Sie sind auf Seite 1von 13

Angewandte

Eine Zeitschrift der Gesellschaft Deutscher Chemiker Chemie www.angewandte.de

Akzeptierter Artikel

Titel: Biological Nanopore Approach for Single-Molecule Protein


Sequencing

Autoren: Zheng-Li Hu, Ming-Zhu Huo, Yi-Lun Ying, and Yi-Tao Long

Dieser Beitrag wurde nach Begutachtung und Überarbeitung sofort als


"akzeptierter Artikel" (Accepted Article; AA) publiziert und kann unter
Angabe der unten stehenden Digitalobjekt-Identifizierungsnummer
(DOI) zitiert werden. Die deutsche Übersetzung wird gemeinsam mit der
endgültigen englischen Fassung erscheinen. Die endgültige englische
Fassung (Version of Record) wird ehestmöglich nach dem Redigieren
und einem Korrekturgang als Early-View-Beitrag erscheinen und kann
sich naturgemäß von der AA-Fassung unterscheiden. Leser sollten
daher die endgültige Fassung, sobald sie veröffentlicht ist, verwenden.
Für die AA-Fassung trägt der Autor die alleinige Verantwortung.

Zitierweise: Angew. Chem. Int. Ed. 10.1002/anie.202013462

Link zur VoR: https://doi.org/10.1002/anie.202013462


Angewandte Chemie 10.1002/ange.202013462

REVIEW
Biological Nanopore Approach for Single-Molecule Protein
Sequencing
Zheng-Li Hu,[a] Ming-Zhu Huo,[a] Yi-Lun Ying*[ab] and Yi-Tao Long*[a]

Accepted Manuscript

1
This article is protected by copyright. All rights reserved.
Angewandte Chemie 10.1002/ange.202013462

REVIEW

Abstract: Proteins are responsible for the occurrence and treatment could be provided, which benefits the unambiguous identification
of many diseases, and therefore protein sequencing will revolutionize of protein variations, such as PTMs.[24–27] Moreover, specific
proteomics and clinical diagnostics. Nanopore electrochemistry has modifications of the nanopore sensing interface could be used to
proved successful for single-molecule DNA sequencing, which optimize nanopore features (e.g. size and charge distribution)
resolves the identities of 4 natural deoxyribonucleotides based on the with an atomic precision, permitting the accurate determination of
current blockages and duration times of their translocations across
the nanopore confinement. However, open challenges still remain for
biological nanopores to sequentially identify each amino acid (AA) of
single proteins due to the inherent complexity of 20 proteinogenic AAs
in charges, volumes, hydrophobicity and structures. Herein, we focus

Accepted Manuscript
on recent exciting advances in biological nanopore approach for
single-molecule protein sequencing (SMPS) from native protein
unfolding, control of peptide translocation, AA identification to
applications in disease detection.

1. Introduction

Function or dysfunction of proteins largely depends on their


primary structures, i.e. the AA sequences. Protein sequencing
would provide important information for disease diagnostics and
treatments. Edman degradation was initially invented to sequence
proteins from N-terminus based on cyclic chemical reactions,
which fails to work without a free α-amino group on the N-terminus
(e.g. N-acetylated proteins) and is difficult to resolve proteins > 50
aa.[1–3] Mass spectrometry (MS) is the current standard method
for protein sequencing but requires enzymatic digestion and/or
fragmentation before detection.[4–6] Protein concentrations vary in
a broad range (~1010) with low copy numbers down to 10-100
molecules in a cell, whereas MS works in a low coverage of
proteome due to a limited dynamic range of mass spectrometers
(~105).[7–9] As such, MS usually requires high-cost equipment and
large amount of samples (e.g. a million copies or an attomole of a
protein) for an accurate detection.[6,10,11] However, enrichment of
proteins based on target-specific biomaterials (e.g. antibody) is
not available for all kinds of proteins.[10,11] Moreover, no methods
exist for protein amplification that analogous to polymerase chain
reaction (PCR) for DNAs. Other drawbacks include insufficient Figure 1. Schematic representation of single-molecule protein sequencing with
sensitivity to identify AAs with identical or similar masses and the a biological nanopore. Top: Illustration of biological nanopore sequencing of a
post-translational modifications (PTMs) of proteins. single translocating protein. A biological nanopore with specific molecular
“brake” site on the nanopore interface is inserted in artificial lipid bilayer.
Recently, nanopores based on electrochemical techniques
Nanopore array is designed to parallel detection of proteins with high-
have developed into a powerful alternative for the realization of throughput. In a typical measurement, (i) a folded protein is unfolded to linear
protein sequencing, because they could provide nano-confined peptide for feasible entry into the nanopore; (ii) then the partially unfolded
space and designable single-molecule sensing interface to protein molecule linearly threads through the nanopore confinement forced by
the electrical field; (iii) eventually the nanopore sensing region identifies single
enable high spatiotemporal resolution and ultimate sensitivity for
AAs residue-by-residue at a controllable pace during the translocation. Bottom:
proteome analysis.[12–15] The interaction of single molecules with Ideal current readouts with distinctive features assigned to each AA
the nanopore sensing interface induces typical ionic current conceptually for calling the linear sequences of single proteins in real time.
modulations, which reflects the composition, charge distribution,
structure and sequence of the translocating molecule.[16–21] A
nanopore, therefore, could directly read the sequences of
individual proteins residue-by-residue, depending on the well- [a] Dr. Zheng-Li Hu, Ms. Ming-Zhu Huo, Prof. Yi-Lun Ying, Prof. Yi-Tao
resolved ionic current alteration (Figure 1). In particular, nanopore Long
State Key Laboratory of Analytical Chemistry for Life Science,
array would contribute to high-throughput parallel detection of
School of Chemistry and Chemical Engineering,
low-abundance proteins, rescuing peptides/proteins from the lack Nanjing University, 163 Xianlin Avenue, Nanjing 210023 (P. R. China)
of in vitro amplification methods.[22,23] This also dictates the bright E-mail: yitaolong@nju.edu.cn, yilunying@nju.edu.cn
prospects to decipher proteins of long-read lengths without the [b] Prof. Yi-Lun Ying
Chemistry and Biomedicine Innovation Center,
needs of labelling and/or enrichment. Besides, a dynamic view of
Nanjing University, 163 Xianlin Avenue, Nanjing 210023 (P. R. China)
protein heterogeneity that is unavailable in an ensemble detection

2
This article is protected by copyright. All rights reserved.
Angewandte Chemie 10.1002/ange.202013462

REVIEW
AAs with high spatial and temporal resolution.[28–31] this, the prevalence of PTMs including phosphorylation,
Different from DNAs, proteins have more complex building acetylation, methylation, glycosylation and ubiquitination, has
blocks-20 proteinogenic AAs and hundreds of variations including made the aim of direct SMPS even more challenging for biological
isomers, enantiomers and PTMs-with very subtle differences in nanopore techniques.[8,46] Hence, a tremendous challenge is to
volumes and structures. These AAs sequentially constitute the improve spatial resolution to provide distinctively featured current
primary structure of a protein, which then folds into higher-order signals for different AAs of the analysed protein.
architectures to display exquisite functionalities. Compared to The first demonstrations have proved the capability of α-HL
recent success of DNA sequencing,[32–35] biological nanopore- nanopores in transporting and identifying peptides (GPP)n with
based protein sequencing is still in an early stage and urgently single, double or collagen-like triple helices.[20,47] A subsequent
needs to address multiple challenges: work detailed interaction kinetics of theα-HL nanopore with
(i) Structured protein unfolding. The commonly used biological cationic α-helical peptides (AAKAA)n.[48] A particular study then
nanopores, such as α-hemolysin (α-HL),[36] Mycobacterium suggested that different translocation velocities of an α-helical
smegmatis porin A (MspA),[37] aerolysin (AeL)[38] and peptide Fmoc-DxAyKz through the α-HL and AeL nanopores are

Accepted Manuscript
Fragaceatoxin C (FraC) nanopores, [39] have too small internal likely caused by the differences in hydrophobic peptide-nanopore
constriction (with diameter of 1-2 nm) to accommodate the large interactions.[49] Another work elucidated that translocations of
secondary, tertiary and quaternary structures of native proteins. positively charged peptides could be facilitated by engineering
Thus, a fundamental requirement for nanopore-based protein electrostatic binding sites of the nanopore sensing interface. [50]
sequencing (NPS) is to unfold the 3D topological structures of a During the last decade, numerous efforts with biological
protein to enable its entry and partition into the nanopore nanopores have been invested into peptide and protein sensing,
confinement in a stretched profile. including conformation/structure characterization, [51–56] variant
(ii) Peptide translocation control. In general, sufficiently long recognition,[24,26,27,57–63] interaction investigation,[64–69] and protein
residence of single molecules should be provided to allow high kinase exploration.[70–74] However, despite all the present
signal-to-noise recording of time-series signals for well-ordering achievements in proteomic analysis, there is still a long way
each AA. As summarised in Figure 2, a peptide traverses across toward SMPS. This minireview focuses on recent progress in
biological nanopores at high velocities (e.g. majority estimated to biological nanopore approach for SMPS, and attempts to provide
< 1 ms aa-1). However, the time-resolution of currently commercial a comprehensive overview of current experimental efforts and
nanopore instruments hinders the accurate distinction of different strategies for overcoming major hurdles of NPS. Finally, we close
elements of single peptides.[40,41] Therefore, a timely subject for it with our insights and perspectives on future developments in the
NPS is to control the translocation of single peptides within the field of NPS.
nanopore sensing interface.
(iii) Amino acid identification. The 20 proteinogenic AAs dictate
volumes ranging from 0.06 (glycine) to 0.23 (tryptophan) nm3 with 2. Unfolding of Structured Proteins
differences as small as 0.001 nm3 (methionine and isoleucine).[42–
44]
They are linked by peptide bonds with an average separation To allow single protein entry into the nanopore confinement, a
of ~0.38 nm to compose linear sequences of proteins. [45] Beyond handful of methods have been proposed to disrupt the folded

Zheng-Li Hu received her B.S. in Chemistry Ming-Zhu Huo received her B.S. in
at Shanghai Normal University in 2014 and Chemistry from Shandong University in
PhD in Analytical Chemistry at East China 2019. She is currently a graduate student
University of Science and Technology working on peptide/protein single-molecule
(ECUST) under the supervision of Prof. Yi- detection under the supervision of Prof. Yi-
Tao Long in 2019. She is currently Tao Long in the School of Chemistry and
performing her postdoctoral research at Chemical Engineering at Nanjing University.
Nanjing University in the fields of biological
nanopores and single-molecule sensing and
sequencing.

Yi-Lun Ying received her B.S. (2009) and Yi-Tao Long received his B.S. (1989) from
PhD (2014) in Analytical Chemistry with ((Author Portrait)) Shandong University, and MSc (1996) and ((Author Portrait))
Prof. Yi-Tao Long after her studying at PhD (1998) from Nanjing University. After
ECUST and University of Birmingham (UK). completing 2-year postdoctoral studies at
Following her postdoctoral research, she Heidelberg University, he worked at the
was appointed as an associate professor at University of Saskatchewan, University of
ECUST. Since 2019, she is a professor and Alberta and University of California, Berkeley.
co-PI at the Chemistry and Biomedicine He then was appointed as a professor at
Innovation Center at Nanjing University. Her ECUST since 2007 and moved to Nanjing
main researches involve electrical-optical University in 2019. His main researches
nanopore sensing and advanced artificial include nanopore electrochemistry,
intelligence for nanopore arrays. biointerfaces nano-spectroelectrochemistry
and integrated biosensors for life science.
((Author Portrait)) ((Author Portrait))

3
This article is protected by copyright. All rights reserved.
Angewandte Chemie 10.1002/ange.202013462

REVIEW
by appending an unstructured polyanion with a ssrA peptide tag
at its C terminal that allows special protein-ClpX binding. ClpX
pulled and linearized Smt3 within the nanopore, resulting in
progressive ionic current decreases followed by a rapid return to
the open pore current. Further study applied unfoldase ClpXP to
modulate sequential unfolding of the structured S2-GT protein
that is composed of one green fluorescent protein (GFP), one titin
fragment (titin I27) and two Smt3.[61] Similarly, incorporation of a
charged polymer tag enables the electrically driven stretching and
traversing of proteins across the nanopore. In recent studies, an
oligonucleotide poly(dC)30 was tagged to support the multistep
unfolding and unidirectional translocation of a model thioredoxin
through the α-HL nanopore (Figure 3e).[99–101]

Accepted Manuscript
3. Controlling of Peptide Translocations

Figure 2. Translocation velocities of peptides through commonly used biological The translocations and trapping times of charged peptides
nanopores. Each data point was estimated from the reported longest residences could be actively or passively controlled by modulating magnitude
of peptides. Related references were shown in the right parentheses, where
and polarity of the applied voltages. As illustated in Figure 4a,
asterisk indicate the capability for identification of single AA difference.
Compared to the results obtained with wild-type (WT) biological nanopores electrophoresis facilated a negatively charged peptide (YEQ) 3,
(open circles),[25,28,29,44,48,54,66,67,69,73,75–81] translocation velocities of peptides while hindered a positively charged peptide (YKQ)3 to enter and
have been remarkably reduced by designing mutant nanopores (red solid migration within the AeL nanopore.[78] In this context, a feasible
circles),[30,70,82,83] introducing dipolar-like peptide structures (orange solid
idea to slow down peptide journey is to modulate charge
triangles),[57,58,84–86] and optimizing solution conditions, such as temperatures
(blue solid diamonds)[87,88] and pH (violet solid pentagons).[89] Ideal velocities in distributions of the molecule itself. [84,86,89,102] In recent works,
suggested region were coloured in light blue. oppositely charged tails were introduced to create a dipolar
feature to control peptide sojourn (e.g. CP2a, N’-(E)12-(N)12-(R)12-
C’) inside the α-HL nanopore via alteration of dielectrophoresis
protein structures. For instance, Congo red (CR) was used as an (Figure 4b).[84,89] A dipolar-like peptide can stall within a nanopore
inhibitor to prevent β-Amyloid 42 (Aβ 42) aggregation into fibrils at a fine applied voltage when dielectrophoresis compensates for
and plaques (Figure 3a).[90] Moreover, an accessible option is to electroosmotic flow (EOF).
use the denaturants to open the ready protein folds. Guanidine Unlike the electrophoretic-driven translocations of DNAs, the
hydrochloride (Gdm-HCl) was employed to unfold the WT combined electrophoresis and EOF accounts for the transports of
maltose-binding protein (MalEwt) and a variant MalE219.[91–93] As peptides/proteins with non-uniform charge distributions.[29,103–106]
Gdm-HCl increasing to ~1.0 M, the α-HL and AeL nanopores Acceptable methods have been proposed to prolong peptide
dictated only long blockades for partially folded proteins, and short residence by balancing the electroosmotic and electrophoretic
blockades for completely unfolded proteins (Figure 3b).[92] driving forces. For instance, the concerted action of EOF and
Previous cases elucidated high-concentration (≥ 2.0 M) urea can electrophoresis decelerated the movement of peptide CAMA P6
degrade proteins.[94] Despite availability and simplicity, a concern (N’-KWKLFKKIGIGKFLQSAKKF-C’) by carefully tuning the
is protein nanopore denaturation under the extreme denaturing solution pH.[103,104] By lowering neutral pH below 5, EOF was
conditions (e.g. >1.5 M Gdm-HCl or 4 M urea).[93,95,96] augmented due to charge modulation of the α-HL nanopore
Alternative strategies were proposed. In Figure 3c, MalE219 induced by protonation of crucial residues (e.g. aspartic acid). The
thermally unfolded when improving temperatures from 20 to 70 ℃, enhanced EOF played against electrophoresis to prevent the
revealing a same melting temperature of ~45 ℃ using the α-HL trans-to-cis oriented motion of CAMA P6, leading to current
and AeL nanopores.[97] However, high temperatures would oscillations of between B1 and B2 states caused by the back-and-
destabilize lipid bilayer and accelerate molecular dynamics. The forth migrations of peptide from pore vestibule to β-barrel domain
fast translocation of peptides hinders the accurate current (Figure 4c).[104] Similarly, several reports have revealed that
recording due to the resolution limit of the existing nanopore proteins could be trapped inside biological nanopores (e.g. ClyA
instruments. Recent studies manifested that the hairpin-like nanopore) for several seconds up to tens of minutes via EOF
structures of peptides were destabilized by regulating protonation enhancement.[105–107]
of AA residues (e.g. histidine) at acidic solution (e.g. pH = 4.5).[53] Another potentially powerful approach is to manipulate the
Additionally, protein folding states would change by modulating local interactions between peptide and nanopore. In Figure 4d,
specific binding between metal ions and AA residues (e.g. Cu2+- the engineered M113F and 2FN (M113F/K147N/T145F) α-HL
histidine).[66] nanopores were devised to decrease the translocation velocity of
To ratchet a protein through the nanopore progressively, early peptide (Y)6 by introducing an additional aromatic interaction.[82] A
attempts incorporated a motor enzyme to mediate the unfolding T232K/K238Q mutant AeL nanopore was recently reported to trap
and translocation of native proteins. For example, a AAA+ tau peptide (N’-SKIGSTENL-C’) for a longer time than the WT AeL
unfoldase ClpX, placed in the trans compartment, was employed nanopore, where the T232K site was designed to increase the
to mechanically denature the Smt3 protein perched above the electrostatic interaction with charged peptides, and the K238Q
vestibule of the α-HL nanopore (Figure 3d).[98] Smt3 was modified site to enhance the repelling barrier (Figure 4e). [30] Moreover, the

4
This article is protected by copyright. All rights reserved.
Angewandte Chemie 10.1002/ange.202013462

REVIEW

Accepted Manuscript
Figure 3. Strategies towards the nanopore unfolding of a single protein. (a) Aggregation transitions of β-Amyloid 42 (Aβ42) in the presence of promoter β-cyclodextrin
(β-CD) and inhibitor Congo red, respectively. (b) Denaturing agent-assisted unfolding of a maltose-binding protein variant (MalE219). (c) Thermal unfolding of
MalE219. A same melting temperature (around 45 °C) was obtained by the AeL and α-HL nanopores. (d) Unfoldase ClpX-mediated protein Smt3 unfolding and
translocation. The model protein contains a Smt3 domain (green) at its N-terminus, a negatively charged flexible polyGSD linker (yellow), and a ClpX-targeting ssrA
tag (red) at its C terminus. During the translocation, (i) Empty α-HL nanopore shows open current of ~34 ± 2 pA; (ii) capture of Smt3 leads to an ionic current drop
to ~14 pA; (iii) ClpX-controlled Smt3 unfolding results in a progressive current ramp reaching ~10 pA and (iv) a terminated current reduction to ~3.8 pA; (i') Smt3
migration into the trans solution with an abrupt increase to the open nanopore current. (e) Co-translocational unfolding of thioredoxin (Trx). A model Trx is attached
to a poly(dC)30 DNA leader via a cysteine at its either N- or C-terminus. The representative current trace indicates the four-step unfolding process: step 1→2, DNA
leader threads into the α-HL nanopore; step 2→3, Trx unfolds partially with linearized polypeptide inside the nanopore; step 3→4, Trx unfolds completely with the
remained polypeptide translocating through the nanopore; and step 4→1, unfold Trx leaves the nanopore. Figures reproduced with permission from: a, ref. 90,
Copyright 2011, American Chemical Society; b, ref. 92, Copyright 2012, American Chemical Society; c, ref. 97, Copyright 2012, American ChemicalFigu Society;
d, ref. 98, Copyright 2013, Nature Publishing Group; e, ref. 100, Copyright 2014, Macmillan Publishers Limited.

unphosphorylated and phosphorylated tau peptides were spatiotemporal sensitivity to identify 20 proteinogenic AAs and
identified with nearly 100% accuracy. To our best knowledge, this diverse PTMs. Recent years have witnessed many encouraging
mutant AeL nanopore gave the slowest translocation velocity of developments in nanopore proteomics toward SMPS. For
peptides (about 5-70 ms aa-1) without modulation of solution example, six arginine peptides (5-10 aa in length) differing by a
environment. Likewise, transport properties of peptides/proteins single AA were resolved independently or in equimolar mixture
could be modulated by manipulating nanopore geometry and using a WT AeL nanopore (Figure 5a). [80] The residence of
diameter of the nanopore sensing region.[28,31,108] cationic peptides were selectively increased by nearly 3 times with
the adoption of an anionic gold cluster Au25(SG)18, accompanying
by a nearly 2-fold reduction in current fluctuations via the
4. Identification of Amino Acids regulation of solution conditions (pH or chaotropic salt).[76]
Another noticeable attempt is the detection of 20 natural AAs
Although sequencing 4 natural deoxyribonucleotides has been using the AeL nanopore (Figure 5b). [44] In this work, a varied
achieved, biological nanopore techniques still lack efficient terminal “X” was chemically attached to an arginine heptapeptide,
which acts as a polycationic carrier to ensure the unidirectional

5
This article is protected by copyright. All rights reserved.
Angewandte Chemie 10.1002/ange.202013462

REVIEW

Accepted Manuscript
Figure 4. Electrical recording of peptide translocation through a biological nanopore. (a) AeL nanopore detection of the negatively charged (YEQ)3 and positively
charged (YKQ)3 peptides. Left: Electrophoresis-facilitated translocations of (YEQ)3 induce large amounts of current blockades. Right: Electrophoresis-hindered
captures of (YKQ)3 cause only a few transient spikes. (b) Engineered dipolar-like peptide CP2a detected using a single α-HL nanopore. The sequence of CP2a is
N’-(E)12-(N)12-(R)12-C’. In neutral solution, the residence time of CP2a increases due to the balanced electrophoretic forces on the oppositely charged segments at
its N- and C-terminus. (c) EOF-modulated translocation of peptide CAMA P6. At acidic pH values, EOF is augmented to oppose electrophoresis, contributing to a
slow-down peptide moving for capable detection of B1 and B2 sub-states of the positively charged CAMA P6. (d) Study of amyloid-β peptide transport through α-HL
nanopores. Left: Molecular graphics illustration of the WT α-HL nanopore (PDB ID code: 7AHL). Right: Typical current traces of peptide (Y)6 traversing across the
WT, M113F and M113F/147N/T145F (2FN) mutant α-HL nanopores, respectively. Left: The residence of (Y)6 is prolonged due to the additional aromatic interactions
between (Y)6 and engineered phenylalanine (F) sites within the nanopore sensing interface. (e) Mapping phosphorylation sites of tau peptides with the T232K/K238Q
mutant AeL nanopore. Upper left: The T232K/K238Q mutant nanopore was designed to introduce an enhanced electrostatic interaction at T232K site and an
increased repelling barrier at K238Q site. Upper right: Well-separated scatter plots of current and duration for tau peptide (green), pS262-tau peptide (red), pT263-
tau peptide (blue), and pS262/pT263-tau peptide (yellow), respectively. Bottom: The typical current traces reveal longer translocation times of WT, pS262- and
pT263-tau peptides in the T232K/K238Q mutant AeL nanopore than WT AeL nanopore. Figures reproduced with permission from: a, ref. 78, Copyright 2019, Wiley-
VCH; b, ref. 84, Copyright 2015, Nature Publishing Group; c, ref. 104, Copyright 2014, Nature Publishing Group; d, ref. 82, Copyright 2009, American Chemical
Society; e, ref. 30, Copyright 2020, WILEY-VCH.

entry and travel of XR7 peptides. To obtain an exquisite sensitivity, Up to now, several groups have realized nanopore sensing of
“X” was electrically confined within the small nanopore sensing peptide/protein variants.[24–26,30,57–60,63,66] As depicted in Figure 5d,
volume for up to tens of milliseconds, allowing AeL identification Cys and homocysteine (Hcy) incorporated to 5’-benzaldehyde
of 13 natural AAs (i.e. R, W, F, K, L, N, T, P, D, A, C, S and G) poly(dA)4 (AHA4) produced readable current responses and long
assigned into small subgroups. Despite satisfied identification of duration (~10 ms) when confined inside the K238Q mutant AeL
the electrically charged family (RR7, KR7, HR7, ER7 and DR7) in a nanopore.[60] Another work adopted a dipolar structure consisting
mixed solution, it is still a great difficulty to recognize all the 20 of a negative E10 N-terminus and a positive R10 C-terminus to
natural AAs in an equimolar mixture. orient and slow down peptide passage across the Fragaceatoxin
At the same time, efforts were devoted to direct identification C (FraC) nanopore, where the balanced EOF and electrophoresis
of single AAs without any attachments. In an initial attempt, single are responsible for the distinction of unmodified, phosphorylated
cysteine (Cys, 0.11 nm3) produced prolonged distinctive current and glycosylated serine (S) in an equilibrium position 11 (from the
blockades (e.g. 0.11 ±0.02 ms at 120 mV) when traversing across N-terminus) (Figure 5e).[57]
a WT AeL nanopore (Figure 5c).[79] While asparagine (Asn, 0.12
nm3) and glutamine (Gln, 0.15 nm3) with larger volumes induced
only undistinguishable current variations, suggesting a relatively 5. Applications in Disease Detection
less dominant role of steric hindrance effect for single AA sensing.
Other factors, especially the specific inter-residue interactions, On the way to the ultimate goal of NPS, increasing studies
should be considered to improve nanopore sensitivity.[43,109] As unravel potential opportunities of nanopore proteomics, especially
mentioned before, the T232K/K238Q mutant AeL nanopore has recently reported applications aiming at disease diagnostics. For
achieved differentiation of non-, mono- and di-phosphorylated tau example, structural transformation of α-synuclein (α-syn) was
peptides by regulating inter-residue electrostatic interactions.[30] probed during its early stage fibrillation, which suggests as a

6
This article is protected by copyright. All rights reserved.
Angewandte Chemie 10.1002/ange.202013462

REVIEW

Accepted Manuscript
Figure 5. Identification of single amino acid differences with a biological nanopore. (a) Detection of arginine peptides of different lengths in an equimolar mixture
using the AeL nanopore. Top right: Setup of the AeL nanopore experiments (not to scale). Top left: Illustration of a typical current blockade event. Below: Histogram
of residual current blockages of arginine peptides consisting of 5, 6, 7, 8, 9, and 10 AAs (from right to left). (b) Recognition of terminated proteinogenic amino acids
(X) in peptide XR7. Left: Mean values of experimental Ib/I0 assigned to twenty XR7 obtained by the AeL nanopore. Right: Constructs of peptide X R7, where an AA of
interest (X) is linked to a polycationic carrier R7. (c) Analysis of single amino acids with the AeL nanopore. Left: Schematic of a single cysteine traversing across an
AeL nanopore. Right: Raw current traces of cysteine (Cys), asparagine (Asn) and glutamine (Gln) (from top to bottom), respectively. The red star denotes a typical
blockades event of Cys in the inset. (d) Discrimination of cysteine (Cys) and homocysteine (Hcy) with the K238Q mutant AeL nanopore. Cys/Hcy is labeled by a
specific 5’-benzaldehyde poly(dA)4 (AHA4) probe. Left: Scheme of K238Q AeL nanopore analysis of AHA4, AHA4-C and AHA4-H. Right: Representative blockade
events of single AHA4-H, AHA4-C and AHA4 (from left to right), respectively. (e) Phosphorylation and glycosylation detection with the FraC nanopore. Left:
Schematic representation of unmodified and modified peptides measured by a FraC nanopore. Right: Histogram of the relative blockade of the unmodified,
phosphorylated and glycosylated peptides, respectively. Peptide sequence is N’-(E)10-XGSGSGSKGS-(R)10-C’, where X represents serine (S) or its variants. Figures
reproduced with permission from: a, ref. 80, Copyright 2018, Nature Publishing Group; b, ref. 44, Copyright 2020, Nature Publishing Group; c, ref. 79, Copyright
2020, The Royal Society of Chemistry; d, ref. 60, Copyright 2019, The Royal Society of Chemistry; e, ref. 57, Copyright 2019, American Chemical Society.

pathological hallmark of Parkinson’s disease (Figure 6a). [110] If protein kinase A (PKA), which resulted in serine phosphorylation
captured inside an α-HL nanopore, the natively unfolded α-syn of the substrate peptide (S-peptide, N’-LRRASLG-C’).[67] In this
monomer either transiently translocated through the nanopore work, PKA activity was quantified by analysing the relative event
lumen, or transformed into partially folded intermediate to exit frequency f/f1 μM of the phosphorylated peptide (P-peptide, N’-
from the nanopore vestibule. Similarly, the α-HL nanopore was LRRApSLG-C’), where f1 μM corresponds to the enhanced event
employed to investigate Alzheimer’s disease (AD)-associated Aβ frequency resulting from the interior label of 1 μM P-peptide.
aggregation, revealing that the amyloidosis process of Aβ is Further nanopore assay manifested that H-89 inhibited PKA
affected by its initial structure features and could be inhibited by activity with a half-maximal inhibitory concentration (IC50) of 104
β-CD.[54,90] The WT and mutated FraC nanopores were adopted nM, while the combination of FsK and IBMX triggered PKA
to detect peptide/protein biomarkers.[28,29] An engineered W116S activation in MCF-7 cell lysates.
FraC nanopore discriminated angiotensin I (Ang I), II (Ang II), III Synergistic binding of Pim-1 kinase to MgATP and a peptide
(Ang III) and IV (Ang IV) peptides simultaneously in a mixture, sensor attached to an engineered α-HL nanopore was measured,
which are key players in regulating blood pressure and the fluid which yielded pseudo-first-order association rate constants k’+2 of
balance (Figure 6b).[28] 60 ± 10 μM for the Pim-1-sensor binary complex and k’+3 of 35 ±
Nanopore sensing of protein kinases is another burgeoning 5 μM for the Pim-1-MgATP-sensor ternary complex (Figure 6d).[72]
area, which is potential therapeutic targets for many diseases The ATP-competitive Pim kinase inhibitor 1 was titrated to inhibit
including cancers and leukemias. As shown in Figure 6c, the AeL Pim-1 against the Pim-1-sensor complex formation. An inhibition
nanopore system was utilized to directly monitor the catalysis of constant of 130 ± 30 nM was obtained for inhibitor 1 in the

7
This article is protected by copyright. All rights reserved.
Angewandte Chemie 10.1002/ange.202013462

REVIEW

Accepted Manuscript
Figure 6. Applications in disease diagnostics using biological nanopores. (a) Characterization of early fibrillation from natively unfolded α-syn monomer to partially
folded α-syn intermediate via an α-HL nanopore. Transient states between block current level Lb and capture blockade current level Lc might be produced by the
elongation/ordering of α-syn intermediate inside nanopore. (b) Simultaneous measurement of angiotensin (Ang) peptide biomarkers with the engineered W116S
FraC nanopores. Ang I, II, III and IV peptides could be discriminated directly by the magnitudes of ionic current blockades. (c) Evaluation of PKA activity through
the aerolysin nanopore. Top: nanopore sensing schematic and current traces of S-peptide and PKA-catalyzed P-peptide. Below: quantitative analysis of PKA
inhibition by the kinase inhibitor H-89, and PKA activation in MCF-7 cells stimulated with Fsk/IBMX. Here, f/f1 μM suggests the relative event frequency of P-peptide
to reduce the frequency errors among different experiments, where f indicates P-peptide event frequency in an analyzed sample, f1 μM represents the increased
event frequency generated by the interior label of 1 μM P-peptide. (d) Investigation of Pim-1 kinase kinetics with an engineered α-HL nanopore sensor. Above:
illustration of the novel heptameric α-HL nanopore containing a single functionalized subunit, which bears a serine/glycine linker, a pseudosubstrate peptide sensor
and a TEV protease recognition site (“TEV RS”). Middle: kinetic model for synergistic binding of Pim-1 and MgATP to the peptide sensor. Bottom: determination of
Pim-1 inhibition caused by an ATP-competitive kinase inhibitor 1 against Pim-1-sensor interaction. Figures reproduced with permission from: a, ref. 110, Copyright
2013, American Chemical Society; b, ref. 28, Copyright 2019, Nature Publishing Group; c, ref. 67, Copyright 2019, American Chemical Society; d, ref. 72, Copyright
2015, Wiley-VCH.

presence of 162 nM Pim-1, 100 μM ATP and 10 mM MgCl2. A alteration of the experimental conditions (e.g. electrical field, pH
more recent work investigated the reversible phosphorylation/ and temperature). The translocation velocities of peptides have
dephosphorylation of the sensor peptide induced by Pim-1 and a been reduced by ~1-4 orders of magnitude with mutant biological
protein phosphatase.[74] In addition, an engineered phi29 nanopores (Figure 2). A direct consequence is the discrimination
nanopore was incorporated with an Epithelial Cell Adhesion of peptides differing by a single AA. However, it is still difficult to
Molecule (EpCAM) peptide probe to specifically detect EpCAM develop a protein sequencer, because the nanopore sensing
antibody (Ab) in the serum, demonstrating the feasibility and volume is too large for AAs (e.g. glycine of 0.06 nm3), such as the
versatility of biological nanopores for disease diagnostics and AeL nanopore with a sensing region of ~2.4 nm and inner
medical utilization.[64] diameter of ~ 1 nm.[44,111,112] Moreover, the interaction difference
between the nanopore and single AAs is not sufficient to produce
the corresponding current identities. In this aspect, site-directed
6. Summary and Outlook manipulation of the nanopore sensing interface has appeared to
improve spatiotemporal sensitivity. Advances in electrochemical
This review has summarised the undergoing methodologies to instruments with lower noise and higher bandwidth would also
promote the developments of NPS, including modification of benefit protein profiling with a higher signal-to-noise ratio.
target peptides/proteins, engineering of biological nanopores, and Compared to biological nanopores, solid-state nanopores

8
This article is protected by copyright. All rights reserved.
Angewandte Chemie 10.1002/ange.202013462

REVIEW
could provide complementary options for peptide/protein sensing Keywords: protein sequencing • biological nanopore • single-
in hash conditions because they have evolved broad molecule sensing interface • nanopore electrochemistry •
environmental tolerances including voltage, temperature, pH and disease diagnostics
denaturants.[113–116] Hence, integrated nanopore arrays could be
developed for high-throughput parallel protein detection by [1] P. Edman, Acta Chem. Scand. 1950, 4, 283–293.
combining biological nanopore sequencer with scalable solid- [2] A. W. Brauer, C. L. Oman, M. N. Margolies, Anal. Biochem. 1984, 137,
state nanopores.[117,118] Moreover, a deep dialogue between 134–142.
nanopore and other on-going methods, such as fluorescent [3] G. Allen, Sequencing of Proteins and Peptides in Laboratory Techniques
fingerprinting and tunneling currents, will expand the opportunities in Biochemistry and Molecular Biology, Vol. 9 (Eds: T. S. Work, R. H.
for SMPS.[119–121] A feasible corporation may lead to a nanopore Burdon), Elsevier, Amsterdam, 1981, pp. 174–186.
[4] J. R. Yates, J. Am. Chem. Soc. 2013, 135, 1629–1640.
coupled multi-compartment device to simultaneously monitor the
[5] J. D. Chapman, D. R. Goodlett, C. D. Masselon, Mass Spectrom. Rev.
transversal tunneling current and longitudinal ionic blocked
2014, 33, 452–470.
current for highly accurate SMPS.[122,123]

Accepted Manuscript
[6] B. T. Chait, Science 2006, 314, 65–66.
Concerted efforts in interdisciplinary areas will open up new [7] R. A. Zubarev, Proteomics 2013, 13, 723–726.
possibility for NPS. Recently, researches in the fields of protein [8] E. A. Ponomarenko, E. V. Poverennaya, E. V. Ilgisonis, M. A. Pyatnitskiy,
engineering and computational enzyme design have collaborated A. T. Kopylov, V. G. Zgoda, A. V. Lisitsa, A. I. Archakov, Int. J. Anal.
to create an enzyme termed as “Edmanase”, which is capable of Chem. 2016, 2016, 7436849.
step-wise cleaving off N-terminal AAs under biocompatible [9] B. Huang, H. Wu, D. Bhaya, A. Grossman, S. Granier, B. K. Kobilka, R.
N. Zare, Science 2007, 315, 81–84.
conditions.[124] This creation provides experimental evidence for
[10] J. P. Savaryn, A. D. Catherman, P. M. Thomas, M. M. Abecassis, N. L.
the feasibility of exopeptidase-based NPS, where the removed
Kelleher, Genome Med. 2013, 5, 53.
AAs are identified one-by-one by the nanopore at a controlled [11] T. E. Angel, U. K. Aryal, S. M. Hengel, E. S. Baker, R. T. Kelly, E. W.
pace. Considering the complexity of large amounts of nanopore Robinson, R. D. Smith, Chem. Soc. Rev. 2012, 41, 3912–3928.
data, machine learning algorithms should be included in the future [12] Y.-L. Ying, Y.-T. Long, J. Am. Chem. Soc. 2019, 141, 15720–15729.
repertoire to efficiently decode current readouts for reconstruction [13] L. Restrepo-Pérez, C. Joo, C. Dekker, Nat. Nanotechnol. 2018, 13, 786–
of the original protein sequences. 796.
Avenues towards SMPS with biological nanopores are full of [14] B. Cressiot, L. Bacri, J. Pelta, Small Methods 2020, 2000090.
[15] A. Asandei, G. Di Muccio, I. Schiopu, L. Mereuta, I. S. Dragomir, M.
challenges and opportunities. Next decade will highlight the cost-
Chinappi, T. Luchian, Small Methods 2020, 1900595.
effective and highly parallelized NPS. Theoretical, methodological
[16] J. J. Kasianowicz, E. Brandin, D. Branton, D. W. Deamer, Proc. Natl.
and technical innovations are on the horizon to improve sensitivity, Acad. Sci. 1996, 93, 13770–13773.
selectivity and reliability of NPS, and therefore would facilitate [17] A. Meller, L. Nivon, D. Branton, Phys. Rev. Lett. 2001, 86, 3435–3438.
label-free, multiplexing and high-throughput profiling of proteome [18] Y. Wang, L.-Q. Gu, K. Tian, Nanoscale 2018, 10, 13857–13866.
in individual cells. If realized, NPS will undoubtedly bring a great [19] S. Howorka, Z. Siwy, Chem. Soc. Rev. 2009, 38, 2360–2384.
revolution to single-cell proteomics in understanding phenotypic [20] Y. T. Long, M. N. Zhang, Sci. China Ser. B-Chem. 2009, 52, 731–733.
identities of cells, unbiased proteome imaging of cellular [21] N. Varongchayakul, J. Song, A. Meller, M. W. Grinstaff, Chem. Soc. Rev.
2018, 47, 8512–8524.
heterogeneity, studying dynamic cellular development and etc.
[22] H. Bayley, Clin. Chem. 2015, 61, 25–31.
The innovative technique of nanopore electrochemistry could also
[23] T. Osaki, H. Suzuki, B. Le Pioufle, S. Takeuchi, Anal. Chem. 2009, 81,
be extended to clinical applications, such as discrimination of 9866–9870.
PTMs and protein biomarkers, offering new insights in protein [24] C. B. Rosen, D. Rodriguez-Larrea, H. Bayley, Nat. Biotechnol. 2014, 32,
function/dysfunction at a single AA level. If combined NPS with 179–181.
single-molecule DNA/RNA sequencing, an unprecedented view [25] Y.-L. Ying, J. Yang, F.-N. Meng, S. Li, M.-Y. Li, Y.-T. Long, Research
will be provided for molecular diagnostics and personalized 2019, 2019, 1050735.
medicine. [26] M. A. Fahie, M. Chen, J. Phys. Chem. B 2015, 119, 10198–10206.
[27] C. Wloka, V. Van Meervelt, D. Van Gelder, N. Danda, N. Jager, C. P.
Williams, G. Maglia, ACS Nano 2017, 11, 4387–4394.
[28] G. Huang, A. Voet, G. Maglia, Nat. Commun. 2019, 10, 835.
Acknowledgements [29] G. Huang, K. Willems, M. Soskine, C. Wloka, G. Maglia, Nat. Commun.
2017, 8, 935.
This work is supported by the National Natural Science [30] S. Li, X. Y. Wu, M. Y. Li, S. C. Liu, Y. L. Ying, Y. T. Long, Small Methods
2020, 2000014.
Foundation of China (21834001 and 21922405), the China
[31] M. Soskine, A. Biesemans, M. De Maeyer, G. Maglia, J. Am. Chem. Soc.
Postdoctoral Science Foundation (2020M680066), the Excellent 2013, 135, 13456–13463.
Research Program of Nanjing University (ZYJH004) and the [32] J. Clarke, H.-C. Wu, L. Jayasinghe, A. Patel, S. Reid, H. Bayley, Nat.
Fundamental Research Funds for the Central Universities Nanotechnol. 2009, 4, 265–270.
(14380239). Yi-Lun Ying is sponsored by National Ten Thousand [33] E. A. Manrao, I. M. Derrington, A. H. Laszlo, K. W. Langford, M. K.
Talent Program for young top-notch talent. We specially thank the Hopper, N. Gillgren, M. Pavlenok, M. Niederweis, J. H. Gundlach, Nat.
kind help on the figure drawing from Kai-Li Xin, Chao-Nan Yang, Biotechnol. 2012, 30, 349–353.
[34] D. Deamer, M. Akeson, D. Branton, Nat. Biotechnol. 2016, 34, 518–524.
Drs. Qiang Zhu and Shao-Chuang Liu.
[35] G. M. Cherf, K. R. Lieberman, H. Rashid, C. E. Lam, K. Karplus, M.
Akeson, Nat. Biotechnol. 2012, 30, 344–348.
[36] L. Song, M. R. Hobaugh, C. Shustak, S. Cheley, H. Bayley, J. E. Gouaux,
Conflict of interest Science 1996, 274, 1859–1866.
[37] M. Faller, M. Niederweis, G. E. Schulz, Science 2004, 303, 1189–1192.
[38] I. Iacovache, S. De Carlo, N. Cirauqui, M. Dal Peraro, F. G. van der Goot,
The authors declare no conflict of interest.
B. Zuber, Nat. Commun. 2016, 7, 12062.

9
This article is protected by copyright. All rights reserved.
Angewandte Chemie 10.1002/ange.202013462

REVIEW
[39] K. Tanaka, J. M. M. Caaveiro, K. Morante, J. M. González-Mañas, K. [75] M. G. Larimi, L. A. Mayse, L. Movileanu, ACS Nano 2019, 13, 4469–
Tsumoto, Nat. Commun. 2015, 6, 6337. 4477.
[40] Z. Gu, Y. L. Ying, Y. T. Long, Sci. China Chem. 2018, 61, 1483–1485. [76] A. E. Chavis, K. T. Brady, G. A. Hatmaker, C. E. Angevine, N.
[41] J. J. Kasianowicz, A. K. Balijepalli, J. Ettedgui, J. H. Forstater, H. Wang, Kothalawala, A. Dass, J. W. F. Robertson, J. E. Reiner, ACS Sensors
H. Zhang, J. W. F. Robertson, Biochim. Biophys. Acta - Biomembr. 2016, 2017, 2, 1319–1328.
1858, 593–606. [77] F. Hu, B. Angelov, S. Li, N. Li, X. Lin, A. Zou, ChemBioChem 2020, DOI
[42] S. J. Perkins, Eur. J. Biochem. 1986, 157, 169–180. 10.1002/cbic.202000119.
[43] G. Di Muccio, A. E. Rossini, D. Di Marino, G. Zollo, M. Chinappi, Sci. Rep. [78] S. Li, C. Cao, J. Yang, Y. T. Long, ChemElectroChem 2019, 6, 126–129.
2019, 9, 13130. [79] B. Yuan, S. Li, Y. L. Ying, Y. T. Long, Analyst 2020, 145, 1179–1183.
[44] H. Ouldali, K. Sarthak, T. Ensslen, F. Piguet, P. Manivet, J. Pelta, J. C. [80] F. Piguet, H. Ouldali, M. Pastoriza-Gallego, P. Manivet, J. Pelta, A.
Behrends, A. Aksimentiev, A. Oukhaled, Nat. Biotechnol. 2020, 38, 176– Oukhaled, Nat. Commun. 2018, 9, 966.
181. [81] Z. Ji, S. Wang, Z. Zhao, Z. Zhou, F. Haque, P. Guo, Small 2016, 12,
[45] W. Timp, G. Timp, Sci. Adv. 2020, 6, eaax8978. 4572–4578.
[46] R. Apweiler, H. Hermjakob, N. Sharon, Biochim. Biophys. Acta - Gen. [82] Q. Zhao, D. A. Jayawardhana, D. Wang, X. Guan, J. Phys. Chem. B 2009,

Accepted Manuscript
Subj. 1999, 1473, 4–8. 113, 3572–3578.
[47] T. C. Sutherland, Y. T. Long, R. I. Stefureac, I. Bediako-Amoa, H. B. [83] J. Wang, J. A. Bafna, S. P. Bhamidimarri, M. Winterhalter, Angew. Chem.
Kraatz, J. S. Lee, Nano Lett. 2004, 4, 1273–1277. Int. Ed. 2019, 58, 4737–4741.
[48] L. Movileanu, J. P. Schmittschmitt, J. M. Scholtz, H. Bayley, Biophys. J. [84] A. Asandei, M. Chinappi, J. K. Lee, C. H. Seo, L. Mereuta, Y. Park, T.
2005, 89, 1030–1045. Luchian, Sci. Rep. 2015, 5, 10419.
[49] R. Stefureac, Y. Long, H.-B. Kraatz, P. Howard, J. S. Lee, Biochemistry [85] A. Asandei, A. E. Rossini, M. Chinappi, Y. Park, T. Luchian, Langmuir
2006, 45, 9172–9179. 2017, 33, 14451–14459.
[50] A. J. Wolfe, M. M. Mohammad, S. Cheley, H. Bayley, L. Movileanu, J. [86] S. Zhao, L. Restrepo-Pérez, M. Soskine, G. Maglia, C. Joo, C. Dekker,
Am. Chem. Soc. 2007, 129, 14034–14041. A. Aksimentiev, ACS Nano 2019, 13, 2398–2409.
[51] V. Van Meervelt, M. Soskine, G. Maglia, ACS Nano 2014, 8, 12826– [87] U. Lamichhane, T. Islam, S. Prasad, H. Weingart, K. R. Mahendran, M.
12835. Winterhalter, Eur. Biophys. J. 2013, 42, 363–369.
[52] N. S. Galenkamp, A. Biesemans, G. Maglia, Nat. Chem. 2020, 12, 481– [88] K. R. Mahendran, U. Lamichhane, M. Romero-Ruiz, S. Nussberger, M.
488. Winterhalter, J. Phys. Chem. Lett. 2013, 4, 78–82.
[53] L. Mereuta, A. Asandei, C. H. Seo, Y. Park, T. Luchian, ACS Appl. Mater. [89] A. Asandei, M. Chinappi, H. K. Kang, C. H. Seo, L. Mereuta, Y. Park, T.
Interfaces 2014, 6, 13242–13256. Luchian, ACS Appl. Mater. Interfaces 2015, 7, 16706–16714.
[54] Y.-X. Hu, Y.-L. Ying, Z. Gu, C. Cao, B.-Y. Yan, H.-F. Wang, Y.-T. Long, [90] H. Y. Wang, Y. L. Ying, Y. Li, H. B. Kraatz, Y. T. Long, Anal. Chem. 2011,
Chem. Commun. 2016, 52, 5542–5545. 83, 1746–1752.
[55] V. Van Meervelt, M. Soskine, S. Singh, G. K. Schuurman-Wolters, H. J. [91] G. Oukhaled, J. Mathé, A. L. Biance, L. Bacri, J. M. Betton, D. Lairez, J.
Wijma, B. Poolman, G. Maglia, J. Am. Chem. Soc. 2017, 139, 18640– Pelta, L. Auvray, Phys. Rev. Lett. 2007, 98, 158101.
18646. [92] C. Merstorf, B. Cressiot, M. Pastoriza-Gallego, A. Oukhaled, J. M. Betton,
[56] S. Wang, Z. Zhou, Z. Zhao, H. Zhang, F. Haque, P. Guo, Biomaterials L. Auvray, J. Pelta, ACS Chem. Biol. 2012, 7, 652–658.
2017, 126, 10–17. [93] M. Pastoriza-Gallego, L. Rabah, G. Gibrat, B. Thiebot, F. G. Van Der
[57] L. Restrepo-Pérez, C. H. Wong, G. Maglia, C. Dekker, C. Joo, Nano Lett. Goot, L. Auvray, J. M. Betton, J. Pelta, J. Am. Chem. Soc. 2011, 133,
2019, 19, 7957–7964. 2923–2931.
[58] L. Restrepo-Pérez, G. Huang, P. R. Bohländer, N. Worp, R. Eelkema, G. [94] M. Pastoriza-Gallego, G. Gibrat, B. Thiebot, J.-M. Betton, J. Pelta,
Maglia, C. Joo, C. Dekker, ACS Nano 2019, 13, 13668–13676. Biochim. Biophys. Acta - Biomembr. 2009, 1788, 1377–1386.
[59] A. J. Boersma, H. Bayley, Angew. Chem. Int. Ed. 2012, 124, 9744–9747. [95] R. K. Bortoleto, R. J. Ward, FEBS Lett. 1999, 459, 438–442.
[60] Y. Lu, X. Y. Wu, Y. L. Ying, Y. T. Long, Chem. Commun. 2019, 55, 9311– [96] M. Pastoriza-Gallego, G. Oukhaled, J. Mathé, B. Thiebot, J. M. Betton,
9314. L. Auvray, J. Pelta, FEBS Lett. 2007, 581, 3371–3376.
[61] J. Nivala, L. Mulroney, G. Li, J. Schreiber, M. Akeson, ACS Nano 2014, [97] L. Payet, M. Martinho, M. Pastoriza-Gallego, J. M. Betton, L. Auvray, J.
8, 12365–12375. Pelta, J. Mathé, Anal. Chem. 2012, 84, 4071–4076.
[62] Z. Ji, P. Guo, Biomaterials 2019, 214, 119222. [98] J. Nivala, D. B. Marks, M. Akeson, Nat. Biotechnol. 2013, 31, 247–250.
[63] M. A. Fahie, B. Yang, M. Mullis, M. A. Holden, M. Chen, Anal. Chem. [99] D. Rodriguez-Larrea, H. Bayley, Nat. Nanotechnol. 2013, 8, 288–295.
2015, 87, 11143–11149. [100] D. Rodriguez-Larrea, H. Bayley, Nat. Commun. 2014, 5, 4841.
[64] S. Wang, F. Haque, P. G. Rychahou, B. M. Evers, P. Guo, ACS Nano [101] J. Feng, P. Martin-Baniandres, M. J. Booth, G. Veggiani, M. Howarth, H.
2013, 7, 9814–9822. Bayley, D. Rodriguez-Larrea, Commun. Biol. 2020, 3, 159.
[65] D. Rotem, L. Jayasinghe, M. Salichou, H. Bayley, J. Am. Chem. Soc. [102] M. Chinappi, T. Luchian, F. Cecconi, Phys. Rev. E - Stat. Nonlinear, Soft
2012, 134, 2781–2787. Matter Phys. 2015, 92, 032714.
[66] I. Schiopu, S. Iftemi, T. Luchian, Langmuir 2015, 31, 387–396. [103] A. Asandei, I. Schiopu, M. Chinappi, C. H. Seo, Y. Park, T. Luchian, ACS
[67] F.-N. Meng, Y.-L. Ying, J. Yang, Y.-T. Long, Anal. Chem. 2019, 91, Appl. Mater. Interfaces 2016, 8, 13166–13179.
9910–9915. [104] L. Mereuta, M. Roy, A. Asandei, J. K. Lee, Y. Park, I. Andricioaei, T.
[68] A. Asandei, A. Apetrei, Y. Park, K. S. Hahm, T. Luchian, Langmuir 2011, Luchian, Sci. Rep. 2014, 4, 3885.
27, 19–24. [105] G. Huang, K. Willems, M. Bartelds, P. Van Dorpe, M. Soskine, G. Maglia,
[69] P. R. Singh, I. Bárcena-Uribarri, N. Modi, U. Kleinekathöfer, R. Benz, M. Nano Lett. 2020, 20, 3819–3827.
Winterhalter, K. R. Mahendran, ACS Nano 2012, 6, 10699–10707. [106] K. Willems, D. Ruić, A. Biesemans, N. S. Galenkamp, P. Van Dorpe, G.
[70] K. Sun, Y. Ju, C. Chen, P. Zhang, E. Sawyer, Y. Luo, J. Geng, Small Maglia, ACS Nano 2019, 13, 9980–9992.
Methods 2020, 1900892. [107] M. Soskine, A. Biesemans, G. Maglia, J. Am. Chem. Soc. 2015, 137,
[71] X. Chen, Y. Zhang, G. Mohammadi Roozbahani, X. Guan, ACS Appl. Bio 5793–5797.
Mater. 2019, 2, 504–509. [108] A. Biesemans, M. Soskine, G. Maglia, Nano Lett. 2015, 15, 6076–6081.
[72] L. Harrington, L. T. Alexander, S. Knapp, H. Bayley, Angew. Chem. Int. [109] M. M. Mohammad, L. Movileanu, Eur. Biophys. J. 2008, 37, 913–925.
Ed. 2015, 127, 8272–8277. [110] H. Y. Wang, Z. Gu, C. Cao, J. Wang, Y. T. Long, Anal. Chem. 2013, 85,
[73] Y. Wang, V. Montana, V. Grubišić, R. F. Stout, V. Parpura, L. Q. Gu, ACS 8254–8261.
Appl. Mater. Interfaces 2015, 7, 184–192. [111] C. Cao, M. Y. Li, N. Cirauqui, Y. Q. Wang, M. Dal Peraro, H. Tian, Y. T.
[74] L. Harrington, L. T. Alexander, S. Knapp, H. Bayley, ACS Nano 2019, 13, Long, Nat. Commun. 2018, 9, 2823.
633–641.

10
This article is protected by copyright. All rights reserved.
Angewandte Chemie 10.1002/ange.202013462

REVIEW
[112] Y. Q. Wang, M. Y. Li, H. Qiu, C. Cao, M. B. Wang, X. Y. Wu, J. Huang,
Y. L. Ying, Y. T. Long, Anal. Chem. 2018, 90, 7790–7794.
[113] C. Dekker, Nat. Nanotechnol. 2007, 2, 209–215.
[114] R. Hu, X. Tong, Q. Zhao, Adv. Healthc. Mater. 2020, 9, 2000933.
[115] L. Xue, H. Yamazaki, R. Ren, M. Wanunu, A. P. Ivanov, J. B. Edel, Nat.
Rev. Mater. 2020, DOI 10.1038/s41578-020-0229-6.
[116] Z. Yuan, C. Wang, X. Yi, Z. Ni, Y. Chen, T. Li, Nanoscale Res. Lett. 2018,
13, 56.
[117] B. Cressiot, S. J. Greive, M. Mojtabavi, A. A. Antson, M. Wanunu, Nat.
Commun. 2018, 9, 4652.
[118] H. Im, N. J. Wittenberg, A. Lesuffleur, N. C. Lindquist, S.-H. Oh, Chem.
Sci. 2010, 1, 688–696.
[119] Y. Yao, M. Docter, J. van Ginkel, D. de Ridder, C. Joo, Phys. Biol. 2015,
12, 55003.

Accepted Manuscript
[120] J. van Ginkel, M. Filius, M. Szczepaniak, P. Tulinski, A. S. Meyer, C. Joo,
Proc. Natl. Acad. Sci. 2018, 115, 3338– 3343.
[121] N. Callahan, J. Tullman, Z. Kelman, J. Marino, Trends Biochem. Sci.
2020, 45, 76–89.
[122] Y. Zhao, B. Ashcroft, P. Zhang, H. Liu, S. Sen, W. Song, J. Im, B. Gyarfas,
S. Manna, S. Biswas, C. Borges, S. Lindsay, Nat. Nanotechnol. 2014, 9,
466–473.
[123] M. Di Ventra, M. Taniguchi, Nat. Nanotechnol. 2016, 11, 117–126.
[124] B. Borgo, J. J. Havranek, Protein Sci. 2015, 24, 571–579.

11
This article is protected by copyright. All rights reserved.
Angewandte Chemie 10.1002/ange.202013462

REVIEW
Entry for the Table of Contents

Accepted Manuscript
Biological nanopore approach is promising for single-molecule protein sequencing (SMPS) by offering single-molecule confined
sensing interface to enable a high spatiotemporal resolution and an ultimate sensitivity. Here, we focus on recent advances in
biological nanopores from protein unfolding, peptide translocation, amino acid identification to diagnostic application, and close with
an outlook of interdisciplinary efforts for SMPS realization.

12
This article is protected by copyright. All rights reserved.

Das könnte Ihnen auch gefallen