scispace - formally typeset
Search or ask a question
Posted ContentDOI

Accessible LAMP-Enabled Rapid Test (ALERT) for detecting SARS-CoV-2

TL;DR: In this article, an accessible LAMP-enabled rapid test (ALERT) was proposed to detect viral RNA from nasal swabs or saliva with high sensitivity (0.1 to 2 viral particles/µl) and specificity (>97% True Negative Rate) utilizing reverse transcription loop-mediated isothermal amplification (RT-LAMP).
Abstract: The COVID-19 pandemic has highlighted bottlenecks in large-scale, frequent testing of populations for infections. PCR-based diagnostic tests are expensive, reliant on expensive centralized labs, can take days to deliver results, and are prone to backlogs and supply shortages. Antigen tests, that bind and detect the surface proteins of a virus, are rapid and inexpensive but suffer from high false negative rates. To address this problem, we have created an inexpensive, simple, and robust 60-minute Do-It-Yourself (DIY) workflow to detect viral RNA from nasal swabs or saliva with high sensitivity (0.1 to 2 viral particles/µl) and specificity (>97% True Negative Rate) utilizing reverse transcription loop-mediated isothermal amplification (RT-LAMP). Our workflow, ALERT (Accessible LAMP-Enabled Rapid Test), incorporates the following features: 1) Increased shelf-life and ambient temperature storage by using wax layers to isolate enzymes from reaction, 2) Improved specificity by using sequence-specific QUASR reporters, 3) Increased sensitivity through use of a magnetic wand to enable pipette-free concentration of sample RNA and cell debris removal, 4) Quality control with a nasopharyngeal-specific mRNA target, and 5) Co-detection of other respiratory viruses, such as Influenza B, by duplexing QUASR-modified RT-LAMP primer sets. The flexible nature of the ALERT workflow allows easy, at-home and point-of-care testing for individuals and higher-throughput processing for centralized labs and hospitals. With minimal effort, SARS-CoV-2-specific primer sets can be swapped out for other targets to repurpose ALERT to detect other viruses, microorganisms or nucleic acid-based markers.

Summary (2 min read)

Introduction

  • Intense pulsed light (IPL) has been used therapeutically in a number of clinical settings, including the treatment of acne vulgaris, birth marks and scars, as well as for hair and tattoo removal, and in skin rejuvenation and wound healing [1].
  • IPL systems deliver polychromatic light ranges between the wavelengths of 530 nm and 1100 nm, which enable the targeting of different chromophores at varying depths in biological tissues [4].
  • Phototherapy outcomes have lacked consistency, and this has been attributed to the diversity of targets and light parameters [11].
  • In contrast to lasers, which are now widely used in medical applications, IPL is not governed by the standard regulations and specifications that ensure accuracy, safety and efficiency of the device.
  • Furthermore, media containing the pH indicator dye, phenol red, produces a specific absorption peak (which is absent from phenol red-free media), and this might have a bearing on the light energy reaching the cultured cells/tissues.

IPL devise energy output

  • An IPL system (iPulse i300, CyDen Ltd., Wales, UK; features as previously described [18] was used in this study.
  • Each measurement was carried out in quintuplicate, and the experiment repeated three times.
  • The foil-enveloped shield was placed in the centre of the sensor unit stage and culture wells of 1mL or 1.5mL volume inserted into the aperture (Figure 1B), with or without phenol red-free or phenol-red-containing Dulbecco’s Modified Eagle’s Medium (DMEM; Gibco Life Technologies, UK).
  • Absorption spectra in the wavelength range of 220 nm-850 nm were recorded, with 2 nm resolution [14].
  • Similarly, each wavelength unit of the phenol red spectrum was converted into a percentage of the total area under the optical density curve (Error! Reference source not found.).

Statistical analysis

  • Data were analysed using Microsoft Excel (2010), IBM SPSS 20 statistics and GraphPad Prism 5 software, and presented as graphs or tables using means plus or minus the standard error of the mean.
  • Parametric data were tested with one-way ANOVA (analysis of variance) to compare means, whilst a Kruskal-Wallis H-test was used as a non-parametric test.
  • Post-hoc tests were used for multiple comparisons in case of significance based on the homogeneity of the variances.
  • Significance was determined at a p-value less than 0.05.

IPL devise output fluence

  • Similarly, higher well volumes (i.e., 1.5 versus 1 mL) were associated with a significant increase in energy loss overall, whilst the delivery of higher energy levels (26.14 and 38.44 J) resulted in a significantly reduced loss (approximately 5-10%) compared with the lowest IPL energy (p < 0.05, Mann–Whitney U test).
  • Moreover, the energy loss due to phenol red in a 1mL volume remained relatively constant when comparing the various energies delivered, and was not statistically significant (p > 0.05, Mann–Whitney U test).
  • The temperature change through the culture system when irradiated with IPL energy was also evaluated (Figure 2C).
  • These data reveal similar trends to those examining energy loss within the system (Figures 2A & 2B); for example, in comparing values for empty versus DMEMfilled wells, unfilled wells are responsible for a high proportion (approximately 60%) of the temperature change.

Culture media absorption

  • Analyses revealed that media samples containing phenol red produced a distinct absorption spectrum spanning 350 nm-602 nm, with an absorption peak at approximately 560 nm, and OD value of 0.53 OD units (Figure 3A).
  • In contrast, the OD values for phenol red-free DMEM, distilled water and PBS were generally very low over the 350 to 850 nm range.
  • The effect of DMEM with or without phenol red on the transmittance spectrum following irradiation with IPL at a fluence of 15.9 J/cm2 was next evaluated (Figure 3B).
  • Phenol red-containing media manifest a low level of transmittance (i.e. increased absorption) over the 530-600nm range, which equated to an approximate 11% energy loss with delivery of 38.5 J.

Discussion

  • An important step in developing in vitro cell culture models to investigate the effects of light therapy is validation of the optical devise itself within the system.
  • A further decrease (by an average of 10%) occurred due to the presence of the plate well bottom, implying that the actual fluencies delivered to the cell monolayer would be approximately 65% of the recorded output.
  • Indeed, their experiments showed that the equipment alone (i.e., culture plate wells) were responsible for more than 50% of the energy lost through the system, with the addition of culture media (with or without phenol red) further increasing energy loss (Figure 2A).
  • The authors data are consistent with published absorption spectra for culture media with and without phenol red [14].
  • In order to investigate this further, the authors examined the temperature change that occurred within the system with IPL irradiation (Figure 2C).

Conclusion

  • This study highlights the importance of testing an optical device within a cell culture model system, as outputs may differ to those published by the manufacturer.
  • Furthermore, the use of phenol-red containing media is valid and physiologically relevant when examining lightculture system interactions.
  • Acknowledgments J. A. M. Alzyoud is grateful to the Hashemite University, Jordan for their financial sponsorship and support.
  • The authors wish to thank CyDen Ltd., in Wales for the IPL light device.

Did you find this useful? Give us your feedback

Figures (3)

Content maybe subject to copyright    Report

Accessible LAMP-Enabled Rapid Test (ALERT) for detecting SARS-CoV-2
Ali Bektaş
1*
, Michael F. Covington
1,8
, Guy Aidelberg
2
, Anibal Arce
3, 5
, Tamara Matute
3,5
, Isaac Núñez
3,5
, Julia Walsh
4
, David
Boutboul
6
, Ariel B. Lindner
2
, Fernán Federici
3, 5
, Anitha Jayaprakash
1,7
1
Oakland Genomics Center, 355 30th St. Oakland, CA, 94609, USA
2
Université de Paris, INSERM U1284, Center for Research and Interdisciplinarity
(CRI), F-75006 Paris, France
3
Institute for Biological and Medical Engineering, Schools of Engineering, Biology and Medicine, Ponticia Universidad
Católica de Chile, Santiago 7820244, Chile
4
School of Public Health, University of California Berkeley, Berkeley, CA, 94720, USA
5
ANID – Millennium Sci-
ence Initiative Program- Millennium Institute for Integrative Biology (iBIO), Santiago, Chile, FONDAP Center for Genome Regulation.
6
Clinical Immunology
Department, U976 HIPI, Hôpital Saint Louis, Université de Paris, Paris, France
7
Girihlet Inc, 355 30th St, Oakland, CA, 94609, USA
8
Amaryllis Nucleics,
355 30th St, Oakland, CA, 94609, USA
Abstract:
The COVID-19 pandemic has highlighted bottlenecks in large-scale, frequent testing of populations for infections. PCR-based
diagnostic tests are expensive, reliant on expensive centralized labs, can take days to deliver results, and are prone to backlogs and
supply shortages. Antigen tests, that bind and detect the surface proteins of a virus, are rapid and inexpensive but suffer from high false
negative rates. To address this problem, we have created an inexpensive, simple, and robust 60-minute Do-It-Yourself (DIY) workow
to detect viral RNA from nasal swabs or saliva with high sensitivity (0.1 to 2 viral particles/µl) and specicity (>97% True Negative Rate)
utilizing reverse transcription loop-mediated isothermal amplication (RT-LAMP).
Our workow, ALERT (Accessible LAMP-Enabled Rapid Test), incorporates the following features: 1) Increased shelf-life and
ambient temperature storage by using wax layers to isolate enzymes from reaction, 2) Improved specicity by using sequence-specic
QUASR reporters, 3) Increased sensitivity through use of a magnetic wand to enable pipette-free concentration of sample RNA and cell
debris removal, 4) Quality control with a nasopharyngeal-specic mRNA target, and 5) Co-detection of other respiratory viruses, such as
Inuenza B, by duplexing QUASR-modied RT-LAMP primer sets.
The exible nature of the ALERT workow allows easy, at-home and point-of-care testing for individuals and higher-throughput
processing for centralized labs and hospitals. With minimal effort, SARS-CoV-2-specic primer sets can be swapped out for other targets
to repurpose ALERT to detect other viruses, microorganisms or nucleic acid-based markers.
*Corresponding author: alibektas@berkeley.edu
Introduction
The COVID-19 pandemic has brought the eld of molecular
diagnostics into the spotlight. Since the initial release of sequence
data for SARS-CoV-2 on January 10, 2020 (Genbank Accession #
MN908947), a positive-sense single-stranded RNA of approximately
29.8 kb, RT-PCR assays have been designed by the WHO, the
US CDC, the Chinese CDC, Institut Pasteur and others [1]. An
unprecedented demand for RT-PCR diagnostics has put a strain on
every aspect of conducting these laboratory-based assays. Despite
the availability of quickly developed protocols (e.g. [2], [3], [4], [5]),
shortages in materials (e.g., swabs, reagents, and consumables)
and of infrastructure (e.g., approved facilities, technicians, and
equipment) have prevented the efcient testing, tracing, and
isolation of infectious individuals. These shortages have led to a
global public health crisis of unforeseen consequences, which is
further exacerbated in the global south.
Conventional RT-PCR-based assays, which require a level of
training, are neither rapid, inexpensive, nor highly scalable without
the use of cost-prohibitive equipment. To address the challenge
of diagnostics at scale there has been an increased emphasis
on simpler detection techniques conducted at the point-of-care
or even in the convenience of one’s own home. Most of these
methods can be broadly categorized into two groups in relation
to their targets, nucleic acids or proteins (i.e. antigen). Amongst
the rapid methods targeting nucleic acids, reverse transcription
loop-mediated isothermal amplication (RT-LAMP)[6] has risen in
prominence during the course of the COVID-19 pandemic. Beyond
its isothermal nature, which eliminates the need for complex
instruments such as thermocyclers necessary for PCR, RT-LAMP
is attractive since it produces an immense amount of amplication
products allowing visual detection by the naked eye via diverse
methods [7]. Since the emergence of COVID-19, multiple primer
sets for SARS-CoV-2 [8] [9] and variations on the RT-LAMP method
have been published [10] and some have received emergency use
authorization by the FDA (Color and Mammoth Biosciences). While
the RT-LAMP workow delivers results faster than RT-PCR and is
highly scalable, it still requires a trained technician to perform the
assay as well as cold shipping and storage of reagents. We have
developed the Accessible LAMP-Enabled Rapid Test (ALERT) to
address some of the shortcomings.
Materials And Methods
Controls and Reference Material
SARS-CoV-2 synthetic RNA controls were purchased from
Twist Biosciences (San Francisco, CA, SKU:102019) and Inuenza
B virus RNA from ATCC (Catalogue Number VR-1813D).
SARS-CoV-2 virus was cultured at the Innovative Genome
Institute’s BSL3 facility at University of California Berkeley and
inactivated in RNAShield (gift of Xammy Hu Nguyenla) at a 2.5e5
PFU/ml concentration. Samples were spiked with virus at units
ranging between 125 PFU and 1000 PFU per RT-LAMP reaction
These samples were processed in BSL2-dedicated rooms following
strict biosafety guidelines.
RNA positive controls for the reactions performed in Chile
were created by in vitro transcription using the Hi-Scribe kit
from NEB (catalogue E2040S) from an amplicon obtained by
PCR amplication of the IDT control for Sars-CoV-2 (catalogue
10006625) using the following primers: NT7_Fw (CGA AAT TAA
TAC GAC TCA CTA TAG GGG CAA CGC GAT GAC GAT GGA
TAG) and T7_Nter_Rv (ACT GAT CAA AAA ACC CCT CAA GAC
CCG TTT AGA GGC CCC AAG GGG TTA TGC TAG TTA GGC
CTG AGT TGA GTC AG).
In vitro transcribed RNA was treated with DNAse I (NEB
M0303S) for 15 minutes at 37°C before purication with Qiagen
RNeasy kit, and serially diluted to the concentrations used.
Sample Collection
Human nasal samples were collected with either
nasopharyngeal (NP) swabs, nasal mid-turbinate swabs (NMT),
or a nasopharynx ush thru (NFT) method. NFT samples were
collected by using a 5 ml dropper lled with water squirted into a
nostril with the head tilted back and the water passing through the
nasopharynx and subsequently spit into a 50-ml Falcon tube.
For collecting nasal mid-turbinate samples, we used a variety
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)
The copyright holder for this preprint this version posted February 20, 2021. ; https://doi.org/10.1101/2021.02.18.21251793doi: medRxiv preprint
NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice.

of swabs due to supply chain shortages, ranging from Q-tip cotton
swabs to ocked nylon nasopharyngeal swabs. We observed that
some sample collection swabs, such as those supplied by Zymo
Research Corporation (C105250), had blue uorescence emission
when excited by UV light thus interfering with the interpretation of
results following our crude sample lysis protocols. We currently use
sterile, ocked oral swabs with a 30mm breakpoint compatible with
1.5ml microcentrifuge tubes (SNT Biotech, www.sntbiotech.com).
Sample Lysis
We experimented with different lysis buffers. Our Proteinase
K extraction method begins by inserting a freshly used NP or NMT
swab into 300 µl of PK Lysis Solution and agitating the swab for
30 seconds. 75 µl of the resulting solution is used and, depending
on the experiment, spiked with inactivated SARS-CoV-2. Samples
were heated at 55°C for 5 min for lysis and 95°C at 10 min for
inactivation. The PK Lysis Solution contained ~15 mAU/ml and a
10 ml stock was prepared by diluting 0.25 ml Proteinase K (Qiagen,
19131) with 9.75 ml water.
In order to process samples already in solution, such as
PBS, nasal swabs (in an unknown solution), and saliva sent by
the XPrize foundation (www.xprize.org/prizes/covidtesting) we
used QuickExtract DNA extraction solution (Lucigen, catalogue #
QE09050) following the protocol by Kellner et al. (2020).
For our DIY workow, our buffer of choice is TCEP/EDTA
following the protocol outlined by Rabe and Cepko (2020) of a
modied HUDSON buffer (Myhrvold et al. 2018). In contrast to
other protocols where a concentrated 100X HUDSON buffer is
added to a liquid sample such as saliva we used a 1X solution
(2.5mM TCEP, 1mM EDTA) for the direct addition of an NMT swab.
Nucleic acid extraction
RNA from NP, NMT, and NFT samples was isolated using
both a QIAamp Viral RNA Mini Kit (Qiagen Catalogue # 52904)
according to the manufacturer’s instructions as well as with a
TRIzol/chloroform method[11]. NP and NMT swabs were inserted
into 1ml of PBS and vortexed prior to extraction while 1 ml of NFT
samples was used directly for extraction.
RNA Isolation with Paramagnetic Beads and Magnetic Wand
For paramagnetic bead-based RNA capture, 0.6 volumes of
a 1X bead solution was added to the lysed sample and uniformly
suspended. After a 5-minute incubation at room temperature the
samples containing the beads were placed on an appropriately
sized magnetic rack and beads were pelleted by the magnet for 5
minutes. All liquid was removed, and a brief wash was conducted
with 100 µl of 85% ethanol. Following the removal of ethanol,
sample tubes were removed from the magnetic rack and the
beads were resuspended in 10-20 µl of water. Depending on the
cartridge design, 5-10 µl of beads in water were directly added
to the cartridge. The 1X bead solution is prepared by combining
one part RNAClean XP beads (Beckman Coulter Life Sciences,
A63987) with ve parts of a concentrated Bead Binding Buffer.
The composition of the Bead Binding Buffer [12] is 20% PEG
8000 (Sigma-Aldrich, P1458), 10 mM Tris HCl, pH 8.0 (Invitrogen,
15568025), 0.05% Tween 20 (Thermo Scientic, J20605AP), 5 mM
Sodium Azide (MP Biomedicals, 0210289125), Sodium Chloride
(Fisher Chemical, S271-500).
To enable a DIY workow, we 3D printed the sheaths for
the anged magnetic wand with a $450 3D printer (Adventurer
3; Flashforge) in as few as 10 minutes using about $0.02 worth
of 1.75 mm diameter PLA lament (Flashforge) per wand. These
anged sheaths accommodate a $0.72 cylindrical grade N42
neodymium magnet (1-inch-long x 1/16-inch diameter; D1X0, K&J
Magnetics, Inc.). The diameter and vertical position of the ange
can be adjusted to accommodate a diverse array of sample tubes.
The diameter of the rest of the sheath is optimal for depositing the
beads in RT-LAMP reaction cartridges that have been prepared
in 0.2 ml strip tubes. If larger tubes are used for the RT-LAMP
reaction cartridge, the diameter of the sheath can be increased
to accommodate more powerful, larger diameter magnets. Using
these designs as a prototype, the magnetic wand sheaths can also
be produced in bulk with injection-molded low-retention plastics.
Capturing the beads with magnetic wands followed a similar
protocol to using a magnetic rack but without the use of pipettes.
After the 5-minute room temperature incubation the magnetic wand
was inserted into the sample/bead solution. A further 5-minute
room temperature incubation was sufcient to allow RNA-bound
beads to be attracted by the wand. The wand, now with beads,
was removed from the sample. At this stage an optional bead wash
can be performed by briey dipping (~2 seconds) the magnetic
wand into a tube with 85% ethanol lled to the same volume as the
sample/bead solution. If an ethanol wash was performed, we let the
beads on the magnetic wand air dry without over-drying beads (~2-
5 minutes). The magnetic wand was placed in a tube containing
eluant (in this case, our RT-LAMP cartridges with 10 µl water on
the topmost layer of wax). The magnet was removed from the wand
and the wand was swirled to release beads into the eluant.
Construction of Wax-layered Cartridges
For a sampling and process control target sequence, we
chose the BPI fold containing family A, member 1 gene (BPIFA1)
as its expression is nasopharyngeal specic [37]. We designed
a LAMP primer set for this gene that traverses an exon-exon
boundary, making the reaction specic to BPIFA1 mRNA. The
SARS-CoV-2 [8][10] and Inuenza B [13] primers were adopted
from the literature but modied for the sequence specic QUASR
(Quenching of Unincorporated Amplication Signal Reporters)
reporting system [38] (Supplemental Table 1).
1-step RT-LAMP
1-step RT-LAMP cartridges using the NA/NB primer set were
prepared by adding 27 µl of RT-LAMP primer mix to the bottom of
a 0.75 ml cryogenic tube (Micronic, Netherlands) followed by 200
µl of melted silicone wax (Siltech D-222, melting point 37°C). After
solidication of this wax layer a small divot was made with a 1 ml
micropipette tip, deposited into which was 3 µl of an enzyme mix
consisting of 1 µl (15 units) of WarmStart RTx (NEB, Ipswich, MA,
M0380) and 2 µl (16 units) of Bst 3.0 (NEB, Ipswich, MA, M0374).
The enzyme droplet was capped by a further addition of 300 µl
of the same silicone wax. In an alternative construction of this
cartridge, we added a layer of 150 µl cold mineral oil (consumer
grade) between the enzyme droplet and silicone wax to shield it
from the thermal shock of hot wax.
For the single wax layer RT-LAMP cartridges using this NA/
NB primer set, the primer mix, prepared in Isothermal Amplication
Buffer (I) (NEB Ipswich, MA, B0537S) contained 2.1 µM each of
NB-FIP and NB-BIP, 0.32 µM each of NB-F3 and NB-B3, 1.3 µM
each of NB-LB and NB-LF-Tx and 2.2 µM of NB-LF-Q. MgSO4
concentration was at 7.8 mM and dNTPs at 1.8 mM. 27 µl of the
primer mix was used as the bottom layer of reaction cartridges
anticipating a 35 µl nal reaction volume together with 1 µl
WarmStart Rtx (NEB, Ipswich, MA, M0380), 2 µl Bst 3.0 (NEB,
Ipswich, MA, M0374) and 5 µl of sample.
For direct addition of paramagnetic beads using the magnetic
wand, we used the NM primer set (Supplemental Table 1) in a 30 µl
nal reaction volume cartridge consisting of (from bottom to top) a
16.5 µl primer mix, a layer of parafn wax (IGI 1250A melting point
of 61.4°C), and an enzyme mix of 1.5 µl (22.5 units) of WarmStart
RTx (NEB, Ipswich, MA, M0380) and 2 µl (16 units) of Bst 2.0
(NEB, Ipswich, MA, M0374). The enzyme was trapped between
two layers of solid wax by adding a nal 4 mm solid, lower-melting-
point silicone wax (Siltech, Silwax D-222, melting point of 37°C)
and melting it down briey at 40°C for 1 min. When the magnetic
2
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)
The copyright holder for this preprint this version posted February 20, 2021. ; https://doi.org/10.1101/2021.02.18.21251793doi: medRxiv preprint

wand was used, 10 µl of molecular biology grade water for bead
deposition was added above this wax layer. (Figure 1a and 1b).
For one-step cartridges using this NM primer set, the 16.5
µl primer mix was prepared with Isothermal Amplication Buffer (I)
(NEB Ipswich, MA, B0537S) with 2.9 µM NM-FIP-F and NM-BIP,
4.35 µM NM-FIP-Q, 1.5 µM NM-LF and NM-LB, and 0.4 µM NM-F3
and NM-B3. MgSO4 was at 12.8 mM and dNTPs at 2.5 mM.
Depending on the primer mixes, 5 µl (for NA/NB) or 10 µl (for
NM) of sample was added to the top layer. Cartridges containing the
NA/NB set were incubated in a 63°C dry block with a heated lid for 1
hr. Cartridges containing the NM primer set, and where the enzyme
was packaged between waxes of two different melting points, tubes
were rst heated to 64°C to allow the mixing of all reagents and
then cooled to 55°C for a 12.5 min reverse transcription reaction,
followed by a 64°C LAMP incubation for 45 min to 80 min.
For the BPIFA1 primer set, standard RT-LAMP primer
concentrations were followed: 1.6 µM FIP and BIP, 0.4 µM LF and
LB-F, 0.2 µM F3 and B3, and 0.6 µM LB-Q. 7 mM MgSO4 and
1.4 mM dNTPs were used in a 25 µl reaction in 1X Isothermal
Amplication Buffer (I) (NEB Ipswich, MA, B0537S) with 0.75 µl
WarmStart Rtx (NEB, Ipswich, MA, M0380) and 1.5 µl Bst 2.0 (NEB
Ipswich, MA, M0538L). BPIFA1 RT-LAMP reactions were incubated
at 63°C for 30 min.
A wax-layered cartridge that is ready-to-run following sample
addition presents a challenge towards the homogenous mixing of
all of its components owing to a pipette-free operation. We found
that the addition of a 2 mm glass bead to the top of the tube allows
for efcient and thorough mixing by both allowing for reagents to
fall through wax layers as well as displacing the bottommost primer
mix.
2-Step RT/LAMP
Cartridges separating the reverse transcription (RT) step
from the loop-mediated isothermal amplication step (LAMP)
were constructed into 8-tube strips using two separate waxes with
distinct melting points. The 10 µl LAMP primer mix, at the bottom
of the cartridge, was sealed from the RT primer mix above it with a
layer of parafn wax (IGI 1250A; melting point of 61.4°C). This seal
was created by adding a roughly 3 mm diameter piece of solid wax
above the LAMP primer mix and melting it at 65°C for 1 min. After
solidication of this wax layer at room temperature, 7 µl of RT primer
mix was added above it. Two drops of a lower melting point silicone
wax (Siltech, Silwax D-222; melting point of 37°C), heated to 50°C
in a water bath, was added on top of the RT primer mix using a 1
ml micropipette and allowed to solidify at room temperature. Finally,
an enzyme mix consisting of 1 µl (15 units) WarmStart RTx (NEB,
Ipswich, MA, M0380) and 2 µl (16 units) of Bst 3.0 (NEB, Ipswich,
MA, M0374) was added as the top layer of the cartridge (Figure 2a-
c).
For the 2-step detection cartridges, we made two separate
primer mixes corresponding to the reverse transcription (RT Mix)
and loop-mediated isothermal amplication (LAMP Mix) steps.
The RT Mix contained the F3 outer primer from the N-A primer
set [8] together with the B3 primer from the N-B primer set, which
we found to improve the detection limit compared to using only
the NB-B3 primer. This may result from DNA polymerase activity
stemming from the inclusion of Bst 3.0 during the RT step. NA-F3
and NB-B3 were at 0.54 mM along with 12.9 mM of MgSO4 and
1.07 mM of dNTPs in NEB Isothermal Amplication Buffer (I) at a
2X concentration. In each cartridge 7 µl of this mix was used in
anticipation of a 15 µl reverse transcription reaction together with 1
µl WarmStart Rtx (NEB, Ipswich, MA, M0380), 2 µl Bst 3.0 (NEB,
Ipswich, MA, M0374), and 5 µl of sample.
The LAMP Mix was composed of the NB primer set published
by Zhang et al. (2020) but modied for the sequence-specic
QUASR reporting system. This mix, with the NEB Isothermal
Amplication Buffer (I) (NEB Ipswich, MA, B0537S) at a 1X
concentration, had primer concentrations of 4 µM NB-FIP and NB-
BIP, 0.625 mM NB-F3 and N-B-B3, 2.5 µM NB-LB and NB-LF-Tx,
and 5 µM of NB-LF-Q. MgSO4 concentration was at 6 mM and
dNTPs at 3.5 mM. The concentration of the LAMP mix is higher
than standard reactions; the nal LAMP reaction volume will be 25
µl when all reagent mixes are combined at the bottom of the tube.
In each cartridge 10 µl of this LAMP mix was used.
The 2-step cartridges (Figure 2a) had 5 µl of sample added to
the enzyme droplet on the top layer of lower melting point silicone
wax, incubated in a thermal cycler at 55°C for 2 minutes allowing
for the enzyme droplet and sample to fall through liquid wax and
meet the RT primer mix. Cartridges were removed from incubation
and the wax allowed to briey solidify in order to allow for mixing of
reagents by manual shaking. After mixing, cartridges were further
incubated for 20 min at 55°C for the RT step to take place (Figure
2b). After this step, the temperature was raised to 63°C for 2 min to
allow the RT reaction to fall through the now liquied parafn wax
layer and meet the LAMP primer mix. The cartridges were again
removed from incubation, wax layers allowed to solidify, and the
RT/LAMP Mixes, now together under a layer of solid silicone and
another of parafn wax, mixed briey by vortex. The LAMP reaction
was then allowed to progress for 45 min at 63°C (Figure 2c).
Duplex RT-LAMP reactions
The nal 25 µl reactions, prepared with 1X Isothermal
Amplication Buffer (I) (NEB Ipswich, MA, B0537S) had 1.6 mM of
both FIP and BIP primer pairs, 0.2 mM of Inuenza B or BPIF1A F3
and B3 primer pairs, 0.25 µM of the NB-F3 and NB-B3 primer pair,
1 µM of both LF and LB primer pairs (LF or LB being modied with
a uorophore) and 2 mM of the corresponding LF or LB quencher
primer, 6 mM MgSO4, 1.6 mM dNTPs, 1 µl (15 units) of WarmStart
RTx (NEB, Ipswich, MA, M0380), and 2 µl (16 units) of Bst 3.0
(NEB, Ipswich, MA, M0374). Reactions were incubated at 63°C for
45 min.
Visualization
End-point reactions were visualized on a UV Transilluminator.
Alternatively, for reactions utilizing uorescein tagged
oligonucleotides we also used the low-cost (<$2) open-source
GMO Detective uorescence detector ([35] and https://github.com/
MakerLabCRI/GMODetective-Detector).
Incubation
Incubations (55°C and 63°C or 64°C) were performed in a
thermalcycler. We have also successfully performed all reactions
in a dry block incubator with a heated lid (Benchmark Scientic,
BSH200-HL).
Analytical Validation for 2-Step RT/LAMP: XPRIZE Blinded
Prociency Test
As a semi-nalist in the XPRIZE Covid Testing challenge [14]
we had the opportunity to participate in a blinded prociency test
alongside 218 other teams. Two separate 96-well sample plates
were mailed to our laboratory in Oakland, CA, with one consisting
of 86 synthetic RNA samples spiked into water (SARS-CoV-2
and 15 other viruses to test cross-reactivity) shipped on dry ice,
and the other plate consisting of 67 samples of different matrices
(phosphate buffered saline (PBS), nasal swabs resuspended in
an unknown buffer, and saliva) spiked with SARS-CoV-2 particles
from Zeptometrix shipped with cold packs. The synthetic RNA plate
was kept at -80ºC and the Zeptometrix plate was kept at 4°C until
analysis. For RNA extraction of the XPRIZE Rapid Covid Testing test
samples, 200 µl of QuickExtract DNA Extraction Solution (Lucigen,
QE09050) was added to 200 µl of each sample. Samples were
incubated for 5 minutes at room temperature prior to inactivation of
the QuickExtract solution at 95ºC for 10 minutes. The samples were
cooled on ice to room temperature prior to adding 240 µl of bead
3
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)
The copyright holder for this preprint this version posted February 20, 2021. ; https://doi.org/10.1101/2021.02.18.21251793doi: medRxiv preprint

Room temperature
64°C
1 2 3 4 5 6 7 8
9 10 11 12 13 14 15 16
17 18 19 20 21 22 23 24
a. b. c.
d.
e.
f.
g.
h.
a.
b.
2000pfu 1000pfu 500pfu 250pfu
125pfu 0pfu
2000pfu 1000pfu 500pfu 250pfu
125pfu 0pfu
Non-template water control
0pfu 125pfu 250pfu 500pfu
200cp Twist 100cp Twist Non-template water
Figure 1. Easy to use ALERT Cartridges. One Step reaction-ready cartridge layering. Cartridge layers at room temperature (A) and at 64°C (B).
Demonstration of magnetic wand. Wand accumulates beads (C and D) and is placed into the reaction cartridge where the magnet is removed to
release beads (E and F). Reagents have dyes added for demonstration purposes: primer mix is orange and the enzyme droplet is black G. Detection
of SARS-CoV-2 using Proteinase K, magnetic wand extraction and a one-step cartridge, NMT swabs spiked with inactivated SARS-CoV-2 H. Same
tubes visualized with the GMO Detective.
0.4ng IB RNA 2000cp Twist 4000cp Twist Non-template water control
9 10 11 12 13 14 15 16
1 2 3 4 5 6 7 8
1 2 3 4 5 6 7 8
9 10 11 12 13 14 15 16
17 18 19 20 21 22
1 2 3 4 5 6 7 8
Room temperature 63°C55°C
a.
b.
c.
a.
b.
c.
a. b. c.
d.
e.
500cp 100cp 50cp
25cp Non-template water control
500cp 100cp 50cp
25cp Non-template water control
0.2ng IB RNA+
NP RNA 2000cp Twist 4000cp Twist NT Water Cnt.
NP RNA +
50cp Twist 25cp Twist Non-template water control
Non-template water control
500pfu
nCoV2
Figure 2. Two Step reaction-ready cartridge layering. Cartridge layers at room temperature (A), 55°C (B) and at 64°C (C). D. 1-Step RT-LAMP LoD
conrmed to be at ~500 copies of Twist Biosciences SARS-CoV-2 control as reported by Zhang et al. 2020 E. 2-step RT/LAMP cartridge with increased
sensitivity with LoD down to 25 copies of Twist Biosciences SARS-CoV-2 control
4
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)
The copyright holder for this preprint this version posted February 20, 2021. ; https://doi.org/10.1101/2021.02.18.21251793doi: medRxiv preprint

solution as described above. After thoroughly mixing the beads into
the samples, beads were allowed to bind RNA for 5 minutes at
room temperature prior to placing the samples on a magnetic rack
for 5 minutes to pellet the RNA-bound beads. The supernatant was
removed from each sample and each set of remaining beads was
washed for 5 minutes with 850 µl 85% ethanol. After removal of
ethanol, the beads were allowed to air dry briey. RNA was eluted
from beads with 10 µl of nuclease-free water and added to the
2-step RT/LAMP cartridges as described above.
Clinical Validation for 2-Step RT/LAMP
In order to validate our detection cartridges with true clinical
samples from patients diagnosed for the presence of absence
of SARS-CoV-2 using established RT-PCR methods we shipped
our v1 2-step RT/LAMP cartridges, at ambient temperature, to
locations willing to share clinical samples. At the Hôpital Saint
Louis (Paris, FR), 18 samples, determined to be positive with RT-
PCR methods approved by local agencies, (Ct values ranging
between 13 and 37.9) were analyzed alongside a negative water
control and Coronavirus NL63 for cross-reactivity. At the Ponticia
Universidad Católica de Chile (Santiago, CL), 10 negative patient
samples and 19 samples, determined to be positive with RT-PCR
using the N1 and N2 primer set designed by the CDC, (Ct values
ranging between 20.25 and 35.34) were analyzed. These clinical
samples were obtained from anonymous patients that attended
the outpatient service of Red Salud UC-CHRISTUS (Santiago,
Chile). All methods were performed in accordance with the relevant
guidelines and regulations. All procedures were approved by the
Ethics Committee of the Ponticia Universidad Católica de Chile.
Results
We have developed a streamlined process for detecting
nucleic acids of RNA viruses, demonstrated as a proof-of-principle
here for the detection of SARS-CoV-2 from nasal mid-turbinate
swabs. Our system incorporates a room temperature stable lysis
buffer (a modied HUDSON buffer or Proteinase K) for the lysis
of virions as well inactivation of RNAses, followed by bead-based
sample concentration in which the beads are captured with a
magnetic wand developed for pipette-free testing. The RT-LAMP
reagents are packaged into a reaction vessel constructed with
layers of waxes in-order to increase shelf-life at room temperature,
provide physical stability for a just-add-sample operation as well as
to minimize risk of cross-contamination.
We were able to achieve a substantial improvement in the
LoD of the rst RT-LAMP primer set published [8] for SARS-CoV-2
by separating the RT reaction from the LAMP reaction using waxes
of different temperatures (2-step cartridges) (Figure 2d and 2e). To
facilitate simpler production and usage of cartridges we ultimately
switched to a 1-step design (Figure 1) using another published
primer set [10] with improved LoD.
Lysis
We used Proteinase K to lyse NMT swabs from 11 distinct
individuals (asymptomatic and presumed to be uninfected with
SARS-CoV-2) spiked with inactivated SARS-CoV-2 ranging from 250
to 1,000 PFU. Lysed samples were concentrated using RNAClean
XP paramagnetic beads and SARS-CoV-2 RNA detected using
a master mix containing the NM primer set. All spiked samples
presented uorescence associated with amplication while non-
spiked samples did not (Figure 4a).
In order to reduce cost and eliminate the 55°C incubation
step of Proteinase K we used the modied HUDSON buffer made
of TCEP and EDTA [15]. NMT swabs were deposited into tubes
with 300 µl 1X HUDSON buffer and spiked with inactivated SARS-
CoV-2 at 0, 125, 250 and 750 PFU. After a brief vortex, samples
were incubated for 5 min at 95ºC. We concentrated these samples
with paramagnetic beads and deposited 10 µl of a 20 µl suspension
of beads in water into a one-step cartridge containing the NM
primer set. All spiked samples presented uorescence associated
with amplication while non-spiked samples did not (Figure 4b).
Magnetic Wand Sample Deposition
In order to produce a streamlined extraction method
compatible with an at-home test we used our 3D-printed magnetic
wand to directly deposit paramagnetic beads into RT-LAMP
reaction cartridges. In its simplest conguration, the magnetic
wand consists of a cylindrical neodymium rare-earth magnet that
slides into a 3D-printed sheath closed on one side. A typical bead
pulldown using a conventional magnetic rack or plate incorporates
a 5-minute incubation with the paramagnetic beads. To make
the magnetic wand more user-friendly with such incubations,
we designed a version of the magnetic wand that incorporates a
ange that rests on the rim of the sample tube and allows the tip
of the wand to be optimally positioned in the sample. The general
workow for magnetic wand use is shown in Figure 3.
To demonstrate the magnetic wand, we used it to transfer
RNA-bound paramagnetic beads into a one-step reaction cartridge
with 10 µl of molecular biology grade water at the top to elute the
beads into. NMT swabs in Proteinase K were spiked with 125 to
1,000 PFU of inactivated SARS-CoV-2 in duplicate and processed
according to our Proteinase K extraction and bead purication
protocol. Paramagnetic beads were recovered with our wands and
directly deposited into the 10µl of water after a brief dip in 100 µl
85% ethanol. All concentrations of virus down to 250 PFU gave
a bright signal while one of the two repeats of 125pfu appeared
Figure 3. Sample concentration with magnetic wand and paramagnetic wand. The paramagnetic beads bind to viral RNA in the lysed sample. The magnetic
wand is used (1) to collect these RNA-bound beads away from the rest of the sample volume and (2) to transfer them to the LAMP reaction cartridge. To
release the beads from the magnetic wand, the magnet is slid from the 3D-printed sheath. In the absence of a magnetic eld, the paramagnetic beads are
no longer magnetized and are able go into solution in water at the top of the LAMP reaction cartridge.
5
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)
The copyright holder for this preprint this version posted February 20, 2021. ; https://doi.org/10.1101/2021.02.18.21251793doi: medRxiv preprint

Citations
More filters
Posted ContentDOI
14 Aug 2020-medRxiv
TL;DR: The design and testing of molecular beacons are described, which allow sequence-specific detection of SARS-CoV-2 genomes with improved discrimination in simple reaction mixtures and how beacons with different fluorescent labels can allow convenient multiplex detection of several amplicons in "single pot" reactions.
Abstract: Background Rapid spread of SARS-CoV-2 has led to a global pandemic, resulting in the need for rapid assays to allow diagnosis and prevention of transmission. Reverse Transcription-Polymerase Chain Reaction (RT-PCR) provides a gold standard assay for SARS-CoV-2 RNA, but tests are expensive and supply chains are potentially fragile, motivating interest in additional assay methods. Reverse Transcription and Loop-Mediated Isothermal Amplification (RT-LAMP) provides an alternative that uses orthogonal and often less expensive reagents without the need for thermocyclers. The presence of SARS-CoV-2 RNA is typically detected using dyes to report bulk amplification of DNA; however a common artifact is nonspecific DNA amplification, which complicates detection. Results Here we describe the design and testing of molecular beacons, which allow sequence-specific detection of SARS-CoV-2 genomes with improved discrimination in simple reaction mixtures. To optimize beacons for RT-LAMP, multiple locked nucleic acid monomers were incorporated to elevate melting temperatures. We also show how beacons with different fluorescent labels can allow convenient multiplex detection of several amplicons in “single pot” reactions, including incorporation of a human RNA LAMP-BEAC assay to confirm sample integrity. Comparison of LAMP-BEAC and RT-qPCR on clinical saliva samples showed good concordance between assays. We also describe custom polymerases for LAMP-BEAC and inexpensive purification procedures. Conclusions LAMP-BEAC thus provides an affordable and simple SARS-CoV-2 RNA assay suitable for population screening.

40 citations

Posted ContentDOI
05 Apr 2021-medRxiv
TL;DR: In this paper, the authors presented an inexpensive, rapid, and robust RT-LAMP based SARS-CoV-2 detection method that is easily scalable, enabling point of care facilities and clinical labs to determine results from patients saliva directly in 30 minutes for less than $2 a sample.
Abstract: Here we present an inexpensive, rapid, and robust RT-LAMP based SARS-CoV-2 detection method that is easily scalable, enabling point of care facilities and clinical labs to determine results from patients saliva directly in 30 minutes for less than $2 a sample The method utilizes a novel combination of widely available reagents that can be prepared in bulk, plated and frozen and remain stable until samples are received This innovation dramatically reduces preparation time, enabling high-throughput automation and testing with time to results (including setup) in less than one hour for 96 patient samples simultaneously when using a 384 well format By utilizing a dual-reporter (phenol red pH indicator for end-point detection and SYTO-9 fluorescent dye for real-time), the assay also provides internal validation of results and redundancy in the event of an instrument malfunction

3 citations

Journal ArticleDOI
01 Dec 2022-Biology
TL;DR: In this paper , the performance of various M-MuLV RTs in reverse-transcription loop-mediated isothermal amplification (RT-LAMP) remains poorly studied, and the comparison of different RTs presented here could help for selecting the optimal enzyme when developing novel LAMP-based diagnostic tests.
Abstract: Simple Summary Reverse transcriptases (RTs) are a family of enzymes synthesizing DNA with RNA as a template, and are used in almost all studies related to RNA. M-MuLV RT is widely used in diagnostics methods, including in reverse-transcription loop-mediated isothermal amplification (RT-LAMP). The performance of various reverse transcriptases in RT-LAMP remains poorly studied. Here, we report the first direct comparison of various M-MuLV RTs in RT-LAMP. The enzymes studied contained different numbers of mutations or an additional Sto7d protein. Several parameters were assessed: optimal reaction temperature, enzyme concentration, reverse transcription time, a minimal amount of RNA template, and tolerance to inhibitors. Mutations increased the optimal temperature of the reverse transcription up to 5–10 °C. All of the RTs were suitable for RT-LAMP with RNA templates in the range of 101–106 copies per reaction. Highly mutated enzymes were more tolerant to most of the inhibitors, but more sensitive to high concentrations of NaCl. The results presented could help select the optimal enzyme for novel LAMP-based diagnostic tests. Abstract Reverse transcriptases (RTs) are a family of enzymes synthesizing DNA using RNA as a template and serving as indispensable tools in studies related to RNA. M-MuLV RT and its analogs are the most commonly used RTs. RTs are widely applied in various diagnostics methods, including reverse-transcription loop-mediated isothermal amplification (RT-LAMP). However, the performance of different RTs in LAMP remains relatively unknown. Here, we report on the first direct comparison of various M-MuLV RTs in RT-LAMP, including enzymes with a different number of mutations and fusions with Sto7d. Several parameters were assessed, namely: optimal reaction temperature, enzyme concentration, reverse transcription time, a minimal amount of RNA template, and tolerance to inhibitors. Mutations increased the optimal reaction temperature from 55 °C to 60–65 °C. All of the RTs were suitable for RT-LAMP with RNA templates in the range of 101–106 copies per reaction. Highly mutated enzymes were 1.5–3-fold more tolerant to whole blood, blood plasma, and guanidinium, but they were two-fold more sensitive to high concentrations of NaCl. The comparison of different RTs presented here could be helpful for selecting the optimal enzyme when developing novel LAMP-based diagnostic tests.
Journal ArticleDOI
25 Apr 2023-Agronomy
TL;DR: In this paper , a review assesses the currently available ready-to-use tools and methodologies that operate via visual estimation, the detection of proteins and DNA/RNA sequences, and the utilization of brand-new innovative approaches, highlighting the availability of solutions that can be used by growers during the process of diagnosing pathogens.
Abstract: Various diseases and pests cause serious damage to vegetable crops during the growing season and after harvesting. Growers attempt to minimize losses by protecting their crops, starting with seed and seedling treatments and followed by monitoring their stands. In many cases, synthetic pesticide treatments are applied. Integrated pest management is currently being employed to minimize the impact of pesticides upon human health and the environment. Over the last few years, “smart” approaches have been developed and adopted in practice to predict, detect, and quantify phytopathogen occurrence and contamination. Our review assesses the currently available ready-to-use tools and methodologies that operate via visual estimation, the detection of proteins and DNA/RNA sequences, and the utilization of brand-new innovative approaches, highlighting the availability of solutions that can be used by growers during the process of diagnosing pathogens.
References
More filters
Journal ArticleDOI
TL;DR: A novel method that amplifies DNA with high specificity, efficiency and rapidity under isothermal conditions that employs a DNA polymerase and a set of four specially designed primers that recognize a total of six distinct sequences on the target DNA.
Abstract: We have developed a novel method, termed loop-mediated isothermal amplification (LAMP), that amplifies DNA with high specificity, efficiency and rapidity under isothermal conditions. This method employs a DNA polymerase and a set of four specially designed primers that recognize a total of six distinct sequences on the target DNA. An inner primer containing sequences of the sense and antisense strands of the target DNA initiates LAMP. The following strand displacement DNA synthesis primed by an outer primer releases a single-stranded DNA. This serves as template for DNA synthesis primed by the second inner and outer primers that hybridize to the other end of the target, which produces a stem–loop DNA structure. In subsequent LAMP cycling one inner primer hybridizes to the loop on the product and initiates displacement DNA synthesis, yielding the original stem–loop DNA and a new stem–loop DNA with a stem twice as long. The cycling reaction continues with accumulation of 109 copies of target in less than an hour. The final products are stem–loop DNAs with several inverted repeats of the target and cauliflower-like structures with multiple loops formed by annealing between alternately inverted repeats of the target in the same strand. Because LAMP recognizes the target by six distinct sequences initially and by four distinct sequences afterwards, it is expected to amplify the target sequence with high selectivity.

6,765 citations

Journal ArticleDOI
TL;DR: The CRISPR-based DETECTR assay provides a visual and faster alternative to the US Centers for Disease Control and Prevention SARS-CoV-2 real-time RT–PCR assay, with 95% positive predictive agreement and 100% negative predictive agreement.
Abstract: An outbreak of betacoronavirus severe acute respiratory syndrome (SARS)-CoV-2 began in Wuhan, China in December 2019. COVID-19, the disease associated with SARS-CoV-2 infection, rapidly spread to produce a global pandemic. We report development of a rapid (<40 min), easy-to-implement and accurate CRISPR–Cas12-based lateral flow assay for detection of SARS-CoV-2 from respiratory swab RNA extracts. We validated our method using contrived reference samples and clinical samples from patients in the United States, including 36 patients with COVID-19 infection and 42 patients with other viral respiratory infections. Our CRISPR-based DETECTR assay provides a visual and faster alternative to the US Centers for Disease Control and Prevention SARS-CoV-2 real-time RT–PCR assay, with 95% positive predictive agreement and 100% negative predictive agreement. SARS-CoV-2 in patient samples is detected in under an hour using a CRISPR-based lateral flow assay.

1,664 citations

Journal ArticleDOI
TL;DR: Real-time monitoring of the LAMP reaction was achieved by real-time measurement of turbidity, which indicated an increase in the turbidity of the reaction mixture according to the production of precipitate correlates with the amount of DNA synthesized.

1,505 citations

Journal ArticleDOI
TL;DR: A simple point-of-care test that is inexpensive enough to use frequently, even if it lacks sensitivity, is proposed for Covid-19 cases.
Abstract: Rethinking Covid-19 Test Sensitivity As Covid-19 cases accelerate or plateau around the world, we urgently need a point-of-care test that is inexpensive enough to use frequently, even if it lacks h...

632 citations

Journal ArticleDOI
TL;DR: Traditional methods and methods based on magnetic particles for nucleic acid purification, and the synthesis of a variety of magnetic particles is presented in more detail.
Abstract: Nucleic acid separation is an increasingly important tool for molecular biology. Before modern technologies could be used, nucleic acid separation had been a time- and work-consuming process based on several extraction and centrifugation steps, often limited by small yields and low purities of the separation products, and not suited for automation and up-scaling. During the last few years, specifically functionalised magnetic particles were developed. Together with an appropriate buffer system, they allow for the quick and efficient purification directly after their extraction from crude cell extracts. Centrifugation steps were avoided. In addition, the new approach provided for an easy automation of the entire process and the isolation of nucleic acids from larger sample volumes. This review describes traditional methods and methods based on magnetic particles for nucleic acid purification. The synthesis of a variety of magnetic particles is presented in more detail. Various suppliers of magnetic particles for nucleic acid separation as well as suppliers offering particle-based kits for a variety of different sample materials are listed. Furthermore, commercially available manual magnetic separators and automated systems for magnetic particle handling and liquid handling are mentioned.

450 citations

Related Papers (5)
Frequently Asked Questions (18)
Q1. What are the contributions in "Accessible lamp-enabled rapid test (alert) for detecting sars-cov-2" ?

To address this problem, the authors have created an inexpensive, simple, and robust 60-minute Do-It-Yourself ( DIY ) workflow to detect viral RNA from nasal swabs or saliva with high sensitivity ( 0. 1 to 2 viral particles/μl ) and specificity ( > 97 % True Negative Rate ) utilizing reverse transcription loop-mediated isothermal amplification ( RT-LAMP ). Their workflow, ALERT ( Accessible LAMP-Enabled Rapid Test ), incorporates the following features: 1 ) Increased shelf-life and ambient temperature storage by using wax layers to isolate enzymes from reaction, 2 ) Improved specificity by using sequence-specific QUASR reporters, 3 ) Increased sensitivity through use of a magnetic wand to enable pipette-free concentration of sample RNA and cell debris removal, 4 ) Quality control with a nasopharyngeal-specific mRNA target, and 5 ) Co-detection of other respiratory viruses, such as Influenza B, by duplexing QUASR-modified RT-LAMP primer sets. 

The tools that are being developed are not only valuable for containing the current pandemic but also for future ones that are sure to emerge and other pathogens already affecting large populations yet receiving scant attention. 

The RT-LAMP reagents are packaged into a reaction vessel constructed with layers of waxes in-order to increase shelf-life at room temperature, provide physical stability for a just-add-sample operation as well as to minimize risk of cross-contamination. 

In molecular diagnostics of pathogens, the sample collection, extraction, and purification steps are as imperative as the final step of molecular detection. 

A typical bead pulldown using a conventional magnetic rack or plate incorporates a 5-minute incubation with the paramagnetic beads. 

The PK Lysis Solution contained ~15 mAU/ml and a 10 ml stock was prepared by diluting 0.25 ml Proteinase K (Qiagen, 19131) with 9.75 ml water. 

After a 5-minute incubation at room temperature the samples containing the beads were placed on an appropriately sized magnetic rack and beads were pelleted by the magnet for 5 minutes. 

A wax-layered cartridge that is ready-to-run following sample addition presents a challenge towards the homogenous mixing of all of its components owing to a pipette-free operation. 

While the RT-LAMP workflow delivers results faster than RT-PCR and is highly scalable, it still requires a trained technician to perform the assay as well as cold shipping and storage of reagents. 

similar to the mineral oil layer used prior to heated lids of thermocyclers, a wax layer above the reaction prevents evaporation and volume loss. 

PCR-based diagnostic tests are expensive, reliant on expensive centralized labs, can take days to deliver results, and are prone to backlogs and supply shortages. 

an RT-LAMP primer set has been designed to detect the saliva specific expression of STATH [24] to work in conjunction with saliva-based detection of SARS-CoV-2. 

but perhaps most crucially, the authors have removed the need for cold storage by utilizing a variety of low melting point waxes to produce reaction-ready cartridges, stable at room temperature for at least 1 month, and potentially up to 3 months. 

The minimal, and reusable, hardware necessary (an incubator and light source) to conduct RT-LAMP reactions with visual results, and the open-source solutions already existing such as the $2 GMO Detective fluorescence visualizer make inexpensive tests a distinct possibility without compromising on the sensitivity delivered by RT-PCR. 

To demonstrate the magnetic wand, the authors used it to transfer RNA-bound paramagnetic beads into a one-step reaction cartridge with 10 µl of molecular biology grade water at the top to elute the beads into. 

1,3,5,7,9,11,13,15,17,19,21 are NMT swabs treated with Proteinase K without being spiked with inactivated SARS-CoV-2. 2,8,14,20 NMT swabs spiked with 1,000 PFU of SARS-CoV-2. NMT swabs spiked with 500 PFU of SARS-CoV-2, 6,12,18 NMT swabs spiked with 250 PFU of SARS-CoV-2. 23.100 copies of synthetic SARSCoV-2 RNA, 24. 50 copies of synthetic SARS-CoV-2 RNA B. TCEP/EDTA lysis efficacy on NMT swabs spiked with inactivated SARS-CoV-2. 

Separating the 1-step RT-LAMP reaction into a 2-step RT/LAMP reaction decreased the LoD to 25 copies of synthetic SARS-CoV-2 RNA (Figure 2e). 

At this stage an optional bead wash can be performed by briefly dipping (~2 seconds) the magnetic wand into a tube with 85% ethanol filled to the same volume as the sample/bead solution.