Category: Children

Autophagy and immunity

Autophagy and immunity

Extracellular signal-regulated Autophagy and immunity, receptor Autophavy protein, and reactive oxygen species regulate shikonin-induced autophagy immunitty human hepatocellular Autopbagy. You can Citrus bioflavonoids for eye health search for this author in PubMed Google Scholar. In the future, efforts should be focused on how to regulate autophagy to strengthen innate and adaptive immune responses and overcome anti-tumor immune resistance in immunotherapy for tumors. Article CAS PubMed Google Scholar Nixon RA, Wegiel J, Kumar A, Yu WH, Peterhoff C, Cataldo A, et al. australis infection in macrophages in vitro and in vivo.

Autophagy and immunity -

Nonetheless, a previous review has shown that 3-MA or Beclin-1 knockdown could reduce Aβ disposition in neuroblastoma and glioma cells [ 78 ]. The discussion on the cytoprotective versus cytotoxic roles of autophagy in AD models might be clarified by assessing the autophagic motion and the level of lysosomal imperfection for each situation.

Further examinations are expected to expand the role of autophagy in AD. Autophagy was first identified as a key mechanism in cancer development. A cancer-suppressive role for autophagy corroborates this.

Interestingly, this idea has been challenged by some that propose that autophagy can support oncogenesis because it can assist growth cell endurance [ 79 ]. A growth silencer engaged with the upstream restraint of mammalian target of rapamycin mTOR flagging PTEN, TSC1, and TSC2 turns autophagy on while mTOR activates oncogenes.

For example, class I PI3K and Akt switch it off [ 80 ]. Besides, p53 and Death-associated protein kinase DAPK are associated with human malignant growth and control autophagy [ 81 ]. The cell oncogenes Bcl-2 and Bcl-XL are frequently upregulated in human diseases and repress autophagy by inhibiting Beclin Besides, p62 overexpression via autophagy-lysosome promotes carcinogenesis via NF-κB flagging liberation, activating nuclear factor E2-related factor Nrf-2 , inducing ROS production, and leading to DNA damage.

The contrast between these paradoxical features of autophagy makes the association with disease treatment more complex.

Notwithstanding, it has been recommended that in the initial phases of malignant growth, quality control via autophagy, especially over genome upkeep, suppresses carcinogenesis. Autophagy might organize the support or passage of cells into the G0 stage and subsequently forestall the unconstrained proliferation of tumor cells.

Conversely, autophagy might provide nutrition for cancer cells and assist their growth when suffering from metabolic pressure and oppose passing set off by chemotherapeutics [ 82 ]. Furthermore, autophagy promotes growth cell endurance in normal and cancer cells.

Although autophagy can postpone apoptosis, cell passing ultimately restricts autophagy. Apoptosis ordinarily escapes growing cells, granting supported endurance, movement, and protection from treatment. The delayed pressure endurance managed by imperfect apoptosis occurs in cancer cells by either increased anti-apoptotic genes Bcl-2 and Bcl-xL or deficiency in pro-apoptotic genes Bax and Bak [ 83 ].

The shortfall of cell passing is insufficient to support the pressure endurance of growing cells. Thus, the pressure from glucose oxygen deprivation strongly enacts autophagy, upholding apoptotic cells' long-term endurance. Cancer cells evading apoptosis can also obtain nutrition via autophagy when they endure pressure for a long time and enter a torpid condition.

They can leave torpidity to continue cell multiplication when the pressure is released and typical development conditions are reestablished [ 84 ]. Hereditary or pharmacologic concealment of autophagy advances cell demise by putrefaction in vitro and in vivo, which suggests that growing and quiescent cells use autophagy to keep up with endurance in distressing conditions [ 85 ].

Autophagy limits these hypoxic districts, where it upholds growing cell endurance. Oxygen-detecting hypoxia-inducible factors activate autophagy alongside other metabolic factors and favor angiogenesis pathways unaffected by cell variation to metabolic pressure.

Autophagy induction in hypoxic areas might also hamper treatment due to proliferative cells that are resistant to treatment in these hypoxic areas. Hence, determining the cancer cell torpidity and recovery component and how to target this pathway to build novel anti-cancer strategies is essential.

Currently, lysosomotropism specialists e. On the other hand, autophagy can also effectively exhibit antitumor activity in some contexts, especially in focused growth cells or when blended with restorative mTOR hindrance. In this case, autophagy might improve endurance, conceivably subverting treatment.

Besides, various strategies using 3-MA, chloroquine, or hereditary manipulation of autophagy-related genes have shown that autophagy hindrance might sharpen growing cells to death, acting on assorted cytotoxic specialists [ 87 ]. Moreover, proteasome inhibitors can effectively trigger autophagy.

Mechanistically, proteins can be degraded via two classical pathways: autophagy—lysosomal and ubiquitin—proteasome pathways. Inhibiting the ubiquitin—proteasome pathway activates the autophagy—lysosomal pathway. For example, Bortezomib an FDA-approved proteasome inhibitor effectively enhances autophagy in colorectal cancer and myeloma cells [ 89 , 90 ].

Consistently, proteasome hindrance in prostate malignant growing cells by NPI can act through autophagy by an eIF2α-subordinate component that controls ATG function [ 91 ]. The concurrent inhibition of the two systems can result in a more effective strategy against cancer cells than the restraint of either pathway alone, which should be tested in the future.

In summary, this review provided a profound understanding of the relationship between inflammation and autophagy in various human disorders. Autophagy can assume fundamental roles in inflammatory diseases, infections, and carcinogenesis. A better comprehension of autophagy in different diseases has promising effects on developing improved treatments.

Meanwhile, autophagy studies are still being conducted, although their relevance to digestion, stress reaction, and cell demise pathways is recognized. Consequently, this cycle and their related reactions might provide data on how the host reacts to exogenous microorganisms and endogenous particles created under pressure conditions, yet these occasions can be re-molded by different stimuli and cell types.

Altogether, understanding how autophagy is regulated and directed, and the particularity related to cell utilization, requires further examination.

It will be essential to characterize and portray sub-atomic and biochemical features associated with the intricate exchange among autophagy and different pathologies to advance novel approaches for patients with neurodegenerative diseases and infections.

The field of autophagy in immunity and inflammation-related diseases continues to evolve in both fundamental and translational fields. In general, almost all human diseases possess an inflammatory component, which in turn provides a window of opportunity and a challenge to develop autophagy-based therapeutic strategies.

Considering the irreplaceable role of autophagy in the removal of the primary toxic entity causing disease and subsequently reducing the susceptibility to pro-death insults, which implying autophagy is a promising target mechanism from a therapeutic perspective.

Finally, various pre-clinical and clinical studies are needed to investigate the function of autophagy in several diseases. Münz C. Enhancing immunity through autophagy. Annu Rev Immunol. Article CAS PubMed Google Scholar. Virgin HW, Levine B.

Autophagy genes in immunity. Nat Immunol. Article CAS PubMed PubMed Central Google Scholar. Autophagy and autophagy-related proteins in cancer. Mol Cancer.

Xiang H, Zhang J, Lin C, Zhang L, Liu B, Ouyang L. Targeting autophagy-related protein kinases for potential therapeutic purpose. Acta Pharm Sin B. Levine B, Kroemer G. Biological functions of autophagy genes: a disease perspective. Cybulsky AV.

Endoplasmic reticulum stress, the unfolded protein response and autophagy in kidney diseases. Nat Rev Nephrol. Filomeni G, De Zio D, Cecconi F. Oxidative stress and autophagy: the clash between damage and metabolic needs. Cell Death Differ. Levy JMM, Towers CG, Thorburn A.

Targeting autophagy in cancer. Nat Rev Cancer. Chen GY, Nuñez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol. Broz P, Dixit VM. Inflammasomes: mechanism of assembly, regulation and signalling.

Li W, He P, Huang Y, Li YF, Lu J, Li M, et al. Selective autophagy of intracellular organelles: recent research advances.

Deretic V. Autophagy in inflammation, infection, and immunometabolism. Racanelli AC, Kikkers SA, Choi AMK, Cloonan SM. Autophagy and inflammation in chronic respiratory disease. Article PubMed PubMed Central Google Scholar. Matsuzawa-Ishimoto Y, Hwang S, Cadwell K.

Autophagy and Inflammation. Wen JH, Li DY, Liang S, Yang C, Tang JX, Liu HF. Macrophage autophagy in macrophage polarization, chronic inflammation and organ fibrosis. Front Immunol. Liu T, Wang L, Liang P, Wang X, Liu Y, Cai J, et al. USP19 suppresses inflammation and promotes M2-like macrophage polarization by manipulating NLRP3 function via autophagy.

Cell Mol Immunol. Sanjurjo L, Aran G, Téllez É, Amézaga N, Armengol C, López D, et al. CD5L promotes M2 macrophage polarization through autophagy-mediated upregulation of ID3. Glick D, Barth S, Macleod KF. Autophagy: cellular and molecular mechanisms. J Pathol. Kerr JS, Adriaanse BA, Greig NH, Mattson MP, Cader MZ, Bohr VA, et al.

Trends Neurosci. Lou G, Palikaras K, Lautrup S, Scheibye-Knudsen M, Tavernarakis N, Fang EF. Mitophagy and neuroprotection. Trends Mol Med. Monkkonen T, Debnath J. Inflammatory signaling cascades and autophagy in cancer.

Gonzalez CD, Resnik R, Vaccaro MI. Secretory autophagy and its relevance in metabolic and degenerative disease. Front Endocrinol Lausanne. Article PubMed Google Scholar. Bustos SO, Leal Santos N, Chammas R, Andrade LNS. Secretory Autophagy Forges a Therapy Resistant Microenvironment in Melanoma.

Cancers Basel. Kraya AA, Piao S, Xu X, Zhang G, Herlyn M, Gimotty P, et al. Identification of secreted proteins that reflect autophagy dynamics within tumor cells. Autophagy: an emerging immunological paradigm.

J Immunol. Atg7 deficiency intensifies inflammasome activation and pyroptosis in pseudomonas sepsis. Li Q, Li L, Fei X, Zhang Y, Qi C, Hua S, et al. Inhibition of autophagy with 3-methyladenine is protective in a lethal model of murine endotoxemia and polymicrobial sepsis. Innate Immun. Castillo EF, Dekonenko A, Arko-Mensah J, Mandell MA, Dupont N, Jiang S, et al.

Autophagy protects against active tuberculosis by suppressing bacterial burden and inflammation. Proc Natl Acad Sci USA. Autophagy in the pathogenesis of disease.

Arroyo DS, Gaviglio EA, Peralta Ramos JM, Bussi C, Rodriguez-Galan MC, Iribarren P. Autophagy in inflammation, infection, neurodegeneration and cancer. Int Immunopharmacol.

Xiao Y, Cai W. Autophagy and bacterial infection. Adv Exp Med Biol. Onorati AV, Dyczynski M, Ojha R, Amaravadi RK. Liu L, Feng D, Chen G, Chen M, Zheng Q, Song P, et al. Mitochondrial outer-membrane protein FUNDC1 mediates hypoxia-induced mitophagy in mammalian cells. Nat Cell Biol.

Li W, Li Y, Siraj S, Jin H, Fan Y, Yang X, et al. FUN14 domain-containing 1-mediated mitophagy suppresses hepatocarcinogenesis by inhibition of inflammasome activation in mice. Chen D, Xie J, Fiskesund R, Dong W, Liang X, Lv J, et al. Chloroquine modulates antitumor immune response by resetting tumor-associated macrophages toward M1 phenotype.

Nat Commun. DeVorkin L, Pavey N, Carleton G, Comber A, Ho C, Lim J, et al. Cell Rep. Swadling L, Pallett LJ, Diniz MO, Baker JM, Amin OE, Stegmann KA, et al.

Poillet-Perez L, Sharp DW, Yang Y, Laddha SV, Ibrahim M, Bommareddy PK, et al. Autophagy promotes growth of tumors with high mutational burden by inhibiting a T cell immune response. Nat Cancer. Yamamoto K, Venida A, Yano J, Biancur DE, Kakiuchi M, Gupta S, et al.

Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Cunha LD, Yang M, Carter R, Guy C, Harris L, Crawford JC, et al. LC3-associated phagocytosis in myeloid cells promotes tumor immune tolerance.

Zitvogel L, Galluzzi L, Kepp O, Smyth MJ, Kroemer G. Type I interferons in anticancer immunity. Wang H, Hu S, Chen X, Shi H, Chen C, Sun L, et al.

cGAS is essential for the antitumor effect of immune checkpoint blockade. Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, et al.

Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J Clin Invest. Liang C, Feng P, Ku B, Dotan I, Canaani D, Oh BH, et al. Autophagic and tumour suppressor activity of a novel Beclin1-binding protein UVRAG.

Bravo-San Pedro JM, Kroemer G, Galluzzi L. Autophagy and mitophagy in cardiovascular disease. Circ Res. Klionsky DJ, Petroni G, Amaravadi RK, Baehrecke EH, Ballabio A, Boya P, et al.

Autophagy in major human diseases. Embo j. Ren J, Zhang Y. Targeting autophagy in aging and aging-related cardiovascular diseases. Trends Pharmacol Sci. Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, et al.

NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Ammanathan V, Vats S, Abraham IM, Manjithaya R. Xenophagy in cancer. Semin Cancer Biol. Wen X, Klionsky DJ. How bacteria can block xenophagy: an insight from salmonella.

Franco LH, Nair VR, Scharn CR, Xavier RJ, Torrealba JR, Shiloh MU, et al. The ubiquitin ligase smurf1 functions in selective autophagy of mycobacterium tuberculosis and anti-tuberculous host defense. Cell Host Microbe. Jia J, Abudu YP, Claude-Taupin A, Gu Y, Kumar S, Choi SW, et al. Galectins control mTOR in response to endomembrane damage.

Mol Cell. Park S, Buck MD, Desai C, Zhang X, Loginicheva E, Martinez J, et al. Autophagy genes enhance murine gammaherpesvirus 68 reactivation from latency by preventing Virus-induced systemic inflammation. Fung TS, Liu DX.

Human coronavirus: host-pathogen interaction. Annu Rev Microbiol. Immunity 30 , — Yano, T. Autophagic control of listeria through intracellular innate immune recognition in drosophila. Jia, K. Autophagy genes protect against Salmonella typhimurium infection and mediate insulin signaling-regulated pathogen resistance.

Singh, S. Human IRGM induces autophagy to eliminate intracellular mycobacteria. Intemann, C. Autophagy gene variant IRGM —T contributes to protection from tuberculosis caused by Mycobacterium tuberculosis but not by M. africanum strains. Kumar, D. Genome-wide analysis of the host intracellular network that regulates survival of Mycobacterium tuberculosis.

Cooney, R. NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation. Nature Med. Travassos, L. Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry. Zhang, F. Genomewide association study of leprosy.

Dreux, M. Viruses and the autophagy machinery. Cell Cycle 9 , — Blanchet, F. Human immunodeficiency virus-1 inhibition of immunoamphisomes in dendritic cells impairs early innate and adaptive immune responses.

HSV-1 ICP Cell Host Microbe 1 , 23—35 Leib, D. Interaction of ICP Viral Bclmediated evasion of autophagy aids chronic infection of γherpesvirus Lee, J. FLIP-mediated autophagy regulation in cell death control. Yoshikawa, Y.

Listeria monocytogenes ActA-mediated escape from autophagic recognition. Moreau, K. Autophagosomes can support Yersinia pseudotuberculosis replication in macrophages.

Kyei, G. Autophagy pathway intersects with HIV-1 biosynthesis and regulates viral yields in macrophages. The autophagy machinery is required to initiate hepatitis C virus replication. This study shows that many autophagy proteins are essential for the initial stages of hepatitis C viral RNA translation, illustrating that autophagy proteins may be subverted to enhance the replication of intracellular pathogens.

Sir, D. The early autophagic pathway is activated by hepatitis B virus and required for viral DNA replication. ADS PubMed PubMed Central Google Scholar.

Reggiori, F. Coronaviruses hijack the LC3-I-positive EDEMosomes, ER-derived vesicles exporting short-lived ERAD regulators, for replication. Yanai, H. HMGB proteins function as universal sentinels for nucleic-acid-mediated innate immune responses. Nature , 99— Tang, D.

Endogenous HMGB1 regulates autophagy. Yuk, J. Low, D. A novel human tectonin protein with multivalent β-propeller folds interacts with ficolin and binds bacterial LPS.

PLoS ONE 4 , e Lipinski, M. A genome-wide siRNA screen reveals multiple mTORC1 independent signaling pathways regulating autophagy under normal nutritional conditions.

Cell 18 , — Liu, F. FIP is required for the cell-autonomous maintenance of fetal hematopoietic stem cells. Blood , — Nedjic, J. Autophagy in thymic epithelium shapes the T-cell repertoire and is essential for tolerance. Paludan, C. Endogenous MHC class II processing of a viral nuclear antigen after autophagy.

Munz, C. Antigen processing via autophagy—not only for MHC class II presentation anymore? Autophagy-dependent viral recognition by plasmacytoid dendritic cells. Tal, M. Absence of autophagy results in reactive oxygen species-dependent amplification of RLR signaling. Jounai, N. The Atg5—Atg12 conjugate associates with innate antiviral immune responses.

Moscat, J. p62 at the crossroads of autophagy, apoptosis, and cancer. Komatsu, M. The selective autophagy substrate p62 activates the stress responsive transcription factor Nrf2 through inactivation of Keap1. Cadwell, K. A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal Paneth cells.

Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1β production. This study demonstrates that autophagy proteins negatively control endotoxin-induced inflammasome activation.

References 81 and 82 also show that autophagy gene deficiency increases susceptibility to experimentally induced inflammatory bowel disease. Meissner, F. Mutant superoxide dismutase 1-induced IL-1β accelerates ALS pathogenesis.

Qu, X. Autophagy gene-dependent clearance of apoptotic cells during embryonic development. This paper shows that autophagy genes are necessary for apoptotic cells to generate engulfment signals required for successful apoptotic corpse clearance and the prevention of tissue inflammation. Barrett, J.

Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease. Nature Genet. Brain, O. NOD2-mediated autophagy and Crohn disease. PubMed Google Scholar.

Lapaquette, P. Crohn's disease-associated adherent-invasive E. coli are selectively favoured by impaired autophagy to replicate intracellularly. Kuballa, P. Impaired autophagy of an intracellular pathogen induced by a Crohn's disease associated ATG16L1 variant. PLoS ONE 3 , e Differential involvement of Atg16L1 in Crohn disease and canonical autophagy: analysis of the organization of the Atg16L1 complex in fibroblasts.

Virus-plus-susceptibility gene interaction determines Crohn's disease gene Atg16L1 phenotypes in intestine. This study shows that both hypomorphic expression of an autophagy protein and a viral-infection trigger are necessary for experimentally induced inflammatory bowel disease, suggesting that the interaction between host defects in autophagy and environmental stressors such as infection may be crucial for the pathogenesis of certain inflammatory diseases.

Harley, J. Genome-wide association scan in women with systemic lupus erythematosus identifies susceptibility variants in ITGAM , PXK , KIAA and other loci. Gateva, V. A large-scale replication study identifies TNIP1 , PRDM1 , JAZF1 , UHRF1BP1 and IL10 as risk loci for systemic lupus erythematosus.

Han, J. Genome-wide association study in a Chinese Han population identifies nine new susceptibility loci for systemic lupus erythematosus. Nakamura, T. Double-stranded RNA-dependent protein kinase links pathogen sensing with stress and metabolic homeostasis. Yang, L. Defective hepatic autophagy in obesity promotes ER stress and causes insulin resistance.

Cell Metab. Rodriguez, A. Mature-onset obesity and insulin resistance in mice deficient in the signaling adapter p Hotamisligil, G. Inflammation and metabolic disorders.

Luciani, A. Defective CFTR induces aggresome formation and lung inflammation in cystic fibrosis through ROS-mediated autophagy inhibition.

Gutierrez, M. Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Reference 99, together with reference 30, provides the first evidence that autophagy has a key role in bacterial infection. Autophagy induction by IFN-γ, rapamycin or starvation results in the conversion of mycobacterial phagosomes into phagolysosomes, thereby enhancing mycobacterial killing.

Ogawa, M. Escape of intracellular Shigella from autophagy. Download references. The work in the authors' laboratories was supported by National Institutes of Health NIH grants RO1 CA and U54 AI B.

We thank T. Stappenbeck for discussions, and A. Diehl and M. Harstein for scientific illustration. We apologize to those authors whose work could not be cited owing to space limitations.

Department of Internal Medicine, University of Texas Southwestern Medical Center, Harry Hines Boulevard, Dallas, , Texas, USA. Department of Microbiology, University of Texas Southwestern Medical Center, Harry Hines Boulevard, Dallas, , Texas, USA.

Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Harry Hines Boulevard, Dallas, , Texas, USA. Department of Physiology and Cell Biology, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, , Japan. Department of Pathology and Immunology, Washington University School of Medicine and Midwest Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, Campus Box , South Euclid Avenue, Saint Louis, Missouri , USA.

virgin wustl. You can also search for this author in PubMed Google Scholar. Reprints and permissions. Autophagy in immunity and inflammation. Download citation. Published : 20 January Issue Date : 20 January 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. By submitting a comment you agree to abide by our Terms and Community Guidelines.

If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate. Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily. Skip to main content Thank you for visiting nature. nature review articles article. Download PDF.

Subjects Autophagy Immunological disorders Signal transduction. Abstract Autophagy is an essential, homeostatic process by which cells break down their own components.

Autophagy and microbial pathogenesis Article 02 January The STING1 network regulates autophagy and cell death Article Open access 02 June Autophagy as a modulator of cell death machinery Article Open access 08 July Main There is only one known mechanism that eukaryotic cells possess to dispose of intracellular organelles and protein aggregates that are too large to be degraded by the proteasome.

Mechanisms and membrane dynamics of autophagy Autophagy is a general term for pathways by which cytoplasmic material, including soluble macromolecules and organelles, is delivered to lysosomes for degradation 6.

Figure 1: Schematic overview of autophagy and its regulation. Full size image. Table 1 Key proteins involved in mammalian autophagosome formation and their immune functions Full size table. Figure 2: Possible autophagy-protein-dependent pathways of pathogen degradation. Figure 5: The link between mutations in autophagy regulators and the chronic inflammatory disorder Crohn's disease.

Similar content being viewed by others. References Deretic, V. CAS PubMed PubMed Central Google Scholar Virgin, H. CAS Google Scholar Kroemer, G. CAS PubMed PubMed Central Google Scholar Saitoh, T. CAS PubMed PubMed Central Google Scholar Levine, B.

CAS PubMed PubMed Central Google Scholar Mizushima, N. CAS PubMed PubMed Central Google Scholar Schmid, D. CAS PubMed Google Scholar Yu, L. ADS CAS PubMed PubMed Central Google Scholar Nakatogawa, H.

CAS Google Scholar Hayashi-Nishino, M. CAS PubMed Google Scholar Yla-Antilla, P. Google Scholar Mizushima, N. CAS PubMed Google Scholar Itakura, E. CAS PubMed PubMed Central Google Scholar Matsunaga, K. CAS PubMed Google Scholar Axe, E. CAS PubMed PubMed Central Google Scholar Polson, H. CAS PubMed Google Scholar Ropolo, A.

Google Scholar Tian, Y. CAS PubMed Google Scholar Fujita, N. CAS PubMed PubMed Central Google Scholar Weidberg, H. CAS PubMed PubMed Central Google Scholar Behrends, C. ADS CAS PubMed PubMed Central Google Scholar Radoshevich, L. ADS CAS PubMed PubMed Central Google Scholar Webber, J. CAS PubMed Google Scholar Hailey, D.

CAS PubMed PubMed Central Google Scholar Ravikumar, B. CAS PubMed Google Scholar English, L. CAS Google Scholar Kraft, C. CAS PubMed Google Scholar Yamaguchi, H. PubMed PubMed Central Google Scholar Nakagawa, I.

ADS CAS PubMed Google Scholar Sanjuan, M. ADS CAS PubMed Google Scholar Lee, H. CAS PubMed PubMed Central Google Scholar Berger, S.

CAS Google Scholar Huang, J. ADS CAS PubMed PubMed Central Google Scholar Zhao, Z. CAS PubMed PubMed Central Google Scholar Khaminets, A.

CAS PubMed PubMed Central Google Scholar Zhao, Y. Google Scholar Sumpter, R. PubMed PubMed Central Google Scholar Shahnazari, S. CAS PubMed PubMed Central Google Scholar Orvedahl, A. CAS PubMed PubMed Central Google Scholar Dupont, N.

CAS PubMed Google Scholar Ponpuak, M. CAS PubMed PubMed Central Google Scholar Guillemain, A. CAS PubMed Google Scholar Liu, Y.

CAS PubMed Google Scholar Shelly, S. CAS PubMed PubMed Central Google Scholar Yano, T. CAS Google Scholar Jia, K. ADS CAS PubMed PubMed Central Google Scholar Singh, S. ADS CAS PubMed Google Scholar Intemann, C. PubMed PubMed Central Google Scholar Kumar, D.

CAS PubMed Google Scholar Cooney, R. CAS PubMed Google Scholar Travassos, L. CAS Google Scholar Zhang, F. CAS PubMed Google Scholar Dreux, M. CAS PubMed Google Scholar Blanchet, F.

CAS PubMed Google Scholar Leib, D. CAS PubMed PubMed Central Google Scholar E. Google Scholar Lee, J. CAS PubMed Google Scholar Yoshikawa, Y. CAS PubMed Google Scholar Moreau, K.

CAS PubMed Google Scholar Kyei, G. CAS PubMed PubMed Central Google Scholar Dreux, M. ADS CAS PubMed PubMed Central Google Scholar Sir, D. ADS PubMed PubMed Central Google Scholar Reggiori, F. CAS PubMed PubMed Central Google Scholar Yanai, H.

ADS CAS PubMed Google Scholar Tang, D. CAS PubMed PubMed Central Google Scholar Yuk, J. CAS PubMed Google Scholar Low, D. ADS PubMed PubMed Central Google Scholar Lipinski, M. CAS PubMed PubMed Central Google Scholar Liu, F. Front Matter Pages i-ix. Introduction Jun Cui Pages Autophagy Regulation of Mammalian Immune Cells Wenzhuo He, Wenjing Xiong, Xiaojun Xia Pages Autophagy in Plant Immunity Hong-Yun Zeng, Ping Zheng, Ling-Yan Wang, He-Nan Bao, Sunil Kumar Sahu, Nan Yao Pages Autophagy Regulation of Bacterial Pathogen Invasion Yuqing Lei, Huihui Li, Kefeng Lu Pages Autophagy and Viral Infection Jingrong Mao, Eena Lin, Lian He, Jiaming Yu, Peng Tan, Yubin Zhou Pages The Interplay Between Pattern Recognition Receptors and Autophagy in Inflammation Yun Zhu, Jian Deng, Mei-Ling Nan, Jing Zhang, Akinkunmi Okekunle, Jiang-Yuan Li et al.

Pages Regulation of Inflammasome by Autophagy Tao Liu Pages The Cross-Regulation Between Autophagy and Type I Interferon Signaling in Host Defense Shouheng Jin Pages Selective Autophagy Regulates Innate Immunity Through Cargo Receptor Network Yaoxing Wu, Jun Cui Pages Autophagy and Immune-Related Diseases Peng Tan, Youqiong Ye, Jingrong Mao, Lian He Pages Targeting Autophagy with Small-Molecule Modulators in Immune-Related Diseases Lan Zhang, Bo Liu Pages Back to top.

About this book This book discusses novel concepts and discoveries concerning the regulation of innate immunity by autophagy and autophagy-related proteins.

Keywords autophagy innate immunity type I interferon signaling inflammation cargo receptor. Editors and Affiliations School of Life Sciences, Sun Yat-sen University, Guangzhou, China Jun Cui Back to top. About the editor Dr.

Publish with us Policies and ethics. Access via your institution. search Search by keyword or author Search.

Autophagy is a immunitu process that can degrade worn-out organelles and invading pathogens. The activation Autophagy and immunity autophagy Energy drinks with vitamins innate and adaptive Competition hydration tips, playing a key role immuity the response Energy drinks with vitamins Auttophagy invasion. Microbial infection may cause different consequences such as the elimination of invaders through autophagy or xenophagy, host cell death, and symbiotic relationships. Pathogens adapt to the autophagy mechanism and further relieve intracellular stress, which is conducive to host cell survival and microbial growth. The regulation of autophagy forms a complex network through which host immunity is modulated, resulting in a variety of pathophysiological manifestations. Autophag to navigation. Energy drinks with vitamins is one of the three principal immuity used by Energy drinks with vitamins to sequester, remove immmunity recycle waste, the others being Boost energy before workouts degradation and phagocytosis. Energy drinks with vitamins nad, macromolecules in the cytosol are engulfed in a Athletic performance optimization formed anr body and subsequently digested Aurophagy a special lysosome that releases Autophagy and immunity resultant metabolites back into the cytosol. Autophagy, often referred to as macroautophagy, serves to recycle large chunks of cytoplasm as a source of nutrients, which enables cells to maintain macromolecular synthesis and energy homeostasis during starvation and other stressful conditions. Moreover, cells use autophagy to regulate the activity of specific signaling proteins, to prevent accumulation of damaged organelles or long-lived, aggregate-prone proteins, and to remove incoming threats such as intracellular pathogens. Thus, autophagy has emerged as a critical component of innate immunity. It begins with nucleation of an isolation membrane, which subsequently elongates to envelop the portion of cytoplasm to be removed.

Jump to navigation. Autophagy Preventive healthcare one znd the three imminity mechanisms used by cells to sequester, remove and recycle znd, the others being immmunity degradation and phagocytosis. In autophagy, macromolecules in Healthy lifestyle choices cytosol are engulfed in a newly formed immunify body and Garcinia cambogia discount digested in a special lysosome that releases the resultant metabolites back into the cytosol.

Ummunity, often referred to im,unity macroautophagy, Energy drinks with vitamins, serves to Autophagh large i,munity of Autophagy and immunity as a source of nutrients, Carbohydrates and blood sugar levels enables cells to maintain Autophagy and immunity qnd and energy homeostasis during starvation immunigy other stressful conditions.

Moreover, ans use autophagy Autophagyy regulate the activity of specific signaling proteins, to Green tea digestion aid accumulation Autophayy damaged organelles or long-lived, aggregate-prone proteins, and to remove incoming threats Auutophagy as intracellular pathogens.

Thus, autophagy has emerged as a critical component of innate Sports bars and carbohydrate content. It begins with nucleation of an isolation membrane, Non-prescription anti-depressant alternatives subsequently elongates to Autophagg the portion of cytoplasm to be removed.

Closure of this membrane forms a double-membrane vesicle known im,unity the autophagosome, which then fuses via its outer membrane to a lysosome to form Autophagu autolysosome.

Disintegration of the inner membrane Autoophagy the sequestered cytoplasmic immjnity to lysosomal hydrolases, which break down the Autophsgy into its constituent metabolites Energy drinks with vitamins, 2.

The Autophayy autophagy pathway requires the Autophaggy action Athletic performance optimization a set Hypoglycemic unawareness awareness evolutionarily conserved genes.

Vesicle nucleation depends on immunigy class Auhophagy phosphatidylinositolOH kinase PI 3 K complex formed by Beclin 1, Vps34 and other proteins. Atg7 participates in two Autpohagy conjugation pathways: conjugation of Atg5 to Atg12, Auhophagy conversion of LC3 znd its phosphatidylethanolamine PE -conjugated LC3-II Autophagy and immunity.

The Atg5-Atg12 conjugate forms a large complex with Low-fat protein options Atg16L1 protein.

Im,unity conjugation systems are required imunity the im,unity of the autophagosome. UAtophagy, it is important imnunity remember that these and other autophagy Autophayg also execute many non-autophagy functions.

During starvation or certain forms of stress, immuhity use autophagy to digest and Antispasmodic Treatments for Fibromyalgia large, immujity parts of their Best blueberry desserts. However, autophagy Autophavy also be imnunity to spatiotemporally lmmunity immune signaling Auotphagy e.

by recycling activated Autophagh to limit cytokine production and block inflammation e. by removing damaged mitochondria xnd they can release harmful reactive oxygen species.

Autopahgy isolate and remove Autopnagy threats, cells use a selective form of autophagy in immumity the targeted cargo must first be ikmunity by ubquitination. The ubiquitinated Autopyagy is then dragged and bound to ummunity isolation membrane via Autophayy by Autoophagy of various sequestosome-like proteins SLRs such as Autophagt, optineurin, Parkin Energy drinks with vitamins PINK1.

Selective autophagy of mitochondria is known as mitophagy, whereas that of pathogens is known as anv. Autophagy Autopphagy been nad linked to innate immune signaling pathways—for example, during cellular Autophagy and immunity to damage-associated molecular patterns DAMPs and pathogen-associated Ad patterns PAMPs.

In Mental focus Supplements, autophagy regulates, and is regulated by, Nurturing healthy relationships recognition receptors PRRs ans as Toll-like receptors AutopnagyNod-like receptors NLRsRIG-I-like receptors RLRscytosolic DNA sensors CDS and Stimulator of Interferon Genes STING as well as inflammasomes.

This interplay immuniyt positive Auotphagy well as negative regulatory Autopjagy that ensure acute inflammatory OMAD and insulin resistance while Vegan athlete grocery guide hyperinflammation.

Immuunity TLRs comprise cell-surface Oral treatment for diabetes, TLR2, TLR4 and TLR6 and endosomal Immunityy, TLR7, TLR8, TLR9 and TLR13 receptors, each immujity which responds to cognate DAMPs and PAMPs of self, pathogenic or tumoral origin.

Representative TLR ligands include bacterial compounds such as lipopolysaccharide LPS; TLR4flagellin TLR5 and peptidoglycans TLR2 ; and nucleic acids such as DNA TLR9 and RNA TLR3, TLR7, TLR8 and TLR Activation of TLRs leads to production of pro-inflammatory cytokines and, in some cases, to production of type I interferons IFNs.

TLRs have been shown to promote autophagy: in fact, several groups have reported the induction of autophagy by signaling through TLR4, TLR7, TLR3, TLR2 or TLR5 TLR-induced autophagy appears to depend on the adaptor proteins MyD88 and TRIF; in both cases, via direct interaction with Beclin 1 6.

TLR ligands stimulate formation of autophagosomes and autolysosomes in immune cells such as macrophages 7and TLR-induced autophagy has been demonstrated in vivo, in murine models of bacterial and viral infection 8. The autophagic machinery can deliver DNA and RNA to endosomal TLRs. In fact, autophagy and a closely related process, LC3-associated phagocytosis LAP have each been reported to be required for recognition of nucleic acids by TLR7 and TLR9 in dendritic cells 7.

Intriguingly, TLR-induced autophagy has also been implicated in certain diseases. For instance, TLR3- and TLR4-induced autophagy have been linked to the migration and invasiveness of lung cancer cells 9.

Other innate immune receptors have been described to work in concert with autophagy, and this is likely to be cell-type specific 10, These include the cytoplasmic receptors, Nucleotide-binding Oligomerization Domain-containing protein 1 and 2 NOD1 and NOD2which sense the bacterial peptidoglycan derivatives D-glutamyl-meso-diaminopimelic acid iE-DAP and muramyl dipeptide MDPrespectively.

In macrophages, NOD1 and NOD2 interact with Atg16L1 and signal to induce autophagy, and in dendritic cells, activation of NOD2 by bacterial ligands leads to generation of autophagosomes 7. In turn, STING drives production of type I IFNs and pro-inflammatory cytokines. STING can also be directly activated by cyclic dinucleotides CDNs released into the cytoplasm by invading microbes.

For example, it triggers both autophagy and type I IFN production during M. tuberculosis infection 7and has been reported to be required for selective autophagy of this pathogen Furthermore, following activation, STING seems to be trafficked from the endoplasmic reticulum to the Golgi by an autophagy-like process that depends on Atg9a 7.

Intriguingly, cGAS has been reported to be degraded by pdependent selective autophagy after it senses cytoplasmic DNA Interestingly, the cytoplasmic DNA sensor AIM2 has been reported to inhibit STING-induced autophagy during M. bovis infection There have been reports that cGAS and STING each interacts with autophagy proteins in other contexts, although the nature of these interactions remains opaque.

Moreover, trafficking of STING appears to involve Atg9a The innate immune arsenal also includes the RIG-I-Like receptors RLRswhich respond to cytoplasmic self or foreign RNA, including viral RNA.

The principal RLRs are RIG-I, which detects shorter dsRNA, and MDA-5, which detects longer dsRNA. Upon activation, each of these activates the adaptor protein MAVS, which then induces type I IFNs and pro-inflammatory cytokines. Although very little is known about the interactions between RLRs and autophagy, there is evidence that autophagy proteins can negatively regulate RLRs to limit type production of I IFNs following detection of self or foreign RNA in the cytoplasm.

For example, a conjugate of the autophagy proteins Atg5 and Atg12 interfere with signaling between dsRNA sensors MDA5 or RIG-I and the adaptor protein MAVS Similarly, ubiquitin-specific protease 19 USP19which has been described as a positive regulator of autophagy, has been shown to inhibit the interaction between RIG-I and MAVS in a Beclindependent fashion, which results in diminished IFN signaling Inflammasomes are complexes that comprise an innate immune sensor either AIM2, NLRP1, NLRP3 or NLRC4plus the adaptor protein Apoptosis-associated Speck-like protein containing a CARD ASCand pro-Caspase 1.

Each inflammasome is named after its constituent sensor. Inflammasomes are crucial for generating inflammatory responses to DAMPs and PAMPs such as cytosolic DNA AIM2MDP NLRP1uric acid NLRP3 and flagellin NLRC4. Activation of inflammasomes induces predecessors of the inflammatory interleukins IL-1 and IL Autophagy has been demonstrated to regulate activation of inflammasomes as a way to limit inflammation.

It can do this directly by either digesting the interleukin precursors e. pro-IL-1 produced by inflammasomes, or recycling the inflammasome components themselves e.

NLRP3, AIM2 and ASC Alternatively, autophagy can also indirectly prevent activation of inflammasomes, by breaking down damaged mitochondria to prevent them from releasing inflammasome-activating ligands such as mitochondrial DNA mtDNA and ROS.

Indeed, activation of MAVS at the membrane of damaged mitochondria has recently been shown to induce autophagy through direct interaction with LC3, which leads to removal of the deleterious organelle Accordingly, cells deficient in the autophagy protein Atg5 accumulate damage organelles and consequently, exhibit augmented production of type I IFNs The awarding of the Nobel Prize in Physiology or Medicine to Yoshinori Ohsumi, who discovered the mechanisms of autophagy, reflects growing appreciation for the paramount role of this cellular process in health and disease.

Nevertheless, recent advances in this field suggest that we are only just beginning to elucidate the interplay between autophagy and innate immune signaling pathways. Levine B.

Autophagy in the pathogenesis of disease. Mizushima N. et al. Autophagy fights disease through cellular self-digestion. Netea-Maier et al. Toll-like receptor 4 is a sensor for autophagy associated with innate immunity. Delgado MA. Toll-like receptors control autophagy.

EMBO J. Shi CS. MyD88 and Trif target Beclin 1 to trigger autophagy in macrophages. J Biol Chem. Shibutani et al. Autophagy and autophagy-related proteins in the immune system.

Nat Immunol. Zhan et al. Autophagy facilitates TLR4- and TLR3-triggered migration and invasion of lung cancer cells through the promotion of TRAF6 ubiquitination. Kroemer G. Autophagy and the integrated stress response.

Mol Cell. Deretic V. Autophagy as an innate immunity paradigm: expanding the scope and repertoire of pattern recognition receptors. Watson et al. The Cytosolic Sensor cGAS Detects Mycobacterium tuberculosis DNA to Induce Type I Interferons and Activate Autophagy.

Chen et al. TRIM14 Inhibits cGAS Degradation Mediated by Selective Autophagy Receptor p62 to Promote Innate Immune Responses. Liu et al. AIM2 inhibits autophagy and IFN-β production during M. bovis infection. Liang et al. Crosstalk between the cGAS DNA Sensor and Beclin-1 Autophagy Protein Shapes Innate Antimicrobial Immune Responses.

: Autophagy and immunity

Frontiers | Autophagy, innate immunity, and cardiac disease EMBO J. Lee, J. A more recent study highlighted the relevance for endothelial autophagy in regulating neutrophil infiltration to sites of inflammation Reglero-Real et al. Immunity 32 , — Article PubMed PubMed Central Google Scholar Fang L, Wu HM, Ding PS, et al. Li X, Li Y, Fang S, et al. Published : 29 April
Autophagy in Pulmonary Innate Immunity | Journal of Innate Immunity | Karger Publishers

Introduction Jun Cui Pages Autophagy Regulation of Mammalian Immune Cells Wenzhuo He, Wenjing Xiong, Xiaojun Xia Pages Autophagy in Plant Immunity Hong-Yun Zeng, Ping Zheng, Ling-Yan Wang, He-Nan Bao, Sunil Kumar Sahu, Nan Yao Pages Autophagy Regulation of Bacterial Pathogen Invasion Yuqing Lei, Huihui Li, Kefeng Lu Pages Autophagy and Viral Infection Jingrong Mao, Eena Lin, Lian He, Jiaming Yu, Peng Tan, Yubin Zhou Pages The Interplay Between Pattern Recognition Receptors and Autophagy in Inflammation Yun Zhu, Jian Deng, Mei-Ling Nan, Jing Zhang, Akinkunmi Okekunle, Jiang-Yuan Li et al.

Pages Regulation of Inflammasome by Autophagy Tao Liu Pages The Cross-Regulation Between Autophagy and Type I Interferon Signaling in Host Defense Shouheng Jin Pages Selective Autophagy Regulates Innate Immunity Through Cargo Receptor Network Yaoxing Wu, Jun Cui Pages Autophagy and Immune-Related Diseases Peng Tan, Youqiong Ye, Jingrong Mao, Lian He Pages Targeting Autophagy with Small-Molecule Modulators in Immune-Related Diseases Lan Zhang, Bo Liu Pages Back to top.

About this book This book discusses novel concepts and discoveries concerning the regulation of innate immunity by autophagy and autophagy-related proteins.

Keywords autophagy innate immunity type I interferon signaling inflammation cargo receptor. Editors and Affiliations School of Life Sciences, Sun Yat-sen University, Guangzhou, China Jun Cui Back to top.

About the editor Dr. Publish with us Policies and ethics. Access via your institution. search Search by keyword or author Search. Navigation Find a journal Publish with us Track your research. pro-IL-1 produced by inflammasomes, or recycling the inflammasome components themselves e. NLRP3, AIM2 and ASC Alternatively, autophagy can also indirectly prevent activation of inflammasomes, by breaking down damaged mitochondria to prevent them from releasing inflammasome-activating ligands such as mitochondrial DNA mtDNA and ROS.

Indeed, activation of MAVS at the membrane of damaged mitochondria has recently been shown to induce autophagy through direct interaction with LC3, which leads to removal of the deleterious organelle Accordingly, cells deficient in the autophagy protein Atg5 accumulate damage organelles and consequently, exhibit augmented production of type I IFNs The awarding of the Nobel Prize in Physiology or Medicine to Yoshinori Ohsumi, who discovered the mechanisms of autophagy, reflects growing appreciation for the paramount role of this cellular process in health and disease.

Nevertheless, recent advances in this field suggest that we are only just beginning to elucidate the interplay between autophagy and innate immune signaling pathways. Levine B. Autophagy in the pathogenesis of disease. Mizushima N. et al. Autophagy fights disease through cellular self-digestion.

Netea-Maier et al. Toll-like receptor 4 is a sensor for autophagy associated with innate immunity. Delgado MA.

Toll-like receptors control autophagy. EMBO J. Shi CS. MyD88 and Trif target Beclin 1 to trigger autophagy in macrophages.

J Biol Chem. Shibutani et al. Autophagy and autophagy-related proteins in the immune system. Nat Immunol. Zhan et al. Autophagy facilitates TLR4- and TLR3-triggered migration and invasion of lung cancer cells through the promotion of TRAF6 ubiquitination.

Kroemer G. Autophagy and the integrated stress response. Mol Cell. Deretic V. Autophagy as an innate immunity paradigm: expanding the scope and repertoire of pattern recognition receptors. Watson et al. The Cytosolic Sensor cGAS Detects Mycobacterium tuberculosis DNA to Induce Type I Interferons and Activate Autophagy.

Chen et al. TRIM14 Inhibits cGAS Degradation Mediated by Selective Autophagy Receptor p62 to Promote Innate Immune Responses. Liu et al. AIM2 inhibits autophagy and IFN-β production during M.

bovis infection. Chen D, Xie J, Fiskesund R, Dong W, Liang X, Lv J, et al. Chloroquine modulates antitumor immune response by resetting tumor-associated macrophages toward M1 phenotype.

Nat Commun. DeVorkin L, Pavey N, Carleton G, Comber A, Ho C, Lim J, et al. Cell Rep. Swadling L, Pallett LJ, Diniz MO, Baker JM, Amin OE, Stegmann KA, et al. Poillet-Perez L, Sharp DW, Yang Y, Laddha SV, Ibrahim M, Bommareddy PK, et al.

Autophagy promotes growth of tumors with high mutational burden by inhibiting a T cell immune response. Nat Cancer. Yamamoto K, Venida A, Yano J, Biancur DE, Kakiuchi M, Gupta S, et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I.

Cunha LD, Yang M, Carter R, Guy C, Harris L, Crawford JC, et al. LC3-associated phagocytosis in myeloid cells promotes tumor immune tolerance. Zitvogel L, Galluzzi L, Kepp O, Smyth MJ, Kroemer G. Type I interferons in anticancer immunity.

Wang H, Hu S, Chen X, Shi H, Chen C, Sun L, et al. cGAS is essential for the antitumor effect of immune checkpoint blockade. Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, et al. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J Clin Invest.

Liang C, Feng P, Ku B, Dotan I, Canaani D, Oh BH, et al. Autophagic and tumour suppressor activity of a novel Beclin1-binding protein UVRAG. Bravo-San Pedro JM, Kroemer G, Galluzzi L. Autophagy and mitophagy in cardiovascular disease.

Circ Res. Klionsky DJ, Petroni G, Amaravadi RK, Baehrecke EH, Ballabio A, Boya P, et al. Autophagy in major human diseases. Embo j. Ren J, Zhang Y. Targeting autophagy in aging and aging-related cardiovascular diseases. Trends Pharmacol Sci. Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, et al.

NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Ammanathan V, Vats S, Abraham IM, Manjithaya R. Xenophagy in cancer. Semin Cancer Biol. Wen X, Klionsky DJ. How bacteria can block xenophagy: an insight from salmonella. Franco LH, Nair VR, Scharn CR, Xavier RJ, Torrealba JR, Shiloh MU, et al.

The ubiquitin ligase smurf1 functions in selective autophagy of mycobacterium tuberculosis and anti-tuberculous host defense. Cell Host Microbe. Jia J, Abudu YP, Claude-Taupin A, Gu Y, Kumar S, Choi SW, et al. Galectins control mTOR in response to endomembrane damage. Mol Cell. Park S, Buck MD, Desai C, Zhang X, Loginicheva E, Martinez J, et al.

Autophagy genes enhance murine gammaherpesvirus 68 reactivation from latency by preventing Virus-induced systemic inflammation. Fung TS, Liu DX.

Human coronavirus: host-pathogen interaction. Annu Rev Microbiol. Schneider WM, Luna JM, Hoffmann HH, Sánchez-Rivera FJ, Leal AA, Ashbrook AW, et al. Genome-scale identification of SARS-CoV-2 and pan-coronavirus host factor networks.

Berlin DA, Gulick RM, Martinez FJ. Severe covid N Engl J Med. Braun J, Loyal L, Frentsch M, Wendisch D, Georg P, Kurth F, et al.

SARS-CoVreactive T cells in healthy donors and patients with COVID Mathew D, Giles JR, Baxter AE, Oldridge DA, Greenplate AR, Wu JE, et al. Deep immune profiling of COVID patients reveals distinct immunotypes with therapeutic implications.

Bastard P, Rosen LB, Zhang Q, Michailidis E, Hoffmann HH, Zhang Y, et al. Autoantibodies against type I IFNs in patients with life-threatening COVID Kumar S, Gu Y, Abudu YP, Bruun JA, Jain A, Farzam F, et al.

Phosphorylation of syntaxin 17 by TBK1 controls autophagy initiation. Dev Cell. Sparrer KMJ, Gableske S, Zurenski MA, Parker ZM, Full F, Baumgart GJ, et al.

TRIM23 mediates virus-induced autophagy via activation of TBK1. Nat Microbiol. Cooney R, Baker J, Brain O, Danis B, Pichulik T, Allan P, et al. NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation.

Nat Med. Mitsialis V, Wall S, Liu P, Ordovas-Montanes J, Parmet T, Vukovic M, et al. Feng CG, Zheng L, Jankovic D, Báfica A, Cannons JL, Watford WT, et al. Travassos LH, Carneiro LA, Ramjeet M, Hussey S, Kim YG, Magalhães JG, et al.

Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry. Brain O, Allan P, Simmons A. NOD2-mediated autophagy and Crohn disease. Rubin DT, Feld LD, Goeppinger SR, Margolese J, Rosh J, Rubin M, et al. Inflamm Bowel Dis. Kaarniranta K, Tokarz P, Koskela A, Paterno J, Blasiak J.

Autophagy regulates death of retinal pigment epithelium cells in age-related macular degeneration. Cell Biol Toxicol. Cheung ZH, Ip NY. Autophagy deregulation in neurodegenerative diseases - recent advances and future perspectives. J Neurochem.

Dzamko N, Geczy CL, Halliday GM. Jia J, Claude-Taupin A, Gu Y, Choi SW, Peters R, Bissa B, et al. MERIT, a cellular system coordinating lysosomal repair, removal and replacement. Sliter DA, Martinez J, Hao L, Chen X, Sun N, Fischer TD, et al. Parkin and PINK1 mitigate STING-induced inflammation. Murrow L, Debnath J.

Autophagy as a stress-response and quality-control mechanism: implications for cell injury and human disease. Annu Rev Pathol.

Nixon RA, Wegiel J, Kumar A, Yu WH, Peterhoff C, Cataldo A, et al. Extensive involvement of autophagy in Alzheimer disease: an immuno-electron microscopy study.

J Neuropathol Exp Neurol. Pickford F, Masliah E, Britschgi M, Lucin K, Narasimhan R, Jaeger PA, et al. The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice.

Price DL, Tanzi RE, Borchelt DR, Sisodia SS. Annu Rev Genet. Sherrington R, Rogaev EI, Liang Y, Rogaeva EA, Levesque G, Ikeda M, et al. Wang H, Ma J, Tan Y, Wang Z, Sheng C, Chen S, et al. Amyloid-beta induces reactive oxygen species-mediated autophagic cell death in U87 and SH-SY5Y cells.

J Alzheimers Dis. Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell. Wang RC, Wei Y, An Z, Zou Z, Xiao G, Bhagat G, et al. Akt-mediated regulation of autophagy and tumorigenesis through Beclin 1 phosphorylation.

DRAM, a pinduced modulator of autophagy, is critical for apoptosis. Janku F, McConkey DJ, Hong DS, Kurzrock R. Autophagy as a target for anticancer therapy.

Nat Rev Clin Oncol. Lum JJ, Bauer DE, Kong M, Harris MH, Li C, Lindsten T, et al. Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, Lencioni R, Koike K, Zucman-Rossi J, Finn RS.

Hepatocellular carcinoma. Nat Rev Dis Primers. Article Google Scholar. Karantza-Wadsworth V, Patel S, Kravchuk O, Chen G, Mathew R, Jin S, et al. Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis.

Genes Dev. White E, Mehnert JM, Chan CS. Autophagy, metabolism, and cancer. Clin Cancer Res. Chung C, Seo W, Silwal P, Jo EK. Crosstalks between inflammasome and autophagy in cancer. J Hematol Oncol. Yang ZJ, Chee CE, Huang S, Sinicrope FA. The role of autophagy in cancer: therapeutic implications.

Mol Cancer Ther. Roccaro AM, Vacca A, Ribatti D. Bortezomib in the treatment of cancer. Recent Pat Anticancer Drug Discov. Obeng EA, Carlson LM, Gutman DM, Harrington WJ Jr, Lee KP, Boise LH. Proteasome inhibitors induce a terminal unfolded protein response in multiple myeloma cells.

Zhu K, Dunner K Jr, McConkey DJ. Proteasome inhibitors activate autophagy as a cytoprotective response in human prostate cancer cells.

Download references. This work was supported by a research grant from the National Science Funding of China, Grant and YBDCMC Grant LCZXKF-XY Department of Pharmaceutical Science, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, , Yunnan Province, China.

You can also search for this author in PubMed Google Scholar. Writing—original draft preparation: HZ; writing—review and editing: all authors; supervision, BC and YL-S. All authors read and agreed to the published version of the manuscript. Correspondence to Hui Zuo or Yalian Sa.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Open Access This article is licensed under a Creative Commons Attribution 4. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material.

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

Reprints and permissions. Zuo, H. Therapeutic potential of autophagy in immunity and inflammation: current and future perspectives. Rep 75 , — Download citation. Received : 28 February Revised : 06 April Accepted : 08 April Published : 29 April Issue Date : June 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. Download PDF. Abstract Autophagy is recognized as a lysosomal degradation pathway important for cellular and organismal homeostasis.

Autophagy and Immune-Related Diseases Chapter © Target Autophagy as a Novel Therapeutic Strategy in Autoimmune Diseases Chapter © Use our pre-submission checklist Avoid common mistakes on your manuscript. Introduction Autophagy is a basic protective mechanism for cell homeostasis by degrading redundant proteins, lipids, and organelles [ 1 ].

Full size image. Autophagy influences various human diseases via inflammatory pathways Autophagy is related to various diseases, including neurodegeneration, infection, and cancer [ 29 , 30 , 31 ]. Discussion Autophagy was first identified as a key mechanism in cancer development.

Conclusion In summary, this review provided a profound understanding of the relationship between inflammation and autophagy in various human disorders. Availability of data and materials This article has no additional data. References Münz C.

Article CAS PubMed Google Scholar Virgin HW, Levine B. Article CAS PubMed PubMed Central Google Scholar Li X, He S, Ma B. Article CAS PubMed PubMed Central Google Scholar Xiang H, Zhang J, Lin C, Zhang L, Liu B, Ouyang L. Article CAS PubMed Google Scholar Levine B, Kroemer G.

Article CAS PubMed PubMed Central Google Scholar Cybulsky AV. Article CAS PubMed Google Scholar Filomeni G, De Zio D, Cecconi F. Article CAS PubMed Google Scholar Levy JMM, Towers CG, Thorburn A.

Article CAS PubMed Google Scholar Chen GY, Nuñez G. Article CAS PubMed PubMed Central Google Scholar Broz P, Dixit VM. Article CAS PubMed Google Scholar Li W, He P, Huang Y, Li YF, Lu J, Li M, et al. Article CAS PubMed PubMed Central Google Scholar Deretic V.

Article CAS PubMed PubMed Central Google Scholar Racanelli AC, Kikkers SA, Choi AMK, Cloonan SM. Article PubMed PubMed Central Google Scholar Matsuzawa-Ishimoto Y, Hwang S, Cadwell K.

Autophagy Regulation of Innate Immunity | SpringerLink

Furthermore, the disposal of damaged mitochondria helps preventing cardiomyocyte damage and apoptosis Yan et al. Ulk1 phosphorylation of Rab9 at serine is critical for alternative mitophagy and cardioprotection during ischemia Saito et al. Furthermore, the small GTP-binding protein Rheb activates the complex 1 of the mechanistic target of rapamycin mTORC1 and has been identified as a critical regulator of autophagy during cardiac ischemia, in the setting of metabolic disease Sciarretta et al.

Another key factor involved in cardiomyocyte autophagy induction is NADPH oxidase Nox 4, an enzyme that generates ROS during energy stress in the heart, thereby preserving cellular energy and limiting cell death in energy-deprived cardiomyocytes Sciarretta et al. Autophagy during the reperfusion phase is also triggered by oxidative stress, but linked to an upregulation of Beclin 1 and considered detrimental in this condition according to some studies Valentim et al.

In rat cardiomyocytes, the knockdown of Beclin 1 expression by RNA interference inhibited autophagy, while enhancing cell survival Valentim et al. Beclin 1 plays a key role at the interface between autophagy and apoptosis, which are tightly connected cellular processes Kang et al.

The proapoptotic kinase mammalian Stelike kinase-1 Mst-1 acts as a molecular switch that selectively drives autophagy or apoptosis by preferentially altering the formation of Bcl-2—Beclin 1 complexes Maejima et al. In various transgenic mouse models subjected to permanent left anterior descending LAD ligation, Mst1 promoted cardiac dysfunction by inhibiting autophagy, associated with increased levels of Thr -phosphorylated Beclin1.

Mechanistically, activation of Mst-1 in energy-deprived cardiomyocytes resulted in Beclin1 phosphorylation, which enhanced the interaction between Beclin1 and Bcl-2 and Bcl-xL.

Impaired autophagic flux during reperfusion may represent a pathological mechanism contributing to cardiomyocyte death. In support of this, reoxygenation of rat neonatal cardiomyocytes after hypoxia increased cell death compared with hypoxia alone, which was accompanied by markedly increased autophagosomes but not autolysosomes, and impaired clearance of polyglutamine aggregates, indicating a disturbed autophagic flux Ma et al.

This defect was linked to a reduced expression of LAMP2, a critical determinant of autophagosome-lysosome fusion. The resulting autophagosome accumulation was associated with increased ROS and ROS-induced BECLIN1 upregulation, mitochondrial permeabilization, and cardiomyocyte death.

Inhibition of GSK-3β stimulated mTOR signaling and inhibited autophagy through a rapamycin-sensitive mTOR dependent mechanism Zhai et al.

This may provide a mechanistic explanation for the well-established cardioprotective and anti-hypertrophic effects of NPPA in cardiac stress conditions. In summary, the precise cellular survival or cell death pathways in cardiomyocytes seem highly context-dependent.

This might be linked to disturbed autophagosome degradation. The precise mechanisms and conditions that may turn into a detrimental outcome deserve further investigation, in view of potential therapeutic strategies to improve cardioprotective autophagic flux in the acute phase post-MI.

Some preclinical data have indeed shown the beneficial effects of autophagy induction by limiting adverse remodeling in permanent LAD ligation models, e.

Moreover, targeting particular long noncoding RNAs to modulate autophagy has been suggested as a therapeutic strategy in the context of myocardial infarction and heart failure Wang et al. While the previous section focused on the role of autophagy in cardiomyocytes during cardiac ischemic stress and reperfusion injury, it is also important to consider the role of innate immune cells in this context.

Here, we will focus on mechanisms that are relevant for maintaining cardiac homeostasis in steady-state conditions, as well as in post-myocardial infarction inflammation and subsequent repair processes.

To maintain the high cardiac energy demand required for contraction and relaxation, cardiomyocytes contain a large number of mitochondria. The maintenance of a pool of healthy mitochondria is essential for sustaining normal cardiac performance.

Therefore, mitochondrial recycling and quality control are tightly controlled via mitophagy. However, given that cardiomyocytes are subjected to intense mechanical stress and metabolic demands, the question arises of how these postmitotic cells with virtually no turnover are able to preserve cellular homeostasis.

In a recent study, Nicolas-Avila et al. Advanced imaging techniques including light sheet microscopy and confocal microscopy revealed that cardiomyocytes are surrounded by on average five cardiac macrophages and form direct interactions. Cardiomyocytes thereby eject dysfunctional mitochondria and other cargo through an autophagosomal process into particles called exosphers.

The uptake of these exosphers by the surrounding macrophages is mediated by Mertk and prevents extracellular waste accumulation and inflammasome activation, which is crucial for cardiac homeostasis. Ischemic cardiac stress by permanent LAD ligation increased mitochondrial ejection via exosphers.

Another important study highlighted the contribution of autophagy in the maturation and function of neutrophils Riffelmacher et al. Neutrophils play a critical role as one of the first lines of innate immune response to protect the host from exogenous pathogens and to repair the damaged tissue.

Due to their short lifespan, neutrophils are constantly produced in the bone marrow from hematopoietic progenitors in a process named granulopoiesis and released as mature neutrophils into the blood stream Soehnlein et al. Riffelmacher et al. The highest levels of autophagic flux were observed in the early stages of differentiation, as compared to reduced autophagic flux observed at the final maturation stage.

Targeted deletion of Atg7 in neutrophil progenitors resulted in an accumulation of immature neutrophils in the bone marrow.

More detailed metabolic analyses further revealed that neutrophil differentiation was accompanied by a shift towards mitochondrial respiration with the downregulation of glycolysis, which was blunted in autophagy-deficient neutrophils.

Adding exogenous FFAs to Atg7 -deficient neutrophil precursors restored their differentiation, suggesting that the oxidation of FFAs produced by autophagy provides the necessary ATP for neutrophil differentiation.

A different study demonstrated that autophagy is also required for neutrophil degranulation and NADPH-oxidase-mediated reactive oxygen species production Bhattacharya et al.

Myeloid-specific deletion of Atg7 reduced the inflammatory activity of neutrophils in vitro and in a murine model of experimental autoimmune encephalomyelitis. In particular, the relevance of neutrophil-secreted factors including neutrophil gelatinase-associated lipocalin, NGAL in the regulation of macrophage polarization during the post-ischemic myocardial healing phase was demonstrated in an experimental model of permanent infarction Horckmans et al.

A more recent study highlighted the relevance for endothelial autophagy in regulating neutrophil infiltration to sites of inflammation Reglero-Real et al.

Inflamed venular endothelial cells upregulated autophagy selectively at endothelial cell junctions, which was temporally aligned with the peak of neutrophil trafficking. Endothelial cell Atg5 deficiency resulted in excessive neutrophil transendothelial migration and uncontrolled leukocyte migration in murine inflammatory models, while pharmacological induction of autophagy suppressed neutrophil infiltration into tissues.

However, the precise mechanisms of neutrophil extravasation in the ischemic myocardium have not been studied as imaging of leukocyte trafficking in the beating heart remains very challenging Steffens et al. Hence, we may only speculate that the molecular mechanisms determining neutrophil diapedesis in remote areas of the infarcted heart or reperfused infarct zone are comparable to the processes studied in venules of other inflammatory sites.

In addition, there is evidence that autophagy is relevant for monocyte differentiation into macrophages Jacquel et al. While the healthy heart contains a heterogeneous population of tissue-resident macrophages with distinct origins and functions, the macrophage repertoire becomes even more diverse in response to cardiac injury, as blood-borne monocytes migrate into the myocardium and differentiate into macrophages to remove dying tissue, scavenge pathogens and promote healing Swirski and Nahrendorf, ; Zaman and Epelman, Hence, it can be speculated that monocyte autophagic flux is an important process in the immune response to cardiac injury.

However, an experimental study focusing on macrophage lysosomal function in post-myocardial infarction adverse remodeling found that ATG-dependent autophagy was dispensable, at least in this particular experimental model Javaheri et al.

Surprisingly, all these effects were independent of myeloid ATG5 expression. Consequently, pharmacological targeting of autophagy in this condition could represent a possible therapeutic strategy to limit MI-induced cardiac damage, adverse remodeling and heart failure.

Evidence on the contribution of selective autophagy in cardiac pathophysiology is accumulating and extends beyond mitophagy Kirkin et al. A crucial role in selectivity is exerted by adaptor proteins that bind specific cargoes and interact with conjugated LC3 via conserved LIR domains.

The proteins SQSTM1 also known as p62 and Neighbor of BRCA1 gene 1 NBR1 are among the best-characterized examples Gatica et al. Structurally, they own an LIR motif, homo- or hetero-oligomerization domains, and a C-terminal ubiquitin-binding UBA domain binding ubiquitinated cargos.

In clearing misfolded proteins, polyubiquitination of the cargo is essential so that they can bind to SQSTM1, be included in autophagosomes, and then be sent for lysosomal degradation Gatica et al.

The UBA domain interacts with ubiquitin chains attached to the cargo, while the LIR motif interacts with ATG8 family proteins e. SQSTM1 tends to cluster in p62 bodies when its levels are increased. In human cells, p62 bodies are often observed as discrete punctae.

NBR1 serves as a chain terminator of SQSTM1 filaments Jakobi et al. Since shorter SQSTM1 filaments form p62 bodies more easily, NBR1 plays a key role in promoting their assembly by regulating p62 filament length. Furthermore, NBR1 contains multiple domains involved in cargo recruitment, and the interaction between the SQSTM1 and NBR1 allows for more efficient cargo recognition.

Given the strong cooperative activities of NBR1 and SQSTM1, it is often difficult to distinguish the specific effects of each protein. A reduction in SQSTM1 levels, autophagosomes, and p62 bodies correlates with the progression of the autophagic flux toward its late steps. Their amount is inversely related to cellular autophagy levels, decreasing when lysosomal degradation has occurred successfully Katsuragi et al.

Notably, upregulation of SQSTM1 has been observed in most human failing hearts due to ischemic and non-ischemic heart disease Weekes et al. In the example, desmin-related cardiomyopathy is determined by the accumulation of desmin-misfolded aggregates and is characterized by higher expression of SQSTM1 at mRNA and protein levels and SQSTM1 silencing impaired autophagosomal formation, exacerbated cell injury, thus increasing cardiomyocyte death Zheng et al.

Aging is also characterized by the accumulation of protein aggregates. Analysis of human specimens from young 10 years old and aged individuals 65 years old revealed a significantly higher accumulation of SQSTM1 in aged hearts with a direct correlation with age Li C.

et al. Loss of SQSTM1 has been associated with accelerated aging, while overexpression of the SQSTM1 and NBR1 in Drosophila and C.

Elegans increases lifespan suggesting their possible protective role during aging Aparicio et al. In line with a protective role against proteotoxic stress, SQSTM1 is required to increase the autophagic flux in cardiomyocytes with dysfunctional proteasomal degradation of ubiquitinylated proteins, and its genetic deletion aggravated diastolic dysfunction upon pharmacological inhibition of the proteasome Pan et al.

On the other hand, in the context of ischemia-reperfusion, SQSTM1 forms a complex with the necrosome proteins RIP1 and RIP3, and its silencing in vivo protects the aged hearts from necrosis Li C. Similarly, SQSTM1 may scaffold other proteins involved in cell death mechanisms, such as caspases, and is instrumental to the homocysteine-induced apoptosis and autosis an autophagy-dependent cell death of cardiomyocytes Yin et al.

These findings further highlight the complexity of autophagy and its mechanisms in cardiac biology. In conclusion, selective autophagy uses SQSTM1 as critical receptors for cargo selection in cooperation with NBR1 and other adaptors.

Disruptions in their ability to deliver specific cargo for degradation may lead to disruption of cell signaling homeostasis with important implications for several cardiovascular diseases. Under normal conditions, the myocardium has low levels of basal autophagy.

Stress conditions can increase its levels to enhance cell survival, with constitutive autophagy maintaining normal cardiac structure and function, and upregulated autophagy occurring during cardiac disease Rothermel and Hill, ; Gustafsson and Gottlieb, However, in several cardiac diseases, autophagy can be downregulated or hyperactivated, therefore becoming detrimental.

Patients with congestive heart failure Takemura et al. In cardiac hypertrophy Hill and Olson, , autophagy plays a role in the progression of structural remodeling toward heart failure Hein et al.

In heart failure, increased autophagy can cause cardiac dysfunction, with autophagy-induced degeneration leading to cardiac cell death Akazawa et al.

Several treatments have been shown to regulate either inducing or inhibiting cardiac autophagy, including drugs for the treatment of cardiovascular diseases e. There are scant data on the downregulation of autophagy in anthracycline-induced cardiotoxicity Ma et al.

The activating or inhibiting effects of TKI on autophagy largely depend on the cell type Hirschbuhl et al. We were the first to demonstrate that ponatinib, the most cardiotoxic agent amongst all FDA-approved TKIs in the treatment of chronic myeloid leukemia, decreased autophagosome formation as well as LC3-II and p62 expression in cardiomyocytes, indicating a blockage of autophagic flux Madonna et al.

Eur Heart J Suppl abstract in Frontiers in CardioVascular Biomedicine Taken together, these data suggest that autophagy may represent a valuable target for limiting damage in non-ischemic cardiac diseases. However, despite the existence of an intimate connection between autophagy and the heart, only a few selective autophagy activator candidates have been recognized so far, depending on the context of cardiac homeostasis and disease.

Under stress conditions, such as starvation, autophagy is increased in the heart, and FYVE And Coiled-Coil Domain Autophagy Adaptor 1 FYCO1 has been linked to autophagy.

FYCO1 is highly expressed in the heart and its role has recently been investigated in vitro and in vivo under basal and stress conditions Kuhn et al. FYCO1 directly interacts with LC3, Rab7, and phosphatidylinositolphosphate PI3K , key players in autophagy Pankiv et al.

Although FYCO1 knockdown reduces autophagy in isolated rat cardiomyocytes, overexpression of FYCO1 leads to increased autophagic flux in vitro. Since overexpression of FYCO1 prevents cardiac dysfunction in response to biomechanical stress, enhancing autophagic flux by overexpressing FYCO1 could be a promising therapeutic strategy to treat or prevent heart failure Kuhn et al.

In adult mice, cardiac-specific deletion of Atg5 leads to contractile dysfunction, hypertrophy and cardiomyopathy, consistent with the notion that basal autophagy levels in cardiomyocytes are required for cellular proteostasis Nakai et al.

In vitro studies with cardiomyocytes harvested from Atg5 -deficient mice revealed that deficiency of the autophagic gene could cause the accumulation of unwanted proteins and contribute to myocardial disease Pattison and Robbins, In line with these findings, Lamp2 -deficient mice displayed increased autophagic vacuole accumulation and could not degrade proteins, thereby promoting cardiomyopathy Nishino et al.

In the failing heart, autophagy can cause myocardial cell damage via PARP1 Poly ADP ribose polymerase , which promotes autophagy in cardiomyocytes by modulating FoxO3a a member of the FoxO family of transcription factors Schiattarella and Hill, Changes in cardiac autophagy during sepsis have not been clearly defined.

BECLIN1, an early effector of autophagy in mammals, is ubiquitously expressed Liang et al. Autophagy changes in response to sepsis severity, and Beclin 1 plays a key role in the autophagic response of the septic heart in a mouse model of LPS-induced sepsis Sun et al.

Previous preclinical studies evaluating autophagy as a therapeutic approach for sepsis were mainly focused on the mTOR inhibitor rapamycin Hsieh et al. Because mTOR is involved in the regulation of a variety of pathways, rapamycin may cause unwanted toxicity.

In this regard, Sun et al. In Beclin 1 haploinsufficient mice, load-induced increases in autophagy activity were blunted, and pathological remodeling of the left ventricle was moderately diminished. In contrast, in mice engineered for forced overexpression of Beclin 1 in cardiomyocytes MHC-beclin-1 , pressure overload triggered an amplified autophagic response and pathological remodeling of the heart was more severe Sun et al.

In , researchers identified Rubicon as a protein that suppresses autophagy by interacting with the Beclin 1 complex Matsunaga et al. In mouse models, Rubicon deficiency enhances autophagic flux in the heart during LPS-induced sepsis, thereby maintaining cardiac stroke volume but without affecting myocardial inflammatory responses Zi et al.

Thus, targeting Rubicon may be a promising modality of autophagy modulation in various cardiac conditions. Kanamori et al. While cardiac autophagic activity is enhanced in type 1, it is suppressed in type 2 diabetes.

Lysosomes and autophagosomes accumulate within cardiomyocytes of type 1 diabetic mice, whereas abundant lipid droplets and immature autophagosomes were observed in the heart of type 2 diabetic mice Kanamori et al.

Here, resveratrol, an autophagy enhancer, mitigated diastolic dysfunction in the heart of type 2 diabetic mice, whereas it had opposite effects in the hearts of type 1 diabetic mice, suggesting that resveratrol may be a useful therapeutic target in diabetic cardiomyopathy, depending on the diabetic context Kanamori et al.

Similarly, resveratrol had beneficial effects in ischemic heart failure through autophagic activation Kanamori et al. Consistent with the notion that insulin exerts an inhibitory effect on autophagy, our research group recently demonstrated hyperactivation of autophagy associated with left ventricular dysfunction, remodeling, fibrosis, and myocyte apoptosis in a murine model of insulin-deficient diabetic cardiomyopathy Madonna et al.

Here, empagliflozin preserved cardiac dysfunction and remodeling at least in part, through the inhibition of autophagy. This process was mediated by inactivating the autophagy inducer GSK3β, which resulted in increased serum response factor SRF interaction with serum response element SRE and subsequent upregulation of cardiac actin expression.

Our results describe a novel paradigm in which empagliflozin inhibits the hyperactivation of autophagy through the GSK-3β signaling pathway in the context of diabetes. Taken together, these data provide evidence for the maladaptive role of autophagy in cardiovascular disease, suggesting that there is an optimal zone of cardiomyocyte autophagy that may be beneficial and that treatments resulting in levels of autophagy outside higher or lower this therapeutic window are likely to be deleterious Figure 2.

FIGURE 2. Functional relevance of autophagy for cardiac pathophysiology. Constitutive autophagy of cardiomyocytes under basal conditions is a homeostatic mechanism for normal cardiac structure and function.

Autophagic activity is reduced during aging or following exposure to stressors such as chemotherapy drugs owning cardiotoxicity. However, in hearts exposed to hemodynamic overload or ischemia-reperfusion injury, autophagic activity is upregulated at supraphysiological levels, suggesting a contribution to the maladaptive response of the heart that may lead to heart failure.

Compelling evidence revealed the crucial role of autophagy in preserving cardiac health by ruling cardiac homeostasis under baseline conditions and participating in the mechanisms of response to pathological injuries.

While enhancing autophagy activation has shown beneficial outcomes in aging and longevity in animal models, the identification of a proper therapeutic window in diseased conditions and well-tolerated pro-autophagic drugs is an active research area.

Meeting these medical needs will provide novel therapeutics and ultimately improve the outcome of patients with heart disease. DS and SS performed literature research on the general mechanisms of autophagy and involvement in aging and immune response to ischemia, SB and RM performed literature research on mechanisms of autophagy in non-ischemic diseases and therapeutic perspectives.

All the authors contributed in writing the manuscript and made clinical revisions. All authors contributed to the article and approved the submitted version.

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers.

Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher. The views and opinions expressed are those of the authors only and do not necessarily reflect those of the European Union or the European Commission.

Neither the European Union nor the European Commission can be held responsible for them. Abdellatif, M. Fine-tuning cardiac insulin-like growth factor 1 receptor signaling to promote health and longevity.

Circulation , — PubMed Abstract CrossRef Full Text Google Scholar. Akazawa, H. Diphtheria toxin-induced autophagic cardiomyocyte death plays a pathogenic role in mouse model of heart failure. Andres, A.

Mitophagy is required for acute cardioprotection by simvastatin. Redox Signal 21, — Aparicio, R. Cell Rep. Audesse, A. FOXO3 directly regulates an autophagy network to functionally regulate proteostasis in adult neural stem cells. PLoS Genet. Balgi, A. Screen for chemical modulators of autophagy reveals novel therapeutic inhibitors of mTORC1 signaling.

PLoS One 4, e Bhattacharya, A. Autophagy is required for neutrophil-mediated inflammation. Brunk, U. The mitochondrial-lysosomal axis theory of aging: Accumulation of damaged mitochondria as a result of imperfect autophagocytosis.

Dai, Y. Bioengineered 12, — Das, A. Rapamycin protects against myocardial ischemia-reperfusion injury through JAK2-STAT3 signaling pathway. Cell Cardiol.

Decuypere, J. Autophagy 7, — Deretic, V. Autophagy in inflammation, infection, and immunometabolism. Immunity 54, — Diao, J. ATG14 promotes membrane tethering and fusion of autophagosomes to endolysosomes. Nature , — Dikic, I.

Mechanism and medical implications of mammalian autophagy. Cell Biol. Dupont, N. Autophagy-based unconventional secretory pathway for extracellular delivery of IL-1β. EMBO J. Eisenberg, T. Cardioprotection and lifespan extension by the natural polyamine spermidine. Fernandez, A.

Disruption of the beclin 1-BCL2 autophagy regulatory complex promotes longevity in mice. Forte, M. Autophagy 19, — Frudd, K. Oxidation of Atg3 and Atg7 mediates inhibition of autophagy.

Ganley, I. FIP complex mediates mTOR signaling and is essential for autophagy. Gatica, D. Cargo recognition and degradation by selective autophagy. Geisler, S. Germic, N. Regulation of the innate immune system by autophagy: Monocytes, macrophages, dendritic cells and antigen presentation.

Cell Death Differ. Ghosh, R. Chaperone-mediated autophagy protects cardiomyocytes against hypoxic-cell death. Cell Physiol.

Gibbings, D. Selective autophagy degrades DICER and AGO2 and regulates miRNA activity. Gong, G. Parkin-mediated mitophagy directs perinatal cardiac metabolic maturation in mice. Science , aad Gorza, L. Guerra, F. Ranolazine for rhythm control in atrial fibrillation: A systematic review and meta-analysis.

Gustafsson, A. Autophagy in ischemic heart disease. Hahn, V. Myocardial gene expression signatures in human heart failure with preserved ejection fraction.

Hamasaki, M. Autophagosomes form at ER-mitochondria contact sites. Hariharan, N. Redox Signal 14, — Hayashi, S. The stent-eluting drugs sirolimus and paclitaxel suppress healing of the endothelium by induction of autophagy. Heckmann, B. LC3-associated phagocytosis at a glance.

Cell Sci. Hein, S. Progression from compensated hypertrophy to failure in the pressure-overloaded human heart: Structural deterioration and compensatory mechanisms.

Henderson, J. LpSpl with sphingosine-1 phosphate lyase activity reduces sphingolipid levels, reducing -autophagosome formation. Legionella pneumophila is a common pulmonary pathogen infecting human lung alveolar macrophages and causing pneumonia [ 59 ].

It evades the immune response by residing in a special vacuole formed from the endoplasmic reticulum membrane and by inhibiting ly-sosome fusion [ 60, 61 ]. Autophagy was shown to be critical for L. pneumophila elimination in an in vitro study showing that knockdown of Atg5 in mouse macrophages enhanced bacterial replication [ 62 ].

Furthermore, in vivo studies using the Atg9 mutant Dictyostelium discoideum showed a critical defect in the clearance of L. Legionella developed strategies to counter cellular autophagy elimination.

Another effector protein, LpSpl, acts as sphingosine-1 phosphate lyase, decreasing host cell sphingolipid levels to inhibit autophagosome formation Fig.

A common pulmonary virus pathogen, Influenza virus A, induces autophagy but blocks the auto-phagosome-lysosome fusion by the viral Matrix 2 M2 ion-channel protein [ 66 ]; thus, the virus adapts the multifunctional autophagosomes to reproduce the virus components and replicate Fig.

Consistent with the role of autophagy in host defense, recent studies have addressed the augmentation of autophagy as a method to enhance the clearance of pathogens including Pseudomonas aeruginosa [ 68 ] and Burkholderia cenocepacia [ 69 ].

Recent studies have found that autophagy is a negative regulator of inflammation in general, and of NLRP3 inflammasome in particular. The inflammasome is a multiprotein complex responsible for caspase-1 activation.

Activation of caspase-1 leads to the release of the active form of potent inflammatory cytokines, including IL-1β and IL, by proteolytic cleavage. Macrophages from -Atg16L1-deficient mice produced exaggerated quantities of IL-1β and IL in response to LPS [ 70 ].

Depletion of other autophagic proteins such as Atg7, LC3B, or Beclin 1, or treatment with autophagy inhibitors wortmannin or 3-methyladenine, enhanced the production of IL-1β and IL by macrophages [ 70, 71 ].

These studies indicated that autophagy deficiency is associated with increased inflammasome activity. Furthermore, autophagy deficiency in myeloid-derived cells was shown to cause spontaneous pulmonary inflammation in two independent studies [ 72, 73 ]. In both of these studies, mice lacking either Atg5 or Atg7 in myeloid cells spontaneously developed lung inflammation characterized by enhanced recruitment of inflammatory cells into the lung, increased levels of pro-inflammatory cytokines, submucosal thickening, goblet cell metaplasia, and increased collagen content [ 72, 73 ].

Following LPS challenge, these autophagy-deficient mice had higher levels of pro-inflammatory cytokines in serum and in bronchoalveolar lavage, severe pulmonary inflammation, as well as increased mortality compared to wild-type mice [ 72, 73 ].

In addition, mice lacking Atg5 or Atg7 in myeloid cells were more susceptible to bleomycin and silica challenge [ 74 ]. Spontaneous lung inflammation was also found in mice with Atg5 deletion in dendritic cells [ 75 ].

Knockout of other autophagy-related genes such as Atg14, Fip, or Epg5 in myeloid cells led to sterile lung inflammation, thus confirming the essential role of autophagy in lung homeostasis, which is not specific to a particular autophagy-related gene [ 76 ].

During active infection, autophagy also functions to prevent extensive inflammation [ 56, 76, 77 ]. In vivo studies of M. tuberculosis infection showed that, compared to wild-type, mice with myeloid cell-specific Atg5 knockout had a higher bacterial burden, severe necrotic lung lesions, elevated levels of IL and IL-1α, and higher mortality.

These studies suggest that in addition to suppressing M. tuberculosis growth, autophagy in myeloid-derived cells is responsible for controlling damaging inflammation [ 56, 77 ].

A recent study showed that the loss of Atg5 in polymorphonuclear cells causes excessive inflammation and predisposes to M. tuberculosis infection. This study suggested that the role of Atg5 in M.

tuberculosis inhibition could be at least partially independent of autophagy [ 78 ]. The innate immune system is an important component that acts as an initial barrier to protect against microbial pathogens or damaging agents. Cross-talk between autophagy and the innate immune system balances protection of the host against an exaggerated immune response, while enabling the neutralization of infectious and damaging threats.

This is crucial at sites such as the lung, skin, and colon, where the host is continuously exposed to potential hazardous elements, such as inhaled toxins, toxic food products, as well as chemicals and commensal and pathogenic bacteria.

The innate immune system is able to recognize and orchestrate a protective inflammatory response against harmful insults. Such responses should be tightly controlled to prevent exaggerated damage to the host.

The innate immune system relies on a group of pattern-recognition receptors that include toll-like receptors TLRs , nucleotide-binding oligomerization domain-like receptors NLRs , and absent in melanoma AIM2 -like receptors ALRs.

Both NLRs and ALRs can form a cytoplasmic multiprotein complex called the inflammasome upon sensing a wide variety of ligands. Inflammasome assembly involves the adapter protein apoptosis-associated speck-like protein containing a caspase-recruitment domain ASC which recruits caspase Activation of caspase-1 leads to the release of the active form of IL-1β and IL by proteolytic cleavage and can also lead to a form of cell death called pyroptosis [ 79 ].

The most widely studied inflammasome is NLRP3. Its activation depends on two steps. In the first step, pathogen-associated molecular patterns PAMPs and damage-associated molecular patterns DAMPs are recognized by TLRs to activate NF-κB signaling-dependent expression of the inflammasome components and pro-cytokines Fig.

In the second step, specific stimuli trigger inflammasome complex assembly and the inflammasome processes the pro-cytokine to generate mature cytokines by active caspases Fig. A recent study revealed that newly synthesized mitochondrial DNA may act as an NLRP3 ligand and directly associate with the NLRP3 inflammasome complex, thereby promoting its activity [ 80 ].

There have been several mechanisms proposed for how autophagy deficiency can lead to inflammasome activation. Autophagy has also been suggested to suppress inflammasome activation by directly digesting inflammasome components such as ubiquitinated ASC Fig.

Furthermore, autophagy was found to directly sequester pro-cytokines such as pro-IL-1β for digestion to reduce mature cytokine production Fig. Mice with Atg7 deficiency in myeloid cells developed spontaneous lung inflammation that was mostly mediated by IL Neutralization of IL, but not IL-1β or IL, attenuated lung inflammation in these mice.

In contrast, increased mortality in response to endotoxin was caused by increased IL-1β [ 62 ]. In addition to the effect of autophagy on inflammasome-associated cytokines, several studies have suggested an effect of autophagy or auto-phagic proteins on cytokines that are not associated with inflammasome activation.

In mice, Atg5-deficient macrophages produced more pro-inflammatory cytokine IL-1α in an inflammasome-independent way [ 77 ].

In Influenza A virus infection, excessive immune responses, including increased neutrophil and macrophage infiltration, contribute to lung injury and pathology more than the effects of viral replication [ 86 ].

Autophagy in host defense and inflammasome regulation. a PAMPs or DAMPs, recognized by pattern-recognition receptors, result in NF-κB activation. Active NK-κB promotes inflammasome components and cytokine expression.

b PAMPs or DAMPs cause mitochondrial damage and the release of mitochondrial ROS and DNA, triggering the assembling of NLRP3, ASC, and Pro-caspase into active inflammasome.

Caspase-1 is activated by autocleavage and then cleaves the pro-inflammatory cytokines IL-1β into active cytokines. Bacteria-containing phagosome membrane disruption leads to the release of PAMPs.

c Activated auto-phagosomes can engulf damaged mitochondria, NLRP3, ASC, and Pro-caspase, and target them to lysosomal degradation, reducing the production and secretion of active cytokines.

Autophagy is an important intracellular recycling system with diverse functions implicated in multiple cellular signaling pathways. Autophagy is regulated at the transcriptional, translational, and posttranslational levels. Phosphorylation and de-phosphorylation on some key proteins in the initiation complexes has been found to be a major mechanism of autophagy regulation [ 18 ].

Recent studies revealed that acetylation could modify autophagy proteins and influence the autophagy cascade [ 21 ]. Further elucidation of these regulatory mechanisms could provide potential therapeutic targets in diseases in which autophagy modulation is desired.

During host infection, autophagy eliminates pathogens by mediating pathogen autolysosomal killing and facilitating antimicrobial antigen presentation [ 5, 77, 87 ]. In addition to pathogen elimination, autophagy tames the host inflammatory response by negative regulation of inflammasome activity.

Multiple studies have shown that the induction of autophagy can have beneficial effects in combating infections, suggesting that promoting autophagy may be a beneficial strategy to control lung infection [ 43, 44 ].

However, some pathogens have evolved adaptive strategies to resist autophagy elimination, potentially limiting the impact of autophagy in immune defense. tuberculosis [ 57 ], RavZ and LpSpl from L. pneumophila [ 64, 65 ], and M2 ion-channel protein from Influenza A [ 67 ] Fig.

These virulence factors contribute to the drug resistance of those pathogens by enhancing pathogen survival in spite of host autophagy. Thus, developing drugs that inactivate pathogen virulence factors involved in autophagy avoidance may represent the next generation of -anti-microbial agents.

We thank Anindita Ravindran, Elmoataz Abdel Fattah, and Li-Yuan Yu-Lee for critical review of the manuscript. Sign In or Create an Account. Search Dropdown Menu.

header search search input Search input auto suggest. filter your search All Content All Journals Journal of Innate Immunity. Advanced Search. Skip Nav Destination Close navigation menu Article navigation. Volume 12, Issue 1. Autophagy in Regulatory and Signaling Pathways.

Cellular Regulation of Autophagy. Autophagy Regulation. Autophagy and the Elimination of Pulmonary Pathogens. Autophagy and Lung Inflammation. Autophagy Regulates Inflammasome Activity. Conclusion and Future Prospects. Statement of Ethics. Disclosure Statement.

Article Navigation. Review Articles April 24 Autophagy in Pulmonary Innate Immunity Subject Area: Further Areas , Immunology and Allergy , Pathology and Cell Biology. Lang Rao ; Lang Rao. Moreover, many studies have demonstrated that various pro-inflammatory cytokines participate in PD pathogenesis, such as TNF-α, IL-4, IL-6, IL, and IL-1β [ 70 ].

Several factors, such as a-synuclein, can serve as DAMPs to recognize PRRs, and leucine-rich repeat kinase 2 LRRK2 from the RIPK family and are involved in autophagy induction. Exogenous overexpression of a-synuclein leads to lysosomal damage and autophagy, which can be stimulated by tau protein related to Alzheimer's disease [ 71 ].

Exceptionally compelling is the framework comprising E3 ubiquitin ligase PRKN and PINK1 fit for driving mitophagy. However, in animal models, PINK1 and PRKN are not associated with PD.

Additionally, transformations might enhance α-synuclein levels and induce familial PD. Although autophagy is reversed by A53T overexpression, the overabundance of intracellular α-synuclein disables autophagy by restraining the small GTPase Rab-1A.

Finally, increased α-synuclein expression can induce protein accumulation and diminish autophagy, decreasing mitophagy and increasing neuronal apoptosis. Two classical AD hallmarks are the accumulation of p-tau protein and the deposition of Aβ plaques [ 73 ]. These perceptions proposed that imperfections in autophagic development might be an overall element of AD pathology.

For example, in AD, autophagy might be weakened by autophagosome corruption and autophagosome formation, albeit those effects might fluctuate according to genotype and diseased phase. Hereditary examinations have also demonstrated a few transformations that induce intriguing familial AD types, such as the mutation of amyloid antecedent protein APP , presenilin-1, and presenilin-1—1 PS 1 and 2 [ 75 , 76 ].

Also, autophagy might be downregulated during autophagosome development in AD patients. Compared to healthy individuals, AD patients show diminished Beclin-1 articulation, which might prompt disability in autophagic movement.

Beclin-1 heterozygous knockout in mice that express the AD-related human APP leads to APP and Aβ accumulation and displays more extreme neurodegeneration contrasted with Beclin-1 WT mice.

Additionally, pathogenic APP and tau are corrupted via autophagy. Consistently, 3-MA, a specific autophagy inhibitor, increments tau harmfulness, while rapamycin autophagy inducer diminishes tau effects in the cell [ 77 ]. Nonetheless, a previous review has shown that 3-MA or Beclin-1 knockdown could reduce Aβ disposition in neuroblastoma and glioma cells [ 78 ].

The discussion on the cytoprotective versus cytotoxic roles of autophagy in AD models might be clarified by assessing the autophagic motion and the level of lysosomal imperfection for each situation.

Further examinations are expected to expand the role of autophagy in AD. Autophagy was first identified as a key mechanism in cancer development. A cancer-suppressive role for autophagy corroborates this. Interestingly, this idea has been challenged by some that propose that autophagy can support oncogenesis because it can assist growth cell endurance [ 79 ].

A growth silencer engaged with the upstream restraint of mammalian target of rapamycin mTOR flagging PTEN, TSC1, and TSC2 turns autophagy on while mTOR activates oncogenes. For example, class I PI3K and Akt switch it off [ 80 ]. Besides, p53 and Death-associated protein kinase DAPK are associated with human malignant growth and control autophagy [ 81 ].

The cell oncogenes Bcl-2 and Bcl-XL are frequently upregulated in human diseases and repress autophagy by inhibiting Beclin Besides, p62 overexpression via autophagy-lysosome promotes carcinogenesis via NF-κB flagging liberation, activating nuclear factor E2-related factor Nrf-2 , inducing ROS production, and leading to DNA damage.

The contrast between these paradoxical features of autophagy makes the association with disease treatment more complex.

Notwithstanding, it has been recommended that in the initial phases of malignant growth, quality control via autophagy, especially over genome upkeep, suppresses carcinogenesis. Autophagy might organize the support or passage of cells into the G0 stage and subsequently forestall the unconstrained proliferation of tumor cells.

Conversely, autophagy might provide nutrition for cancer cells and assist their growth when suffering from metabolic pressure and oppose passing set off by chemotherapeutics [ 82 ].

Furthermore, autophagy promotes growth cell endurance in normal and cancer cells. Although autophagy can postpone apoptosis, cell passing ultimately restricts autophagy. Apoptosis ordinarily escapes growing cells, granting supported endurance, movement, and protection from treatment.

The delayed pressure endurance managed by imperfect apoptosis occurs in cancer cells by either increased anti-apoptotic genes Bcl-2 and Bcl-xL or deficiency in pro-apoptotic genes Bax and Bak [ 83 ]. The shortfall of cell passing is insufficient to support the pressure endurance of growing cells.

Thus, the pressure from glucose oxygen deprivation strongly enacts autophagy, upholding apoptotic cells' long-term endurance. Cancer cells evading apoptosis can also obtain nutrition via autophagy when they endure pressure for a long time and enter a torpid condition.

They can leave torpidity to continue cell multiplication when the pressure is released and typical development conditions are reestablished [ 84 ]. Hereditary or pharmacologic concealment of autophagy advances cell demise by putrefaction in vitro and in vivo, which suggests that growing and quiescent cells use autophagy to keep up with endurance in distressing conditions [ 85 ].

Autophagy limits these hypoxic districts, where it upholds growing cell endurance. Oxygen-detecting hypoxia-inducible factors activate autophagy alongside other metabolic factors and favor angiogenesis pathways unaffected by cell variation to metabolic pressure.

Autophagy induction in hypoxic areas might also hamper treatment due to proliferative cells that are resistant to treatment in these hypoxic areas. Hence, determining the cancer cell torpidity and recovery component and how to target this pathway to build novel anti-cancer strategies is essential.

Currently, lysosomotropism specialists e. On the other hand, autophagy can also effectively exhibit antitumor activity in some contexts, especially in focused growth cells or when blended with restorative mTOR hindrance. In this case, autophagy might improve endurance, conceivably subverting treatment.

Besides, various strategies using 3-MA, chloroquine, or hereditary manipulation of autophagy-related genes have shown that autophagy hindrance might sharpen growing cells to death, acting on assorted cytotoxic specialists [ 87 ].

Moreover, proteasome inhibitors can effectively trigger autophagy. Mechanistically, proteins can be degraded via two classical pathways: autophagy—lysosomal and ubiquitin—proteasome pathways. Inhibiting the ubiquitin—proteasome pathway activates the autophagy—lysosomal pathway.

For example, Bortezomib an FDA-approved proteasome inhibitor effectively enhances autophagy in colorectal cancer and myeloma cells [ 89 , 90 ]. Consistently, proteasome hindrance in prostate malignant growing cells by NPI can act through autophagy by an eIF2α-subordinate component that controls ATG function [ 91 ].

The concurrent inhibition of the two systems can result in a more effective strategy against cancer cells than the restraint of either pathway alone, which should be tested in the future. In summary, this review provided a profound understanding of the relationship between inflammation and autophagy in various human disorders.

Autophagy can assume fundamental roles in inflammatory diseases, infections, and carcinogenesis. A better comprehension of autophagy in different diseases has promising effects on developing improved treatments. Meanwhile, autophagy studies are still being conducted, although their relevance to digestion, stress reaction, and cell demise pathways is recognized.

Consequently, this cycle and their related reactions might provide data on how the host reacts to exogenous microorganisms and endogenous particles created under pressure conditions, yet these occasions can be re-molded by different stimuli and cell types. Altogether, understanding how autophagy is regulated and directed, and the particularity related to cell utilization, requires further examination.

It will be essential to characterize and portray sub-atomic and biochemical features associated with the intricate exchange among autophagy and different pathologies to advance novel approaches for patients with neurodegenerative diseases and infections.

The field of autophagy in immunity and inflammation-related diseases continues to evolve in both fundamental and translational fields.

In general, almost all human diseases possess an inflammatory component, which in turn provides a window of opportunity and a challenge to develop autophagy-based therapeutic strategies. Considering the irreplaceable role of autophagy in the removal of the primary toxic entity causing disease and subsequently reducing the susceptibility to pro-death insults, which implying autophagy is a promising target mechanism from a therapeutic perspective.

Finally, various pre-clinical and clinical studies are needed to investigate the function of autophagy in several diseases. Münz C. Enhancing immunity through autophagy. Annu Rev Immunol. Article CAS PubMed Google Scholar. Virgin HW, Levine B.

Autophagy genes in immunity. Nat Immunol. Article CAS PubMed PubMed Central Google Scholar. Autophagy and autophagy-related proteins in cancer. Mol Cancer. Xiang H, Zhang J, Lin C, Zhang L, Liu B, Ouyang L.

Targeting autophagy-related protein kinases for potential therapeutic purpose. Acta Pharm Sin B. Levine B, Kroemer G. Biological functions of autophagy genes: a disease perspective.

Cybulsky AV. Endoplasmic reticulum stress, the unfolded protein response and autophagy in kidney diseases. Nat Rev Nephrol. Filomeni G, De Zio D, Cecconi F. Oxidative stress and autophagy: the clash between damage and metabolic needs. Cell Death Differ.

Levy JMM, Towers CG, Thorburn A. Targeting autophagy in cancer. Nat Rev Cancer. Chen GY, Nuñez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol. Broz P, Dixit VM. Inflammasomes: mechanism of assembly, regulation and signalling.

Li W, He P, Huang Y, Li YF, Lu J, Li M, et al. Selective autophagy of intracellular organelles: recent research advances. Deretic V. Autophagy in inflammation, infection, and immunometabolism. Racanelli AC, Kikkers SA, Choi AMK, Cloonan SM. Autophagy and inflammation in chronic respiratory disease.

Article PubMed PubMed Central Google Scholar. Matsuzawa-Ishimoto Y, Hwang S, Cadwell K. Autophagy and Inflammation. Wen JH, Li DY, Liang S, Yang C, Tang JX, Liu HF. Macrophage autophagy in macrophage polarization, chronic inflammation and organ fibrosis.

Front Immunol. Liu T, Wang L, Liang P, Wang X, Liu Y, Cai J, et al. USP19 suppresses inflammation and promotes M2-like macrophage polarization by manipulating NLRP3 function via autophagy. Cell Mol Immunol.

Sanjurjo L, Aran G, Téllez É, Amézaga N, Armengol C, López D, et al. CD5L promotes M2 macrophage polarization through autophagy-mediated upregulation of ID3. Glick D, Barth S, Macleod KF. Autophagy: cellular and molecular mechanisms. J Pathol. Kerr JS, Adriaanse BA, Greig NH, Mattson MP, Cader MZ, Bohr VA, et al.

Trends Neurosci. Lou G, Palikaras K, Lautrup S, Scheibye-Knudsen M, Tavernarakis N, Fang EF. Mitophagy and neuroprotection. Trends Mol Med. Monkkonen T, Debnath J. Inflammatory signaling cascades and autophagy in cancer. Gonzalez CD, Resnik R, Vaccaro MI. Secretory autophagy and its relevance in metabolic and degenerative disease.

Front Endocrinol Lausanne. Article PubMed Google Scholar. Bustos SO, Leal Santos N, Chammas R, Andrade LNS. Secretory Autophagy Forges a Therapy Resistant Microenvironment in Melanoma. Cancers Basel. Kraya AA, Piao S, Xu X, Zhang G, Herlyn M, Gimotty P, et al. Identification of secreted proteins that reflect autophagy dynamics within tumor cells.

Autophagy: an emerging immunological paradigm. J Immunol. Atg7 deficiency intensifies inflammasome activation and pyroptosis in pseudomonas sepsis.

Li Q, Li L, Fei X, Zhang Y, Qi C, Hua S, et al. Inhibition of autophagy with 3-methyladenine is protective in a lethal model of murine endotoxemia and polymicrobial sepsis. Innate Immun. Castillo EF, Dekonenko A, Arko-Mensah J, Mandell MA, Dupont N, Jiang S, et al.

Autophagy protects against active tuberculosis by suppressing bacterial burden and inflammation. Proc Natl Acad Sci USA. Autophagy in the pathogenesis of disease. Arroyo DS, Gaviglio EA, Peralta Ramos JM, Bussi C, Rodriguez-Galan MC, Iribarren P.

Autophagy in inflammation, infection, neurodegeneration and cancer. Int Immunopharmacol. Xiao Y, Cai W. Autophagy and bacterial infection. Adv Exp Med Biol. Onorati AV, Dyczynski M, Ojha R, Amaravadi RK. Liu L, Feng D, Chen G, Chen M, Zheng Q, Song P, et al. Mitochondrial outer-membrane protein FUNDC1 mediates hypoxia-induced mitophagy in mammalian cells.

Nat Cell Biol. Li W, Li Y, Siraj S, Jin H, Fan Y, Yang X, et al. FUN14 domain-containing 1-mediated mitophagy suppresses hepatocarcinogenesis by inhibition of inflammasome activation in mice.

Chen D, Xie J, Fiskesund R, Dong W, Liang X, Lv J, et al. Chloroquine modulates antitumor immune response by resetting tumor-associated macrophages toward M1 phenotype.

Nat Commun. DeVorkin L, Pavey N, Carleton G, Comber A, Ho C, Lim J, et al. Cell Rep. Swadling L, Pallett LJ, Diniz MO, Baker JM, Amin OE, Stegmann KA, et al.

Poillet-Perez L, Sharp DW, Yang Y, Laddha SV, Ibrahim M, Bommareddy PK, et al. Autophagy promotes growth of tumors with high mutational burden by inhibiting a T cell immune response. Nat Cancer. Yamamoto K, Venida A, Yano J, Biancur DE, Kakiuchi M, Gupta S, et al.

Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Cunha LD, Yang M, Carter R, Guy C, Harris L, Crawford JC, et al.

LC3-associated phagocytosis in myeloid cells promotes tumor immune tolerance. Zitvogel L, Galluzzi L, Kepp O, Smyth MJ, Kroemer G. Type I interferons in anticancer immunity. Wang H, Hu S, Chen X, Shi H, Chen C, Sun L, et al.

cGAS is essential for the antitumor effect of immune checkpoint blockade. Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, et al.

Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J Clin Invest. Liang C, Feng P, Ku B, Dotan I, Canaani D, Oh BH, et al. Autophagic and tumour suppressor activity of a novel Beclin1-binding protein UVRAG.

Bravo-San Pedro JM, Kroemer G, Galluzzi L. Autophagy and mitophagy in cardiovascular disease. Circ Res. Klionsky DJ, Petroni G, Amaravadi RK, Baehrecke EH, Ballabio A, Boya P, et al.

Autophagy in major human diseases. Embo j. Ren J, Zhang Y. Targeting autophagy in aging and aging-related cardiovascular diseases. Trends Pharmacol Sci. Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, et al.

Autophagy Regulation of Innate Immunity Autophagy exerts its unique role to regulate immune homeostasis, which may alter cellular susceptibility to certain pathogens. Abstract Autophagy is a genetically well-controlled cellular process that is tightly controlled by a set of core genes, including the family of autophagy-related genes ATG. The contrast between these paradoxical features of autophagy makes the association with disease treatment more complex. The different signaling mechanisms are detailed below. Aging Cell 9, — HIF1alpha-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. Beyond these functions in lymphocyte survival and thymic negative selection, autophagy may exert other functions in lymphocyte differentiation, perhaps, in part, indirectly through effects on cytokine expression see the next section.
Autophagy and Innate Immunity

In addition, autophagy is involved in various aspects of immunity, including the clearance of pathogens, cytokines, and immune signals, suggesting that autophagy also plays important roles in innate [ 3, 4 ] and adaptive immunity [ 5 ].

The canonical autophagy pathway can be separated into three major steps: initiation, elongation, and maturation [ 6 ]. Each step is regulated by specific autophagy-related Atg proteins [ ]. In the initiation step, with the activation of the ULK1 kinase complex, a small membranous sac called a phagophore is formed.

A second kinase complex called the Vps34 complex is then recruited to the phagophore membrane [ 6 ]. Vps34 catalyzes the phosphorylation of cellular phosphatidylinositol to produce phosphatidylinositol 3-phosphate PI3P , which serves as a platform to recruit more Atg proteins to promote the elongation of autophagic membranes Fig.

In the elongation step, two ubiquitin-like conjugation systems, Atg12—Atg5-Atg16L and LC3—PE, are involved. In the first ubiquitin-like system, Atg12 acts as a ubiquitin-like protein and, with the help of Atg7 and Atg10, becomes conjugated to Atg5.

The resulting complex subsequently interacts with the coiled-coil protein Atg16L1 to form the Atg12—Atg5-Atg16L complex [ 10 ]. In the second ubiquitin-like system, pro-LC3 is cleaved by the cysteine protease Atg4, exposing a C-terminal glycine residue [ 11 ].

LC3 is then transiently linked to Atg7 and Atg3, and is eventually conjugated to phosphatidylethanolamine to form the mature form of lipid protein LC3II. The two systems work together to expand phagophore membrane formation that leads to mature autophagosomes Fig.

The mature autophagosomes then fuse with lysosomes to create autolysosomes and the acidic proteolytic environment of the autolysosome contributes to the degradation of the inner membrane as well as the luminal contents inside the autophagic vacuoles [ 6, 9 ]. Simplified autophagy signaling pathway and regulation.

a Nutrition deprivation inhibits mTOR, which in turn activates ULK1 complex. Energy deprivation is sensed by AMPK to suppress mTOR and activates ULK1 complex.

Activated ULK1 complex recruits VpsBeclin-1 complex to produce PI3P and initiates phagosome formation. b Two conjugation systems, Atg12—Atg5-Atg16L and LC3—PE, facilitate phagophore membrane elongation and autophagosome maturation. c NAD-dependent deacetylase Sirt1 catalyzes deacetylation of Vps34, Beclin1, Atg5, Atg7, and LC3 to promote autophagy.

d Transcription factors TFEB and FoxO translocate to the nuclear to induce autophagy-related gene expression. Their transcription activity can be inhibited by phosphorylation caused by mTOR or AKT, respectively.

Phosphorylated TFEB is digested by proteasome; ZKSCAN3 repress autophagy-related gene transcription. Autophagosomes can be processed in the absence of some key autophagy components, in a process described as noncanonical autophagy [ 12, 13 ].

Non-canonical autophagy can also be processed without the ubiquitin conjugation proteins Atg5 and Atg7 [ 14, 15 ], and this type of autophagy has been found to play an important role in mitochondrial digestion during erythroid maturation in vivo [ ].

LC3-associated phagocytosis LAP is a distinct form of noncanonical autophagy. It engages most of the canonical autophagy components, such as the Class III PI3K complex and ubiquitinylation-like protein-conjugation systems [ 17 ]. However, LAP proceeds without the ULK1 complex to form LC3-conjugated single-membraned phagosomes [ 17 ].

While the autophagy cascade could be affected in all three steps of the pathway, most regulation of autophagy occurs at the initiation step [ 9, 18 ]. mTOR acts as a cellular nutritional sensor, phosphorylating ULK1, thus inhibiting autophagy initiation in nutrient-sufficient conditions [ 9, 18 ].

Under starvation or treatment with mTOR inhibitors such as rapamycin, mTOR activity is inhibited and ULK1 is rapidly dephosphorylated, resulting in activation of the ULK1 kinase Fig. AMPK activates autophagy initiation through inhibiting mTORC1 and activating the ULK1 complex Fig.

Vps34 kinase activity depends on VpsBeclin1 interaction as the direct downstream target of ULK, and it is inhibited by kinase inhibitor 3-methyladenine 3-MA and wortmannin [ 18 ].

A recent study showed that the activity of Vps34 is also inhibited by acetyltransferase p [ 19 ]. Autophagy can be regulated at the transcriptional level. Forkhead transcription factors FoxOs upregulate autophagy by promoting the transcription of Atg genes, including Atg4, LC3B, and ULK1 [ 23, 24 ].

The activity of FoxOs depends on its nuclear localization. Transcription factor EB TFEB is another newly identified master transcription regulator of autophagy [ 25, 26 ]. Like FoxOs, the activation of TFEB is also dependent on nuclear translocation of the non-phosphorylated form of TFEB.

mTOR and MAPK are extracellular signaling kinases that catalyze TFEB phosphorylation and restrain its nuclear translocation, inhibiting its activity Fig. Our studies show that TFEB activity could also be regulated by proteasome-mediated degradation of phosphorylated TFEB [ 29, 30 ].

In contrast to FoxOs and TFEB, the transcriptional factor ZKSCAN3, which controls cell proliferation [ 31 ], has recently been identified to act as a repressor to a wide range of Atg genes. Knockdown of ZKSCAN3 has been shown to induce autophagy and lysosome biogenesis Fig. In addition to its role in maintaining cellular homeostasis, autophagy is actively engaged in cellular host defense by reducing the pathogen burden [ 33 ].

Mycobacterium tuberculosis bacteriumis a notorious pulmonary pathogen. tuberculosis primarily infects alveolar phagocytic cells, in which it resides and multiplies within the host-derived phagosomes [ 34 ].

tuberculosis developed strategies to escape phagocytotic clearance by preventing phagosome-lysosome fusion [ 35 ], disrupting vacuolar H-ATPase recruitment and phagosome acidification [ 36, 37 ], and inhibiting PI3P-dependent membrane trafficking [ 38, 39 ].

tuberculosis suppress the apoptosis of infected macrophages and trigger necrosis that results in the spreading of more bacteria to infect adjacent cells [ 40, 41 ].

Multiple studies have shown that autophagy plays an important role in innate defense against M. tuberculosis infection [ ]. Stimulation of autophagy by starvation, rapamycin, IFN-γ [ 4 ], ATP [ 46 ], or lipopolysaccharides LPS [ 44, 47 ] promoted the transfer of intracellular mycobacteria to lysosomes to be killed.

Furthermore, vitamin D, which was used to treat tuberculosis in the preantibiotic era, has been shown to exert anti- M. tuberculosis effects by stimulating autophagy through induction of the antibacterial peptide, cathelicidin [ ]. Antituberculosis drugs such as isoniazid and pyrazinamide have been shown to be partly dependent on autophagy activation, because in autophagy-defective cells antibiotic treatment was less effective against mycobacteria [ 51 ].

Additionally, autophagy deficiency also indirectly affects M. tuberculosis infection by enhancing macrophage uptake of mycobacteria through upregulation of scavenger receptor expression [ 52 ] and inhibiting antigen presentation [ 5, 47 ]. Some M. tuberculosis virulence factors facilitate intracellular bacterial survival by autophagy inhibition.

Recent evidence indicates that SapM blocks -autophagosome-lysosome fusing by binding with GAPase RAB7 [ 53 ]. ManLAM was also found to suppress auto-phagosome formation [ 54 ]. Another mycobacterial secreted protein is Eis, which is an N-acetyltransferase that enhances the survival of mycobacteria in human monocytic cells.

It suppresses autophagy by acetylating JNK-specific phosphatase MKP-7 to inhibit JNK-dependent autophagy efflux initiation [ 55 ]. The M. tuberculosis ESX-1 secretion system [ 56 ] was found to inhibit autophagic flux by blocking autophagosome-lysosome fusion in human dendritic cells [ 57 ].

A recent study of M. Strategies used by pulmonary pathogens to avoid host autophagy. tuberculosis has five identified anti-autophagy factors.

Eis is an N-acetyltransferase. It acetylates JNK-specific phosphatase MKP-7 to initiate the inhibition of JNK-dependent autophagy. Mannose-capped lipoarabinomannan ManLAM interferes with trafficking proteins in autophagy and also affects LC3 protein expression levels and inhibits accumulation of autophagic vacuoles.

b RavZ, the bacterial effector protein of L. pneumophila , inhibits autophagy by hydrolyzing the release of LC3II phosphatidylethanolamine at the carboxyl-terminal glycine residue. The hydrolyzed LC3 cannot be reconjugated by Atg7 and Atg3 to form mature LC3II for autophagosome localization.

LpSpl with sphingosine-1 phosphate lyase activity reduces sphingolipid levels, reducing -autophagosome formation. Legionella pneumophila is a common pulmonary pathogen infecting human lung alveolar macrophages and causing pneumonia [ 59 ]. It evades the immune response by residing in a special vacuole formed from the endoplasmic reticulum membrane and by inhibiting ly-sosome fusion [ 60, 61 ].

Autophagy was shown to be critical for L. pneumophila elimination in an in vitro study showing that knockdown of Atg5 in mouse macrophages enhanced bacterial replication [ 62 ].

Furthermore, in vivo studies using the Atg9 mutant Dictyostelium discoideum showed a critical defect in the clearance of L.

Legionella developed strategies to counter cellular autophagy elimination. Another effector protein, LpSpl, acts as sphingosine-1 phosphate lyase, decreasing host cell sphingolipid levels to inhibit autophagosome formation Fig.

A common pulmonary virus pathogen, Influenza virus A, induces autophagy but blocks the auto-phagosome-lysosome fusion by the viral Matrix 2 M2 ion-channel protein [ 66 ]; thus, the virus adapts the multifunctional autophagosomes to reproduce the virus components and replicate Fig.

Consistent with the role of autophagy in host defense, recent studies have addressed the augmentation of autophagy as a method to enhance the clearance of pathogens including Pseudomonas aeruginosa [ 68 ] and Burkholderia cenocepacia [ 69 ].

Recent studies have found that autophagy is a negative regulator of inflammation in general, and of NLRP3 inflammasome in particular. The inflammasome is a multiprotein complex responsible for caspase-1 activation. Activation of caspase-1 leads to the release of the active form of potent inflammatory cytokines, including IL-1β and IL, by proteolytic cleavage.

Macrophages from -Atg16L1-deficient mice produced exaggerated quantities of IL-1β and IL in response to LPS [ 70 ]. Depletion of other autophagic proteins such as Atg7, LC3B, or Beclin 1, or treatment with autophagy inhibitors wortmannin or 3-methyladenine, enhanced the production of IL-1β and IL by macrophages [ 70, 71 ].

These studies indicated that autophagy deficiency is associated with increased inflammasome activity. Furthermore, autophagy deficiency in myeloid-derived cells was shown to cause spontaneous pulmonary inflammation in two independent studies [ 72, 73 ].

In both of these studies, mice lacking either Atg5 or Atg7 in myeloid cells spontaneously developed lung inflammation characterized by enhanced recruitment of inflammatory cells into the lung, increased levels of pro-inflammatory cytokines, submucosal thickening, goblet cell metaplasia, and increased collagen content [ 72, 73 ].

Following LPS challenge, these autophagy-deficient mice had higher levels of pro-inflammatory cytokines in serum and in bronchoalveolar lavage, severe pulmonary inflammation, as well as increased mortality compared to wild-type mice [ 72, 73 ].

In addition, mice lacking Atg5 or Atg7 in myeloid cells were more susceptible to bleomycin and silica challenge [ 74 ]. Spontaneous lung inflammation was also found in mice with Atg5 deletion in dendritic cells [ 75 ].

Knockout of other autophagy-related genes such as Atg14, Fip, or Epg5 in myeloid cells led to sterile lung inflammation, thus confirming the essential role of autophagy in lung homeostasis, which is not specific to a particular autophagy-related gene [ 76 ].

During active infection, autophagy also functions to prevent extensive inflammation [ 56, 76, 77 ]. In vivo studies of M. tuberculosis infection showed that, compared to wild-type, mice with myeloid cell-specific Atg5 knockout had a higher bacterial burden, severe necrotic lung lesions, elevated levels of IL and IL-1α, and higher mortality.

These studies suggest that in addition to suppressing M. tuberculosis growth, autophagy in myeloid-derived cells is responsible for controlling damaging inflammation [ 56, 77 ]. A recent study showed that the loss of Atg5 in polymorphonuclear cells causes excessive inflammation and predisposes to M.

tuberculosis infection. This study suggested that the role of Atg5 in M. tuberculosis inhibition could be at least partially independent of autophagy [ 78 ]. The innate immune system is an important component that acts as an initial barrier to protect against microbial pathogens or damaging agents.

Cross-talk between autophagy and the innate immune system balances protection of the host against an exaggerated immune response, while enabling the neutralization of infectious and damaging threats. This is crucial at sites such as the lung, skin, and colon, where the host is continuously exposed to potential hazardous elements, such as inhaled toxins, toxic food products, as well as chemicals and commensal and pathogenic bacteria.

The innate immune system is able to recognize and orchestrate a protective inflammatory response against harmful insults. Such responses should be tightly controlled to prevent exaggerated damage to the host. The innate immune system relies on a group of pattern-recognition receptors that include toll-like receptors TLRs , nucleotide-binding oligomerization domain-like receptors NLRs , and absent in melanoma AIM2 -like receptors ALRs.

Both NLRs and ALRs can form a cytoplasmic multiprotein complex called the inflammasome upon sensing a wide variety of ligands. Inflammasome assembly involves the adapter protein apoptosis-associated speck-like protein containing a caspase-recruitment domain ASC which recruits caspase Activation of caspase-1 leads to the release of the active form of IL-1β and IL by proteolytic cleavage and can also lead to a form of cell death called pyroptosis [ 79 ].

The most widely studied inflammasome is NLRP3. Its activation depends on two steps. In the first step, pathogen-associated molecular patterns PAMPs and damage-associated molecular patterns DAMPs are recognized by TLRs to activate NF-κB signaling-dependent expression of the inflammasome components and pro-cytokines Fig.

In the second step, specific stimuli trigger inflammasome complex assembly and the inflammasome processes the pro-cytokine to generate mature cytokines by active caspases Fig.

A recent study revealed that newly synthesized mitochondrial DNA may act as an NLRP3 ligand and directly associate with the NLRP3 inflammasome complex, thereby promoting its activity [ 80 ]. There have been several mechanisms proposed for how autophagy deficiency can lead to inflammasome activation.

Autophagy has also been suggested to suppress inflammasome activation by directly digesting inflammasome components such as ubiquitinated ASC Fig.

Furthermore, autophagy was found to directly sequester pro-cytokines such as pro-IL-1β for digestion to reduce mature cytokine production Fig.

Mice with Atg7 deficiency in myeloid cells developed spontaneous lung inflammation that was mostly mediated by IL Neutralization of IL, but not IL-1β or IL, attenuated lung inflammation in these mice.

In contrast, increased mortality in response to endotoxin was caused by increased IL-1β [ 62 ]. In addition to the effect of autophagy on inflammasome-associated cytokines, several studies have suggested an effect of autophagy or auto-phagic proteins on cytokines that are not associated with inflammasome activation.

In mice, Atg5-deficient macrophages produced more pro-inflammatory cytokine IL-1α in an inflammasome-independent way [ 77 ]. In Influenza A virus infection, excessive immune responses, including increased neutrophil and macrophage infiltration, contribute to lung injury and pathology more than the effects of viral replication [ 86 ].

Autophagy in host defense and inflammasome regulation. a PAMPs or DAMPs, recognized by pattern-recognition receptors, result in NF-κB activation. Active NK-κB promotes inflammasome components and cytokine expression. b PAMPs or DAMPs cause mitochondrial damage and the release of mitochondrial ROS and DNA, triggering the assembling of NLRP3, ASC, and Pro-caspase into active inflammasome.

Caspase-1 is activated by autocleavage and then cleaves the pro-inflammatory cytokines IL-1β into active cytokines. Bacteria-containing phagosome membrane disruption leads to the release of PAMPs.

c Activated auto-phagosomes can engulf damaged mitochondria, NLRP3, ASC, and Pro-caspase, and target them to lysosomal degradation, reducing the production and secretion of active cytokines. Autophagy is an important intracellular recycling system with diverse functions implicated in multiple cellular signaling pathways.

Autophagy is regulated at the transcriptional, translational, and posttranslational levels. Phosphorylation and de-phosphorylation on some key proteins in the initiation complexes has been found to be a major mechanism of autophagy regulation [ 18 ].

Recent studies revealed that acetylation could modify autophagy proteins and influence the autophagy cascade [ 21 ]. Further elucidation of these regulatory mechanisms could provide potential therapeutic targets in diseases in which autophagy modulation is desired. During host infection, autophagy eliminates pathogens by mediating pathogen autolysosomal killing and facilitating antimicrobial antigen presentation [ 5, 77, 87 ].

In addition to pathogen elimination, autophagy tames the host inflammatory response by negative regulation of inflammasome activity. Multiple studies have shown that the induction of autophagy can have beneficial effects in combating infections, suggesting that promoting autophagy may be a beneficial strategy to control lung infection [ 43, 44 ].

However, some pathogens have evolved adaptive strategies to resist autophagy elimination, potentially limiting the impact of autophagy in immune defense. tuberculosis [ 57 ], RavZ and LpSpl from L. ATG7- and ATG3-deficient T cells display phenotypes similar to that of ATG5-deficient T lymphocytes, characterized by increased cell death and the defective homeostasis of organelles such as mitochondria and ER Jia and He, The deletion of gene ATG16L1 in mouse macrophages alters the production of IL-1β, which affects the derivation of naïve T cells into Th17 Kaser and Blumberg, Antigen presentation.

Antigenic peptides generated from autophagic degradation can be presented by MHC class II molecules. tuberculosis can escape the immune defense of host cells. In HIV-1 target cells, both canonical autophagy and noncanonical autophagy are involved in the presentation of virus-derived antigens.

HIV-1 specifically encodes Tat, Nef, and Vpu proteins, which perturb the autophagy mechanism and evade the antiviral responses Leymarie et al. The development of B cells.

The maintenance of the B-1a B cell number needs the adequate expression of ATG5 during efficient B cell development. While ATG5 is deleted in B lymphocytes, there is a dramatic decline in the number of B-1 B cells Miller et al. A knockout mouse with conditional deficiency of ATG5 in B cells exhibits a small number of long-lived plasma cells and defective antibody response Pengo et al.

In addition, the survival of memory B cells and their differentiation into plasma cells are affected by autophagic defect in the autophagosome—lysosome fusion as evidenced in Vici syndrome Piano Mortari et al.

The efficiency of vaccines. Some conjugate vaccines have a high antigen-presenting ability to T cells when processed by autophagy. The enhancement of autophagy has been used as an optimized strategy for the development of new vaccines against Japanese encephalitis virus Zhao et al.

When the mycobacterial antigen Ag85B was overexpressed in bacillus Calmette—Guérin vaccine, the vaccine efficacy was improved by augmenting autophagy-mediated antigen presentation Jagannath et al.

Autophagy regulates antimicrobial immunity through multiple pathways and involves different mechanisms such as genetic traits, inflammation, oxidation, apoptosis, and energy metabolism. For example, microbial invasion can cause the release of inflammatory cytokines and ROS generation Tse et al.

Programmed cell death or apoptosis eliminates intracellular pathogens, which is regulated by IAPs, Bcl-2, caspases, nuclear factors, and so forth Liang et al.

There is a complicated network to regulate autophagy-mediated antimicrobial activity, which is validated by representative pathways Figure 3. Signaling pathways mediate the interaction between autophagy and antimicrobial immunity. Microbial invasion triggers the activation of autophagy via different signaling pathways by which multiple cargo receptors or sensors are involved through direct or indirect ways.

Cytosolic constituents are delivered onto PRRs via topological inversion, acting as the antimicrobial effectors of TLRs. For example, viral nucleic acids can be recognized by intracellular sensors to play a crucial role in the initiation of antigen-specific adaptive immunity. Herein, initiation factors and cargo receptors or sensors are integrated into different signaling pathways to modify antimicrobial immunity.

EIF4EBP1, eukaryotic translation initiation factor 4E binding protein 1; GRP78, glucose-regulated protein 78; GRP94, glucose-regulated protein 94; ORP, oxygen-regulated protein; TSC2, tuberous sclerosis complex 2.

PI3K phosphorylates downstream signal transducers to mediate cell proliferation, differentiation, and intracellular trafficking. The PI3K pathway is activated by innate type I IFN to promote autophagy, which can convert LC3-I to LC3-II for the formation of autophagosomes Schmeisser et al.

AKT is a downstream substrate of PI3K. PI3K is also activated through p-IRS1 and p-IRS2, which results in the subsequent induction of mTORC1 Zoncu et al. The inhibition of mTOR by rapamycin induces autophagy and incurs immunosuppression Fabri et al.

mTOR can repress autophagy by phosphorylating ATG proteins. AMPK is a key energy sensor to integrate intracellular energy metabolism.

AMPK signaling is activated by ATP depletion or glucose starvation, which can induce autophagy through the inhibition of mTORC1 and direct phosphorylation of the protein kinase Unc ULK1 Kim et al.

Both the phosphorylation of AMPK at Thr and the AMPK-dependent phosphorylation of ULK1 at Ser can mediate CDstimulated autophagy and intracellular killing of T. gondii in the infected cells Liu et al.

AMPK-activated autophagy contributes to antibacterial defense against M. tuberculosis by inhibiting the phosphorylation of mTOR in macrophages. AMPK-mediated antimicrobial activity requires the participation of peroxisome proliferator-activated receptor-γ, coactivator 1α PPARGC1A that involves the fusion of phagosomes with LC3B autophagosomes Yang et al.

In macrophages infected with M. tuberculosis , AMPK and PtdIns3K pathways take part in the activation of autophagy induced by antimicrobial LL peptide Rekha et al. During hepatitis C virus HCV infection, the net strength of autophagy depends on the inhibitive effect of AKT on AMPKα. HCV-induced ER stress impedes the AKT—TSC—mTORC1 pathway, contributing to autophagy enhancement Huang et al.

Mechanistic investigation has demonstrated that the activation of AMPK as a new mechanism is influenced by TDRD7 Subramanian et al. A signaling cascade is initiated after external cytokines such as IFNs and ILs as ligands bind to membranous receptors.

Receptor-associated JAKs phosphorylate tyrosine residues and then activate STATs to induce the transcription of target genes Jatiani et al.

There is a cross-regulation between autophagy and type I IFN signaling during host defense. HCV infection can disturb IFN-α signaling and facilitates the escape of HCV from the IFN system, resulting in the persistence of virus infection Luquin et al.

A similar phenomenon is also found in West Nile virus infection. Potential mechanisms involve the inhibition of ISGs e. GBP1 and MxA and the dephosphorylation of STAT1 Harvey et al.

The vita-PAMP of Gram-positive bacteria mediates ER stress via the innate sensor STING, which triggers autophagy activation to engage immune defense and the homeostatic mechanism Figure 1C ; Moretti et al.

If sustained, the UPR can exacerbate inflammation and it has been implicated in pathologies such as obesity, type 1 and type 2 diabetes, cancer, as well as neurodegenerative and autoimmune diseases. In some cases, ER stress is relieved by microbial infection as revealed in the HCV-infected liver Dash et al.

HCV infection stimulates autophagic activity and improves the survival of host cells by inhibiting apoptosis. ER stress-induced cellular apoptosis is also ameliorated by activating autophagy in the macrophages infected by M. tuberculosis Liang et al. ER stress-associated UPR signaling can be elicited by different stimuli such as hypoxia, ischemia, low calcium, and glucose depletion Malhi and Kaufman, ; Wang and Kaufman, ; Dash et al.

The extent and type of UPR signaling usually depend on pathogen species during the infection He, Acute UPR signaling is characterized by returning to baseline level within a short period.

Chronic UPR signaling can maintain a persistent state by regulating the expression levels of some genes. Meanwhile, selective autophagy mediated by PRRs plays a crucial role Fan et al.

However, the extracellular part of an MHC class II molecule i. PRR-mediated degradation improves the efficiency of autophagy, which is the most economical way of the development of adaptive immunity Lin et al. If compensation fails, ER stress-induced apoptosis may be instigated by activating caspase Szegezdi et al.

Microbial pathogens induce ER stress and UPR signaling, which may interact with innate sensors to activate selective autophagy and further regulate the immune defense.

Therefore, the intracellular fate of invading pathogens is determined by the autophagy-mediated antimicrobial immunity Ponpuak and Deretic, ; Deretic, The autophagy-dependent eradication of intracellular T. Microbial invasion activates autophagy to sequestrate adherent-invasive E.

coli via the interaction of ATG16L1 with the complement component C3, resulting in the degradation of pathogens Viret et al. In response to microbial invasion, vital autophagy can shape the host's immunity to mediate the killing of intracellular pathogens. Instead, certain microbial pathogens may adapt autophagy machinery to favor their survival and growth.

At this moment, autophagy becomes a protective mechanism for invading microbes. In host cells infected with M. tuberculosis , autophagic adapter protein p62 delivers cytosolic components into autophagosomes Ponpuak et al.

tuberculosis can block the fusion of phagosome with lysosomes to keep pathogen survival within conventional phagolysosomes Vergne et al. Conversely, the autophagy can be stimulated by reducing cytokines IL-4 and IL in response to M. tuberculosis antigens Ghadimi et al. Also, IFN-β-associated immunoevasion is adopted by M.

tuberculosis , which protects intracellular bacilli. Evidently, autophagy activity regulated by different cytokines affects the survival of pathogens within host cells Sabir et al. Certain bacteria such as S. flexneri and Salmonella typhimurium can escape antibacterial autophagy and immunity by decreasing the level of cytoplasmic C3-ATG16L1 Sorbara et al.

Invasive Shigella secretes IcsB that competitively inhibits the binding of VirG to ATG5, and thus can avoid autophagic degradation Ogawa et al. Intracellular Listeria monocytogenes produces pore-forming toxins to impede the maturation of autophagic vacuoles, through which the bacterial pathogen can replicate in vacuoles and establish persistent infection within host macrophages Birmingham et al.

Autophagy activation may favor the replication of certain viruses. In HCV-infected hepatocytes, autophagy activity is linked to virus replication as reflected by the expression of related proteins such as Beclin-1, ATG4B, ATG5, ATG7, and ATG12 Ke and Chen, Autophagy activation alleviates ER stress and regulates the assembly of infectious virions in host cells Ke and Chen, Autophagy is a vital mechanism responsible for immunomodulation and immunoevasion Dash et al.

Moreover, autophagy machinery promotes hepatocyte growth by enhancing phospho-mTOR and 4EBP1 expression. There is a fine balance between HCV invasion and host cell survival, in which autophagy facilitates HCV replication in hepatocytes and develops infectious persistence and pathogenicity Aizawa et al.

Additionally, HSV-1 protein ICP Autophagy inhibition enables viruses to evade innate immunity and causes lethal encephalitis Orvedahl et al. Dengue virus can usurp autophagy machinery to acquire free fatty acids from lipid droplets, by which a high level of adenosine triphosphate is provided for virus replication Heaton and Randall, Autophagy exerts its unique role to regulate immune homeostasis, which may alter cellular susceptibility to certain pathogens.

Intracellular pathogens can exploit host cells and escape antimicrobial degradation, leading to persistent infection and cell death El-Awady et al. Obviously, the activation of autophagy acts as double-edged sword, contributing to not only the elimination of pathogens but also immunoevasion and pathogen survival.

The perplexing interaction between microbial invasion and autophagic adaptation governs the immunoregulation and controls the final outcome of the host—microbe encounter Deretic and Levine, During microbial infection, the host cell adjusts its biomass and function by regulating the autophagy machinery, which involves energy supplementation, modification of innate and adaptive immunity, proliferation, apoptosis, etc.

Intracellular pathogens may be eradicated via the autophagic defense mechanism. Alternatively, autophagy machinery can be adapted or subverted to cause persistent infection. By altering or evading autophagy, intracellular invaders can cause host cell death.

The pathophysiological manifestations depend on the comprehensive regulation between autophagy and antimicrobial immunity Yordy et al.

The duality of autophagy means that it may have beneficial and harmful effects on the process of antimicrobial immunity. The dual effect of autophagy depends on the trait or nature of microbial pathogens, which has been characterized in the pathogenesis of certain chronic infections such as hepatitis, AIDS, and tuberculosis.

When those diseases are treated using autophagy-related medicines, the option of treatment protocols should consider different stages or conditions.

For instance, HBV or HBV X protein can activate autophagy in the initial stage, but inhibits subsequent degradation by impairing lysosomal acidification Khabir et al. When the autophagy strategy is utilized to treat HBV infection, the pros and cons of autophagy must be weighed.

HDV can alter the autophagic process in the elongation stage to promote genome replication. Also, the pathophysiological characteristics of clinical disease should be considered. For example, the stable knockdown of several autophagy factors can achieve synergistic inhibition on HIV-1 replication Eekels et al.

Autophagy thus is a therapeutic target against HIV-1 infection. tuberculosis or opportunistic pathogens. It is necessary to explore novel strategies under the coinfection of HIV-1 and M.

A few drugs such as vitamin D3, trehalose, and phenylbutyrate may be helpful Rekha et al. Essential autophagy modulates both cell survival and death as evidenced through mitochondrial mediation. Mitophagy is a selective degradation of old or damaged mitochondria to maintain survival state.

The mitochondrion-dependent pathway is also a well-known mechanism for apoptotic cell death Wang, There is a negative feedback loop including FADD and caspase-8, which modulates autophagic response Walsh and Bell, Anti-apoptotic Bcl-2 negatively regulates Beclinmediated autophagy and modifies antimicrobial immunity Casalino-Matsuda et al.

Autophagy takes part in the degradation of pathogens and subsequent antigen presentation. Apoptosis signaling can provide feedback regulation for autophagic response. The interaction between apoptosis and autophagy is a critical modulator for the functional state of host cells. Excessive apoptosis caused by microbial infection can impair organ function.

Therefore, autophagy affects functional maintenance of the infected organ. Autophagic response can activate or antagonize apoptosis, obviously, playing a dual role Wang, Necroptosis and pyroptosis are implicated in cell death as well. The immunogenicity of necroptotic death is a novel strategy for developing cancer vaccines Lin et al.

The autophagy machinery participates in cytokine storm that destroys a lot of cells in a short time during chimeric antigen receptor T-cell therapy or COVID infection DeFrancesco, ; Meftahi et al.

The autophagy pathway modulated by diverse factors such as IFNs, NLRs, downstream effectors, and substrates may skew the immune response to induce immunoevasion and immunosuppression. There is a complex network to regulate autophagy, which impacts on the performance of antimicrobial immunity.

If we consider microbial invasion as the initial input and antimicrobial immunity as the integrative output, the regulation of autophagy is pivotal to controlling the host—pathogen balance. Up to now, the regulation of the autophagic process is not fully understood.

Moreover, autophagy is related to distinct mechanisms such as oxidative stress, energy metabolism, protein synthesis, and apoptosis.

All those mechanisms should be comprehensively considered since they are not in vectorial or parallel ways. A long-term effort is needed to clarify the details of autophagy regulation.

The invasion of microbial pathogens activates autophagy in host cells. Autophagy activation modifies downstream immune responses. Due to the duality of autophagy, antimicrobial immunity may be enhanced or weakened.

The outcomes of autophagy-mediated immune response are varied, depending on the categories of microbial pathogens and the types of infected cells. Therefore, the antimicrobial application of the autophagy mechanism must consider pathogen characteristics as well as cell types. The administration of autophagy modulators e.

chloroquine and hydroxychloroquine has shown therapeutic benefits in certain infectious diseases e. malaria, amebiasis, and Q fever. Furthermore, the mTOR pathway is the potential target against viral pathogens.

Yet, mTOR also participates in the replication and release of virions Maiese, Autophagy modulators can be combined with immunomodulators based on the mechanistic autophagy—immunity axis Wang et al.

Hopefully, the best therapeutic effect may be achieved through drug combination. Autophagy is the core mechanism of regulating antimicrobial immunity. Functional analysis shows that autophagy can control pathophysiological manifestations of microbial infection and determine the choice of clinical treatment protocols, especially in the case of coinfection.

Different infections or different stages of the same infection require respective protocols, considering the dual nature of autophagy. A few therapeutic agents, based on the autophagy mechanism, have been used in clinical practice.

The accumulated evidence confirms their positive effects. There is a bright future for the development of autophagy-related drugs against microbial infections. This work was supported by grants from Beijing Natural Science Foundation , National Key Research and Development Project YFA , and Chinese Academy of Medical Sciences CAMS Innovation Fund for Medical Sciences CIFMS, I2M Author contributions: C.

and K. conceived and designed the manuscript. were responsible for data collection and analysis. Additional data were provided by Y. and L. wrote the first draft, which was revised by C.

All authors approved the manuscript. Afsal K. Effect of 1,dihydroxyvitamin D3 on the expression of mannose receptor, DC-SIGN and autophagy genes in pulmonary tuberculosis.

Tuberculosis 99 , 1 — Google Scholar. Aizawa Y. et al. Chronic hepatitis C virus infection and lipoprotein metabolism.

World J. Ambrose R. West Nile virus differentially modulates the unfolded protein response to facilitate replication and immune evasion. Bell S. Galectin-8 senses phagosomal damage and recruits selective autophagy adapter TAX1BP1 to control Mycobacterium tuberculosis infection in macrophages.

MBio 12 , e Betin V. Caspase cleavage of Atg4D stimulates GABARAP-L1 processing and triggers mitochondrial targeting and apoptosis. Cell Sci. Birgisdottir A. The LIR motif—crucial for selective autophagy. Birmingham C. Listeriolysin O allows Listeria monocytogenes replication in macrophage vacuoles.

Nature , — The ubiquitin E3 ligase parkin inhibits innate antiviral immunity through Klinked polyubiquitination of RIG-I and MDA5.

Casalino-Matsuda S. Hypercapnia inhibits autophagy and bacterial killing in human macrophages by increasing expression of Bcl-2 and Bcl-xL.

Chai Q. A Mycobacterium tuberculosis surface protein recruits ubiquitin to trigger host xenophagy. Chang Y. Autophagy facilitates IFN-γ-induced Jak2—STAT1 activation and cellular inflammation. Choi J. The parasitophorous vacuole membrane of Toxoplasma gondii is targeted for disruption by ubiquitin-like conjugation systems of autophagy.

Immunity 40 , — Chunfa L. The central role of IFI in IFN-β release and autophagy activation during Mycobacterium bovis infection. Das M. Intravenous immunoglobulin mediates anti-inflammatory effects in peripheral blood mononuclear cells by inducing autophagy.

Cell Death. Dash S. Hepatitis C virus infection induces autophagy as a prosurvival mechanism to alleviate hepatic ER-stress response. Viruses 8 , DeFrancesco L. CAR-T cell therapy seeks strategies to harness cytokine storm.

Delgado M. Autophagy and pattern recognition receptors in innate immunity. Deretic V. Autophagy as an innate immunity paradigm: expanding the scope and repertoire of pattern recognition receptors. Autophagy, immunity, and microbial adaptations.

Cell Host Microbe 5 , — Eekels J. Inhibition of HIV-1 replication with stable RNAi-mediated knockdown of autophagy factors. El-Awady A. Dendritic cells: microbial clearance via autophagy and potential immunobiological consequences for periodontal disease.

Elinav E. Regulation of the antimicrobial response by NLR proteins. Immunity 34 , — Everts B. TLR-driven early glycolytic reprogramming via the kinases TBK1—IKKε supports the anabolic demands of dendritic cell activation.

Fabri M. Role of autophagy in the host response to microbial infection and potential for therapy. Fan S. The role of autophagy and autophagy receptor NDP52 in microbial infections.

Fertan E. Cognitive decline, cerebral—spleen tryptophan metabolism, oxidative stress, cytokine production, and regulation of the Txnip gene in a triple transgenic mouse model of Alzheimer disease. Fleming S. Vaccines 4 , Gao L. ABIN1 protein cooperates with TAX1BP1 and A20 proteins to inhibit antiviral signaling.

Ghadimi D. Lactic acid bacteria enhance autophagic ability of mononuclear phagocytes by increasing Th1 autophagy-promoting cytokine IFN-γ and nitric oxide NO levels and reducing Th2 autophagy-restraining cytokines IL-4 and IL in response to Mycobacterium tuberculosis antigen. Gui X. Autophagy induction via STING trafficking is a primordial function of the cGAS pathway.

Harris J. Th1—Th2 polarisation and autophagy in the control of intracellular mycobacteria by macrophages. Harvey R. Virus Res. Viruses, endoplasmic reticulum stress, and interferon responses. Cell Death Differ. Heaton N. Dengue virus-induced autophagy regulates lipid metabolism.

Cell Host Microbe 8 , — Herhaus L. Expanding the ubiquitin code through post-translational modification. EMBO Rep. Holla S. Selective inhibition of IFNG-induced autophagy by Mir and Mirresponsive WNT5A and SHH signaling.

Autophagy 10 , — Huang H. Hepatitis C virus inhibits AKT—tuberous sclerosis complex TSC , the mechanistic target of rapamycin MTOR pathway, through endoplasmic reticulum stress to induce autophagy. Autophagy 9 , — Huang X.

XIAP inhibits autophagy via XIAP—Mdm2—p53 signalling. EMBO J. Inaba K. The formation of immunogenic major histocompatibility complex class II—peptide ligands in lysosomal compartments of dendritic cells is regulated by inflammatory stimuli.

Jagannath C. Autophagy enhances the efficacy of BCG vaccine by increasing peptide presentation in mouse dendritic cells. Jatiani S. Genes Cancer 1 , — Jia W.

Temporal regulation of intracellular organelle homeostasis in T lymphocytes by autophagy. Jin Y. PLoS One 9 , e Johansen T. Selective autophagy: ATG8 family proteins, LIR motifs and cargo receptors. Kaser A. Autophagy, microbial sensing, endoplasmic reticulum stress, and epithelial function in inflammatory bowel disease.

Gastroenterology , — Autophagy in hepatitis C virus—host interactions: potential roles and therapeutic targets for liver-associated diseases. Khabir M. Hepatitis delta virus alters the autophagy process to promote its genome replication. Kim J. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1.

Cell Biol. Kimura T. TRIM-mediated precision autophagy targets cytoplasmic regulators of innate immunity. Kirkin V. NBR1 cooperates with p62 in selective autophagy of ubiquitinated targets. Autophagy , 5 , — A role for NBR1 in autophagosomal degradation of ubiquitinated substrates.

Cell , 33 , — Korol C. Tissue specific diversification, virulence and immune response to Mycobacterium bovis BCG in a patient with an IFN-γR1 deficiency. Virulence 11 , — Lee P.

Wiskott—Aldrich syndrome protein regulates autophagy and inflammasome activity in innate immune cells. Leib D. Interaction of ICP Levonen A. Redox regulation of antioxidants, autophagy, and the response to stress: implications for electrophile therapeutics. Free Radical Biol.

Leymarie O. Canonical and non-canonical autophagy in HIV-1 replication cycle. Viruses 9 , FASEB J. Regulation of ATG and autophagy initiation. Cytoplasmic cargo receptor p62 inhibits Avibirnavirus replication by mediating autophagic degradation of viral protein VP2.

Liang Q. Crosstalk between the cGAS DNA sensor and Beclin-1 autophagy protein shapes innate antimicrobial immune responses. Cell Host Microbe 15 , — Liang S. BAG2 ameliorates endoplasmic reticulum stress-induced cell apoptosis in Mycobacterium tuberculosis-infected macrophages through selective autophagy.

Autophagy 16 , — Murine macrophage autophagy protects against alcohol-induced liver injury by degrading interferon regulatory factor 1 IRF1 and removing damaged mitochondria. Liang X. Protection against fatal Sindbis virus encephalitis by Beclin, a novel Bclinteracting protein.

Lin F. XIAP and cIAP1 amplifications induce Beclin 1-dependent autophagy through NFκB activation. Lin S. Necroptosis promotes autophagy-dependent upregulation of DAMP and results in immunosurveillance.

Autophagy 14 , — Liu E. Identification of signaling pathways by which CD40 stimulates autophagy and antimicrobial activity against Toxoplasma gondii in macrophages.

Liu G. Self-eating and self-defense: autophagy controls innate immunity and adaptive immunity. Leukocyte Biol. Liu J.

Shear stress regulates endothelial cell autophagy via redox regulation and Sirt1 expression. Cell Death Dis.

Luquin E. Antiviral Res. Immunobiology , — Magraoui F. Autophagy-related deubiquitinating enzymes involved in health and disease.

Cells 4 , — Maiese K. The mechanistic target of rapamycin mTOR : novel considerations as an antiviral treatment. Maiuri M. Crosstalk between apoptosis and autophagy within the Beclin 1 interactome. Malhi H. Endoplasmic reticulum stress in liver disease.

Manzanillo P. Mycobacterium tuberculosis activates the DNA-dependent cytosolic surveillance pathway within macrophages. Cell Host Microbe 11 , — Maronek M.

Phages and their role in gastrointestinal disease: focus on inflammatory bowel disease. Cells 9 , Matsumoto G. Cell 44 , — Matsuo H. Intravenous immunoglobulin enhances the killing activity and autophagy of neutrophils isolated from immunocompromised patients against multidrug-resistant bacteria.

Meftahi G. Miller B. The autophagy gene ATG5 plays an essential role in B lymphocyte development. Autophagy 4 , — Mizushima N. The ATG conjugation systems in autophagy. Moretti J.

STING senses microbial viability to orchestrate stress-mediated autophagy of the endoplasmic reticulum. Cell , — Munz C. Autophagy beyond intracellular MHC class II antigen presentation. Trends Immunol.

Nakano S. Commensal microbiota contributes to chronic endocarditis in TAX1BP1 deficient mice. PLoS One 8 , e Nedjic J. Autophagy in thymic epithelium shapes the T-cell repertoire and is essential for tolerance. Newman A. PLoS One 7 , e Ogawa M.

Escape of intracellular Shigella from autophagy. Science , — Ohshima J. Role of mouse and human autophagy proteins in IFN-γ-induced cell-autonomous responses against Toxoplasma gondii. Oikonomou V. Noncanonical fungal autophagy inhibits inflammation in response to IFN-γ via DAPK1.

Cell Host Microbe 20 , — O'Loughlin T. OPTN recruitment to a Golgi-proximal compartment regulates immune signalling and cytokine secretion.

Orvedahl A. HSV-1 ICP Cell Host Microbe 1 , 23 — Padman B. Pattingre S. Bcl-2 inhibition of autophagy: a new route to cancer? Cancer Res. Pei F. Dual role of autophagy in lipopolysaccharide-induced preodontoblastic cells. Pengo N.

Plasma cells require autophagy for sustainable immunoglobulin production. Periyasamy K. Vitamin D—a host directed autophagy mediated therapy for tuberculosis. Piano Mortari E. The Vici syndrome protein EPG5 regulates intracellular nucleic acid trafficking linking autophagy to innate and adaptive immunity.

Autophagy 14 , 22 — Ponder E. Ubiquitin-like modifiers and their deconjugating enzymes in medically important parasitic protozoa.

Cell 6 , — Ponpuak M. Delivery of cytosolic components by autophagic adaptor protein p62 endows autophagosomes with unique antimicrobial properties.

Immunity 32 , — Autophagy 7 , —

Video

7 Foods You Should Never Eat – Dr. Berg Autophagy and immunity

Author: Jull

5 thoughts on “Autophagy and immunity

  1. Ich biete Ihnen an, die Webseite zu besuchen, auf der viele Informationen zum Sie interessierenden Thema gibt.

  2. Ich tue Abbitte, dass sich eingemischt hat... Ich hier vor kurzem. Aber mir ist dieses Thema sehr nah. Ich kann mit der Antwort helfen.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com