Liu L, Tian Y, Buettner R, Hsieh MY, Horne D, Dellinger TH, Han ES, Jove R, et al.

Article PubMed PubMed Central CAS Google Scholar Pasquier E, Andre N, Street J, Chougule A, Rekhi B, Ghosh J, Philip DS, Meurer M, MacKenzie KL, Kavallaris M, et al.

Article PubMed PubMed Central Google Scholar Pessetto ZY, Ma Y, Hirst JJ, von Mehren M, Weir SJ, Godwin AK. Article CAS PubMed PubMed Central Google Scholar Schweitzer BI, Dicker AP, Bertino JR. CAS PubMed Google Scholar Gangjee A, Kurup S, Namjoshi O. Article CAS PubMed Google Scholar de Castro MA, Bunt G, Wouters FS.

Article PubMed PubMed Central CAS Google Scholar Zhou J, Tan SH, Nicolas V, Bauvy C, Yang ND, Zhang J, Xue Y, Codogno P, Shen HM. Article CAS PubMed PubMed Central Google Scholar Rink L, Skorobogatko Y, Kossenkov AV, Belinsky MG, Pajak T, Heinrich MC, Blanke CD, von Mehren M, Ochs MF, Eisenberg B, et al.

Article CAS PubMed PubMed Central Google Scholar Tarn C, Merkel E, Canutescu AA, Shen W, Skorobogatko Y, Heslin MJ, Eisenberg B, Birbe R, Patchefsky A, Dunbrack R, et al.

Article CAS PubMed Google Scholar Wang CM, Huang K, Zhou Y, Du CY, Ye YW, Fu H, Zhou XY, Shi YQ. Article CAS PubMed Google Scholar Puccio CA, Mittelman A, Lichtman SM, Silver RT, Budman DR, Ahmed T, Feldman EJ, Coleman M, Arnold PM, Arlin ZA, et al. Article CAS PubMed Google Scholar Shim JS, Liu JO.

Article CAS PubMed PubMed Central Google Scholar Bernstein WB, Dennis PA. Article PubMed PubMed Central Google Scholar Dolma S, Selvadurai HJ, Lan X, Lee L, Kushida M, Voisin V, Whetstone H, So M, Aviv T, Park N, et al.

Article CAS PubMed Google Scholar Heuillet E, Petitet F, Mignani S, Malleron JL, Lavayre J, Neliat G, Doble A, Blanchard JC. Article CAS PubMed Google Scholar Shchors K, Massaras A, Hanahan D.

Article CAS PubMed Google Scholar Kametaka S, Okano T, Ohsumi M, Ohsumi Y. Article CAS PubMed Google Scholar Cao Y, Klionsky DJ. Article CAS PubMed Google Scholar Liang XH, Kleeman LK, Jiang HH, Gordon G, Goldman JE, Berry G, Herman B, Levine B. CAS PubMed PubMed Central Google Scholar Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, Rosen J, Eskelinen EL, Mizushima N, Ohsumi Y, et al.

Article CAS PubMed PubMed Central Google Scholar Thorburn A. Article PubMed PubMed Central CAS Google Scholar White E, DiPaola RS. Article PubMed PubMed Central Google Scholar Jia G, Kong R, Ma ZB, Han B, Wang YW, Pan SH, Li YH, Sun B.

Article PubMed PubMed Central CAS Google Scholar Wei Y, Sinha S, Levine B. Article CAS PubMed PubMed Central Google Scholar Shimizu S, Yoshida T, Tsujioka M, Arakawa S.

Article PubMed PubMed Central CAS Google Scholar Dong LH, Cheng S, Zheng Z, Wang L, Shen Y, Shen ZX, Chen SJ, Zhao WL. Article CAS PubMed PubMed Central Google Scholar Gatto F, Nielsen J. Article PubMed PubMed Central CAS Google Scholar Ashley EA, Dhorda M, Fairhurst RM, Amaratunga C, Lim P, Suon S, Sreng S, Anderson JM, Mao S, Sam B, et al.

Article PubMed PubMed Central CAS Google Scholar Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al. Article CAS PubMed PubMed Central Google Scholar Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, Walczak H, Vandenabeele P.

Article CAS PubMed Google Scholar Gao M, Monian P, Pan Q, Zhang W, Xiang J, Jiang X. Article CAS PubMed Google Scholar Bridges RJ, Natale NR, Patel SA. Article CAS PubMed PubMed Central Google Scholar Lewerenz J, Hewett SJ, Huang Y, Lambros M, Gout PW, Kalivas PW, Massie A, Smolders I, Methner A, Pergande M, et al.

Article CAS PubMed PubMed Central Google Scholar Dale J, Alcorn N, Capell H, Madhok R. Article CAS PubMed Google Scholar Dai L, Cao Y, Chen Y, Parsons C, Qin Z. Article PubMed PubMed Central CAS Google Scholar Yoshikawa M, Tsuchihashi K, Ishimoto T, Yae T, Motohara T, Sugihara E, Onishi N, Masuko T, Yoshizawa K, Kawashiri S, et al.

Article CAS PubMed Google Scholar Buckingham SC, Campbell SL, Haas BR, Montana V, Robel S, Ogunrinu T, Sontheimer H. Article CAS PubMed PubMed Central Google Scholar Sehm T, Fan Z, Ghoochani A, Rauh M, Engelhorn T, Minakaki G, Dorfler A, Klucken J, Buchfelder M, Eyupoglu IY, et al.

PubMed PubMed Central Google Scholar Shitara K, Doi T, Nagano O, Imamura CK, Ozeki T, Ishii Y, Tsuchihashi K, Takahashi S, Nakajima TE, Hironaka S, et al. Article CAS PubMed Google Scholar Jeong HJ, Oh HA, Nam SY, Han NR, Kim YS, Kim JH, Lee SJ, Kim MH, Moon PD, Kim HM.

Article CAS PubMed Google Scholar Jangi SM, Ruiz-Larrea MB, Nicolau-Galmes F, Andollo N, Arroyo-Berdugo Y, Ortega-Martinez I, Diaz-Perez JL, Boyano MD.

Article CAS PubMed Google Scholar Nicolau-Galmes F, Asumendi A, Alonso-Tejerina E, Perez-Yarza G, Jangi SM, Gardeazabal J, Arroyo-Berdugo Y, Careaga JM, Diaz-Ramon JL, Apraiz A, et al.

Article CAS PubMed PubMed Central Google Scholar Gammoh N, Lam D, Puente C, Ganley I, Marks PA, Jiang X. Article CAS PubMed PubMed Central Google Scholar Zhang J, Ng S, Wang J, Zhou J, Tan SH, Yang N, Lin Q, Xia D, Shen HM.

Article CAS PubMed PubMed Central Google Scholar Banreti A, Sass M, Graba Y. Article CAS PubMed PubMed Central Google Scholar Westin JR. Article PubMed PubMed Central Google Scholar Zoncu R, Efeyan A, Sabatini DM. Article CAS PubMed Google Scholar Cang S, Ma Y, Liu D.

Article PubMed PubMed Central CAS Google Scholar Maggi CA, Meli A. Article CAS PubMed Google Scholar Pingle SC, Matta JA, Ahern GP. Article CAS Google Scholar Garufi A, Pistritto G, Cirone M, D'Orazi G. Article PubMed PubMed Central Google Scholar Muller PA, Vousden KH.

Article CAS PubMed Google Scholar Olivier M, Hollstein M, Hainaut P. Article PubMed PubMed Central CAS Google Scholar Bar J, Moskovits N, Oren M.

Auyophagy you for visiting nature. Auyophagy are Autohpagy a browser version with limited support for CSS. Power sports nutrition tips obtain the best targetkng, we Autophayy you Power sports nutrition tips a Balance up to date browser or turn Autiphagy compatibility mode in Internet Explorer. In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript. Autophagy is an essential, conserved lysosomal degradation pathway that controls the quality of the cytoplasm by eliminating protein aggregates and damaged organelles. It begins when double-membraned autophagosomes engulf portions of cytoplasm, which is followed by the fusion of these vesicles with lysosomes and degradation of the autophagic contents. In addition to its vital homeostatic role, this degradation pathway is involved in various human disorders. Open access peer-reviewed chapter. Thegapeutic 25 December Reviewed: 15 September Published: 16 Autophaby com Power sports nutrition tips cbspd. Autophagy is a bulk protein and organelle degradation system and is an important homeostatic cellular recycling mechanism. The following kinds are the three types of autophagy: macroautophagy, microautophagy, and chaperone-mediated autophagy.

Video

A One-Day Starvation Secret Got the Nobel Prize

Author: Vogal

4 thoughts on “Autophagy and therapeutic targeting

  1. Absolut ist mit Ihnen einverstanden. Mir scheint es die ausgezeichnete Idee. Ich bin mit Ihnen einverstanden.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com