bims-proteo Biomed News
on Proteostasis
Issue of 2021‒02‒28
forty-five papers selected by
Eric Chevet
INSERM


  1. Nat Commun. 2021 Feb 26. 12(1): 1322
      The ubiquitin-proteasome system (UPS) and autophagy are two major quality control processes whose impairment is linked to a wide variety of diseases. The coordination between UPS and autophagy remains incompletely understood. Here, we show that ubiquitin ligase UBE3C and deubiquitinating enzyme TRABID reciprocally regulate K29/K48-branched ubiquitination of VPS34. We find that this ubiquitination enhances the binding of VPS34 to proteasomes for degradation, thereby suppressing autophagosome formation and maturation. Under ER and proteotoxic stresses, UBE3C recruitment to phagophores is compromised with a concomitant increase of its association with proteasomes. This switch attenuates the action of UBE3C on VPS34, thereby elevating autophagy activity to facilitate proteostasis, ER quality control and cell survival. Specifically in the liver, we show that TRABID-mediated VPS34 stabilization is critical for lipid metabolism and is downregulated during the pathogenesis of steatosis. This study identifies a ubiquitination type on VPS34 and elucidates its cellular fate and physiological functions in proteostasis and liver metabolism.
    DOI:  https://doi.org/10.1038/s41467-021-21715-1
  2. Biochem Soc Trans. 2021 Feb 26. pii: BST20200694. [Epub ahead of print]
      The accumulation of misfolded proteins is associated with numerous degenerative conditions, cancers and genetic diseases. These pathological imbalances in protein homeostasis (termed proteostasis), result from the improper triage and disposal of damaged and defective proteins from the cell. The ubiquitin-proteasome system is a key pathway for the molecular control of misfolded cytosolic proteins, co-opting a cascade of ubiquitin ligases to direct terminally damaged proteins to the proteasome via modification with chains of the small protein, ubiquitin. Despite the evidence for ubiquitination in this critical pathway, the precise complement of ubiquitin ligases and deubiquitinases that modulate this process remains under investigation. Whilst chaperones act as the first line of defence against protein misfolding, the ubiquitination machinery has a pivotal role in targeting terminally defunct cytosolic proteins for destruction. Recent work points to a complex assemblage of chaperones, ubiquitination machinery and subcellular quarantine as components of the cellular arsenal against proteinopathies. In this review, we examine the contribution of these pathways and cellular compartments to the maintenance of the cytosolic proteome. Here we will particularly focus on the ubiquitin code and the critical enzymes which regulate misfolded proteins in the cytosol, the molecular point of origin for many neurodegenerative and genetic diseases.
    Keywords:  cytosolic proteins; molecular chaperones; protein misfolding; protein quality control; ubiquitin; ubiquitin proteasome system
    DOI:  https://doi.org/10.1042/BST20200694
  3. Nat Commun. 2021 02 22. 12(1): 1220
      Attachment of ubiquitin (Ub) to proteins is one of the most abundant and versatile of all posttranslational modifications and affects outcomes in essentially all physiological processes. RING E3 ligases target E2 Ub-conjugating enzymes to the substrate, resulting in its ubiquitination. However, the mechanism by which a ubiquitin chain is formed on the substrate remains elusive. Here we demonstrate how substrate binding can induce a specific RING topology that enables self-ubiquitination. By analyzing a catalytically trapped structure showing the initiation of TRIM21 RING-anchored ubiquitin chain elongation, and in combination with a kinetic study, we illuminate the chemical mechanism of ubiquitin conjugation. Moreover, biochemical and cellular experiments show that the topology found in the structure can be induced by substrate binding. Our results provide insights into ubiquitin chain formation on a structural, biochemical and cellular level with broad implications for targeted protein degradation.
    DOI:  https://doi.org/10.1038/s41467-021-21443-6
  4. J Cancer Immunol (Wilmington). 2020 ;2(3): 103-115
      Endoplasmic reticulum oxidoreductin-1 alpha (ERO1α) was originally shown to be an endoplasmic reticulum (ER) resident protein undergoing oxidative cycles in concert with protein disulfide isomerase (PDI) to promote proper protein folding and to maintain homeostasis within the ER. ERO1α belongs to the flavoprotein family containing a flavin adenine dinucleotide utilized in transferring of electrons during oxidation-reduction cycles. This family is used to maintain redox potentials and protein homeostasis within the ER. ERO1α's location and function has since been shown to exist beyond the ER. Originally thought to exist solely in the ER, it has since been found to exist in the golgi apparatus, as well as in exosomes purified from patient samples. Besides aiding in protein folding of transmembrane and secretory proteins in conjunction with PDI, ERO1α is also known for formation of de novo disulfide bridges. Public databases, such as the Cancer Genome Atlas (TCGA) and The Protein Atlas, reveal ERO1α as a poor prognostic marker in multiple disease settings. Recent evidence indicates that ERO1α expression in tumor cells is a critical determinant of metastasis. However, the impact of increased ERO1α expression in tumor cells extends into the tumor microenvironment. Secretory proteins requiring ERO1α expression for proper folding have been implicated as being involved in immune escape through promotion of upregulation of programmed death ligand-1 (PD-L1) and stimulation of polymorphonuclear myeloid derived suppressor cells (PMN-MDSC's) via secretion of granulocytic colony stimulating factor (G-CSF). Hereby, ERO1α plays a pivotal role in cancer progression and potentially immune escape; making ERO1α an emerging attractive putative target for the treatment of cancer.
    Keywords:  Cancer; Cancer therapeutics; ER stress; ERO1; Immune resistance
    DOI:  https://doi.org/10.33696/cancerimmunol.2.023
  5. Biochemistry. 2021 Feb 26.
      DCAF16 is a substrate recognition component of Cullin-RING E3 ubiquitin ligases that can be targeted by electrophilic PROTACs (proteolysis targeting chimeras) to promote the nuclear-restricted degradation of proteins. The endogenous protein substates of DCAF16 remain unknown. In this study, we compared the protein content of DCAF16-wild type and DCAF16-knockout (KO) cells by untargeted mass spectrometry-based proteomics, identifying the Tudor domain-containing protein Spindlin-4 (SPIN4) as a protein with a level that was substantially increased in cells lacking DCAF16. Very few other proteomic changes were found in DCAF16-KO cells, pointing to a specific relationship between DCAF16 and SPIN4. Consistent with this hypothesis, we found that DCAF16 interacts with and ubiquitinates SPIN4, but not other related SPIN proteins, and identified a conserved lysine residue unique to SPIN4 that is involved in DCAF16 binding. Finally, we provide evidence that SPIN4 preferentially binds trimethylated histone H3K4 over other modified histone modifications. These results, taken together, indicate that DCAF16 and SPIN4 form a dedicated E3 ligase-substrate complex that regulates the turnover and presumed functions of SPIN4 in human cells.
    DOI:  https://doi.org/10.1021/acs.biochem.1c00067
  6. Curr Opin Cell Biol. 2021 Feb 18. pii: S0955-0674(21)00008-9. [Epub ahead of print]71 1-6
      The endoplasmic reticulum (ER) is the main harbor for newly synthesized proteins in eukaryotic cells. Through a continuous membrane network of sheets and tubules, the ER hosts secretory proteins, integral membrane proteins, and luminal proteins of the endomembrane system. These proteins are translated by ribosomes outside the ER and require subsequent integration into or translocation across the lipid bilayer of the ER. They are then modified post-translationally and folded in the ER. Some of these proteins are packaged into coat protein complex II-coated vesicles for export. Here, we review recent advances in understanding the mechanism of protein translocation and transmembrane domain insertion in the ER, summarize new insights into selective cargo packaging, and discuss the roles of ER morphological dynamics in these processes.
    Keywords:  COPII; Cargo receptor; Endoplasmic reticulum; Membrane insertion; Protein translocation; Retrotranslocation
    DOI:  https://doi.org/10.1016/j.ceb.2021.01.008
  7. Autophagy. 2021 Feb 25. 1-15
      Glioma is the most common primary malignant brain tumor with poor survival and limited therapeutic options. The non-psychoactive phytocannabinoid cannabidiol (CBD) has been shown to be effective against glioma; however, the molecular target and mechanism of action of CBD in glioma are poorly understood. Here we investigated the molecular mechanisms underlying the antitumor effect of CBD in preclinical models of human glioma. Our results showed that CBD induced autophagic rather than apoptotic cell death in glioma cells. We also showed that CBD induced mitochondrial dysfunction and lethal mitophagy arrest, leading to autophagic cell death. Mechanistically, calcium flux induced by CBD through TRPV4 (transient receptor potential cation channel subfamily V member 4) activation played a key role in mitophagy initiation. We further confirmed TRPV4 levels correlated with both tumor grade and poor survival in glioma patients. Transcriptome analysis and other results demonstrated that ER stress and the ATF4-DDIT3-TRIB3-AKT-MTOR axis downstream of TRPV4 were involved in CBD-induced mitophagy in glioma cells. Lastly, CBD and temozolomide combination therapy in patient-derived neurosphere cultures and mouse orthotopic models showed significant synergistic effect in both controlling tumor size and improving survival. Altogether, these findings showed for the first time that the antitumor effect of CBD in glioma is caused by lethal mitophagy and identified TRPV4 as a molecular target and potential biomarker of CBD in glioma. Given the low toxicity and high tolerability of CBD, we therefore propose CBD should be tested clinically for glioma, both alone and in combination with temozolomide.Abbreviations: 4-PBA: 4-phenylbutyrate; AKT: AKT serine/threonine kinase; ATF4: activating transcription factor 4; Baf-A1: bafilomycin A1; CANX: calnexin; CASP3: caspase 3; CAT: catalase; CBD: cannabidiol; CQ: chloroquine; DDIT3: DNA damage inducible transcript 3; ER: endoplasmic reticulum; GBM: glioblastoma multiforme; GFP: green fluorescent protein; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MTOR: mechanistic target of rapamycin kinase; PARP1: poly(ADP-ribose) polymerase; PINK1: PTEN induced kinase 1; PRKN: parkin RBR E3 ubiquitin protein ligase; SLC8A1: solute carrier family 8 member A1; SQSTM1: sequestosome 1; TCGA: The cancer genome atlas; TEM: transmission electron microscopy; TMZ: temozolomide; TRIB3: tribbles pseudokinase 3; TRPC: transient receptor potential cation channel subfamily C; TRPV4: transient receptor potential cation channel subfamily V member 4.
    Keywords:  Cannabidiol; cannabinoid; glioblastoma; glioma; mitophagy; trpv4
    DOI:  https://doi.org/10.1080/15548627.2021.1885203
  8. FEBS J. 2021 Feb 24.
      The transmembrane α-helices of membrane proteins are in general highly hydrophobic, and they enter the lipid bilayer through a lateral gate in the Sec61 translocon. However, some transmembrane α-helices are less hydrophobic and form membrane channels or substrate-binding pockets. Insertion of these amphipathic transmembrane α-helices into the membrane requires the specific membrane-embedded insertase called the endoplasmic reticulum membrane complex (EMC), which is a multi-subunit chaperone distinct from the GET insertase complex. Four recent cryo-EM studies on the eukaryotic EMC have revealed their remarkable architectural conservation from yeast to human; a general consensus on the substrate transmembrane helix-binding pocket; and the evolutionary link to the prokaryotic insertases of the tail-anchored membrane proteins. These structures provide a solid framework for future mechanistic investigation.
    Keywords:  Cryo-EM; insertase; membrane protein biogenesis; membrane protein folding; structural biology
    DOI:  https://doi.org/10.1111/febs.15786
  9. J Biol Chem. 2021 Feb 22. pii: S0021-9258(21)00226-X. [Epub ahead of print] 100453
      Collagen is the most abundant protein in humans. It has a characteristic triple-helix structure and is heavily post-translationally modified. The complex biosynthesis of collagen involves processing by many enzymes and chaperones in the rough endoplasmic reticulum. Lysyl hydroxylase 1 (LH1) is required to hydroxylate lysine for cross-linking and carbohydrate attachment within collagen triple helical sequences. Additionally, a recent study of prolyl 3-hydroxylase 3 (P3H3) demonstrated that this enzyme may be critical for LH1 activity, however the details surrounding its involvement remain unclear. If P3H3 is an LH1 chaperone that is critical for LH1 activity, P3H3 and LH1 null mice should display a similar deficiency in lysyl hydroxylation. To test this hypothesis, we compared the amount and location of hydroxylysine in the triple helical domains of type V and I collagen from P3H3 null, LH1 null and wild type mice. The amount of hydroxylysine in type V collagen was reduced in P3H3 null mice, but surprisingly type V collagen from LH1 null mice contained as much hydroxylysine as type V collagen from wild type mice. In type I collagen, our results indicate that LH1 plays a global enzymatic role in lysyl hydroxylation. P3H3 is also involved in lysyl hydroxylation, particularly at cross-link formation sites, but is not required for all lysyl hydroxylation sites. In summary, our study suggests that LH1 and P3H3 likely have two distinct mechanisms to recognize different collagen types and to distinguish cross-link formation sites from other sites in type I collagen.
    Keywords:  Collagen; Endoplasmic reticulum; Lysyl hydroxylase; Molecular chaperone; Post-translational modifications; Prolyl hydroxylase
    DOI:  https://doi.org/10.1016/j.jbc.2021.100453
  10. Biol Proced Online. 2021 Feb 22. 23(1): 8
      The proteostasis network includes all the factors that control the function of proteins in their native state and minimize their non-functional or harmful reactions. The molecular chaperones, the important mediator in the proteostasis network can be considered as any protein that contributes to proper folding and assembly of other macromolecules, through maturating of unfolded or partially folded macromolecules, refolding of stress-denatured proteins, and modifying oligomeric assembly, otherwise it leads to their proteolytic degradation. Viruses that use the hosts' gene expression tools and protein synthesis apparatus to survive and replicate, are obviously protected by such a host chaperone system. This means that many viruses use members of the hosts' chaperoning system to infect the target cells, replicate, and spread. During viral infection, increase in endoplasmic reticulum (ER) stress due to high expression of viral proteins enhances the level of heat shock proteins (HSPs) and induces cell apoptosis or necrosis. Indeed, evidence suggests that ER stress and the induction of unfolded protein response (UPR) may be a major aspect of the corona-host virus interaction. In addition, several clinical reports have confirmed the autoimmune phenomena in COVID-19-patients, and a strong association between this autoimmunity and severe SARS-CoV-2 infection. Part of such autoimmunity is due to shared epitopes among the virus and host. This article reviews the proteostasis network and its relationship to the immune system in SARS-CoV-2 infection.
    Keywords:  COVID-19; Chaperone; Coronavirus; Proteostasis; SARS-CoV-2
    DOI:  https://doi.org/10.1186/s12575-021-00145-9
  11. Autophagy. 2021 Feb 25. 1-3
      Neurons are long-lived cells that communicate via release of neurotransmitter at specialized contacts termed synapses. The maintenance of neuronal health and the regulation of synaptic function requires the efficient removal of damaged or dispensable proteins and organelles from synapses. How macroautophagy/autophagy contributes to neuronal and synaptic protein turnover, and what its main physiological substrates are in healthy neurons is largely unknown. We have now shown that loss of neuronal autophagy facilitates presynaptic neurotransmission by controlling the axonal endoplasmic reticulum and, thereby, axonal and synaptic calcium homeostasis.
    Keywords:  Autophagy; SV protein; calcium; endoplasmic reticulum; er-phagy; neurotransmission; reticulophagy; ryanodine receptor; synapse
    DOI:  https://doi.org/10.1080/15548627.2021.1893569
  12. J Biol Chem. 2021 Feb 18. pii: S0021-9258(21)00214-3. [Epub ahead of print] 100441
      The targeting and insertion of tail-anchored (TA) integral membrane proteins (IMP) into the correct membrane is critical for cellular homeostasis. The fungal protein Sgt2, and its human homolog SGTA, is the entry point for clients to the Guided Entry of Tail-anchored protein (GET) pathway, which targets ER-bound TA IMPs. Consisting of three structurally independent domains, the C-terminus of Sgt2 binds to the hydrophobic transmembrane domain (TMD) of clients. However, the exact binding interface within Sgt2 and molecular details that underlie its binding mechanism and client preference are not known. Here, we reveal the mechanism of Sgt2 binding to hydrophobic clients, including TA IMPs. Through sequence analysis, biophysical characterization, and a series of capture assays, we establish that the Sgt2 C-terminal domain is flexible but conserved, and sufficient for client binding. A molecular model for this domain reveals a helical hand forming a hydrophobic groove approximately 15Å long, that is consistent with our observed higher affinity for client TMDs with a hydrophobic face and a minimal length of 11 residues. This work places Sgt2 into a broader family of TPR-containing co-chaperone proteins, demonstrating structural and sequence-based similarities to the DP domains in the yeast Hsp90 and Hsp70 coordinating protein, Sti1.
    Keywords:  Hop; Protein targeting; Sti1; Tail-anchored proteins; co-chaperones
    DOI:  https://doi.org/10.1016/j.jbc.2021.100441
  13. Cell Physiol Biochem. 2021 Feb 25. 55(S1): 89-105
      Cell volume is one of the most aggressively defended physiological set points in biology. Changes in intracellular ion and water concentrations, which are induced by changes in metabolism or environmental exposures, disrupt protein folding, enzymatic activity, and macromolecular assemblies. To counter these challenges, cells and organisms have evolved multifaceted, evolutionarily conserved molecular mechanisms to restore cell volume and repair stress induced damage. However, many unanswered questions remain regarding the nature of cell volume 'sensing' as well as the molecular signaling pathways involved in activating physiological response mechanisms. Unbiased genetic screening in the model organism C. elegans is providing new and unexpected insights into these questions, particularly questions relating to the hypertonic stress response (HTSR) pathway. One surprising characteristic of the HTSR pathway in C. elegans is that it is under strong negative regulation by proteins involved in protein homeostasis and the extracellular matrix (ECM). The role of the ECM in particular highlights the importance of studying the HTSR in the context of a live organism where native ECM-tissue associations are preserved. A second novel and recently discovered characteristic is that the HTSR is regulated at the post-transcriptional level. The goal of this review is to describe these discoveries, to provide context for their implications, and to raise outstanding questions to guide future research.
    Keywords:  Hypertonic stress; Osmoregulation; Stress response; Compatible solute
    DOI:  https://doi.org/10.33594/000000332
  14. Autophagy. 2021 Feb 25. 1-3
      Phase-separated droplets with liquid-like properties can be degraded by macroautophagy/autophagy, but the mechanism underlying this degradation is poorly understood. We have recently derived a physical model to investigate the interaction between autophagic membranes and such droplets, uncovering that intrinsic wetting interactions underlie droplet-membrane contacts. We found that the competition between droplet surface tension and the increasing tendency of growing membrane sheets to bend determines whether a droplet is completely engulfed or isolated in a piecemeal fashion, a process we term fluidophagy. Intriguingly, we found that another critical parameter of droplet-membrane interactions, the spontaneous curvature of the membrane, determines whether the droplet is degraded by autophagy or - counterintuitively - serves as a platform from which autophagic membranes expand into the cytosol. We also discovered that the interaction of membrane-associated LC3 with the LC3-interacting region (LIR) found in the autophagic cargo receptor protein SQSTM1/p62 and many other autophagy-related proteins influences the preferred bending directionality of forming autophagosomes in living cells. Our study provides a physical account of how droplet-membrane wetting underpins the structure and fate of forming autophagosomes.
    Keywords:  Autophagy; condensate; droplet; isolation membrane; membrane; p62; phase separation; piecemeal autophagy; wetting
    DOI:  https://doi.org/10.1080/15548627.2021.1887548
  15. J Pathol. 2021 Feb 27.
      Tamoxifen (TAM) resistance is a significant clinical challenge in endocrine therapies for estrogen receptor (ER)-positive breast cancer patients. Cullin 4B (CUL4B), which acts as a scaffold protein in CUL4B-RING ubiquitin ligase complexes (CRL4B), is frequently overexpressed in cancer and represses tumor suppressors through diverse epigenetic mechanisms. However, the role and the underlying mechanisms of CUL4B in regulating drug resistance remain unknown. Here, we showed that CUL4B promotes TAM resistance in breast cancer cells through a miR-32-5p/ER-α36 axis. We found that upregulation of CUL4B correlated with decreased TAM sensitivity of breast cancer cells, and knockdown of CUL4B or expression of a dominant-negative CUL4B mutant restored the response to TAM in TAM-resistant MCF7-TAMR and T47D-TAMR cells. Mechanistically, we demonstrated that CUL4B renders breast cancer cells TAM resistant by upregulating ER-α36 expression, which was mediated by downregulation of miR-32-5p. We further showed that CRL4B epigenetically represses the transcription of miR-32-5p by catalyzing monoubiquitination at H2AK119 and coordinating with PRC2 and HDAC complexes to promote trimethylation at H3K27 at the promoter of miR-32-5p. Pharmacologic or genetic inhibition of CRL4B/PRC2/HDAC complexes significantly increased TAM sensitivity in breast cancer cells in vitro and in vivo. Taken together, our findings thus establish a critical role for the CUL4B-miR-32-5p-ER-α36 axis in the regulation of TAM resistance and have important therapeutic implications of combined application of TAM and the inhibitors of CRL4B/PRC2/HDAC complex in breast cancer treatment. This article is protected by copyright. All rights reserved.
    Keywords:  CUL4B; ER-α36; breast cancer; miR-32-5p; tamoxifen resistance
    DOI:  https://doi.org/10.1002/path.5657
  16. J Biol Chem. 2021 Feb 19. pii: S0021-9258(21)00213-1. [Epub ahead of print] 100440
      Obesity associates with inflammation, insulin resistance and higher blood lipids. It is unclear if immune responses facilitate lipid breakdown and release from adipocytes via lipolysis in a separate way from hormones or adrenergic signals. We found that an ancient component of ER stress, inositol-requiring protein 1 (IRE1), discriminates inflammation-induced adipocyte lipolysis versus lipolysis from adrenergic or hormonal stimuli. Our data show that inhibiting IRE1 kinase activity was sufficient to block adipocyte-autonomous lipolysis from multiple inflammatory ligands, including bacterial components, certain cytokines, and thapsigargin-induced ER stress. IRE1-mediated lipolysis was specific for inflammatory triggers since IRE1 kinase activity was dispensable for isoproterenol and cAMP-induced lipolysis in adipocytes and mouse adipose tissue. IRE1 RNase activity was not associated with inflammation-induced adipocyte lipolysis. Inhibiting IRE1 kinase activity blocked NF-κB activation, interleukin-6 secretion, and adipocyte-autonomous lipolysis from inflammatory ligands. Inflammation-induced lipolysis mediated by IRE1 occurred independently from changes in insulin signaling in adipocytes, suggesting that inflammation can promote IRE1-mediated lipolysis independent of adipocyte insulin resistance. We found no role for canonical unfolded protein responses or ABL kinases in linking ER stress to IRE1-mediated lipolysis. Adiponectin-Cre-mediated IRE1 knockout in mice showed that adipocyte IRE1 was required for inflammatory ligand-induced lipolysis in adipose tissue explants and that adipocyte IRE1 was required for approximately half of the increase in blood triglycerides after a bacterial endotoxin-mediated inflammatory stimulus in vivo. Together, our results show that IRE1 propagates an inflammation-specific lipolytic program independent from hormonal or adrenergic regulation. Targeting IRE1 kinase activity may benefit metabolic syndrome and inflammatory lipid disorders.
    Keywords:  ER stress; adipocyte; cytokine; endocrinology; immunometabolism; inflammation; lipid; lipolysis; metabolic syndrome; obesity
    DOI:  https://doi.org/10.1016/j.jbc.2021.100440
  17. Autophagy. 2021 Feb 26. 1-17
      Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two clinically distinct classes of neurodegenerative disorders. Yet, they share a range of genetic, cellular, and molecular features. Hexanucleotide repeat expansions (HREs) in the C9orf72 gene and the accumulation of toxic protein aggregates in the nervous systems of the affected individuals are among such common features. Though the mechanisms by which HREs cause toxicity is not clear, the toxic gain of function due to transcribed HRE RNA or dipeptide repeat proteins (DPRs) produced by repeat-associated non-AUG translation together with a reduction in C9orf72 expression are proposed as the contributing factors for disease pathogenesis in ALS and FTD. In addition, several recent studies point toward alterations in protein homeostasis as one of the root causes of the disease pathogenesis. In this review, we discuss the effects of the C9orf72 HRE in the autophagy-lysosome pathway based on various recent findings. We suggest that dysfunction of the autophagy-lysosome pathway synergizes with toxicity from C9orf72 repeat RNA and DPRs to drive disease pathogenesis.Abbreviation: ALP: autophagy-lysosome pathway; ALS: amyotrophic lateral sclerosis; AMPK: AMP-activated protein kinase; ATG: autophagy-related; ASO: antisense oligonucleotide; C9orf72: C9orf72-SMCR8 complex subunit; DENN: differentially expressed in normal and neoplastic cells; DPR: dipeptide repeat protein; EIF2A/eIF2α: eukaryotic translation initiation factor 2A; ER: endoplasmic reticulum; FTD: frontotemporal dementia; GAP: GTPase-activating protein; GEF: guanine nucleotide exchange factor; HRE: hexanucleotide repeat expansion; iPSC: induced pluripotent stem cell; ISR: integrated stress response; M6PR: mannose-6-phosphate receptor, cation dependent; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MN: motor neuron; MTORC1: mechanistic target of rapamycin kinase complex 1; ND: neurodegenerative disorder; RAN: repeat-associated non-ATG; RB1CC1/FIP200: RB1 inducible coiled-coil 1; SLC66A1/PQLC2: solute carrier family 66 member 1; SMCR8: SMCR8-C9orf72 complex subunit; SQSTM1/p62: sequestosome 1; STX17: syntaxin 17; TARDBP/TDP-43: TAR DNA binding protein; TBK1: TANK binding kinase 1; TFEB: transcription factor EB; ULK1: unc-51 like autophagy activating kinase 1; UPS: ubiquitin-proteasome system; WDR41: WD repeat domain 41.
    Keywords:  Amyotrophic lateral sclerosis (ALS); autophagy; axonal transport; c9orf72; dipeptide repeat protein (DPR); frontotemporal dementia (FTD); lysosome; smcr8; wdr41
    DOI:  https://doi.org/10.1080/15548627.2021.1872189
  18. Oncogene. 2021 Feb 24.
      Protein ubiquitination is a critical regulator of cellular homeostasis. Aberrations in the addition or removal of ubiquitin can result in the development of cancer and key components of the ubiquitination machinery serve as oncogenes or tumour suppressors. An emerging target in the development of cancer therapeutics are the deubiquitinase (DUB) enzymes that remove ubiquitin from protein substrates. Whether this class of enzyme plays a role in cervical cancer has not been fully explored. By interrogating the cervical cancer data from the TCGA consortium, we noted that the DUB USP13 is amplified in ~15% of cervical cancer cases. We confirmed that USP13 expression was increased in cervical cancer cell lines, cytology samples from patients with cervical disease and in cervical cancer tissue. Depletion of USP13 inhibited cervical cancer cell proliferation. Mechanistically, USP13 bound to, deubiquitinated and stabilised Mcl-1, a pivotal member of the anti-apoptotic BCL-2 family. Furthermore, reduced Mcl-1 expression partially contributed to the observed proliferative defect in USP13 depleted cells. Importantly, the expression of USP13 and Mcl-1 proteins correlated in cervical cancer tissue. Finally, we demonstrated that depletion of USP13 expression or inhibition of USP13 enzymatic activity increased the sensitivity of cervical cancer cells to the BH3 mimetic inhibitor ABT-263. Together, our data demonstrates that USP13 is a potential oncogene in cervical cancer that functions to stabilise the pro-survival protein Mcl-1, offering a potential therapeutic target for these cancers.
    DOI:  https://doi.org/10.1038/s41388-021-01679-8
  19. J Neurosci. 2021 Feb 22. pii: JN-RM-1074-20. [Epub ahead of print]
      Reversible modification of AMPARs with ubiquitin regulates receptor levels at synapses and controls synaptic strength. The conserved deubiquitinating enzyme (DUB) USP-46 removes ubiquitin from AMPARs and protects them from degradation in both C. elegans and mammals. Although DUBs are critical for diverse physiological processes, the mechanisms that regulate DUBs, especially in the nervous system, are not well understood. We and others previously showed that the WD40-repeat proteins WDR-48 and WDR-20 bind to and stimulate the catalytic activity of USP-46. Here, we identify an activity-dependent mechanism that regulates WDR-20 expression and show that WDR-20 works together with USP-46 and WDR-48 to promote surface levels of the C. elegans AMPAR GLR-1. usp-46, wdr-48 and wdr-20 loss-of-function mutants exhibit reduced levels of GLR-1 at the neuronal surface and corresponding defects in GLR-1-mediated behavior. Increased expression of WDR-20, but not WDR-48, is sufficient to increase GLR-1 surface levels in an usp-46-dependent manner. Loss of usp-46, wdr-48 and wdr-20 function reduces the rate of local GLR-1 insertion in neurites, whereas overexpression of wdr-20 is sufficient to increase the rate of GLR-1 insertion. Genetic manipulations that chronically reduce or increase glutamate signaling result in reciprocal alterations in wdr-20 transcription and homeostatic compensatory changes in surface GLR-1 levels that are dependent on wdr-20 This study identifies wdr-20 as a novel activity-regulated gene that couples chronic changes in synaptic activity with increased local insertion and surface levels of GLR-1 via the deubiquitinating enzyme USP-46.SIGNIFICANCE STATEMENTDeubiquitinating enzymes (DUBs) are critical regulators of synapse development and function; however the regulatory mechanisms that control their various physiological functions are not well understood. This study identifies a novel role for the DUB Ubiquitin-Specific Protease-46 (USP-46) and its associated regulatory protein WD40-Repeat Protein-20 (WDR-20) in regulating local insertion of glutamate receptors into the neuronal cell surface. This work also identifies WDR-20 as an activity-regulated gene that couples chronic changes in synaptic activity with homeostatic compensatory increases in surface levels of GLR-1 via USP-46. Given that 35% of USP family DUBs associate with WDR proteins, understanding the mechanisms by which WDR proteins regulate USP-46 could have implications for a large number of DUBs in other cell types.
    DOI:  https://doi.org/10.1523/JNEUROSCI.1074-20.2021
  20. Cell Stress Chaperones. 2021 Feb 22.
      Stalled mRNA translation results in the production of incompletely synthesized proteins that are targeted for degradation by ribosome-associated quality control (RQC). Here we investigated the fate of defective proteins translated from stall-inducing, nonstop mRNA that escape ubiquitylation by the RQC protein LTN1. We found that nonstop protein products accumulated in nucleoli and this localization was driven by polylysine tracts produced by translation of the poly(A) tails of nonstop mRNA. Nucleolar sequestration increased the solubility of invading proteins but disrupted nucleoli, altering their dynamics, morphology, and resistance to stress in cell culture and intact flies. Our work elucidates how stalled translation may affect distal cellular processes and may inform studies on the pathology of diseases caused by failures in RQC and characterized by nucleolar stress.
    Keywords:  LTN1; Nonstop mRNA; Nucleolus; Phase separation; Protein quality control; Ribosome-associated quality control (RQC)
    DOI:  https://doi.org/10.1007/s12192-021-01200-w
  21. J Chem Inf Model. 2021 Feb 24.
      Proteolysis-targeting chimaeras (PROTACs) are molecules that combine a target-binding warhead with an E3 ligase-recruiting moiety; by drawing the target protein into a ternary complex with the E3 ligase, PROTACs induce target protein degradation. While PROTACs hold exciting potential as chemical probes and as therapeutic agents, development of a PROTAC typically requires synthesis of numerous analogs to thoroughly explore variations on the chemical linker; without extensive trial and error, it is unclear how to link the two protein-recruiting moieties to promote formation of a productive ternary complex. Here, we describe a structure-based computational method for evaluating the suitability of a given linker for ternary complex formation. Our method uses Rosetta to dock the protein components and then builds the PROTAC from its component fragments into each binding mode; complete models of the ternary complex are then refined. We apply this approach to retrospectively evaluate multiple PROTACs from the literature, spanning diverse target proteins. We find that modeling ternary complex formation is sufficient to explain both activity and selectivity reported for these PROTACs, implying that other cellular factors are not key determinants of activity in these cases. We further find that interpreting PROTAC activity is best approached using an ensemble of structures of the ternary complex rather than a single static conformation and that members of a structurally conserved protein family can be recruited by the same PROTAC through vastly different binding modes. To encourage adoption of these methods and promote further analyses, we disseminate both the computational methods and the models of ternary complexes.
    DOI:  https://doi.org/10.1021/acs.jcim.0c01451
  22. Angew Chem Int Ed Engl. 2021 Feb 23.
      The 90 kDa heat shock protein (Hsp90) is a molecular chaperone that processes nascent polypeptides into their biologically active conformations. Many of these proteins contribute to the progression of cancer, and consequently, inhibition of the Hsp90 protein folding machinery represents an innovative approach toward cancer chemotherapy. However, clinical trials with Hsp90 N-terminal inhibitors have encountered deleterious side effects and toxicities, which appear to result from the pan -inhibition of all four Hsp90 isoforms. Therefore, the development of isoform-selective Hsp90 inhibitors is sought to delineate the pathological role played by each isoform. Herein, we describe a structure-based approach that was used to design the first Hsp90a-selective inhibitors, which exhibit > 50-fold selectivity versus other Hsp90 isoforms.
    Keywords:  Hsp90, cancer, isoform-selective, neurodegeneration, inhibitor
    DOI:  https://doi.org/10.1002/anie.202015422
  23. J Cell Sci. 2021 Feb 23. pii: jcs.249276. [Epub ahead of print]
      A genome-wide screen recently identified SEC24A as a novel mediator of thapsigargin-induced cell death in HAP1 cells. Here, we determined the cellular mechanism and specificity of SEC24A-mediated cytotoxicity. Measurement of calcium levels using organelle-specific fluorescent indicator dyes showed that calcium efflux from endoplasmic reticulum (ER) and influx into mitochondria were significantly impaired in SEC24A knockout cells. Furthermore, SEC24A knockout cells also showed ∼44% less colocalization of mitochondria and peripheral tubular ER. Knockout of SEC24A, but not its paralogs SEC24B, SEC24C, or SEC24D, rescued HAP1 cells from cell death induced by three different inhibitors of Sarcoplasmic/Endoplasmic Reticulum Ca2+ ATPase (SERCA) but not from cell death induced by a topoisomerase inhibitor. Thapsigargin-treated SEC24A knockout cells showed a ∼2.5-fold increase in autophagic flux and ∼10-fold reduction in apoptosis compared to wild-type cells. Taken together, our findings indicate that SEC24A plays a previously unrecognized role in regulating association and calcium flux between the ER and mitochondria, thereby impacting processes dependent on mitochondrial calcium levels, including autophagy and apoptosis.
    Keywords:  Apoptosis; Autophagy; Calcium; ER stress; Mitochondrial-associated membranes; SEC24A; SERCA; Thapsigargin
    DOI:  https://doi.org/10.1242/jcs.249276
  24. EMBO J. 2021 Feb 25. e104888
      Endoplasmic reticulum (ER) calcium (Ca2+ ) stores are critical to proteostasis, intracellular signaling, and cellular bioenergetics. Through forward genetic screening in mice, we identified two members of a new complex, Pacs1 and Wdr37, which are required for normal ER Ca2+ handling in lymphocytes. Deletion of Pacs1 or Wdr37 caused peripheral lymphopenia that was linked to blunted Ca2+ release from the ER after antigen receptor stimulation. Pacs1-deficient cells showed diminished inositol triphosphate receptor expression together with increased ER and oxidative stress. Mature Pacs1-/- B cells proliferated and died in vivo under lymphocyte replete conditions, indicating spontaneous loss of cellular quiescence. Disruption of Pacs1-Wdr37 did not diminish adaptive immune responses, but potently suppressed lymphoproliferative disease models by forcing loss of quiescence. Thus, Pacs1-Wdr37 plays a critical role in stabilizing lymphocyte populations through ER Ca2+ handling and presents a new target for lymphoproliferative disease therapy.
    Keywords:  Pacs1; Wdr37; calcium homeostasis; lymphocyte quiescence; lymphoproliferative disease
    DOI:  https://doi.org/10.15252/embj.2020104888
  25. Proc Natl Acad Sci U S A. 2021 Mar 02. pii: e2014188118. [Epub ahead of print]118(9):
      Tau protein plays an important role in the biology of stress granules and in the stress response of neurons, but the nature of these biochemical interactions is not known. Here we show that the interaction of tau with RNA and the RNA binding protein TIA1 is sufficient to drive phase separation of tau at physiological concentrations, without the requirement for artificial crowding agents such as polyethylene glycol (PEG). We further show that phase separation of tau in the presence of RNA and TIA1 generates abundant tau oligomers. Prior studies indicate that recombinant tau readily forms oligomers and fibrils in vitro in the presence of polyanionic agents, including RNA, but the resulting tau aggregates are not particularly toxic. We discover that tau oligomers generated during copartitioning with TIA1 are significantly more toxic than tau aggregates generated by incubation with RNA alone or phase-separated tau complexes generated by incubation with artificial crowding agents. This pathway identifies a potentially important source for generation of toxic tau oligomers in tau-related neurodegenerative diseases. Our results also reveal a general principle that phase-separated RBP droplets provide a vehicle for coassortment of selected proteins. Tau selectively copartitions with TIA1 under physiological conditions, emphasizing the importance of TIA1 for tau biology. Other RBPs, such as G3BP1, are able to copartition with tau, but this happens only in the presence of crowding agents. This type of selective mixing might provide a basis through which membraneless organelles bring together functionally relevant proteins to promote particular biological activities.
    Keywords:  Alzheimer's disease; RNA binding proteins; fibrillar tau; liquid–liquid phase separation (LLPS); oligomeric tau
    DOI:  https://doi.org/10.1073/pnas.2014188118
  26. Nat Rev Cardiol. 2021 Feb 22.
      Cardiovascular diseases (CVDs), such as ischaemic heart disease, cardiomyopathy, atherosclerosis, hypertension, stroke and heart failure, are among the leading causes of morbidity and mortality worldwide. Although specific CVDs and the associated cardiometabolic abnormalities have distinct pathophysiological and clinical manifestations, they often share common traits, including disruption of proteostasis resulting in accumulation of unfolded or misfolded proteins in the endoplasmic reticulum (ER). ER proteostasis is governed by the unfolded protein response (UPR), a signalling pathway that adjusts the protein-folding capacity of the cell to sustain the cell's secretory function. When the adaptive UPR fails to preserve ER homeostasis, a maladaptive or terminal UPR is engaged, leading to the disruption of ER integrity and to apoptosis. ER stress functions as a double-edged sword, with long-term ER stress resulting in cellular defects causing disturbed cardiovascular function. In this Review, we discuss the distinct roles of the UPR and ER stress response as both causes and consequences of CVD. We also summarize the latest advances in our understanding of the importance of the UPR and ER stress in the pathogenesis of CVD and discuss potential therapeutic strategies aimed at restoring ER proteostasis in CVDs.
    DOI:  https://doi.org/10.1038/s41569-021-00511-w
  27. J Lipid Res. 2021 Feb 22. pii: S0022-2275(21)00032-8. [Epub ahead of print] 100051
      Oxysterols are oxidized derivatives of cholesterol that play regulatory roles in lipid biosynthesis and homeostasis. How oxysterol signaling coordinates different lipid classes such as sterols and triglycerides remain incompletely understood. Here, we show that 4β-hydroxycholesterol (4β-HC), a liver and serum abundant oxysterol of poorly defined function, is a potent and selective inducer of the master lipogenic transcription factor, Sterol Regulatory Element Binding Protein 1c (SREBP1c), but not the related steroidogenic transcription factor SREBP2. By correlating tracing of lipid synthesis with lipogenic gene expression profiling, we found that 4β-HC acts as a putative agonist for Liver X receptor (LXR), a sterol sensor and transcriptional regulator previously linked to SREBP1c activation. Unique among the oxysterol agonists of LXR, 4β-HC induced expression of the lipogenic program downstream of SREBP1c and triggered de novo lipogenesis both in primary hepatocytes and in mouse liver. Additionally, 4β-HC acted in parallel to insulin-PI3K-dependent signaling to stimulate triglyceride synthesis and lipid droplet accumulation. Thus, 4β-HC is an endogenous regulator of de novo lipogenesis through the LXR-SREBP1c axis.
    DOI:  https://doi.org/10.1016/j.jlr.2021.100051
  28. Front Cell Dev Biol. 2021 ;9 627700
      The endoplasmic reticulum (ER) forms direct membrane contact sites with the plasma membrane (PM) in eukaryotic cells. These ER-PM contact sites play essential roles in lipid homeostasis, ion dynamics, and cell signaling, which are carried out by protein-protein or protein-lipid interactions. Distinct tethering factors dynamically control the architecture of ER-PM junctions in response to intracellular signals or external stimuli. The physiological roles of ER-PM contact sites are dependent on a variety of regulators that individually or cooperatively perform functions in diverse cellular processes. This review focuses on proteins functioning at ER-PM contact sites and highlights the recent progress in their mechanisms and physiological roles.
    Keywords:  endoplasmic reticulum (ER); enzyme; lipid transfer; membrane contact sites (MCSs); plasma membrane; tether
    DOI:  https://doi.org/10.3389/fcell.2021.627700
  29. Sci Signal. 2021 Feb 23. pii: eabd2639. [Epub ahead of print]14(671):
      Ribosome biogenesis in eukaryotes requires the coordinated production and assembly of 80 ribosomal proteins and four ribosomal RNAs (rRNAs), and its rate must be synchronized with cellular growth. Here, we showed that the Microprocessor complex, which mediates the first step of microRNA processing, potentiated the transcription of ribosomal protein genes by eliminating DNA/RNA hybrids known as R-loops. Nutrient deprivation triggered the nuclear export of Drosha, a key component of the Microprocessor complex, and its subsequent degradation by the E3 ubiquitin ligase Nedd4, thereby reducing ribosomal protein production and protein synthesis. In mouse erythroid progenitors, conditional deletion of Drosha led to the reduced production of ribosomal proteins, translational inhibition of the mRNA encoding the erythroid transcription factor Gata1, and impaired erythropoiesis. This phenotype mirrored the clinical presentation of human "ribosomopathies." Thus, the Microprocessor complex plays a pivotal role in synchronizing protein synthesis capacity with cellular growth rate and is a potential drug target for anemias caused by ribosomal insufficiency.
    DOI:  https://doi.org/10.1126/scisignal.abd2639
  30. Mol Cell Endocrinol. 2021 Feb 23. pii: S0303-7207(21)00062-9. [Epub ahead of print] 111218
      Maternal obesity malprograms offspring obesity and associated metabolic disorder. As a common phenomenon in obesity, endoplasmic reticulum (ER) stress also presents early prior to the development. Here, we investigate metabolic effect of early activated hypothalamic ER stress in offspring exposed to maternal obesogenic environment and the underlying mechanism in ICR mice model. We found higher body weight, hyperphagia and defective hypothalamic feeding-circuit in the offspring born to obese dams, with hypothalamic ER stress, and even more comprehensive cell proteotoxic stress were induced during the early postnatal period. However, neonatal inhibition of hypothalamic ER stress worsened the metabolic end. We believe that the uncoordinated interaction between the unfolded protein response and the heat shock response, regulated by heat shock protein 70, might be responsible for the malformed hypothalamic feeding circuit of the offspring exposure to maternal obesogenic conditions and were linked with deleterious metabolism in adulthood, especially when exposure to high-energy conditions.
    Keywords:  Feeding circuit development; Heat shock response; Hypothalamic ER stress; Maternal obesity; Unfolded protein response
    DOI:  https://doi.org/10.1016/j.mce.2021.111218
  31. Cell Mol Life Sci. 2021 Feb 25.
      Innate immunity is an evolutionary ancient defence strategy that serves to eliminate infectious agents while maintaining host health. It involves a complex network of sensors, signaling proteins and immune effectors that detect the danger, then relay and execute the immune programme. Post-translational modifications relying on conserved ubiquitin and ubiquitin-like proteins are an integral part of the system. Studies using invertebrate models of infection, such as the nematode Caenorhabditis elegans, have greatly contributed to our understanding of how ubiquitin-related processes act in immune sensing, regulate immune signaling pathways, and participate to host defence responses. This review highlights the interest of working with a genetically tractable model organism and illustrates how C. elegans has been used to identify ubiquitin-dependent immune mechanisms, discover novel ubiquitin-based resistance strategies that mediate pathogen clearance, and unravel the role of ubiquitin-related processes in tolerance, preserving host fitness during pathogen attack. Special emphasis is placed on processes that are conserved in mammals.
    Keywords:  Host–pathogen interaction; Proteostasis; SUMOylation; Ubiquitination; Unfolded protein response
    DOI:  https://doi.org/10.1007/s00018-021-03787-w
  32. Dev Cell. 2021 Feb 15. pii: S1534-5807(21)00074-5. [Epub ahead of print]
      Cell-cell fusion is a frequent and essential event during development, and its dysregulation causes diseases ranging from infertility to muscle weakness. Fusing cells need to repeatedly remodel their plasma membrane through orchestrated formation and disassembly of actin filaments, but how the dynamic reorganization of the cortical actin cytoskeleton is controlled is still poorly understood. Here, we identified a ubiquitin-dependent toggle switch that establishes reversible actin bundling during mammalian cell fusion. We found that EPS8-IRSp53 complexes stabilize cortical actin bundles at sites of cell contact to promote close membrane alignment. EPS8 monoubiquitylation by CUL3KCTD10 displaces EPS8-IRSp53 from membranes and counteracts actin bundling, a dual activity that restricts actin bundling to allow paired cells to progress with fusion. We conclude that cytoskeletal rearrangements during development are precisely controlled by ubiquitylation, raising the possibility of modulating the efficiency of cell-cell fusion for therapeutic benefit.
    Keywords:  CUL3; EPS8; actin; actin bundle; cell fusion; monoubiquitylation; ubiquitin
    DOI:  https://doi.org/10.1016/j.devcel.2021.01.016
  33. Nucleic Acids Res. 2021 Feb 22. pii: gkab081. [Epub ahead of print]
      The major clinical problem in human cancer is metastasis. Metastases are the cause of 90% of human cancer deaths. TAp63 is a critical suppressor of tumorigenesis and metastasis. ΔNp63 acts as a dominant-negative inhibitor to block the function of p53 and TAp63. Although several ubiquitin E3 ligases have been reported to regulate p63 stability, the mechanism of p63 regulation remains partially understood. Herein, we show that CHIP, an E3 ligase with a U-box domain, physically interacts with p63 and promotes p63 degradation. Notably, Hsp70 depletion by siRNA stabilizes TAp63 in H1299 cells and destabilizes ΔNp63 in SCC9 cells. Loss of Hsp70 results in a reduction in the TAp63-CHIP interaction in H1299 cells and an increase in the interaction between ΔNp63 and CHIP in SCC9 cells. Our results reveal that Hsp70 acts as a molecular switch to control CHIP-mediated ubiquitination and degradation of p63 isoforms. Furthermore, regulation of p63 by the Hsp70-CHIP axis contributes to the migration and invasion of tumor cells. Hence, our findings demonstrate that Hsp70 is a crucial regulator of CHIP-mediated ubiquitination and degradation of p63 isoforms and identify a new pathway for maintaining TAp63 or ΔNp63 stability in cancers.
    DOI:  https://doi.org/10.1093/nar/gkab081
  34. Cell Stress Chaperones. 2021 Feb 24.
      Pheochromocytomas and paragangliomas (PCPGs) are catecholamine-producing neuroendocrine tumors. Accumulating evidences indicate that the blockade of antioxidative pathways might be a novel therapeutic approach to the treatment of PCPG. NIX has been confirmed to play a key role in maintaining redox homeostasis in tumors, while the function of NIX in PCPG remains unclear. In this study, the analyses of the disease-free survival (DFS) showed that high NIX protein level is related to poor prognosis in patients of PCPG. Consistent with this, high level of NIX protein upregulates the level of p-NF-κB and promotes the migration of PC12 cells. In NIX-over-expressing PC12 cells, the level of reactive oxygen species (ROS) is decreased while trolox-equivalent antioxidant capacity (TEAC) increased. But in NIX-silencing cells, ROS level is increased, while TEAC reversely reduced, consequently antioxidase and phase II enzymes of NRF2 signaling were activated, and elevated endoplasmic reticulum (ER) stress was observed. Additionally, the apoptosis induced by luminespib/NVP-AUY922, an inhibitor of heat shock protein 90 (HSP90, a cellular stress response factor), was enhanced in NIX-silencing cells but reduced in the NIX-over-expressing cells. All of these results indicated that high NIX protein level enhances antioxidant capacity of PC12 cells and reduces the apoptosis caused by cell stress, such as induced by luminespib/NVP-AUY922. Therefore, luminespib/NVP-AUY922 might be effective only for PCPG with low NIX level, while targeting NIX could be a further supplement to the therapeutic treatment strategy for PCPG patients with high NIX protein level.
    Keywords:  HSP90 inhibitor; Luminespib/NVP-AUY922; NIX; PC12 cells; PCPG; ROS
    DOI:  https://doi.org/10.1007/s12192-021-01193-6
  35. Nat Commun. 2021 02 23. 12(1): 1248
      Mutations in human equilibrative nucleoside transporter 3 (ENT3) encoded by SLC29A3 results in anemia and erythroid hypoplasia, suggesting that ENT3 may regulate erythropoiesis. Here, we demonstrate that lysosomal ENT3 transport of taurine-conjugated bile acids (TBA) facilitates TBA chemical chaperone function and alleviates endoplasmic reticulum (ER) stress in expanding mouse hematopoietic stem and progenitor cells (HSPCs). Slc29a3-/- HSPCs accumulate less TBA despite elevated levels of TBA in Slc29a3-/- mouse plasma and have elevated basal ER stress, reactive oxygen species (ROS), and radiation-induced apoptosis. Reintroduction of ENT3 allows for increased accumulation of TBA into HSPCs, which results in TBA-mediated alleviation of ER stress and erythroid apoptosis. Transplanting TBA-preconditioned HSPCs expressing ENT3 into Slc29a3-/- mice increase bone marrow repopulation capacity and erythroid pool size and prevent early mortalities. Together, these findings suggest a putative role for a facilitative lysosomal transporter in the bile acid regulation of ER stress in mouse HSPCs which may have implications in erythroid biology, the treatment of anemia observed in ENT3-mutated human genetic disorders, and nucleoside analog drug therapy.
    DOI:  https://doi.org/10.1038/s41467-021-21451-6
  36. Bioorg Med Chem Lett. 2021 Feb 17. pii: S0960-894X(21)00083-4. [Epub ahead of print] 127857
      Heat shock protein 90 (Hsp90) is an essential molecular chaperone that performs vital stress-related and housekeeping functions in cells and is a current therapeutic target for diseases such as cancers. Particularly, the development of Hsp90 C-terminal domain (CTD) inhibitors is highly desirable as inhibitors that target the N-terminal nucleotide-binding domain may cause unwanted biological effects. Herein, we report on the discovery of two drug-like novel Hsp90 CTD inhibitors by using virtual screening and intrinsic protein fluorescence quenching binding assays, paving the way for future development of new therapies that employ molecular chaperone inhibitors.
    Keywords:  Cancer; Hsp90; Inhibitor; Molecular chaperone; Virtual screening
    DOI:  https://doi.org/10.1016/j.bmcl.2021.127857
  37. Biochim Biophys Acta Mol Basis Dis. 2021 Feb 19. pii: S0925-4439(21)00037-5. [Epub ahead of print] 166104
      Depolarized/damaged mitochondria aggregate at the perinuclear region prior to mitophagy in cells treated with mitochondrial stressors. However, the cellular mechanism(s) by which damaged mitochondria are transported and remain aggregated at the perinuclear region is unknown. Here, we demonstrate that mitofusins (Mfn1/2) are post-translationally modified by SUMO2 (Small Ubiquitin-related Modifier 2) in Human embryonic kidney 293 (Hek293) cells treated with protonophore CCCP and proteasome inhibitor MG132, both known mitochondrial stressors. SUMOylation of Mfn1/2 is not for their proteasomal degradation but facilitate mitochondrial congression at the perinuclear region in CCCP- and MG132-treated cells. Additionally, congressed mitochondria (mito-aggresomes) colocalize with LC3, ubiquitin, and SUMO2 in CCCP-treated cells. Knowing that SUMO functions as a "molecular glue" to facilitate protein-protein interactions, we propose that SUMOylation of Mfn1/2 may congress, glues, and confines damaged mitochondria to the perinuclear region thereby, protectively quarantining them from the heathy mitochondrial network until their removal via mitophagy in cells.
    Keywords:  26S Proteasome; Autophagy; Mitochondria; Mitofusin; Mitophagy; Small Ubiquitin Modifier (SUMO); ubiquitin
    DOI:  https://doi.org/10.1016/j.bbadis.2021.166104
  38. Front Oncol. 2020 ;10 619727
      Cancer progression involves a variety of pro-tumorigenic biological processes including cell proliferation, migration, invasion, and survival. A cellular pathway implicated in these pro-tumorigenic processes is autophagy, a catabolic route used for recycling of cytoplasmic components to generate macromolecular building blocks and energy, under stress conditions, to remove damaged cellular constituents to adapt to changing nutrient conditions and to maintain cellular homeostasis. During autophagy, cells form a double-membrane sequestering a compartment termed the phagophore, which matures into an autophagosome. Following fusion with the lysosome, the cargo is degraded inside the autolysosomes and the resulting macromolecules released back into the cytosol for reuse. Cancer cells use this recycling system during cancer progression, however the key autophagy players involved in this disease is unclear. Accumulative evidences show that autophagy receptors, crucial players for selective autophagy, are overexpressed during cancer progression, yet the mechanisms whereby pro-tumorigenic biological processes are modulated by these receptors remains unknown. In this review, we summarized the most important findings related with the pro-tumorigenic role of autophagy receptors p62/SQSTM1, NBR1, NDP52, and OPTN in cancer progression. In addition, we showed the most relevant cargos degraded by these receptors that have been shown to function as critical regulators of pro-tumorigenic processes. Finally, we discussed the role of autophagy receptors in the context of the cellular pathways implicated in this disease, such as growth factors signaling, oxidative stress response and apoptosis. In summary, we highlight that autophagy receptors should be considered important players of cancer progression, which could offer a niche for the development of novel diagnosis and cancer treatment strategies.
    Keywords:  aggressiveness; autophagy; autophagy receptors; cancer progression; metastasis
    DOI:  https://doi.org/10.3389/fonc.2020.619727
  39. Plant Cell. 2021 Feb 22. pii: koab062. [Epub ahead of print]
      Oxygen deprivation caused by flooding activates acclimation responses to stress and restricts plant growth. After experiencing flooding stress, plants must restore normal growth; however, which genes are dynamically and precisely controlled by flooding stress remains largely unknown. Here, we show that the Arabidopsis thaliana ubiquitin E3 ligase SUBMERGENCE RESISTANT1 (SR1) regulates the stability of the transcription factor WRKY33 to modulate the submergence response. SR1 physically interacts with WRKY33 in vivo and in vitro and controls its ubiquitination and proteasomal degradation. Both the sr1 mutant and WRKY33 overexpressors exhibited enhanced submergence tolerance and enhanced expression of hypoxia-responsive genes. Genetic experiments showed that WRKY33 functions downstream of SR1 during the submergence response. Submergence induced the phosphorylation of WRKY33, which enhanced the activation of RAP2.2, a positive regulator of hypoxia-response genes. Phosphorylated WRKY33 and RAP2.2 were degraded by SR1 and the N-degron pathway during reoxygenation, respectively. Taken together, our findings reveal that the on-and-off module SR1-WRKY33-RAP2.2 is connected to the well-known N-degron pathway to regulate acclimation to submergence in Arabidopsis. These two different but related modulation cascades precisely balance submergence acclimation with normal plant growth.
    DOI:  https://doi.org/10.1093/plcell/koab062
  40. Mol Cell Proteomics. 2021 Feb 19. pii: S1535-9476(21)00039-6. [Epub ahead of print] 100066
      Plague is a zoonotic disease that primarily infects rodents via fleabite. Transmission from flea to host niches requires rapid adaption of Yersinia pestis to the outer environments to establish infection. Here, quantitative proteome and secretome analysis of Y. pestis grown under conditions mimicking the two typical niches, i.e. the mammalian host (Mh) and the flea vector (Fv), were performed to understand the adaption strategies of this deadly pathogen. A secretome of Y. pestis containing 308 proteins has been identified using TMT-labeling mass spectrometry analysis. Although some proteins are known to be secreted, such as the type III secretion substrates, PsaA and F1 antigen, most of them were found to be secretory proteins for the first time. Comparative proteomic analysis showed membrane proteins, chaperonins and stress response proteins are significantly up-regulated under the Mh condition, among which the previously uncharacterized proteins YP_3416∼YP_3418 are remarkable because they cannot only be secreted but also translocated into HeLa cells by Y. pestis. We further demonstrated that the purified YP_3416 and YP_3418 exhibited E3 ubiquitin ligase activity in in vitro ubiquitination assay and yp_3416∼3418 deletion mutant of Y. pestis showed significant virulence attenuation in mice. Taken together, our results represent the first Y. pestis secretome, which will promote the better understanding of Y. pestis pathogenesis, as well as the development of new strategies for treatment and prevention of plague.
    Keywords:  E3 ubiquitin ligases; Yersinia pestis; comparative proteomics; secretome; virulence
    DOI:  https://doi.org/10.1016/j.mcpro.2021.100066
  41. Curr Biol. 2021 Feb 17. pii: S0960-9822(21)00146-9. [Epub ahead of print]
      Mutations in WDR45 and WDR45B cause the human neurological diseases β-propeller protein-associated neurodegeneration (BPAN) and intellectual disability (ID), respectively. WDR45 and WDR45B, along with WIPI1 and WIPI2, belong to a WD40 repeat-containing phosphatidylinositol-3-phosphate (PI(3)P)-binding protein family. Their yeast homolog Atg18 forms a complex with Atg2 and is required for autophagosome formation in part by tethering isolation membranes (IMs) (autophagosome precursor) to the endoplasmic reticulum (ER) to supply lipid for IM expansion in the autophagy pathway. The exact functions of WDR45/45B are unclear. We show here that WDR45/45B are specifically required for neural autophagy. In Wdr45/45b-depleted cells, the size of autophagosomes is decreased, and this is rescued by overexpression of ATG2A, providing in vivo evidence for the lipid transfer activity of ATG2-WIPI complexes. WDR45/45B are dispensable for the closure of autophagosomes but essential for the progression of autophagosomes into autolysosomes. WDR45/45B interact with the tether protein EPG5 and target it to late endosomes/lysosomes to promote autophagosome maturation. In the absence of Wdr45/45b, formation of the fusion machinery, consisting of SNARE proteins and EPG5, is dampened. BPAN- and ID-related mutations of WDR45/45B fail to rescue the autophagy defects in Wdr45/45b-deficient cells, possibly due to their impaired binding to EPG5. Promoting autophagosome maturation by inhibiting O-GlcNAcylation increases SNARE complex formation and facilitates the fusion of autophagosomes with late endosomes/lysosomes in Wdr45/45b double knockout (DKO) cells. Thus, our results uncover a novel function of WDR45/45B in autophagosome-lysosome fusion and provide molecular insights into the development of WDR45/WDR45B mutation-associated diseases.
    Keywords:  BPAN; ID; WDR45; WDR45B; autophagy
    DOI:  https://doi.org/10.1016/j.cub.2021.01.081
  42. J Biol Chem. 2021 Feb 19. pii: S0021-9258(21)00223-4. [Epub ahead of print] 100450
      Proteasome-mediated substrate degradation is an essential process that relies on the coordinated actions of ubiquitin (Ub), shuttle proteins containing Ub-like (UBL) domains, and the proteasome. Proteinaceous substrates are tagged with polyUb and shuttle proteins and these signals are then recognized by the proteasome, which subsequently degrades the substrate. To date, three proteasomal receptors have been identified, as well as multiple shuttle proteins and numerous types of polyUb chains that signal for degradation. While the components of this pathway are well-known, our understanding of their interplay is unclear - especially in the context of Rpn1, the largest proteasomal subunit. Here, using nuclear magnetic resonance (NMR) spectroscopy in combination with competition assays, we show that Rpn1 associates with UBL-containing proteins and polyUb chains, while exhibiting a preference for shuttle protein Rad23. Rpn1 appears to contain multiple Ub/UBL-binding sites, theoretically as many as one for each of its hallmark proteasome/cyclosome repeats. Remarkably, we also find that binding sites on Rpn1 can be shared among Ub and UBL species, while proteasomal receptors Rpn1 and Rpn10 can compete with each other for binding of shuttle protein Dsk2. Taken together, our results rule out the possibility of exclusive recognition sites on Rpn1 for individual Ub/UBL signals and further emphasize the complexity of the redundancy-laden proteasomal degradation pathway.
    Keywords:  Rpn1; UBA domain; UBL domain; proteasome; ubiquitin
    DOI:  https://doi.org/10.1016/j.jbc.2021.100450
  43. bioRxiv. 2021 Feb 16. pii: 2021.02.16.431318. [Epub ahead of print]
      The SARS-CoV-2 virus causes severe acute respiratory syndrome (COVID-19) and has rapidly created a global pandemic. Patients that survive may face a slow recovery with long lasting side effects that can afflict different organs. SARS-CoV-2 primarily infects epithelial airway cells that express the host entry receptor Angiotensin Converting Enzyme 2 (ACE2) which binds to spike protein trimers on the surface of SARS-CoV-2 virions. However, SARS-CoV-2 can spread to other tissues even though they are negative for ACE2. To gain insight into the molecular constituents that might influence SARS-CoV-2 tropism, we determined which additional host factors engage with the viral spike protein in disease-relevant human bronchial epithelial cells (16HBEo - ). We found that spike recruited the extracellular proteins laminin and thrombospondin and was retained in the endoplasmatic reticulum (ER) by the proteins DJB11 and FBX2 which support re-folding or degradation of nascent proteins in the ER. Because emerging mutations of the spike protein potentially impact the virus tropism, we compared the interactome of D614 spike with that of the rapidly spreading G614 mutated spike. More D614 than G614 spike associated with the proteins UGGT1, calnexin, HSP7A and GRP78/BiP which ensure glycosylation and folding of proteins in the ER. In contrast to G614 spike, D614 spike was endoproteolytically cleaved, and the N-terminal S1 domain was degraded in the ER even though C-terminal 'S2 only' proteoforms remained present. D614 spike also bound more laminin than G614 spike, which suggested that extracellular laminins may function as co-factor for an alternative, 'S2 only' dependent virus entry. Because the host interactome determines whether an infection is productive, we developed a novel proteome-based cell type set enrichment analysis (pCtSEA). With pCtSEA we determined that the host interactome of the spike protein may extend the tropism of SARS-CoV-2 beyond mucous epithelia to several different cell types, including macrophages and epithelial cells in the nephron. An 'S2 only' dependent, alternative infection of additional cell types with SARS-CoV-2 may impact vaccination strategies and may provide a molecular explanation for a severe or prolonged progression of disease in select COVID-19 patients.
    DOI:  https://doi.org/10.1101/2021.02.16.431318
  44. Cell Chem Biol. 2021 Feb 07. pii: S2451-9456(21)00043-X. [Epub ahead of print]
      There is a critical need to develop small-molecule inhibitors of mucin-type O-linked glycosylation. The best-known reagent currently is benzyl-GalNAc, but it is effective only at millimolar concentrations. This article demonstrates that Ac5GalNTGc, a peracetylated C-2 sulfhydryl-substituted GalNAc, fulfills this unmet need. When added to cultured leukocytes, breast cells, and prostate cells, Ac5GalNTGc increased cell-surface VVA binding by ∼10-fold, indicating truncation of O-glycan biosynthesis. Cytometry, mass spectrometry, and western blot analysis of HL-60 promyelocytes demonstrated that 50-80 μM Ac5GalNTGc prevented elaboration of 30%-60% of the O-glycans beyond the Tn-antigen (GalNAcα1-Ser/Thr) stage. The effect of the compound on N-glycans and glycosphingolipids was small. Glycan inhibition induced by Ac5GalNTGc resulted in 50%-80% reduction in leukocyte sialyl-Lewis X expression and L-/P-selectin-mediated rolling under flow conditions. Ac5GalNTGc was pharmacologically active in mouse. It reduced neutrophil infiltration to sites of inflammation by ∼60%. Overall, Ac5GalNTGc may find diverse applications as a potent inhibitor of O-glycosylation.
    Keywords:  O-glycan; cell adhesion; glycosylation; inflammation; inhibitor; mucin; neutrophil; selectin; sialyl-Lewis X; small molecule
    DOI:  https://doi.org/10.1016/j.chembiol.2021.01.017
  45. Autophagy. 2021 Feb 08. 1-382
    Daniel J Klionsky, Amal Kamal Abdel-Aziz, Sara Abdelfatah, Mahmoud Abdellatif, Asghar Abdoli, Steffen Abel, Hagai Abeliovich, Marie H Abildgaard, Yakubu Princely Abudu, Abraham Acevedo-Arozena, Iannis E Adamopoulos, Khosrow Adeli, Timon E Adolph, Annagrazia Adornetto, Elma Aflaki, Galila Agam, Anupam Agarwal, Bharat B Aggarwal, Maria Agnello, Patrizia Agostinis, Javed N Agrewala, Alexander Agrotis, Patricia V Aguilar, S Tariq Ahmad, Zubair M Ahmed, Ulises Ahumada-Castro, Sonja Aits, Shu Aizawa, Yunus Akkoc, Tonia Akoumianaki, Hafize Aysin Akpinar, Ahmed M Al-Abd, Lina Al-Akra, Abeer Al-Gharaibeh, Moulay A Alaoui-Jamali, Simon Alberti, Elísabet Alcocer-Gómez, Cristiano Alessandri, Muhammad Ali, M Abdul Alim Al-Bari, Saeb Aliwaini, Javad Alizadeh, Eugènia Almacellas, Alexandru Almasan, Alicia Alonso, Guillermo D Alonso, Nihal Altan-Bonnet, Dario C Altieri, Élida M C Álvarez, Sara Alves, Cristine Alves da Costa, Mazen M Alzaharna, Marialaura Amadio, Consuelo Amantini, Cristina Amaral, Susanna Ambrosio, Amal O Amer, Veena Ammanathan, Zhenyi An, Stig U Andersen, Shaida A Andrabi, Magaiver Andrade-Silva, Allen M Andres, Sabrina Angelini, David Ann, Uche C Anozie, Mohammad Y Ansari, Pedro Antas, Adam Antebi, Zuriñe Antón, Tahira Anwar, Lionel Apetoh, Nadezda Apostolova, Toshiyuki Araki, Yasuhiro Araki, Kohei Arasaki, Wagner L Araújo, Jun Araya, Catherine Arden, Maria-Angeles Arévalo, Sandro Arguelles, Esperanza Arias, Jyothi Arikkath, Hirokazu Arimoto, Aileen R Ariosa, Darius Armstrong-James, Laetitia Arnauné-Pelloquin, Angeles Aroca, Daniela S Arroyo, Ivica Arsov, Rubén Artero, Dalia Maria Lucia Asaro, Michael Aschner, Milad Ashrafizadeh, Osnat Ashur-Fabian, Atanas G Atanasov, Alicia K Au, Patrick Auberger, Holger W Auner, Laure Aurelian, Riccardo Autelli, Laura Avagliano, Yenniffer Ávalos, Sanja Aveic, Célia Alexandra Aveleira, Tamar Avin-Wittenberg, Yucel Aydin, Scott Ayton, Srinivas Ayyadevara, Maria Azzopardi, Misuzu Baba, Jonathan M Backer, Steven K Backues, Dong-Hun Bae, Ok-Nam Bae, Soo Han Bae, Eric H Baehrecke, Ahruem Baek, Seung-Hoon Baek, Sung Hee Baek, Giacinto Bagetta, Agnieszka Bagniewska-Zadworna, Hua Bai, Jie Bai, Xiyuan Bai, Yidong Bai, Nandadulal Bairagi, Shounak Baksi, Teresa Balbi, Cosima T Baldari, Walter Balduini, Andrea Ballabio, Maria Ballester, Salma Balazadeh, Rena Balzan, Rina Bandopadhyay, Sreeparna Banerjee, Sulagna Banerjee, Ágnes Bánréti, Yan Bao, Mauricio S Baptista, Alessandra Baracca, Cristiana Barbati, Ariadna Bargiela, Daniela Barilà, Peter G Barlow, Sami J Barmada, Esther Barreiro, George E Barreto, Jiri Bartek, Bonnie Bartel, Alberto Bartolome, Gaurav R Barve, Suresh H Basagoudanavar, Diane C Bassham, Robert C Bast, Alakananda Basu, Henri Batoko, Isabella Batten, Etienne E Baulieu, Bradley L Baumgarner, Jagadeesh Bayry, Rupert Beale, Isabelle Beau, Florian Beaumatin, Luiz R G Bechara, George R Beck, Michael F Beers, Jakob Begun, Christian Behrends, Georg M N Behrens, Roberto Bei, Eloy Bejarano, Shai Bel, Christian Behl, Amine Belaid, Naïma Belgareh-Touzé, Cristina Bellarosa, Francesca Belleudi, Melissa Belló Pérez, Raquel Bello-Morales, Jackeline Soares de Oliveira Beltran, Sebastián Beltran, Doris Mangiaracina Benbrook, Mykolas Bendorius, Bruno A Benitez, Irene Benito-Cuesta, Julien Bensalem, Martin W Berchtold, Sabina Berezowska, Daniele Bergamaschi, Matteo Bergami, Andreas Bergmann, Laura Berliocchi, Clarisse Berlioz-Torrent, Amélie Bernard, Lionel Berthoux, Cagri G Besirli, Sebastien Besteiro, Virginie M Betin, Rudi Beyaert, Jelena S Bezbradica, Kiran Bhaskar, Ingrid Bhatia-Kissova, Resham Bhattacharya, Sujoy Bhattacharya, Shalmoli Bhattacharyya, Md Shenuarin Bhuiyan, Sujit Kumar Bhutia, Lanrong Bi, Xiaolin Bi, Trevor J Biden, Krikor Bijian, Viktor A Billes, Nadine Binart, Claudia Bincoletto, Asa B Birgisdottir, Geir Bjorkoy, Gonzalo Blanco, Ana Blas-Garcia, Janusz Blasiak, Robert Blomgran, Klas Blomgren, Janice S Blum, Emilio Boada-Romero, Mirta Boban, Kathleen Boesze-Battaglia, Philippe Boeuf, Barry Boland, Pascale Bomont, Paolo Bonaldo, Srinivasa Reddy Bonam, Laura Bonfili, Juan S Bonifacino, Brian A Boone, Martin D Bootman, Matteo Bordi, Christoph Borner, Beat C Bornhauser, Gautam Borthakur, Jürgen Bosch, Santanu Bose, Luis M Botana, Juan Botas, Chantal M Boulanger, Michael E Boulton, Mathieu Bourdenx, Benjamin Bourgeois, Nollaig M Bourke, Guilhem Bousquet, Patricia Boya, Peter V Bozhkov, Luiz H M Bozi, Tolga O Bozkurt, Doug E Brackney, Christian H Brandts, Ralf J Braun, Gerhard H Braus, Roberto Bravo-Sagua, José M Bravo-San Pedro, Patrick Brest, Marie-Agnès Bringer, Alfredo Briones-Herrera, V Courtney Broaddus, Peter Brodersen, Jeffrey L Brodsky, Steven L Brody, Paola G Bronson, Jeff M Bronstein, Carolyn N Brown, Rhoderick E Brown, Patricia C Brum, John H Brumell, Nicola Brunetti-Pierri, Daniele Bruno, Robert J Bryson-Richardson, Cecilia Bucci, Carmen Buchrieser, Marta Bueno, Laura Elisa Buitrago-Molina, Simone Buraschi, Shilpa Buch, J Ross Buchan, Erin M Buckingham, Hikmet Budak, Mauricio Budini, Geert Bultynck, Florin Burada, Joseph R Burgoyne, M Isabel Burón, Victor Bustos, Sabrina Büttner, Elena Butturini, Aaron Byrd, Isabel Cabas, Sandra Cabrera-Benitez, Ken Cadwell, Jingjing Cai, Lu Cai, Qian Cai, Montserrat Cairó, Jose A Calbet, Guy A Caldwell, Kim A Caldwell, Jarrod A Call, Riccardo Calvani, Ana C Calvo, Miguel Calvo-Rubio Barrera, Niels Os Camara, Jacques H Camonis, Nadine Camougrand, Michelangelo Campanella, Edward M Campbell, François-Xavier Campbell-Valois, Silvia Campello, Ilaria Campesi, Juliane C Campos, Olivier Camuzard, Jorge Cancino, Danilo Candido de Almeida, Laura Canesi, Isabella Caniggia, Barbara Canonico, Carles Cantí, Bin Cao, Michele Caraglia, Beatriz Caramés, Evie H Carchman, Elena Cardenal-Muñoz, Cesar Cardenas, Luis Cardenas, Sandra M Cardoso, Jennifer S Carew, Georges F Carle, Gillian Carleton, Silvia Carloni, Didac Carmona-Gutierrez, Leticia A Carneiro, Oliana Carnevali, Julian M Carosi, Serena Carra, Alice Carrier, Lucie Carrier, Bernadette Carroll, A Brent Carter, Andreia Neves Carvalho, Magali Casanova, Caty Casas, Josefina Casas, Chiara Cassioli, Eliseo F Castillo, Karen Castillo, Sonia Castillo-Lluva, Francesca Castoldi, Marco Castori, Ariel F Castro, Margarida Castro-Caldas, Javier Castro-Hernandez, Susana Castro-Obregon, Sergio D Catz, Claudia Cavadas, Federica Cavaliere, Gabriella Cavallini, Maria Cavinato, Maria L Cayuela, Paula Cebollada Rica, Valentina Cecarini, Francesco Cecconi, Marzanna Cechowska-Pasko, Simone Cenci, Victòria Ceperuelo-Mallafré, João J Cerqueira, Janete M Cerutti, Davide Cervia, Vildan Bozok Cetintas, Silvia Cetrullo, Han-Jung Chae, Andrei S Chagin, Chee-Yin Chai, Gopal Chakrabarti, Oishee Chakrabarti, Tapas Chakraborty, Trinad Chakraborty, Mounia Chami, Georgios Chamilos, David W Chan, Edmond Y W Chan, Edward D Chan, H Y Edwin Chan, Helen H Chan, Hung Chan, Matthew T V Chan, Yau Sang Chan, Partha K Chandra, Chih-Peng Chang, Chunmei Chang, Hao-Chun Chang, Kai Chang, Jie Chao, Tracey Chapman, Nicolas Charlet-Berguerand, Samrat Chatterjee, Shail K Chaube, Anu Chaudhary, Santosh Chauhan, Edward Chaum, Frédéric Checler, Michael E Cheetham, Chang-Shi Chen, Guang-Chao Chen, Jian-Fu Chen, Liam L Chen, Leilei Chen, Lin Chen, Mingliang Chen, Mu-Kuan Chen, Ning Chen, Quan Chen, Ruey-Hwa Chen, Shi Chen, Wei Chen, Weiqiang Chen, Xin-Ming Chen, Xiong-Wen Chen, Xu Chen, Yan Chen, Ye-Guang Chen, Yingyu Chen, Yongqiang Chen, Yu-Jen Chen, Yue-Qin Chen, Zhefan Stephen Chen, Zhi Chen, Zhi-Hua Chen, Zhijian J Chen, Zhixiang Chen, Hanhua Cheng, Jun Cheng, Shi-Yuan Cheng, Wei Cheng, Xiaodong Cheng, Xiu-Tang Cheng, Yiyun Cheng, Zhiyong Cheng, Zhong Chen, Heesun Cheong, Jit Kong Cheong, Boris V Chernyak, Sara Cherry, Chi Fai Randy Cheung, Chun Hei Antonio Cheung, King-Ho Cheung, Eric Chevet, Richard J Chi, Alan Kwok Shing Chiang, Ferdinando Chiaradonna, Roberto Chiarelli, Mario Chiariello, Nathalia Chica, Susanna Chiocca, Mario Chiong, Shih-Hwa Chiou, Abhilash I Chiramel, Valerio Chiurchiù, Dong-Hyung Cho, Seong-Kyu Choe, Augustine M K Choi, Mary E Choi, Kamalika Roy Choudhury, Norman S Chow, Charleen T Chu, Jason P Chua, John Jia En Chua, Hyewon Chung, Kin Pan Chung, Seockhoon Chung, So-Hyang Chung, Yuen-Li Chung, Valentina Cianfanelli, Iwona A Ciechomska, Mariana Cifuentes, Laura Cinque, Sebahattin Cirak, Mara Cirone, Michael J Clague, Robert Clarke, Emilio Clementi, Eliana M Coccia, Patrice Codogno, Ehud Cohen, Mickael M Cohen, Tania Colasanti, Fiorella Colasuonno, Robert A Colbert, Anna Colell, Miodrag Čolić, Nuria S Coll, Mark O Collins, María I Colombo, Daniel A Colón-Ramos, Lydie Combaret, Sergio Comincini, Márcia R Cominetti, Antonella Consiglio, Andrea Conte, Fabrizio Conti, Viorica Raluca Contu, Mark R Cookson, Kevin M Coombs, Isabelle Coppens, Maria Tiziana Corasaniti, Dale P Corkery, Nils Cordes, Katia Cortese, Maria do Carmo Costa, Sarah Costantino, Paola Costelli, Ana Coto-Montes, Peter J Crack, Jose L Crespo, Alfredo Criollo, Valeria Crippa, Riccardo Cristofani, Tamas Csizmadia, Antonio Cuadrado, Bing Cui, Jun Cui, Yixian Cui, Yong Cui, Emmanuel Culetto, Andrea C Cumino, Andrey V Cybulsky, Mark J Czaja, Stanislaw J Czuczwar, Stefania D'Adamo, Marcello D'Amelio, Daniela D'Arcangelo, Andrew C D'Lugos, Gabriella D'Orazi, James A da Silva, Hormos Salimi Dafsari, Ruben K Dagda, Yasin Dagdas, Maria Daglia, Xiaoxia Dai, Yun Dai, Yuyuan Dai, Jessica Dal Col, Paul Dalhaimer, Luisa Dalla Valle, Tobias Dallenga, Guillaume Dalmasso, Markus Damme, Ilaria Dando, Nico P Dantuma, April L Darling, Hiranmoy Das, Srinivasan Dasarathy, Santosh K Dasari, Srikanta Dash, Oliver Daumke, Adrian N Dauphinee, Jeffrey S Davies, Valeria A Dávila, Roger J Davis, Tanja Davis, Sharadha Dayalan Naidu, Francesca De Amicis, Karolien De Bosscher, Francesca De Felice, Lucia De Franceschi, Chiara De Leonibus, Mayara G de Mattos Barbosa, Guido R Y De Meyer, Angelo De Milito, Cosimo De Nunzio, Clara De Palma, Mauro De Santi, Claudio De Virgilio, Daniela De Zio, Jayanta Debnath, Brian J DeBosch, Jean-Paul Decuypere, Mark A Deehan, Gianluca Deflorian, James DeGregori, Benjamin Dehay, Gabriel Del Rio, Joe R Delaney, Lea M D Delbridge, Elizabeth Delorme-Axford, M Victoria Delpino, Francesca Demarchi, Vilma Dembitz, Nicholas D Demers, Hongbin Deng, Zhiqiang Deng, Joern Dengjel, Paul Dent, Donna Denton, Melvin L DePamphilis, Channing J Der, Vojo Deretic, Albert Descoteaux, Laura Devis, Sushil Devkota, Olivier Devuyst, Grant Dewson, Mahendiran Dharmasivam, Rohan Dhiman, Diego di Bernardo, Manlio Di Cristina, Fabio Di Domenico, Pietro Di Fazio, Alessio Di Fonzo, Giovanni Di Guardo, Gianni M Di Guglielmo, Luca Di Leo, Chiara Di Malta, Alessia Di Nardo, Martina Di Rienzo, Federica Di Sano, George Diallinas, Jiajie Diao, Guillermo Diaz-Araya, Inés Díaz-Laviada, Jared M Dickinson, Marc Diederich, Mélanie Dieudé, Ivan Dikic, Shiping Ding, Wen-Xing Ding, Luciana Dini, Jelena Dinić, Miroslav Dinic, Albena T Dinkova-Kostova, Marc S Dionne, Jörg H W Distler, Abhinav Diwan, Ian M C Dixon, Mojgan Djavaheri-Mergny, Ina Dobrinski, Oxana Dobrovinskaya, Radek Dobrowolski, Renwick C J Dobson, Jelena Đokić, Serap Dokmeci Emre, Massimo Donadelli, Bo Dong, Xiaonan Dong, Zhiwu Dong, Gerald W Dorn Ii, Volker Dotsch, Huan Dou, Juan Dou, Moataz Dowaidar, Sami Dridi, Liat Drucker, Ailian Du, Caigan Du, Guangwei Du, Hai-Ning Du, Li-Lin Du, André du Toit, Shao-Bin Duan, Xiaoqiong Duan, Sónia P Duarte, Anna Dubrovska, Elaine A Dunlop, Nicolas Dupont, Raúl V Durán, Bilikere S Dwarakanath, Sergey A Dyshlovoy, Darius Ebrahimi-Fakhari, Leopold Eckhart, Charles L Edelstein, Thomas Efferth, Eftekhar Eftekharpour, Ludwig Eichinger, Nabil Eid, Tobias Eisenberg, N Tony Eissa, Sanaa Eissa, Miriam Ejarque, Abdeljabar El Andaloussi, Nazira El-Hage, Shahenda El-Naggar, Anna Maria Eleuteri, Eman S El-Shafey, Mohamed Elgendy, Aristides G Eliopoulos, María M Elizalde, Philip M Elks, Hans-Peter Elsasser, Eslam S Elsherbiny, Brooke M Emerling, N C Tolga Emre, Christina H Eng, Nikolai Engedal, Anna-Mart Engelbrecht, Agnete S T Engelsen, Jorrit M Enserink, Ricardo Escalante, Audrey Esclatine, Mafalda Escobar-Henriques, Eeva-Liisa Eskelinen, Lucile Espert, Makandjou-Ola Eusebio, Gemma Fabrias, Cinzia Fabrizi, Antonio Facchiano, Francesco Facchiano, Bengt Fadeel, Claudio Fader, Alex C Faesen, W Douglas Fairlie, Alberto Falcó, Bjorn H Falkenburger, Daping Fan, Jie Fan, Yanbo Fan, Evandro F Fang, Yanshan Fang, Yognqi Fang, Manolis Fanto, Tamar Farfel-Becker, Mathias Faure, Gholamreza Fazeli, Anthony O Fedele, Arthur M Feldman, Du Feng, Jiachun Feng, Lifeng Feng, Yibin Feng, Yuchen Feng, Wei Feng, Thais Fenz Araujo, Thomas A Ferguson, Álvaro F Fernández, Jose C Fernandez-Checa, Sonia Fernández-Veledo, Alisdair R Fernie, Anthony W Ferrante, Alessandra Ferraresi, Merari F Ferrari, Julio C B Ferreira, Susan Ferro-Novick, Antonio Figueras, Riccardo Filadi, Nicoletta Filigheddu, Eduardo Filippi-Chiela, Giuseppe Filomeni, Gian Maria Fimia, Vittorio Fineschi, Francesca Finetti, Steven Finkbeiner, Edward A Fisher, Paul B Fisher, Flavio Flamigni, Steven J Fliesler, Trude H Flo, Ida Florance, Oliver Florey, Tullio Florio, Erika Fodor, Carlo Follo, Edward A Fon, Antonella Forlino, Francesco Fornai, Paola Fortini, Anna Fracassi, Alessandro Fraldi, Brunella Franco, Rodrigo Franco, Flavia Franconi, Lisa B Frankel, Scott L Friedman, Leopold F Fröhlich, Gema Frühbeck, Jose M Fuentes, Yukio Fujiki, Naonobu Fujita, Yuuki Fujiwara, Mitsunori Fukuda, Simone Fulda, Luc Furic, Norihiko Furuya, Carmela Fusco, Michaela U Gack, Lidia Gaffke, Sehamuddin Galadari, Alessia Galasso, Maria F Galindo, Sachith Gallolu Kankanamalage, Lorenzo Galluzzi, Vincent Galy, Noor Gammoh, Boyi Gan, Ian G Ganley, Feng Gao, Hui Gao, Minghui Gao, Ping Gao, Shou-Jiang Gao, Wentao Gao, Xiaobo Gao, Ana Garcera, Maria Noé Garcia, Verónica E Garcia, Francisco García-Del Portillo, Vega Garcia-Escudero, Aracely Garcia-Garcia, Marina Garcia-Macia, Diana García-Moreno, Carmen Garcia-Ruiz, Patricia García-Sanz, Abhishek D Garg, Ricardo Gargini, Tina Garofalo, Robert F Garry, Nils C Gassen, Damian Gatica, Liang Ge, Wanzhong Ge, Ruth Geiss-Friedlander, Cecilia Gelfi, Pascal Genschik, Ian E Gentle, Valeria Gerbino, Christoph Gerhardt, Kyla Germain, Marc Germain, David A Gewirtz, Elham Ghasemipour Afshar, Saeid Ghavami, Alessandra Ghigo, Manosij Ghosh, Georgios Giamas, Claudia Giampietri, Alexandra Giatromanolaki, Gary E Gibson, Spencer B Gibson, Vanessa Ginet, Edward Giniger, Carlotta Giorgi, Henrique Girao, Stephen E Girardin, Mridhula Giridharan, Sandy Giuliano, Cecilia Giulivi, Sylvie Giuriato, Julien Giustiniani, Alexander Gluschko, Veit Goder, Alexander Goginashvili, Jakub Golab, David C Goldstone, Anna Golebiewska, Luciana R Gomes, Rodrigo Gomez, Rubén Gómez-Sánchez, Maria Catalina Gomez-Puerto, Raquel Gomez-Sintes, Qingqiu Gong, Felix M Goni, Javier González-Gallego, Tomas Gonzalez-Hernandez, Rosa A Gonzalez-Polo, Jose A Gonzalez-Reyes, Patricia González-Rodríguez, Ing Swie Goping, Marina S Gorbatyuk, Nikolai V Gorbunov, Kıvanç Görgülü, Roxana M Gorojod, Sharon M Gorski, Sandro Goruppi, Cecilia Gotor, Roberta A Gottlieb, Illana Gozes, Devrim Gozuacik, Martin Graef, Markus H Gräler, Veronica Granatiero, Daniel Grasso, Joshua P Gray, Douglas R Green, Alexander Greenhough, Stephen L Gregory, Edward F Griffin, Mark W Grinstaff, Frederic Gros, Charles Grose, Angelina S Gross, Florian Gruber, Paolo Grumati, Tilman Grune, Xueyan Gu, Jun-Lin Guan, Carlos M Guardia, Kishore Guda, Flora Guerra, Consuelo Guerri, Prasun Guha, Carlos Guillén, Shashi Gujar, Anna Gukovskaya, Ilya Gukovsky, Jan Gunst, Andreas Günther, Anyonya R Guntur, Chuanyong Guo, Chun Guo, Hongqing Guo, Lian-Wang Guo, Ming Guo, Pawan Gupta, Shashi Kumar Gupta, Swapnil Gupta, Veer Bala Gupta, Vivek Gupta, Asa B Gustafsson, David D Gutterman, Ranjitha H B, Annakaisa Haapasalo, James E Haber, Aleksandra Hać, Shinji Hadano, Anders J Hafrén, Mansour Haidar, Belinda S Hall, Gunnel Halldén, Anne Hamacher-Brady, Andrea Hamann, Maho Hamasaki, Weidong Han, Malene Hansen, Phyllis I Hanson, Zijian Hao, Masaru Harada, Ljubica Harhaji-Trajkovic, Nirmala Hariharan, Nigil Haroon, James Harris, Takafumi Hasegawa, Noor Hasima Nagoor, Jeffrey A Haspel, Volker Haucke, Wayne D Hawkins, Bruce A Hay, Cole M Haynes, Soren B Hayrabedyan, Thomas S Hays, Congcong He, Qin He, Rong-Rong He, You-Wen He, Yu-Ying He, Yasser Heakal, Alexander M Heberle, J Fielding Hejtmancik, Gudmundur Vignir Helgason, Vanessa Henkel, Marc Herb, Alexander Hergovich, Anna Herman-Antosiewicz, Agustín Hernández, Carlos Hernandez, Sergio Hernandez-Diaz, Virginia Hernandez-Gea, Amaury Herpin, Judit Herreros, Javier H Hervás, Daniel Hesselson, Claudio Hetz, Volker T Heussler, Yujiro Higuchi, Sabine Hilfiker, Joseph A Hill, William S Hlavacek, Emmanuel A Ho, Idy H T Ho, Philip Wing-Lok Ho, Shu-Leong Ho, Wan Yun Ho, G Aaron Hobbs, Mark Hochstrasser, Peter H M Hoet, Daniel Hofius, Paul Hofman, Annika Höhn, Carina I Holmberg, Jose R Hombrebueno, Chang-Won Hong Yi-Ren Hong, Lora V Hooper, Thorsten Hoppe, Rastislav Horos, Yujin Hoshida, I-Lun Hsin, Hsin-Yun Hsu, Bing Hu, Dong Hu, Li-Fang Hu, Ming Chang Hu, Ronggui Hu, Wei Hu, Yu-Chen Hu, Zhuo-Wei Hu, Fang Hua, Jinlian Hua, Yingqi Hua, Chongmin Huan, Canhua Huang, Chuanshu Huang, Chuanxin Huang, Chunling Huang, Haishan Huang, Kun Huang, Michael L H Huang, Rui Huang, Shan Huang, Tianzhi Huang, Xing Huang, Yuxiang Jack Huang, Tobias B Huber, Virginie Hubert, Christian A Hubner, Stephanie M Hughes, William E Hughes, Magali Humbert, Gerhard Hummer, James H Hurley, Sabah Hussain, Salik Hussain, Patrick J Hussey, Martina Hutabarat, Hui-Yun Hwang, Seungmin Hwang, Antonio Ieni, Fumiyo Ikeda, Yusuke Imagawa, Yuzuru Imai, Carol Imbriano, Masaya Imoto, Denise M Inman, Ken Inoki, Juan Iovanna, Renato V Iozzo, Giuseppe Ippolito, Javier E Irazoqui, Pablo Iribarren, Mohd Ishaq, Makoto Ishikawa, Nestor Ishimwe, Ciro Isidoro, Nahed Ismail, Shohreh Issazadeh-Navikas, Eisuke Itakura, Daisuke Ito, Davor Ivankovic, Saška Ivanova, Anand Krishnan V Iyer, José M Izquierdo, Masanori Izumi, Marja Jäättelä, Majid Sakhi Jabir, William T Jackson, Nadia Jacobo-Herrera, Anne-Claire Jacomin, Elise Jacquin, Pooja Jadiya, Hartmut Jaeschke, Chinnaswamy Jagannath, Arjen J Jakobi, Johan Jakobsson, Bassam Janji, Pidder Jansen-Dürr, Patric J Jansson, Jonathan Jantsch, Sławomir Januszewski, Alagie Jassey, Steve Jean, Hélène Jeltsch-David, Pavla Jendelova, Andreas Jenny, Thomas E Jensen, Niels Jessen, Jenna L Jewell, Jing Ji, Lijun Jia, Rui Jia, Liwen Jiang, Qing Jiang, Richeng Jiang, Teng Jiang, Xuejun Jiang, Yu Jiang, Maria Jimenez-Sanchez, Eun-Jung Jin, Fengyan Jin, Hongchuan Jin, Li Jin, Luqi Jin, Meiyan Jin, Si Jin, Eun-Kyeong Jo, Carine Joffre, Terje Johansen, Gail V W Johnson, Simon A Johnston, Eija Jokitalo, Mohit Kumar Jolly, Leo A B Joosten, Joaquin Jordan, Bertrand Joseph, Dianwen Ju, Jeong-Sun Ju, Jingfang Ju, Esmeralda Juárez, Delphine Judith, Gábor Juhász, Youngsoo Jun, Chang Hwa Jung, Sung-Chul Jung, Yong Keun Jung, Heinz Jungbluth, Johannes Jungverdorben, Steffen Just, Kai Kaarniranta, Allen Kaasik, Tomohiro Kabuta, Daniel Kaganovich, Alon Kahana, Renate Kain, Shinjo Kajimura, Maria Kalamvoki, Manjula Kalia, Danuta S Kalinowski, Nina Kaludercic, Ioanna Kalvari, Joanna Kaminska, Vitaliy O Kaminskyy, Hiromitsu Kanamori, Keizo Kanasaki, Chanhee Kang, Rui Kang, Sang Sun Kang, Senthilvelrajan Kaniyappan, Tomotake Kanki, Thirumala-Devi Kanneganti, Anumantha G Kanthasamy, Arthi Kanthasamy, Marc Kantorow, Orsolya Kapuy, Michalis V Karamouzis, Md Razaul Karim, Parimal Karmakar, Rajesh G Katare, Masaru Kato, Stefan H E Kaufmann, Anu Kauppinen, Gur P Kaushal, Susmita Kaushik, Kiyoshi Kawasaki, Kemal Kazan, Po-Yuan Ke, Damien J Keating, Ursula Keber, John H Kehrl, Kate E Keller, Christian W Keller, Jongsook Kim Kemper, Candia M Kenific, Oliver Kepp, Stephanie Kermorgant, Andreas Kern, Robin Ketteler, Tom G Keulers, Boris Khalfin, Hany Khalil, Bilon Khambu, Shahid Y Khan, Vinoth Kumar Megraj Khandelwal, Rekha Khandia, Widuri Kho, Noopur V Khobrekar, Sataree Khuansuwan, Mukhran Khundadze, Samuel A Killackey, Dasol Kim, Deok Ryong Kim, Do-Hyung Kim, Dong-Eun Kim, Eun Young Kim, Eun-Kyoung Kim, Hak-Rim Kim, Hee-Sik Kim, Hyung-Ryong Kim, Jeong Hun Kim, Jin Kyung Kim, Jin-Hoi Kim, Joungmok Kim, Ju Hwan Kim, Keun Il Kim, Peter K Kim, Seong-Jun Kim, Scot R Kimball, Adi Kimchi, Alec C Kimmelman, Tomonori Kimura, Matthew A King, Kerri J Kinghorn, Conan G Kinsey, Vladimir Kirkin, Lorrie A Kirshenbaum, Sergey L Kiselev, Shuji Kishi, Katsuhiko Kitamoto, Yasushi Kitaoka, Kaio Kitazato, Richard N Kitsis, Josef T Kittler, Ole Kjaerulff, Peter S Klein, Thomas Klopstock, Jochen Klucken, Helene Knævelsrud, Roland L Knorr, Ben C B Ko, Fred Ko, Jiunn-Liang Ko, Hotaka Kobayashi, Satoru Kobayashi, Ina Koch, Jan C Koch, Ulrich Koenig, Donat Kögel, Young Ho Koh, Masato Koike, Sepp D Kohlwein, Nur M Kocaturk, Masaaki Komatsu, Jeannette König, Toru Kono, Benjamin T Kopp, Tamas Korcsmaros, Gözde Korkmaz, Viktor I Korolchuk, Mónica Suárez Korsnes, Ali Koskela, Janaiah Kota, Yaichiro Kotake, Monica L Kotler, Yanjun Kou, Michael I Koukourakis, Evangelos Koustas, Attila L Kovacs, Tibor Kovács, Daisuke Koya, Tomohiro Kozako, Claudine Kraft, Dimitri Krainc, Helmut Krämer, Anna D Krasnodembskaya, Carole Kretz-Remy, Guido Kroemer, Nicholas T Ktistakis, Kazuyuki Kuchitsu, Sabine Kuenen, Lars Kuerschner, Thomas Kukar, Ajay Kumar, Ashok Kumar, Deepak Kumar, Dhiraj Kumar, Sharad Kumar, Shinji Kume, Caroline Kumsta, Chanakya N Kundu, Mondira Kundu, Ajaikumar B Kunnumakkara, Lukasz Kurgan, Tatiana G Kutateladze, Ozlem Kutlu, SeongAe Kwak, Ho Jeong Kwon, Taeg Kyu Kwon, Yong Tae Kwon, Irene Kyrmizi, Albert La Spada, Patrick Labonté, Sylvain Ladoire, Ilaria Laface, Frank Lafont, Diane C Lagace, Vikramjit Lahiri, Zhibing Lai, Angela S Laird, Aparna Lakkaraju, Trond Lamark, Sheng-Hui Lan, Ane Landajuela, Darius J R Lane, Jon D Lane, Charles H Lang, Carsten Lange, Ülo Langel, Rupert Langer, Pierre Lapaquette, Jocelyn Laporte, Nicholas F LaRusso, Isabel Lastres-Becker, Wilson Chun Yu Lau, Gordon W Laurie, Sergio Lavandero, Betty Yuen Kwan Law, Helen Ka-Wai Law, Rob Layfield, Weidong Le, Herve Le Stunff, Alexandre Y Leary, Jean-Jacques Lebrun, Lionel Y W Leck, Jean-Philippe Leduc-Gaudet, Changwook Lee, Chung-Pei Lee, Da-Hye Lee, Edward B Lee, Erinna F Lee, Gyun Min Lee, He-Jin Lee, Heung Kyu Lee, Jae Man Lee, Jason S Lee, Jin-A Lee, Joo-Yong Lee, Jun Hee Lee, Michael Lee, Min Goo Lee, Min Jae Lee, Myung-Shik Lee, Sang Yoon Lee, Seung-Jae Lee, Stella Y Lee, Sung Bae Lee, Won Hee Lee, Ying-Ray Lee, Yong-Ho Lee, Youngil Lee, Christophe Lefebvre, Renaud Legouis, Yu L Lei, Yuchen Lei, Sergey Leikin, Gerd Leitinger, Leticia Lemus, Shuilong Leng, Olivia Lenoir, Guido Lenz, Heinz Josef Lenz, Paola Lenzi, Yolanda León, Andréia M Leopoldino, Christoph Leschczyk, Stina Leskelä, Elisabeth Letellier, Chi-Ting Leung, Po Sing Leung, Jeremy S Leventhal, Beth Levine, Patrick A Lewis, Klaus Ley, Bin Li, Da-Qiang Li, Jianming Li, Jing Li, Jiong Li, Ke Li, Liwu Li, Mei Li, Min Li, Min Li, Ming Li, Mingchuan Li, Pin-Lan Li, Ming-Qing Li, Qing Li, Sheng Li, Tiangang Li, Wei Li, Wenming Li, Xue Li, Yi-Ping Li, Yuan Li, Zhiqiang Li, Zhiyong Li, Zhiyuan Li, Jiqin Lian, Chengyu Liang, Qiangrong Liang, Weicheng Liang, Yongheng Liang, YongTian Liang, Guanghong Liao, Lujian Liao, Mingzhi Liao, Yung-Feng Liao, Mariangela Librizzi, Pearl P Y Lie, Mary A Lilly, Hyunjung J Lim, Thania R R Lima, Federica Limana, Chao Lin, Chih-Wen Lin, Dar-Shong Lin, Fu-Cheng Lin, Jiandie D Lin, Kurt M Lin, Kwang-Huei Lin, Liang-Tzung Lin, Pei-Hui Lin, Qiong Lin, Shaofeng Lin, Su-Ju Lin, Wenyu Lin, Xueying Lin, Yao-Xin Lin, Yee-Shin Lin, Rafael Linden, Paula Lindner, Shuo-Chien Ling, Paul Lingor, Amelia K Linnemann, Yih-Cherng Liou, Marta M Lipinski, Saška Lipovšek, Vitor A Lira, Natalia Lisiak, Paloma B Liton, Chao Liu, Ching-Hsuan Liu, Chun-Feng Liu, Cui Hua Liu, Fang Liu, Hao Liu, Hsiao-Sheng Liu, Hua-Feng Liu, Huifang Liu, Jia Liu, Jing Liu, Julia Liu, Leyuan Liu, Longhua Liu, Meilian Liu, Qin Liu, Wei Liu, Wende Liu, Xiao-Hong Liu, Xiaodong Liu, Xingguo Liu, Xu Liu, Xuedong Liu, Yanfen Liu, Yang Liu, Yang Liu, Yueyang Liu, Yule Liu, J Andrew Livingston, Gerard Lizard, Jose M Lizcano, Senka Ljubojevic-Holzer, Matilde E LLeonart, David Llobet-Navàs, Alicia Llorente, Chih Hung Lo, Damián Lobato-Márquez, Qi Long, Yun Chau Long, Ben Loos, Julia A Loos, Manuela G López, Guillermo López-Doménech, José Antonio López-Guerrero, Ana T López-Jiménez, Óscar López-Pérez, Israel López-Valero, Magdalena J Lorenowicz, Mar Lorente, Peter Lorincz, Laura Lossi, Sophie Lotersztajn, Penny E Lovat, Jonathan F Lovell, Alenka Lovy, Péter Lőw, Guang Lu, Haocheng Lu, Jia-Hong Lu, Jin-Jian Lu, Mengji Lu, Shuyan Lu, Alessandro Luciani, John M Lucocq, Paula Ludovico, Micah A Luftig, Morten Luhr, Diego Luis-Ravelo, Julian J Lum, Liany Luna-Dulcey, Anders H Lund, Viktor K Lund, Jan D Lünemann, Patrick Lüningschrör, Honglin Luo, Rongcan Luo, Shouqing Luo, Zhi Luo, Claudio Luparello, Bernhard Lüscher, Luan Luu, Alex Lyakhovich, Konstantin G Lyamzaev, Alf Håkon Lystad, Lyubomyr Lytvynchuk, Alvin C Ma, Changle Ma, Mengxiao Ma, Ning-Fang Ma, Quan-Hong Ma, Xinliang Ma, Yueyun Ma, Zhenyi Ma, Ormond A MacDougald, Fernando Macian, Gustavo C MacIntosh, Jeffrey P MacKeigan, Kay F Macleod, Sandra Maday, Frank Madeo, Muniswamy Madesh, Tobias Madl, Julio Madrigal-Matute, Akiko Maeda, Yasuhiro Maejima, Marta Magarinos, Poornima Mahavadi, Emiliano Maiani, Kenneth Maiese, Panchanan Maiti, Maria Chiara Maiuri, Barbara Majello, Michael B Major, Elena Makareeva, Fayaz Malik, Karthik Mallilankaraman, Walter Malorni, Alina Maloyan, Najiba Mammadova, Gene Chi Wai Man, Federico Manai, Joseph D Mancias, Eva-Maria Mandelkow, Michael A Mandell, Angelo A Manfredi, Masoud H Manjili, Ravi Manjithaya, Patricio Manque, Bella B Manshian, Raquel Manzano, Claudia Manzoni, Kai Mao, Cinzia Marchese, Sandrine Marchetti, Anna Maria Marconi, Fabrizio Marcucci, Stefania Mardente, Olga A Mareninova, Marta Margeta, Muriel Mari, Sara Marinelli, Oliviero Marinelli, Guillermo Mariño, Sofia Mariotto, Richard S Marshall, Mark R Marten, Sascha Martens, Alexandre P J Martin, Katie R Martin, Sara Martin, Shaun Martin, Adrián Martín-Segura, Miguel A Martín-Acebes, Inmaculada Martin-Burriel, Marcos Martin-Rincon, Paloma Martin-Sanz, José A Martina, Wim Martinet, Aitor Martinez, Ana Martinez, Jennifer Martinez, Moises Martinez Velazquez, Nuria Martinez-Lopez, Marta Martinez-Vicente, Daniel O Martins, Joilson O Martins, Waleska K Martins, Tania Martins-Marques, Emanuele Marzetti, Shashank Masaldan, Celine Masclaux-Daubresse, Douglas G Mashek, Valentina Massa, Lourdes Massieu, Glenn R Masson, Laura Masuelli, Anatoliy I Masyuk, Tetyana V Masyuk, Paola Matarrese, Ander Matheu, Satoaki Matoba, Sachiko Matsuzaki, Pamela Mattar, Alessandro Matte, Domenico Mattoscio, José L Mauriz, Mario Mauthe, Caroline Mauvezin, Emanual Maverakis, Paola Maycotte, Johanna Mayer, Gianluigi Mazzoccoli, Cristina Mazzoni, Joseph R Mazzulli, Nami McCarty, Christine McDonald, Mitchell R McGill, Sharon L McKenna, BethAnn McLaughlin, Fionn McLoughlin, Mark A McNiven, Thomas G McWilliams, Fatima Mechta-Grigoriou, Tania Catarina Medeiros, Diego L Medina, Lynn A Megeney, Klara Megyeri, Maryam Mehrpour, Jawahar L Mehta, Alfred J Meijer, Annemarie H Meijer, Jakob Mejlvang, Alicia Meléndez, Annette Melk, Gonen Memisoglu, Alexandrina F Mendes, Delong Meng, Fei Meng, Tian Meng, Rubem Menna-Barreto, Manoj B Menon, Carol Mercer, Anne E Mercier, Jean-Louis Mergny, Adalberto Merighi, Seth D Merkley, Giuseppe Merla, Volker Meske, Ana Cecilia Mestre, Shree Padma Metur, Christian Meyer, Hemmo Meyer, Wenyi Mi, Jeanne Mialet-Perez, Junying Miao, Lucia Micale, Yasuo Miki, Enrico Milan, Małgorzata Milczarek, Dana L Miller, Samuel I Miller, Silke Miller, Steven W Millward, Ira Milosevic, Elena A Minina, Hamed Mirzaei, Hamid Reza Mirzaei, Mehdi Mirzaei, Amit Mishra, Nandita Mishra, Paras Kumar Mishra, Maja Misirkic Marjanovic, Roberta Misasi, Amit Misra, Gabriella Misso, Claire Mitchell, Geraldine Mitou, Tetsuji Miura, Shigeki Miyamoto, Makoto Miyazaki, Mitsunori Miyazaki, Taiga Miyazaki, Keisuke Miyazawa, Noboru Mizushima, Trine H Mogensen, Baharia Mograbi, Reza Mohammadinejad, Yasir Mohamud, Abhishek Mohanty, Sipra Mohapatra, Torsten Möhlmann, Asif Mohmmed, Anna Moles, Kelle H Moley, Maurizio Molinari, Vincenzo Mollace, Andreas Buch Møller, Bertrand Mollereau, Faustino Mollinedo, Costanza Montagna, Mervyn J Monteiro, Andrea Montella, L Ruth Montes, Barbara Montico, Vinod K Mony, Giacomo Monzio Compagnoni, Michael N Moore, Mohammad A Moosavi, Ana L Mora, Marina Mora, David Morales-Alamo, Rosario Moratalla, Paula I Moreira, Elena Morelli, Sandra Moreno, Daniel Moreno-Blas, Viviana Moresi, Benjamin Morga, Alwena H Morgan, Fabrice Morin, Hideaki Morishita, Orson L Moritz, Mariko Moriyama, Yuji Moriyasu, Manuela Morleo, Eugenia Morselli, Jose F Moruno-Manchon, Jorge Moscat, Serge Mostowy, Elisa Motori, Andrea Felinto Moura, Naima Moustaid-Moussa, Maria Mrakovcic, Gabriel Muciño-Hernández, Anupam Mukherjee, Subhadip Mukhopadhyay, Jean M Mulcahy Levy, Victoriano Mulero, Sylviane Muller, Christian Münch, Ashok Munjal, Pura Munoz-Canoves, Teresa Muñoz-Galdeano, Christian Münz, Tomokazu Murakawa, Claudia Muratori, Brona M Murphy, J Patrick Murphy, Aditya Murthy, Timo T Myöhänen, Indira U Mysorekar, Jennifer Mytych, Seyed Mohammad Nabavi, Massimo Nabissi, Péter Nagy, Jihoon Nah, Aimable Nahimana, Ichiro Nakagawa, Ken Nakamura, Hitoshi Nakatogawa, Shyam S Nandi, Meera Nanjundan, Monica Nanni, Gennaro Napolitano, Roberta Nardacci, Masashi Narita, Melissa Nassif, Ilana Nathan, Manabu Natsumeda, Ryno J Naude, Christin Naumann, Olaia Naveiras, Fatemeh Navid, Steffan T Nawrocki, Taras Y Nazarko, Francesca Nazio, Florentina Negoita, Thomas Neill, Amanda L Neisch, Luca M Neri, Mihai G Netea, Patrick Neubert, Thomas P Neufeld, Dietbert Neumann, Albert Neutzner, Phillip T Newton, Paul A Ney, Ioannis P Nezis, Charlene C W Ng, Tzi Bun Ng, Hang T T Nguyen, Long T Nguyen, Hong-Min Ni, Clíona Ní Cheallaigh, Zhenhong Ni, M Celeste Nicolao, Francesco Nicoli, Manuel Nieto-Diaz, Per Nilsson, Shunbin Ning, Rituraj Niranjan, Hiroshi Nishimune, Mireia Niso-Santano, Ralph A Nixon, Annalisa Nobili, Clevio Nobrega, Takeshi Noda, Uxía Nogueira-Recalde, Trevor M Nolan, Ivan Nombela, Ivana Novak, Beatriz Novoa, Takashi Nozawa, Nobuyuki Nukina, Carmen Nussbaum-Krammer, Jesper Nylandsted, Tracey R O'Donovan, Seónadh M O'Leary, Eyleen J O'Rourke, Mary P O'Sullivan, Timothy E O'Sullivan, Salvatore Oddo, Ina Oehme, Michinaga Ogawa, Eric Ogier-Denis, Margret H Ogmundsdottir, Besim Ogretmen, Goo Taeg Oh, Seon-Hee Oh, Young J Oh, Takashi Ohama, Yohei Ohashi, Masaki Ohmuraya, Vasileios Oikonomou, Rani Ojha, Koji Okamoto, Hitoshi Okazawa, Masahide Oku, Sara Oliván, Jorge M A Oliveira, Michael Ollmann, James A Olzmann, Shakib Omari, M Bishr Omary, Gizem Önal, Martin Ondrej, Sang-Bing Ong, Sang-Ging Ong, Anna Onnis, Juan A Orellana, Sara Orellana-Muñoz, Maria Del Mar Ortega-Villaizan, Xilma R Ortiz-Gonzalez, Elena Ortona, Heinz D Osiewacz, Abdel-Hamid K Osman, Rosario Osta, Marisa S Otegui, Kinya Otsu, Christiane Ott, Luisa Ottobrini, Jing-Hsiung James Ou, Tiago F Outeiro, Inger Oynebraten, Melek Ozturk, Gilles Pagès, Susanta Pahari, Marta Pajares, Utpal B Pajvani, Rituraj Pal, Simona Paladino, Nicolas Pallet, Michela Palmieri, Giuseppe Palmisano, Camilla Palumbo, Francesco Pampaloni, Lifeng Pan, Qingjun Pan, Wenliang Pan, Xin Pan, Ganna Panasyuk, Rahul Pandey, Udai B Pandey, Vrajesh Pandya, Francesco Paneni, Shirley Y Pang, Elisa Panzarini, Daniela L Papademetrio, Elena Papaleo, Daniel Papinski, Diana Papp, Eun Chan Park, Hwan Tae Park, Ji-Man Park, Jong-In Park, Joon Tae Park, Junsoo Park, Sang Chul Park, Sang-Youel Park, Abraham H Parola, Jan B Parys, Adrien Pasquier, Benoit Pasquier, João F Passos, Nunzia Pastore, Hemal H Patel, Daniel Patschan, Sophie Pattingre, Gustavo Pedraza-Alva, Jose Pedraza-Chaverri, Zully Pedrozo, Gang Pei, Jianming Pei, Hadas Peled-Zehavi, Joaquín M Pellegrini, Joffrey Pelletier, Miguel A Peñalva, Di Peng, Ying Peng, Fabio Penna, Maria Pennuto, Francesca Pentimalli, Cláudia Mf Pereira, Gustavo J S Pereira, Lilian C Pereira, Luis Pereira de Almeida, Nirma D Perera, Ángel Pérez-Lara, Ana B Perez-Oliva, María Esther Pérez-Pérez, Palsamy Periyasamy, Andras Perl, Cristiana Perrotta, Ida Perrotta, Richard G Pestell, Morten Petersen, Irina Petrache, Goran Petrovski, Thorsten Pfirrmann, Astrid S Pfister, Jennifer A Philips, Huifeng Pi, Anna Picca, Alicia M Pickrell, Sandy Picot, Giovanna M Pierantoni, Marina Pierdominici, Philippe Pierre, Valérie Pierrefite-Carle, Karolina Pierzynowska, Federico Pietrocola, Miroslawa Pietruczuk, Claudio Pignata, Felipe X Pimentel-Muiños, Mario Pinar, Roberta O Pinheiro, Ronit Pinkas-Kramarski, Paolo Pinton, Karolina Pircs, Sujan Piya, Paola Pizzo, Theo S Plantinga, Harald W Platta, Ainhoa Plaza-Zabala, Markus Plomann, Egor Y Plotnikov, Helene Plun-Favreau, Ryszard Pluta, Roger Pocock, Stefanie Pöggeler, Christian Pohl, Marc Poirot, Angelo Poletti, Marisa Ponpuak, Hana Popelka, Blagovesta Popova, Helena Porta, Soledad Porte Alcon, Eliana Portilla-Fernandez, Martin Post, Malia B Potts, Joanna Poulton, Ted Powers, Veena Prahlad, Tomasz K Prajsnar, Domenico Praticò, Rosaria Prencipe, Muriel Priault, Tassula Proikas-Cezanne, Vasilis J Promponas, Christopher G Proud, Rosa Puertollano, Luigi Puglielli, Thomas Pulinilkunnil, Deepika Puri, Rajat Puri, Julien Puyal, Xiaopeng Qi, Yongmei Qi, Wenbin Qian, Lei Qiang, Yu Qiu, Joe Quadrilatero, Jorge Quarleri, Nina Raben, Hannah Rabinowich, Debora Ragona, Michael J Ragusa, Nader Rahimi, Marveh Rahmati, Valeria Raia, Nuno Raimundo, Namakkal-Soorappan Rajasekaran, Sriganesh Ramachandra Rao, Abdelhaq Rami, Ignacio Ramírez-Pardo, David B Ramsden, Felix Randow, Pundi N Rangarajan, Danilo Ranieri, Hai Rao, Lang Rao, Rekha Rao, Sumit Rathore, J Arjuna Ratnayaka, Edward A Ratovitski, Palaniyandi Ravanan, Gloria Ravegnini, Swapan K Ray, Babak Razani, Vito Rebecca, Fulvio Reggiori, Anne Régnier-Vigouroux, Andreas S Reichert, David Reigada, Jan H Reiling, Theo Rein, Siegfried Reipert, Rokeya Sultana Rekha, Hongmei Ren, Jun Ren, Weichao Ren, Tristan Renault, Giorgia Renga, Karen Reue, Kim Rewitz, Bruna Ribeiro de Andrade Ramos, S Amer Riazuddin, Teresa M Ribeiro-Rodrigues, Jean-Ehrland Ricci, Romeo Ricci, Victoria Riccio, Des R Richardson, Yasuko Rikihisa, Makarand V Risbud, Ruth M Risueño, Konstantinos Ritis, Salvatore Rizza, Rosario Rizzuto, Helen C Roberts, Luke D Roberts, Katherine J Robinson, Maria Carmela Roccheri, Stephane Rocchi, George G Rodney, Tiago Rodrigues, Vagner Ramon Rodrigues Silva, Amaia Rodriguez, Ruth Rodriguez-Barrueco, Nieves Rodriguez-Henche, Humberto Rodriguez-Rocha, Jeroen Roelofs, Robert S Rogers, Vladimir V Rogov, Ana I Rojo, Krzysztof Rolka, Vanina Romanello, Luigina Romani, Alessandra Romano, Patricia S Romano, David Romeo-Guitart, Luis C Romero, Montserrat Romero, Joseph C Roney, Christopher Rongo, Sante Roperto, Mathias T Rosenfeldt, Philip Rosenstiel, Anne G Rosenwald, Kevin A Roth, Lynn Roth, Steven Roth, Kasper M A Rouschop, Benoit D Roussel, Sophie Roux, Patrizia Rovere-Querini, Ajit Roy, Aurore Rozieres, Diego Ruano, David C Rubinsztein, Maria P Rubtsova, Klaus Ruckdeschel, Christoph Ruckenstuhl, Emil Rudolf, Rüdiger Rudolf, Alessandra Ruggieri, Avnika Ashok Ruparelia, Paola Rusmini, Ryan R Russell, Gian Luigi Russo, Maria Russo, Rossella Russo, Oxana O Ryabaya, Kevin M Ryan, Kwon-Yul Ryu, Maria Sabater-Arcis, Ulka Sachdev, Michael Sacher, Carsten Sachse, Abhishek Sadhu, Junichi Sadoshima, Nathaniel Safren, Paul Saftig, Antonia P Sagona, Gaurav Sahay, Amirhossein Sahebkar, Mustafa Sahin, Ozgur Sahin, Sumit Sahni, Nayuta Saito, Shigeru Saito, Tsunenori Saito, Ryohei Sakai, Yasuyoshi Sakai, Jun-Ichi Sakamaki, Kalle Saksela, Gloria Salazar, Anna Salazar-Degracia, Ghasem H Salekdeh, Ashok K Saluja, Belém Sampaio-Marques, Maria Cecilia Sanchez, Jose A Sanchez-Alcazar, Victoria Sanchez-Vera, Vanessa Sancho-Shimizu, J Thomas Sanderson, Marco Sandri, Stefano Santaguida, Laura Santambrogio, Magda M Santana, Giorgio Santoni, Alberto Sanz, Pascual Sanz, Shweta Saran, Marco Sardiello, Timothy J Sargeant, Apurva Sarin, Chinmoy Sarkar, Sovan Sarkar, Maria-Rosa Sarrias, Surajit Sarkar, Dipanka Tanu Sarmah, Jaakko Sarparanta, Aishwarya Sathyanarayan, Ranganayaki Sathyanarayanan, K Matthew Scaglione, Francesca Scatozza, Liliana Schaefer, Zachary T Schafer, Ulrich E Schaible, Anthony H V Schapira, Michael Scharl, Hermann M Schatzl, Catherine H Schein, Wiep Scheper, David Scheuring, Maria Vittoria Schiaffino, Monica Schiappacassi, Rainer Schindl, Uwe Schlattner, Oliver Schmidt, Roland Schmitt, Stephen D Schmidt, Ingo Schmitz, Eran Schmukler, Anja Schneider, Bianca E Schneider, Romana Schober, Alejandra C Schoijet, Micah B Schott, Michael Schramm, Bernd Schröder, Kai Schuh, Christoph Schüller, Ryan J Schulze, Lea Schürmanns, Jens C Schwamborn, Melanie Schwarten, Filippo Scialo, Sebastiano Sciarretta, Melanie J Scott, Kathleen W Scotto, A Ivana Scovassi, Andrea Scrima, Aurora Scrivo, David Sebastian, Salwa Sebti, Simon Sedej, Laura Segatori, Nava Segev, Per O Seglen, Iban Seiliez, Ekihiro Seki, Scott B Selleck, Frank W Sellke, Joshua T Selsby, Michael Sendtner, Serif Senturk, Elena Seranova, Consolato Sergi, Ruth Serra-Moreno, Hiromi Sesaki, Carmine Settembre, Subba Rao Gangi Setty, Gianluca Sgarbi, Ou Sha, John J Shacka, Javeed A Shah, Dantong Shang, Changshun Shao, Feng Shao, Soroush Sharbati, Lisa M Sharkey, Dipali Sharma, Gaurav Sharma, Kulbhushan Sharma, Pawan Sharma, Surendra Sharma, Han-Ming Shen, Hongtao Shen, Jiangang Shen, Ming Shen, Weili Shen, Zheni Shen, Rui Sheng, Zhi Sheng, Zu-Hang Sheng, Jianjian Shi, Xiaobing Shi, Ying-Hong Shi, Kahori Shiba-Fukushima, Jeng-Jer Shieh, Yohta Shimada, Shigeomi Shimizu, Makoto Shimozawa, Takahiro Shintani, Christopher J Shoemaker, Shahla Shojaei, Ikuo Shoji, Bhupendra V Shravage, Viji Shridhar, Chih-Wen Shu, Hong-Bing Shu, Ke Shui, Arvind K Shukla, Timothy E Shutt, Valentina Sica, Aleem Siddiqui, Amanda Sierra, Virginia Sierra-Torre, Santiago Signorelli, Payel Sil, Bruno J de Andrade Silva, Johnatas D Silva, Eduardo Silva-Pavez, Sandrine Silvente-Poirot, Rachel E Simmonds, Anna Katharina Simon, Hans-Uwe Simon, Matias Simons, Anurag Singh, Lalit P Singh, Rajat Singh, Shivendra V Singh, Shrawan K Singh, Sudha B Singh, Sunaina Singh, Surinder Pal Singh, Debasish Sinha, Rohit Anthony Sinha, Sangita Sinha, Agnieszka Sirko, Kapil Sirohi, Efthimios L Sivridis, Panagiotis Skendros, Aleksandra Skirycz, Iva Slaninová, Soraya S Smaili, Andrei Smertenko, Matthew D Smith, Stefaan J Soenen, Eun Jung Sohn, Sophia P M Sok, Giancarlo Solaini, Thierry Soldati, Scott A Soleimanpour, Rosa M Soler, Alexei Solovchenko, Jason A Somarelli, Avinash Sonawane, Fuyong Song, Hyun Kyu Song, Ju-Xian Song, Kunhua Song, Zhiyin Song, Leandro R Soria, Maurizio Sorice, Alexander A Soukas, Sandra-Fausia Soukup, Diana Sousa, Nadia Sousa, Paul A Spagnuolo, Stephen A Spector, M M Srinivas Bharath, Daret St Clair, Venturina Stagni, Leopoldo Staiano, Clint A Stalnecker, Metodi V Stankov, Peter B Stathopulos, Katja Stefan, Sven Marcel Stefan, Leonidas Stefanis, Joan S Steffan, Alexander Steinkasserer, Harald Stenmark, Jared Sterneckert, Craig Stevens, Veronika Stoka, Stephan Storch, Björn Stork, Flavie Strappazzon, Anne Marie Strohecker, Dwayne G Stupack, Huanxing Su, Ling-Yan Su, Longxiang Su, Ana M Suarez-Fontes, Carlos S Subauste, Selvakumar Subbian, Paula V Subirada, Ganapasam Sudhandiran, Carolyn M Sue, Xinbing Sui, Corey Summers, Guangchao Sun, Jun Sun, Kang Sun, Meng-Xiang Sun, Qiming Sun, Yi Sun, Zhongjie Sun, Karen K S Sunahara, Eva Sundberg, Katalin Susztak, Peter Sutovsky, Hidekazu Suzuki, Gary Sweeney, J David Symons, Stephen Cho Wing Sze, Nathaniel J Szewczyk, Anna Tabęcka-Łonczynska, Claudio Tabolacci, Frank Tacke, Heinrich Taegtmeyer, Marco Tafani, Mitsuo Tagaya, Haoran Tai, Stephen W G Tait, Yoshinori Takahashi, Szabolcs Takats, Priti Talwar, Chit Tam, Shing Yau Tam, Davide Tampellini, Atsushi Tamura, Chong Teik Tan, Eng-King Tan, Ya-Qin Tan, Masaki Tanaka, Motomasa Tanaka, Daolin Tang, Jingfeng Tang, Tie-Shan Tang, Isei Tanida, Zhipeng Tao, Mohammed Taouis, Lars Tatenhorst, Nektarios Tavernarakis, Allen Taylor, Gregory A Taylor, Joan M Taylor, Elena Tchetina, Andrew R Tee, Irmgard Tegeder, David Teis, Natercia Teixeira, Fatima Teixeira-Clerc, Kumsal A Tekirdag, Tewin Tencomnao, Sandra Tenreiro, Alexei V Tepikin, Pilar S Testillano, Gianluca Tettamanti, Pierre-Louis Tharaux, Kathrin Thedieck, Arvind A Thekkinghat, Stefano Thellung, Josephine W Thinwa, V P Thirumalaikumar, Sufi Mary Thomas, Paul G Thomes, Andrew Thorburn, Lipi Thukral, Thomas Thum, Michael Thumm, Ling Tian, Ales Tichy, Andreas Till, Vincent Timmerman, Vladimir I Titorenko, Sokol V Todi, Krassimira Todorova, Janne M Toivonen, Luana Tomaipitinca, Dhanendra Tomar, Cristina Tomas-Zapico, Sergej Tomić, Benjamin Chun-Kit Tong, Chao Tong, Xin Tong, Sharon A Tooze, Maria L Torgersen, Satoru Torii, Liliana Torres-López, Alicia Torriglia, Christina G Towers, Roberto Towns, Shinya Toyokuni, Vladimir Trajkovic, Donatella Tramontano, Quynh-Giao Tran, Leonardo H Travassos, Charles B Trelford, Shirley Tremel, Ioannis P Trougakos, Betty P Tsao, Mario P Tschan, Hung-Fat Tse, Tak Fu Tse, Hitoshi Tsugawa, Andrey S Tsvetkov, David A Tumbarello, Yasin Tumtas, María J Tuñón, Sandra Turcotte, Boris Turk, Vito Turk, Bradley J Turner, Richard I Tuxworth, Jessica K Tyler, Elena V Tyutereva, Yasuo Uchiyama, Aslihan Ugun-Klusek, Holm H Uhlig, Marzena Ułamek-Kozioł, Ilya V Ulasov, Midori Umekawa, Christian Ungermann, Rei Unno, Sylvie Urbe, Elisabet Uribe-Carretero, Suayib Üstün, Vladimir N Uversky, Thomas Vaccari, Maria I Vaccaro, Björn F Vahsen, Helin Vakifahmetoglu-Norberg, Rut Valdor, Maria J Valente, Ayelén Valko, Richard B Vallee, Angela M Valverde, Greet Van den Berghe, Stijn van der Veen, Luc Van Kaer, Jorg van Loosdregt, Sjoerd J L van Wijk, Wim Vandenberghe, Ilse Vanhorebeek, Marcos A Vannier-Santos, Nicola Vannini, M Cristina Vanrell, Chiara Vantaggiato, Gabriele Varano, Isabel Varela-Nieto, Máté Varga, M Helena Vasconcelos, Somya Vats, Demetrios G Vavvas, Ignacio Vega-Naredo, Silvia Vega-Rubin-de-Celis, Guillermo Velasco, Ariadna P Velázquez, Tibor Vellai, Edo Vellenga, Francesca Velotti, Mireille Verdier, Panayotis Verginis, Isabelle Vergne, Paul Verkade, Manish Verma, Patrik Verstreken, Tim Vervliet, Jörg Vervoorts, Alexandre T Vessoni, Victor M Victor, Michel Vidal, Chiara Vidoni, Otilia V Vieira, Richard D Vierstra, Sonia Viganó, Helena Vihinen, Vinoy Vijayan, Miquel Vila, Marçal Vilar, José M Villalba, Antonio Villalobo, Beatriz Villarejo-Zori, Francesc Villarroya, Joan Villarroya, Olivier Vincent, Cecile Vindis, Christophe Viret, Maria Teresa Viscomi, Dora Visnjic, Ilio Vitale, David J Vocadlo, Olga V Voitsekhovskaja, Cinzia Volonté, Mattia Volta, Marta Vomero, Clarissa Von Haefen, Marc A Vooijs, Wolfgang Voos, Ljubica Vucicevic, Richard Wade-Martins, Satoshi Waguri, Kenrick A Waite, Shuji Wakatsuki, David W Walker, Mark J Walker, Simon A Walker, Jochen Walter, Francisco G Wandosell, Bo Wang, Chao-Yung Wang, Chen Wang, Chenran Wang, Chenwei Wang, Cun-Yu Wang, Dong Wang, Fangyang Wang, Feng Wang, Fengming Wang, Guansong Wang, Han Wang, Hao Wang, Hexiang Wang, Hong-Gang Wang, Jianrong Wang, Jigang Wang, Jiou Wang, Jundong Wang, Kui Wang, Lianrong Wang, Liming Wang, Maggie Haitian Wang, Meiqing Wang, Nanbu Wang, Pengwei Wang, Peipei Wang, Ping Wang, Ping Wang, Qing Jun Wang, Qing Wang, Qing Kenneth Wang, Qiong A Wang, Wen-Tao Wang, Wuyang Wang, Xinnan Wang, Xuejun Wang, Yan Wang, Yanchang Wang, Yanzhuang Wang, Yen-Yun Wang, Yihua Wang, Yipeng Wang, Yu Wang, Yuqi Wang, Zhe Wang, Zhenyu Wang, Zhouguang Wang, Gary Warnes, Verena Warnsmann, Hirotaka Watada, Eizo Watanabe, Maxinne Watchon, Anna Wawrzyńska, Timothy E Weaver, Grzegorz Wegrzyn, Ann M Wehman, Huafeng Wei, Lei Wei, Taotao Wei, Yongjie Wei, Oliver H Weiergräber, Conrad C Weihl, Günther Weindl, Ralf Weiskirchen, Alan Wells, Runxia H Wen, Xin Wen, Antonia Werner, Beatrice Weykopf, Sally P Wheatley, J Lindsay Whitton, Alexander J Whitworth, Katarzyna Wiktorska, Manon E Wildenberg, Tom Wileman, Simon Wilkinson, Dieter Willbold, Brett Williams, Robin S B Williams, Roger L Williams, Peter R Williamson, Richard A Wilson, Beate Winner, Nathaniel J Winsor, Steven S Witkin, Harald Wodrich, Ute Woehlbier, Thomas Wollert, Esther Wong, Jack Ho Wong, Richard W Wong, Vincent Kam Wai Wong, W Wei-Lynn Wong, An-Guo Wu, Chengbiao Wu, Jian Wu, Junfang Wu, Kenneth K Wu, Min Wu, Shan-Ying Wu, Shengzhou Wu, Shu-Yan Wu, Shufang Wu, William K K Wu, Xiaohong Wu, Xiaoqing Wu, Yao-Wen Wu, Yihua Wu, Ramnik J Xavier, Hongguang Xia, Lixin Xia, Zhengyuan Xia, Ge Xiang, Jin Xiang, Mingliang Xiang, Wei Xiang, Bin Xiao, Guozhi Xiao, Hengyi Xiao, Hong-Tao Xiao, Jian Xiao, Lan Xiao, Shi Xiao, Yin Xiao, Baoming Xie, Chuan-Ming Xie, Min Xie, Yuxiang Xie, Zhiping Xie, Zhonglin Xie, Maria Xilouri, Congfeng Xu, En Xu, Haoxing Xu, Jing Xu, JinRong Xu, Liang Xu, Wen Wen Xu, Xiulong Xu, Yu Xue, Sokhna M S Yakhine-Diop, Masamitsu Yamaguchi, Osamu Yamaguchi, Ai Yamamoto, Shunhei Yamashina, Shengmin Yan, Shian-Jang Yan, Zhen Yan, Yasuo Yanagi, Chuanbin Yang, Dun-Sheng Yang, Huan Yang, Huang-Tian Yang, Hui Yang, Jin-Ming Yang, Jing Yang, Jingyu Yang, Ling Yang, Liu Yang, Ming Yang, Pei-Ming Yang, Qian Yang, Seungwon Yang, Shu Yang, Shun-Fa Yang, Wannian Yang, Wei Yuan Yang, Xiaoyong Yang, Xuesong Yang, Yi Yang, Ying Yang, Honghong Yao, Shenggen Yao, Xiaoqiang Yao, Yong-Gang Yao, Yong-Ming Yao, Takahiro Yasui, Meysam Yazdankhah, Paul M Yen, Cong Yi, Xiao-Ming Yin, Yanhai Yin, Zhangyuan Yin, Ziyi Yin, Meidan Ying, Zheng Ying, Calvin K Yip, Stephanie Pei Tung Yiu, Young H Yoo, Kiyotsugu Yoshida, Saori R Yoshii, Tamotsu Yoshimori, Bahman Yousefi, Boxuan Yu, Haiyang Yu, Jun Yu, Jun Yu, Li Yu, Ming-Lung Yu, Seong-Woon Yu, Victor C Yu, W Haung Yu, Zhengping Yu, Zhou Yu, Junying Yuan, Ling-Qing Yuan, Shilin Yuan, Shyng-Shiou F Yuan, Yanggang Yuan, Zengqiang Yuan, Jianbo Yue, Zhenyu Yue, Jeanho Yun, Raymond L Yung, David N Zacks, Gabriele Zaffagnini, Vanessa O Zambelli, Isabella Zanella, Qun S Zang, Sara Zanivan, Silvia Zappavigna, Pilar Zaragoza, Konstantinos S Zarbalis, Amir Zarebkohan, Amira Zarrouk, Scott O Zeitlin, Jialiu Zeng, Ju-Deng Zeng, Eva Žerovnik, Lixuan Zhan, Bin Zhang, Donna D Zhang, Hanlin Zhang, Hong Zhang, Hong Zhang, Honghe Zhang, Huafeng Zhang, Huaye Zhang, Hui Zhang, Hui-Ling Zhang, Jianbin Zhang, Jianhua Zhang, Jing-Pu Zhang, Kalin Y B Zhang, Leshuai W Zhang, Lin Zhang, Lisheng Zhang, Lu Zhang, Luoying Zhang, Menghuan Zhang, Peng Zhang, Sheng Zhang, Wei Zhang, Xiangnan Zhang, Xiao-Wei Zhang, Xiaolei Zhang, Xiaoyan Zhang, Xin Zhang, Xinxin Zhang, Xu Dong Zhang, Yang Zhang, Yanjin Zhang, Yi Zhang, Ying-Dong Zhang, Yingmei Zhang, Yuan-Yuan Zhang, Yuchen Zhang, Zhe Zhang, Zhengguang Zhang, Zhibing Zhang, Zhihai Zhang, Zhiyong Zhang, Zili Zhang, Haobin Zhao, Lei Zhao, Shuang Zhao, Tongbiao Zhao, Xiao-Fan Zhao, Ying Zhao, Yongchao Zhao, Yongliang Zhao, Yuting Zhao, Guoping Zheng, Kai Zheng, Ling Zheng, Shizhong Zheng, Xi-Long Zheng, Yi Zheng, Zu-Guo Zheng, Boris Zhivotovsky, Qing Zhong, Ao Zhou, Ben Zhou, Cefan Zhou, Gang Zhou, Hao Zhou, Hong Zhou, Hongbo Zhou, Jie Zhou, Jing Zhou, Jing Zhou, Jiyong Zhou, Kailiang Zhou, Rongjia Zhou, Xu-Jie Zhou, Yanshuang Zhou, Yinghong Zhou, Yubin Zhou, Zheng-Yu Zhou, Zhou Zhou, Binglin Zhu, Changlian Zhu, Guo-Qing Zhu, Haining Zhu, Hongxin Zhu, Hua Zhu, Wei-Guo Zhu, Yanping Zhu, Yushan Zhu, Haixia Zhuang, Xiaohong Zhuang, Katarzyna Zientara-Rytter, Christine M Zimmermann, Elena Ziviani, Teresa Zoladek, Wei-Xing Zong, Dmitry B Zorov, Antonio Zorzano, Weiping Zou, Zhen Zou, Zhengzhi Zou, Steven Zuryn, Werner Zwerschke, Beate Brand-Saberi, X Charlie Dong, Chandra Shekar Kenchappa, Zuguo Li, Yong Lin, Shigeru Oshima, Yueguang Rong, Judith C Sluimer, Christina L Stallings, Chun-Kit Tong.
      In 2008, we published the first set of guidelines for standardizing research in autophagy. Since then, this topic has received increasing attention, and many scientists have entered the field. Our knowledge base and relevant new technologies have also been expanding. Thus, it is important to formulate on a regular basis updated guidelines for monitoring autophagy in different organisms. Despite numerous reviews, there continues to be confusion regarding acceptable methods to evaluate autophagy, especially in multicellular eukaryotes. Here, we present a set of guidelines for investigators to select and interpret methods to examine autophagy and related processes, and for reviewers to provide realistic and reasonable critiques of reports that are focused on these processes. These guidelines are not meant to be a dogmatic set of rules, because the appropriateness of any assay largely depends on the question being asked and the system being used. Moreover, no individual assay is perfect for every situation, calling for the use of multiple techniques to properly monitor autophagy in each experimental setting. Finally, several core components of the autophagy machinery have been implicated in distinct autophagic processes (canonical and noncanonical autophagy), implying that genetic approaches to block autophagy should rely on targeting two or more autophagy-related genes that ideally participate in distinct steps of the pathway. Along similar lines, because multiple proteins involved in autophagy also regulate other cellular pathways including apoptosis, not all of them can be used as a specific marker for bona fide autophagic responses. Here, we critically discuss current methods of assessing autophagy and the information they can, or cannot, provide. Our ultimate goal is to encourage intellectual and technical innovation in the field.
    Keywords:  Autophagosome; LC3; cancer; flux; lysosome; macroautophagy; neurodegeneration; phagophore; stress; vacuole
    DOI:  https://doi.org/10.1080/15548627.2020.1797280