• Aucun résultat trouvé

Interfering with outer membrane biogenesis to fight Gram-negative bacterial pathogens

N/A
N/A
Protected

Academic year: 2021

Partager "Interfering with outer membrane biogenesis to fight Gram-negative bacterial pathogens"

Copied!
6
0
0

Texte intégral

(1)

HAL Id: hal-03039249

https://hal.archives-ouvertes.fr/hal-03039249

Submitted on 3 Dec 2020

HAL is a multi-disciplinary open access archive for the deposit and dissemination of sci-entific research documents, whether they are pub-lished or not. The documents may come from teaching and research institutions in France or abroad, or from public or private research centers.

L’archive ouverte pluridisciplinaire HAL, est destinée au dépôt et à la diffusion de documents scientifiques de niveau recherche, publiés ou non, émanant des établissements d’enseignement et de recherche français ou étrangers, des laboratoires publics ou privés.

Interfering with outer membrane biogenesis to fight

Gram-negative bacterial pathogens

Raffaele Ieva

To cite this version:

Raffaele Ieva. Interfering with outer membrane biogenesis to fight Gram-negative bacterial pathogens. Virulence, Taylor & Francis, 2017, 8, pp.1049 - 1052. �10.1080/21505594.2017.1296617�. �hal-03039249�

(2)

Full Terms & Conditions of access and use can be found at

https://www.tandfonline.com/action/journalInformation?journalCode=kvir20

Virulence

ISSN: 2150-5594 (Print) 2150-5608 (Online) Journal homepage: https://www.tandfonline.com/loi/kvir20

Interfering with outer membrane biogenesis to

fight Gram-negative bacterial pathogens

Raffaele Ieva

To cite this article: Raffaele Ieva (2017) Interfering with outer membrane biogenesis to fight Gram-negative bacterial pathogens, Virulence, 8:7, 1049-1052, DOI: 10.1080/21505594.2017.1296617 To link to this article: https://doi.org/10.1080/21505594.2017.1296617

© 2017 Taylor & Francis

Accepted author version posted online: 17 Feb 2017.

Published online: 17 Mar 2017. Submit your article to this journal

Article views: 872

View related articles

View Crossmark data

(3)

EDITORIAL

Interfering with outer membrane biogenesis to fight Gram-negative

bacterial pathogens

Raffaele Ieva

Laboratoire de Microbiologie et de Genetique Moleculaires (LMGM), Centre de Biologie Integrative (CBI), Universite de Toulouse, CNRS, UPS, Toulouse, France

ARTICLE HISTORYReceived 10 February 2017; Accepted 10 February 2017

KEYWORDSantivirulence drugs; bacterial envelope; BAM; OMP; outer membrane;Yersinia enterocolitica

The multilayer envelope of Gram-negative bacteria con-sists of an inner and an outer membrane (OM) separated by the periplasmic space and a thin cell wall. The exter-nal leaflet of the OM is mainly composed of lipopolysac-charide (LPS). Thanks to the highly compacted saturated acyl chains of its lipidic moiety, and the covalently linked core oligosaccharide and O-antigen polysaccharide, the LPS forms an effective hydrophobic and hydrophilic bar-rier that protects these microorganisms from external insults, including several antibiotics.1 The OM further harbours an arsenal of protein assemblies that promote interaction with the colonized environment, biofilm for-mation, as well as adhesion to and invasion of host tis-sues.2 Thus accurate biogenesis of the OM is critical for bacterial survival. Pathogens with defects in OM func-tionality are likely to succumb in the competition with bacteria of the surrounding microbiota or may be attenu-ated in their ability to infect the host. Recently, due to difficulties in combating infectious diseases with tradi-tional antibiotics, much effort has been put into develop-ing novel antivirulence drugs that can specifically interfere with mechanisms of pathogenesis.3,4,5 As these

medicaments do not compromise essential functions of bacterial cells, but rather impair the overall fitness of pathogenic strains in the context of the infection process, they are likely to generate a low selective pressure for suppressor mutations. As part of this effort, a great deal of research work is currently focused on deciphering the intricate network of cellular processes that ensure ef fi-cient OM biogenesis in bacteria and its impact on dis-eases. In this issue of Virulence, Weirich et al. shed light on how loss of activity of non-essential proteins involved in OM biogenesis influences the pathogenic potential of

Yersinia enterocolitica,6 an enteropathogen that can be

transmitted from farm animals to humans.7

Two main classes of proteins populate the OM: 1) Lipoproteins, which associate to the membrane via a lipi-dated amino(N)-terminal cysteine residue; 2) Integral OM proteins (OMPs) that span the lipid bilayer by adopting b-barrel structures of variable size (8–26 b-strands) and may additionally carry soluble domains that ultimately localize in the periplasm or on the cell surface.8Both lipoproteins and OMPs are synthesized in the cytosol, transported across the inner membrane and shuttled through the periplasm to their site of assembly at the OM. Due to the amphipathic character of their membrane spanning domains, OMPs are aggregation-prone and their transport across the periplasm is aided by chaperones.9 Some OMPs are preferentially cha-peroned by SurA, while other OMPs can be transported via a pathway assisted by the chaperone Skp in coopera-tion with the protease DegP.10,11In all cases, precursors are delivered to theb-barrel assembly machinery (BAM) for assembly into the OM. BAM is a pentamer in Escheri-chia coli and possibly in other Enterobacteriaceae,12,13,14

which include several pathogens (strains of E. coli, Sal-monella enterica, Shigella flexneri, Klebsiella pneumonia, Yersinia species). The central and essential component BamA spans through the OM via a carboxy(C)-terminal b-barrel domain and interacts via an N-terminal, peri-plasm-disposed domain with partner subunits. BamA associates with the lipoproteins BamBCDE, of which only BamD is essential.13,14 Numerous lines of evidence indicate that the folding and assembly of distinct OMPs are differentially impacted by the lack of a periplasmic chaperone or a non-essential BAM lipoprotein.

CONTACT Raffaele Ieva raffaele.ieva@ibcg.biotoul.fr Laboratoire de Microbiologie et de Genetique Moleculaires (LMGM), Universite de Toulouse, CNRS,

31062 Toulouse, France.

Comment on: Weirich J, et al. Identifying components required for OMP biogenesis as novel targets for antiinfective drugs. Virulence 2017; 1-20; PMID:28118090; https://doi.org/10.1080/21505594.2016.1278333

© 2017 Taylor & Francis

VIRULENCE

2017, VOL. 8, NO. 7, 1049–1052

(4)

However, it still remains rather elusive how periplasmic chaperones or BAM lipoproteins contribute to the assembly of different OMPs, despite numerous genetic, biochemical and structural studies.15 Of note, while BamA is widely conserved across different phyla of bac-teria, the set of bacterial lipoproteins or periplasmic fac-tors that stably associate with BamA may differ.14 It is tempting to speculate that the heterogeneity of the BAM complex in different families of bacteria reflects specific requirements in OMP biogenesis and niches coloniza-tion, and that these differences could potentially be exploited in antivirulence therapies to selectively target different microorganisms.

Weirich et al. describe a multi-approach analysis of the consequences produced by the loss of non-essential factors involved in OMP biogenesis in Y. enterocoli-tica.6 The main determinants that contribute to its

pathogenesis are the LPS, the type III secretion system and its effector proteins, and the OMPs Ail (attach-ment and invasion locus), YadA (Yersinia adhesin A) and Inv (invasin).16 The pathogenic potential of Y. enterocolitica strains can be estimated in a mouse infection model, providing the means to assess the contribution of different cellular factors to the infec-tion process. Weirich et al. focus their analysis on the loss of SurA, Skp, DegP, BamB, BamC and BamE. Theyfind that losses of BamB or SurA and, to a lower extent, Skp enhance the cell susceptibility to concen-trations of antibiotics and detergents that are normally tolerated by wild type cells.6 The observed chemical susceptibility of these mutant cells underlines, together with previousfindings, the potential of pharmacologi-cal inactivation of BamB and SurA to enhance the potency of traditional antibiotics.6,17,18,19,20,21 Impor-tantly, by combining semi-quantitative proteomics and measurements of mRNA levels, Weirich at al. further investigate how losses of BamB, SurA and Skp influ-ence the steady-state levels of proteins of the OM. Accumulation of OMP assembly intermediates in the periplasm induces the envelope stress-activated sE response,22which causes upregulation of OMP biogen-esis factors and downregulation of mRNA levels encoding abundant OMPs.23Variations in the levels of

proteins encoded by genes controlled by sE are diffi-cult to interpret as these variations may result from additive effects owing to regulation of mRNA levels and proteostasis. The collected data set published by Weirich et al. is of great interest as it contributes to reveal the regulatory and proteostatic pathways that govern OM biogenesis in Y. enterocolitica. Further-more, comparisons of the data set published here and results obtained using similar approaches on E. coli may help to understand differential strategies adopted

by diverse, albeit evolutionarily related, pathogens in responding to envelope stress. In E. coli for instance, transcripts encoding BAM subunits are activated upon loss of SurA11 via the sE response.23 In Y. enterocoli-tica cells lacking SurA or BamB, the levels of tran-scripts encoding BAM subunits are unaffected, while the amounts of transcripts encoding other OMPs (OmpF and LamB) are reduced, which is most proba-bly due tosE activation.6 The hypothesis that Yersinia species have a reduced sE regulon23 may explain the

observed differential regulation of BAM transcripts. One key finding reported by Weirich et al. concerns the differential effects of lack of SurA or BamB on dis-tinct OMPs, among which autotransporters, a subclass of the type V secretion system. Autotransporters are vir-ulence factors consisting generally of a C-terminal b-bar-rel domain (b-domain) that promotes translocation across the OM of a covalently linked N-terminal passen-ger domain.24,25,26Inv is an inverse autotransporter with a N-terminal b-domain. YadA is a trimeric autotrans-porter. Theb-domains of both Inv and YadA ultimately fold into 12-strand b-barrels, however in the case of YadA 3 identical monomers each contribute 4b-strands to thefinal structure. Weirich et al. find that the levels of Inv detected on the cell surface are reduced in surA¡and bamB¡cells. The levels of the corresponding transcript

are unaffected in surA¡cells,6indicating that biogenesis of Inv strongly depends on SurA. This chaperon has been shown to specifically interact with the passenger domain of a classical autotransporter.27 SurA was also suggested to extensively interact with the passenger domain of an inverse autotransporter.28 At least in the case of classical autotransporters, completion of b-domain assembly into the OM is licensed upon secre-tion of the passenger domain.27,29,30Thus further experi-ments will be necessary to establish whether, in the case of Y. enterocolitica Inv, SurA is mainly required to main-tain the passenger domain in a secretion competent con-formation or to chaperone its b-domain across the periplasm. On the other hand, the data presented by Weirich et al. clearly show that biogenesis of YadA does not depend on SurA nor BamB. The reason for this dif-ferential dependence may relate to the low number of b-strands of the YadA protein monomer.6 This is an

interesting hypothesis in line with the observation that TolC, another trimeric OMP (each TolC monomer con-tributes 4b-strands to a 12-strand b-barrel) is indepen-dent of SurA and BamB.11,31As suggested by the authors and elsewhere, it is conceivable that the small size of the amphipathicb-domain determines a low dependence on the biogenesis factors SurA and BamB.6,32,33In addition, the proposed cooperation of multiple BAM complexes in folding and assembling trimeric b-barrel structures32

(5)

might generate a protein rich environment in which YadA can efficiently assemble even in the absence of SurA or BamB.6

Most convincingly, Weirich et al. show that loss of SurA or BamB (or to a lower extent Skp) attenuates the virulence of Y. enterocolitica in both orogastric and sys-temic mouse infection models. Several factors are likely to contribute to these phenotypes at different steps of the infection process. Besides Inv, also the iron uptake recep-tor (FyuA) is reduced in all mutants tested in mouse infection.6 Furthermore, the authors show that the LPS of bamB¡cells lacks O-antigen,6a feature that might sig-nificantly contribute to its serum susceptibility.34,35 The

authors further comment that although the overall bio-genesis on YadA is not affected,6 minor changes in pas-senger domain properties, due to the altered landscape of OM components, may influence its ability to bind complement and promote serum resistance. In sum-mary, Weirich et al. present a multifaceted analysis to identify targets of the OM biogenesis machineries for antivirulence drugs that can attenuate Y. enterocolitica and, potentially, other enterobacterial pathogens. Similar approaches should be extended to mutant strains lacking other components involved in OMP biogenesis, maintenance and in envelope stress response pathways.23,36,37,38,39,40

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Acknowledgments

The author thanks Drs. Peter Redder and Harris Bernstein for discussion.

Funding

This work was supported by the ATIP-Avenir program (CNRS).

References

[1] Delcour AH. Outer membrane permeability and antibiotic resistance. Biochim Biophys Acta 2009; 1794:808-16; PMID:19100346; https://doi.org/10.1016/ j.bbapap.2008.11.005

[2] Costa TR, Felisberto-Rodrigues C, Meir A, Prevost MS, Redzej A, Trokter M, Waksman G. Secretion systems in Gram-negative bacteria: structural and mechanistic insights. Nat Rev Microbiol 2015; 13:343-59; PMID:25978706; https://doi.org/10.1038/nrmicro3456 [3] Rasko DA, Sperandio V. Anti-virulence strategies to

combat bacteria-mediated disease. Nat Rev Drug Discov

2010; 9:117-28; PMID:20081869; https://doi.org/10.1038/ nrd3013

[4] Baron C. Antivirulence drugs to target bacterial secre-tion systems. Curr Opin Microbiol 2010; 13:100-5; PMID:20079679; https://doi.org/10.1016/j.mib.2009. 12.003

[5] Steadman D, Lo A, Waksman G, Remaut H. Bacterial surface appendages as targets for novel antibacterial ther-apeutics. Future Microbiol 2014; 9:887-900; PMID: 25156378; https://doi.org/10.2217/fmb.14.46

[6] Weirich J, Br€autigam C, M€uhlenkamp M, Franz-Wachtel M, Macek B, Meuskens I, Skurnik M, Leskinen K, Bohn E, Autenrieth I, et al. Identifying components required for OMP biogenesis as novel targets for antiinfective drugs. Virulence 2017; 1-20; PMID:28118090; https:// doi.org/10.1080/21505594.2016.1278333

[7] Bottone EJ. Yersinia enterocolitica: the charisma contin-ues. Clin Microbiol Rev 1997; 10:257-76; PMID:9105754 [8] Fairman JW, Noinaj N, Buchanan SK. The structural

biology of b-barrel membrane proteins: a summary of recent reports. Curr Opin Struct Biol 2011; 21:523-31; PMID:21719274; https://doi.org/10.1016/j.sbi.2011. 05.005

[9] Plummer AM, Fleming KG. From Chaperones to the Membrane with a BAM!. Trends Biochem Sci 2016; 41:872-82; PMID:27450425; https://doi.org/10.1016/j. tibs.2016.06.005

[10] Sklar JG, Wu T, Kahne D, Silhavy TJ. Defining the roles of the periplasmic chaperones SurA, Skp, and DegP in Escherichia coli. Genes Dev 2007; 21:2473-84; PMID:17908933; https://doi.org/10.1101/gad. 1581007

[11] Vertommen D, Ruiz N, Leverrier P, Silhavy TJ, Collet JF. Characterization of the role of the Escherichia coli peri-plasmic chaperone SurA using differential proteomics. Proteomics 2009; 9:2432-43; PMID:19343722; https:// doi.org/10.1002/pmic.200800794

[12] Wu T, Malinverni J, Ruiz N, Kim S, Silhavy TJ, Kahne D. Identification of a multicomponent complex required for outer membrane biogenesis in Escherichia coli. Cell 2005; 121:235-45; PMID:15851030; https://doi.org/ 10.1016/j.cell.2005.02.015

[13] Sklar JG, Wu T, Gronenberg LS, Malinverni JC, Kahne D, Silhavy TJ. Lipoprotein SmpA is a component of the YaeT complex that assembles outer membrane proteins in Escherichia coli. Proc Natl Acad Sci U S A 2007; 104:6400-5; PMID:17404237; https://doi.org/10.1073/ pnas.0701579104

[14] Webb CT, Heinz E, Lithgow T. Evolution of the b-barrel assembly machinery. Trends Microbiol 2012; 20:612-20; PMID:22959613; https://doi.org/10.1016/j. tim.2012.08.006

[15] Bakelar J, Buchanan SK, Noinaj N. Structural snapshots of the b-barrel assembly machinery. FEBS J 2016; PMID:27862971

[16] Leo JC, Skurnik M. Adhesins of human pathogens from the genus Yersinia. Adv Exp Med Biol 2011; 715:1-15; PMID:21557054

[17] Ruiz N, Falcone B, Kahne D, Silhavy TJ. Chemical con-ditionality: a genetic strategy to probe organelle assembly. Cell 2005; 121:307-17; PMID:15851036; https://doi.org/ 10.1016/j.cell.2005.02.014

(6)

[18] Southern SJ, Scott AE, Jenner DC, Ireland PM, Norville IH, Sarkar-Tyson M. Survival protein A is essential for virulence in Yersinia pestis. Microb Pathog 2016; 92:50-3; PMID:26724738; https://doi.org/10.1016/j.micpath. 2015.12.013

[19] Hsieh PF, Hsu CR, Chen CT, Lin TL, Wang JT. The Klebsiella pneumoniae YfgL (BamB) lipoprotein contrib-utes to outer membrane protein biogenesis, type-1 fim-briae expression, anti-phagocytosis, and in vivo virulence. Virulence 2016; 7:587-601; PMID:27029012; https://doi.org/10.1080/21505594.2016.1171435

[20] Fardini Y, Trotereau J, Bottreau E, Souchard C, Velge P, Virlogeux-Payant I. Investigation of the role of the BAM complex and SurA chaperone in outer-membrane protein biogenesis and type III secretion system expression in Sal-monella. Microbiology 2009; 155:1613-22; PMID: 19372159; https://doi.org/10.1099/mic.0.025155-0 [21] Obi IR, Nordfelth R, Francis MS. Varying dependency of

periplasmic peptidylprolyl cis-trans isomerases in pro-moting Yersinia pseudotuberculosis stress tolerance and pathogenicity. Biochem J 2011; 439:321-32; PMID: 21726196; https://doi.org/10.1042/BJ20110767

[22] Walsh NP, Alba BM, Bose B, Gross CA, Sauer RT. OMP peptide signals initiate the envelope-stress response by acti-vating DegS protease via relief of inhibition mediated by its PDZ domain. Cell 2003; 113:61-71; PMID:12679035; https://doi.org/10.1016/S0092-8674(03)00203-4

[23] Rhodius VA, Suh WC, Nonaka G, West J, Gross CA. Con-served and variable functions of the sigmaE stress response in related genomes. PLoS Biol 2006; 4:e2; PMID:16336047; https://doi.org/10.1371/journal.pbio.0040002

[24] van Ulsen P, Rahman Su, Jong WS, Daleke-Schermer-horn MH, Luirink J. Type V secretion: from biogenesis to biotechnology. Biochim Biophys Acta 2014; 1843:1592-611; PMID:24269841; https://doi.org/10.1016/j.bbamcr. 2013.11.006

[25] Drobnak I, Braselmann E, Chaney JL, Leyton DL, Bern-stein HD, Lithgow T, Luirink J, Nataro JP, Clark PL. Of linkers and autochaperones: an unambiguous nomencla-ture to identify common and uncommon themes for autotransporter secretion. Mol Microbiol 2015; 95:1-16; PMID:25345653; https://doi.org/10.1111/mmi.12838 [26] Fan E, Chauhan N, Udatha DB, Leo JC, Linke D. Type V

Secretion Systems in Bacteria. Kudva I, Cornick N, Plummer P, Zhang Q, Nicholson T, Bannantine J, Bel-laire B (ed) 2016; p 305-335; Virulence Mechanisms of Bacterial Pathogens. ASM Press, Washington, DC [27] Ieva R, Tian P, Peterson JH, Bernstein HD. Sequential

and spatially restricted interactions of assembly factors with an autotransporter beta domain. Proc Natl Acad Sci U S A 2011; 108:E383-91; PMID:21646511; https://doi. org/10.1073/pnas.1103827108

[28] Oberhettinger P, Leo JC, Linke D, Autenrieth IB, Sch€utz MS. The inverse autotransporter intimin exports its pas-senger domain via a hairpin intermediate. J Biol Chem 2015; 290:1837-49; PMID:25488660; https://doi.org/ 10.1074/jbc.M114.604769

[29] Sauri A, Soprova Z, Wickstr€om D, de Gier JW, Van der Schors RC, Smit AB, Jong WS, Luirink J. The Bam

(Omp85) complex is involved in secretion of the auto-transporter haemoglobin protease. Microbiology 2009; 155:3982-91; PMID:19815580; https://doi.org/10.1099/ mic.0.034991-0

[30] Pavlova O, Peterson JH, Ieva R, Bernstein HD. Mechanis-tic link betweenb barrel assembly and the initiation of autotransporter secretion. Proc Natl Acad Sci U S A 2013; 110:E938-47; PMID:23431155; https://doi.org/ 10.1073/pnas.1219076110

[31] Charlson ES, Werner JN, Misra R. Differential effects of yfgL mutation on Escherichia coli outer membrane pro-teins and lipopolysaccharide. J Bacteriol 2006; 188:7186-94; PMID:17015657; https://doi.org/10.1128/JB.00571-06 [32] Mahoney TF, Ricci DP, Silhavy TJ. Classifying b-Bar-rel Assembly Substrates by Manipulating Essential Bam Complex Members. J Bacteriol 2016; 198:1984-92; PMID:27161117; https://doi.org/10.1128/JB.00263-16

[33] Palomino C, Marın E, Fernandez LA. The fimbrial usher FimD follows the SurA-BamB pathway for its assembly in the outer membrane of Escherichia coli. J Bacteriol 2011; 193:5222-30; PMID:21784935; https://doi.org/ 10.1128/JB.05585-11

[34] Biedzka-Sarek M, Venho R, Skurnik M. Role of YadA, Ail, and Lipopolysaccharide in Serum Resistance of Yer-sinia enterocolitica Serotype O:3. Infect Immun 2005; 73:2232-44; PMID:15784567; https://doi.org/10.1128/ IAI.73.4.2232-2244.2005

[35] Bengoechea JA, Najdenski H, Skurnik M. Lipopolysac-charide O antigen status of Yersinia enterocolitica O:8 is essential for virulence and absence of O antigen affects the expression of other Yersinia virulence factors. Mol Microbiol 2004; 52:451-69; PMID:15066033; https://doi. org/10.1111/j.1365-2958.2004.03987.x

[36] Selkrig J, Mosbahi K, Webb CT, Belousoff MJ, Perry AJ, Wells TJ, Morris F, Leyton DL, Totsika M, Phan MD, et al. Discovery of an archetypal protein transport system in bacterial outer membranes. Nat Struct Mol Biol 2012; 19:506-10; PMID:22466966; https://doi.org/10.1038/ nsmb.2261

[37] Bury-Mone S, Nomane Y, Reymond N, Barbet R, Jacquet E, Imbeaud S, Jacq A, Bouloc P. Global analysis of extrac-ytoplasmic stress signaling in Escherichia coli. PLoS Genet 2009; 5:e1000651; PMID:19763168; https://doi. org/10.1371/journal.pgen.1000651

[38] Gennaris A, Ezraty B, Henry C, Agrebi R, Vergnes A, Oheix E, Bos J, Leverrier P, Espinosa L, Szewczyk J, et al. Repairing oxidized proteins in the bacterial envelope using respiratory chain electrons. Nature 2015; 528:409-12; PMID:26641313; https://doi.org/10.1038/nature15764 [39] Cho SH, Szewczyk J, Pesavento C, Zietek M, Banzhaf M, Roszczenko P, Asmar A, Laloux G, Hov AK, Leverrier P, et al. Detecting envelope stress by monitoring b-barrel assembly. Cell 2014; 159:1652-64; PMID:25525882; https://doi.org/10.1016/j.cell.2014.11.045

[40] Konovalova A, Mitchell AM, Silhavy TJ. A lipoprotein/ b-barrel complex monitors lipopolysaccharide integrity transducing information across the outer membrane. Elife 2016; 5:e15276; PMID:27282389

Références

Documents relatifs

[r]

If similar processes played a role in formation of minerals around microfossils from the Kakontwe, Taishir and Rasthof formations, originally siliceous tests would be converted

This inverse relationship strongly suggests that the single event upsets were not primarily caused by solar energetic protons, as we have shown in Figure 1 that more

L’origine de cette expression vient moins d’un hypothétique partage des compétences (Giusti, 2012a) entre militaires (la topographie de surface, les formes du terrain),

présentées. Les températures et les masses de fluide frigorigène cyclés expérimentales et théoriques sont comparables et en accord appréciable. On constate tout~fois

In addition, no general correlation scheme exists, so that it is impossible for .a designer to take burnout data for water, for instance, and use it to

We use the composite fermion approach for theoretical studies of the Coulomb drag between two parallel layers of two-dimensional electron gases in the quantum

Dans l'air humide, les faibles contraintes de croissance peuvent être expliquées par la relaxation des contraintes après le processus de pré-transition en raison