ArticlePDF Available

Methamphetamine facilitates HIV infection of primary human monocytes through inhibiting cellular viral restriction factors

Authors:

Abstract and Figures

Background Methamphetamine (METH), a potent addictive psychostimulant, is highly prevalent in HIV-infected individuals. Clinically, METH use is implicated in alteration of immune system and increase of HIV spread/replication. Therefore, it is of importance to examine whether METH has direct effect on HIV infection of monocytes, the major target and reservoir cells for the virus. Results METH-treated monocytes were more susceptible to HIV infection as evidenced by increased levels of viral proteins (p24 and Pr55Gag) and expression of viral GAG gene. In addition, using HIV Bal with luciferase reporter gene (HIV Bal-eLuc), we showed that METH-treated cells expressed higher luciferase activities than untreated monocytes. Mechanistically, METH inhibited the expression of IFN-λ1, IRF7, STAT1, and the antiviral IFN-stimulated genes (ISGs: OAS2, GBP5, ISG56, Viperin and ISG15). In addition, METH down-regulated the expression of the HIV restriction microRNAs (miR-28, miR-29a, miR-125b, miR-146a, miR-155, miR-223, and miR-382). Conclusions METH compromises the intracellular anti-HIV immunity and facilitates HIV replication in primary human monocytes.
Content may be subject to copyright.
Liuetal. Cell Biosci (2021) 11:194
https://doi.org/10.1186/s13578-021-00703-4
RESEARCH
Methamphetamine facilitates HIV
infection ofprimary human monocytes
throughinhibiting cellular viral restriction
factors
Yu Liu1†, Feng‑Zhen Meng1†, Xu Wang1,2, Peng Wang1,2, Jin‑Biao Liu1, Wen‑Hui Hu1, Won‑Bin Young1 and
Wen‑Zhe Ho1,2*
Abstract
Background: Methamphetamine (METH), a potent addictive psychostimulant, is highly prevalent in HIV‑infected
individuals. Clinically, METH use is implicated in alteration of immune system and increase of HIV spread/replication.
Therefore, it is of importance to examine whether METH has direct effect on HIV infection of monocytes, the major
target and reservoir cells for the virus.
Results: METH‑treated monocytes were more susceptible to HIV infection as evidenced by increased levels of viral
proteins (p24 and Pr55Gag) and expression of viral GAG gene. In addition, using HIV Bal with luciferase reporter gene
(HIV Bal‑eLuc), we showed that METH‑treated cells expressed higher luciferase activities than untreated monocytes.
Mechanistically, METH inhibited the expression of IFN‑λ1, IRF7, STAT1, and the antiviral IFN‑stimulated genes (ISGs:
OAS2, GBP5, ISG56, Viperin and ISG15). In addition, METH down‑regulated the expression of the HIV restriction microR‑
NAs (miR‑28, miR‑29a, miR‑125b, miR‑146a, miR‑155, miR‑223, and miR‑382).
Conclusions: METH compromises the intracellular anti‑HIV immunity and facilitates HIV replication in primary
human monocytes.
Keywords: Methamphetamine, Human immunodeficiency virus, Interferon‑stimulated genes, Monocytes
© The Author(s) 2021. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which
permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the
original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or
other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line
to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this
licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/. The Creative Commons Public Domain Dedication waiver (http:// creat iveco
mmons. org/ publi cdoma in/ zero/1. 0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
Introduction
METH is one of the most widely abused illicit drugs
among HIV-infected individuals. METH use and HIV
infection frequently coexist due to the association of
METH use with engagement of high-risk behaviors
[13]. ere is a high prevalence of HIV infection in
METH using population [4, 5]. Among men who have
sex with men, those who use METH are more suscepti-
ble to HIV infection than non-users [610]. Clinically,
METH use has been implicated in HIV disease progres-
sion [11]. Active METH users with HIV infection display
higher levels of viral load than non-users [12]. In addi-
tion, METH users have delayed viral suppression after
initiation of antiretroviral therapy (ART), higher levels of
blood HIV RNA, increased frequency of drug resistance
mutations and accelerated progression to AIDS [1317].
METH abuse contributes to CD4+ T cells depletion,
inflammation/immune activation, and the promotion of
HIV entry and disease progression [18].
Open Access
Cell & Bioscience
*Correspondence: wenzheho@temple.edu
Yu Liu and Feng‑Zhen Meng contributed equally to this work
1 Department of Pathology and Laboratory Medicine, Temple University
Lewis Katz School of Medicine, 3500 N Broad St., Philadelphia, PA 19140,
USA
Full list of author information is available at the end of the article
Page 2 of 9
Liuetal. Cell Biosci (2021) 11:194
Cells of monocyte/macrophage lineage are crucial
in initial HIV infection and implicated in the immu-
nopathogenesis of HIV disease. Monocytes are among
the first and major cell types infected by HIV and serve
as reservoirs for the virus. However, unlike tissue mac-
rophages and invitro monocyte-derived macrophages
which are highly susceptible to HIV infection, periph-
eral blood monocytes are refractory to HIV infec-
tion invivo and invitro, and only a small percentage
of monocytes harbor the virus [19, 20]. Despite of
their relative resistance to HIV infection, monocytes
are involved in HIV infection of the central nervous
system (CNS) as they can bring the virus to the brain
[21]. A study reported that HIV-infected monocytes
are more likely to cross the blood brain barrier (BBB) as
compared to uninfected monocytes [22]. Among HIV-
infected METH users, HIV-associated neurocognitive
disorders (HAND) are more frequent and severe [23,
24]. A recent study demonstrated that METH could
enhance HIV infection of neural progenitor cells, a
possible mechanism for the impairment or disruption
of neurocognitive functioning in HIV-infected indi-
viduals with NeuroAIDS [25]. Several studies showed
that elevated extracellular CNS dopamine by METH
abuse could facilitate uninfected and HIV-infected
CD14+CD16+ monocytes transmigration across the
BBB, resulting in the propagation of viral reservoirs and
inflammation in the CNS which contribute to the devel-
opment of HAND [26, 27]. us, it is of great interest
to determine the direct impact of METH on suscepti-
bility of peripheral blood monocytes to HIV infection.
In addition, it is critical to understand the pathologi-
cal effects of METH on the specific intracellular innate
immunity against HIV in monocytes.
Results
METH enhances HIV infection
We first examined cytotoxicity effect of METH on
monocytes. As shown in Additional file 1: Fig. S1,
METH at the concentration as high as 1000μM had
little effect on cell viability. We then studied whether
METH could enhance susceptibility of monocytes
to HIV infection. As demonstrated in Fig. 1A and B,
METH treatment of monocytes dose-dependently
increased the expression of both intracellular and
extracellular HIV GAG gene expression. In addition,
METH-treated monocytes showed higher levels of
HIV (p24 and Pr55Gag) proteins than untreated cells
(Fig.1C and D). e enhancing effect of METH on HIV
p24 protein production was dose-dependent (Fig.1E).
As shown in Fig.1F, METH treatment enhanced lucif-
erase activity in HIV Bal-eLuc-infected cells.
METH suppresses theJAK/STAT signaling pathway
To study the mechanisms by which METH enhances
HIV infection of monocytes (Fig.1), we examined the
effect of METH on IFNs. As shown in Fig.2A, while it
had little effect on IFN-α/β expression, METH treat-
ment significantly suppressed IFN-λ1 expression in
monocytes. In addition, METH inhibited the expression
of phosphorylated IRF7 in a time-dependent fashion
(Fig.2B).
IRF7 is a key regulator for both type I and type III
IFNs during viral infections [28, 29]. The phosphoryla-
tion of IRF7 would directly trigger the transcription of
IFNs and the downstream antiviral signaling, includ-
ing the activation of JAK/STAT pathway and the pro-
duction of ISGs. We also examined whether METH
has a negative impact on the expression of STAT fam-
ily members including STAT1, STAT2 and STAT3,
the crucial factors in JAK/STAT signaling pathway
[30, 31]. As shown in Fig.2C, while METH treatment
of monocytes had little effect on STAT2 and STAT3
expression, it significantly inhibited STAT1 expres-
sion at both mRNA and protein levels, and reduced
the phosphorylation of STAT1. We next examined the
effect of METH on the expression of the intracellu-
lar antiviral ISGs. As shown in Fig.2D, METH dose-
dependently inhibited the expression of the antiviral
ISGs (OAS2, GBP5, ISG56, Viperin, ISG15) at 24 h
post-treatment. In addition, the Western blot analy-
sis demonstrated that METH-treated monocytes had
lower protein levels of the antiviral ISGs than the
untreated cells (Fig.2E).
METH inhibits HIV restriction miRNAs
Our earlier study showed that monocytes contain sig-
nificantly higher levels of the HIV restriction miRNAs
than monocyte-derived macrophages, which explains
why monocytes are refractory to HIV infection [32]. We
thus investigated whether METH negatively influences
the expression of the HIV restriction miRNAs in mono-
cytes. As shown in Fig.2F, METH treatment of mono-
cytes suppressed the expression of the intracellular HIV
restriction miRNAs (miR-28, miR-29a, miR-125b, miR-
146a, miR-155, miR-223, and miR-382). In addition, we
observed lower levels of these HIV restriction miRNAs in
the supernatants of monocyte cultures 36h after METH
treatment compared to those in untreated cells (Fig.2G).
Discussion
Although METH use has been linked to HIV transmis-
sion and infection, its pathological effects on the host
cell-mediated specific innate immunity against HIV
Page 3 of 9
Liuetal. Cell Biosci (2021) 11:194
Fig. 1 METH enhances HIV infection of primary human monocytes. AE Monocytes isolated from human peripheral blood were treated with
METH for 24 h and then infected with HIV Bal strain overnight. Cells were washed with PBS three times and cultured in the presence of METH for
72 h. RNAs extracted from cells (A) and the cell‑free supernatants (B) were subjected to the real‑time PCR with HIV GAG gene primers. C, D Proteins
of cells and culture supernatants were analyzed by Western blot using the antibodies against HIV proteins (p24 and Pr55Gag) and GAPDH. E The
cell‑free supernatants were subjected to ELISA assay to quantitatively determine p24 protein level. F Monocytes were treated with METH (150 μM)
for 24 h and then infected with HIV Bal‑eLuc overnight. Cells were washed with PBS three times and cultured in the presence of METH for 24 h or
48 h prior to luminescence assay. Data shown were the mean ± SD of three independent experiments with monocytes from three different donors
(*P < 0.05, **P < 0.01)
Page 4 of 9
Liuetal. Cell Biosci (2021) 11:194
infection remain to be determined. e earlier stud-
ies reported that METH could enhance HIV infec-
tion of several cell types, including dendritic cells [33],
macrophages [34, 35], CD4+ T cells [36, 37], micro-
glia [38], and neural progenitor cells [25]. However, it
is unclear whether METH facilitates HIV infection of
primary human monocytes. In the present study, we
demonstrated that METH treatment of the monocytes
significantly enhanced HIV infection/replication at
both intracellular and extracellular levels (Fig. 1). To
investigate the underlying mechanisms of METH-medi-
ated HIV enhancement in monocytes, we examined
that the impact of METH on the expression of IFNs-
JAK/STAT signaling pathways. We found that although
METH treatment of monocytes had little effect on
IFN-α and IFN-β expression, it significantly suppressed
IFN-λ1 expression (Fig.2A). IFN-λ can induce type I
IFN-like antiviral response and inhibition of HIV [39,
40]. It is likely that IFN-λ inhibition by METH can result
in reduction of ISGs. e following results indicated
Fig. 2 METH inhibits viral restriction factors. AE Monocytes from human peripheral blood were treated with METH (150 μM) for the indicated
times or at the indicated concentrations for 24 h. A The cellular RNAs were extracted and subjected to the real‑time PCR for IFN‑α, IFN‑β, and IFN‑λ1
mRNA expression, the culture supernatants were collected and subjected to ELISA for IFN‑λ1 protein expression. BE The cellular RNAs or proteins
were extracted and subjected to the real‑time PCR or Western blot assays. F, G Monocytes were treated with METH (150 μM) for the indicated times.
Cellular miRNAs were quantified by the real‑time PCR. RNU48 was used as a control gene. miRNAs in the culture supernatants were quantified by
the real‑time PCR. Synthetic Caenorhabditis elegans miRNA‑39 (cel‑miR‑39) was used as a spiked‑in control miRNA for normalization. Data shown
were the mean ± SD of three independent experiments with monocytes from three different donors (*P < 0.05, **P < 0.01)
Page 5 of 9
Liuetal. Cell Biosci (2021) 11:194
that METH treatment of the cells down-regulated the
expression of the antiviral ISGs, including OAS2, GBP5,
ISG56, Viperin, and ISG15 (Fig.2D and E). ese ISGs
are known to have the ability to restrict HIV replication
at different steps of viral replication cycle [4145]. For
instance, Krapp etal. demonstrated that the expression
of GBP5 could interfere with the processing and virion
incorporation of the HIV envelope glycoprotein, which
remarkably reduce virion incorporation of mature
gp120 and enhance virion-associated immature gp160
precursor, leading to the inhibition of HIV infectivity
[42]. Okumura etal. showed that ISG15 impaired the
interaction between HIV GAG and tumor susceptibility
gene 101 (Tsg101), and suppressed HIV virion release
[46].
Given that the IFN regulatory factors (IRFs) are
responsible for IFN-JAK/STAT signaling pathway and
the production of the antiviral ISGs, we also exam-
ined the impact of METH on IRFs, particularly IRF1,
IRF3, IRF5 and IRF7, the key players in regulating the
expression of antiviral ISGs and producing an antiviral
state [47]. We found that while METH had little effect
on IRF1, IRF3, IRF5, it specifically suppressed IRF7
expression in monocytes at both mRNA and protein
levels (Fig.2B). IRF7 has a key role for the production
of both type I and type III IFNs during viral infections
[28, 29]. e phosphorylation of IRF7 would directly
trigger the transcription of IFNs and the downstream
antiviral signaling, including the activation of JAK/
STAT pathway. erefore, it is likely that IRF7 suppres-
sion by METH is a possible mechanism for STAT1 inhi-
bition in METH-treated monocytes (Fig. 2C). STAT1
is a crucial regulatory factor in IFNs-mediated induc-
tion of antiviral ISGs [48, 49]. While exact mechanism
by which METH inhibits IRF and STAT remain to be
determined, it is possible that down-regulation of IFN-
λ1 has a negative impact on both IRF7 and STAT1
expression.
In addition to the HIV restriction factors of pro-
tein nature, a cluster of HIV restriction miRNAs have
also been shown to be a contributor to HIV latency in
resting CD4+ T cells [50, 51]. These cellular miRNAs
interact with the 3’-termini of HIV RNA, resulting in
the transcriptional inefficiency and post-transcrip-
tional suppression [52, 53]. Our earlier study showed
that primary human monocytes expressed much
higher levels of miRNAs (miRNA-382, -223, -150,
and -28) than monocyte-derived macrophages, and
the suppression of these miRNAs facilitates HIV-1
infectivity, which provide direct evidence that HIV
restriction miRNAs have a key role in protecting
monocytes/macrophages from HIV-1 infection [32].
Several studies reported that METH use altered the
miRNA expression in human serum or animal model
[54, 55]. We thus determined whether METH could
regulate the expression of the miRNAs that are impli-
cated in HIV infection and persistence. Among the
cellular miRNAs, the miR-29 family members (miR-
29a, miR-29b, and miR-29c) suppress HIV replication
by targeting a highly conserved region of HIV [56].
Importantly, our early study documented that the lev-
els of the cellular HIV restriction miRNAs arenega-
tively correlated with susceptibility of monocytes and
macrophages to HIV infection [32]. We also reported
that ART failed to restore the levels of several HIV
restriction miRNAs in PBMCs of HIV-infected men
who have sex with men who used METH [57]. There-
fore, it is clinically relevant to seek the direct evi-
dence of how METH negatively impacts on the HIV
restriction miRNAs. Our observation that METH
could significantly suppress the expression of the HIV
restriction miRNAs (Fig.2F and G) provide not only
direct evidence for our invivo finding [57], but also an
additional mechanism for METH-mediated enhance-
ment of HIV infection.
It is suggested that METH could facilitate HIV entry
into the cells through up-regulation CXCR4 and CCR5,
the key coreceptors for HIV entry into target cells [33,
34]. However, in contrast to these earlier studies, we did
not observe the enhancing effect of METH on CCR5
expression in monocytes (Additional file 2: Fig. S2).
METH also had no significant impact on the expression
of CD4. is conflicting finding could be due to the
difference in use of different cell types: while we used
primary human monocytes, the previous studies used
monocyte-derived dendritic cells [33] and macrophages
[34] which are more susceptible to HIV infection com-
pared to monocytes [32].
In summary, we demonstrate that METH can enhance
HIV infection of primary human in monocytes through
the inhibition of the multiple cellular antiviral factors
(IFN-λ1, ISGs, and miRNAs). While it is possible that
there are additional mechanisms involved in the METH
action on HIV enhancement, compromising the intra-
cellular immunity against HIV should be responsible
for much of METH-mediated HIV enhancement in
monocytes. ese findings suggest that METH use is a
contributing factor for HIV infection and persistence
in monocytes. As HIV-infected individuals are living
longer with ART and many of infected individuals are
METH users, to further identify the pathological role of
METH in HIV-infected reservoir cells is necessary for
Page 6 of 9
Liuetal. Cell Biosci (2021) 11:194
understanding the mechanisms of HIV persistence and
developing strategies for the viral eradication.
Conclusions
METH compromises the intracellular anti-HIV immu-
nity and facilitates HIV replication in primary human
monocytes.
Materials andmethods
Cells andreagents
Purified human peripheral blood monocytes were
obtained from Human Immunology Core at the Uni-
versity of Pennsylvania (Philadelphia, PA, USA). The
Core has the Institutional Review Board approval for
blood collection from healthy donors. These blood
samples were screened for all normal viral bloodborne
pathogens and certified to be pathogen free. The pro-
tocol of monocyte isolation was described previously
[34]. Briefly, after the initial purification, greater
than 97% of the cells were monocytes, as determined
by nonspecific esterase staining and flow cytometry
analysis using monoclonal antibody against CD14,
the marker specific for monocytes and macrophages.
Freshly isolated monocytes were cultured in 1640
RPMI (Gibco, New York, USA) medium supplemented
with 10% fetal bovine serum (Corning, New York,
USA), 1% MEM NEAA (Gibco, New York, USA), 1%
L-Glutamine (Gibco, New York, USA) and 1% penicil-
lin–streptomycin solution (Lonza, Walkersville, GA,
USA). Rabbit antibodies against OAS2, GBP5, ISG56,
Viperin, ISG15, IRF7, p-IRF7, STAT1, p-STAT1 and
GAPDH were purchased from Cell Signaling Technol-
ogy (Danvers, MA, USA). Mouse anti-HIV p24 anti-
body was purchased from Abcam (Abcam, Cambridge,
UK). METH was purchased from Sigma Aldrich (St
Louis, MO, USA). METH powder was dissolved in
sterile endotoxin-free water (HyPure Cell culture
grade water, GE Healthcare Life Science, Logan, UT,
USA) and stored at 4. Trichloroacetic acid (TCA)
and acetone were purchased from Sigma-Aldrich. All
antibodies and reagents for flow cytometry assay were
purchased from BD Bioscience (BD Bioscience, San
Jose, CA, USA).
HIV infection andMETH treatment
HIV Bal strain was obtained from AIDS Reagent Pro-
gram (NIH, Bethesda, MD), HIV Bal with luciferase
reporter gene (HIV Bal-eLuc) was generated by Dr. Won-
Bin Young [58]. Freshly isolated and purified monocytes
were treated with METH at clinically relevant concentra-
tions [5962] (0, 100, 150, and 250μM) for 24h before
being infected with HIV Bal strain (p24 60ng/106 cells)
or HIV Bal-eLuc (p24 60ng/106 cells) overnight. e cells
were then washed three times with plain RPMI to remove
any unabsorbed virus and cultured in the presence of
METH.
MTS assay
e cytotoxic effect of METH on monocytes was
evaluated by MTS (3-(4, 5-dimethylthiazol-2-yl)-
5-(3carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetra-
zolium, innersalt) assay. Freshly isolated human blood
monocytes (2 × 104 cells/well) were placed in 96-well
round bottom plates, and treated with different con-
centrations of METH (0, 100, 150, 250, 400, 600, and
1000μM) for 96h. e cells were then incubated with
CellTiter 96® AQueous One Solution Reagent (Promega
Corporation, Madison, WI) containing MTS and phena-
zine ethosulfate for 4h at 37. Absorbance at 490nm
was measured by a plate reader (SpectraMax i3, Molecu-
lar Devices, Sunnyvale, CA, USA).
ELISA
HIV p24 protein levels in monocyte culture supernatants
were determined by ELISA kit from Abnova (Taipei, Tai-
wan) as instructed by the manufacturer. IFN-λ1 protein
levels in monocyte culture supernatants were determined
by human IL-29 (IFN lambda1) ELISA kit from Invitro-
gen (Invitrogen, CA, USA) according to the manufactur-
er’s instruction.
Flow cytometry assay
Monocytes from human peripheral blood were treated
with METH (150μM) for 24h. en the cells were col-
lected and washed with a cell staining buffer prior to
staining with PE mouse anti-human CD4 antibody and
PE mouse anti-human CCR5 antibody, respectively.
PE-isotype IgG antibody-stained cells were used as the
negative control. e stained cells were measured by a
FACSCanto II (BD Bioscience, CA, USA) and analyzed
using FlowJo software (Tree Star Inc., Ashland, OR,
USA).
Western blot assay
Proteins from monocytes and culture supernatants
were determined by Western blot assay for viral pro-
teins (p24 and Pr55Gag) expression. Monocytes were
lysed with RIPA lysis buffer supplemented with pro-
tease/phosphatase inhibitors (Sigma Aldrich, St Louis,
MO). Proteins from the culture supernatants were
extracted by the TCA/acetone precipitation method.
Briefly, 0.5mL of culture supernatants were precipi-
tated with 0.5mL of 20% TCA at 20°C for 1h and
Page 7 of 9
Liuetal. Cell Biosci (2021) 11:194
then centrifuged at 11,500rpm for 15min at 4°C. After
three washes with 1mL of ice-cold acetone, the pellet
was lysed with Western blot lysis buffer. e protein
concentrations were determined by the bicinchoninic
acid (BCA) assay (ChemCruz, Dallas, TX). e blots
were incubated with primary antibodies in 5% nonfat
milk in PBS overnight at 4, then washed with PBS
containing 0.5% Tween. e blots were further incu-
bated with horseradish peroxidase-conjugated sec-
ond antibodies at room temperature for an hour, then
washed with PBST. e blots were developed with
enhanced chemiluminescence (Amersham, Bucks, UK)
and then exposed to an iBright 1500 imaging analyzer
(Invitrogen, CA,USA).
RNA andmicroRNA extraction andquantication
Freshly isolated monocytes in 48-well plates were
treated with or without METH (100, 150, and 250μM)
for different time points (0, 6, 12, and 24 h). Total
RNAs were extracted with Tri-reagent (Molecular
Research Center, OH, USA). RNA (1μg) was subjected
to reverse transcriptase PCR using reagents from Pro-
mega (Promega, WI, USA). e cDNA sample was then
subjected to the real-time PCR using iQ SYBR Green
Supermix (Bio-Rad Laboratories, CA, USA). All values
were normalized to GAPDH mRNA. e sequences of
oligonucleotide primers used in this study are listed
in Table1. Extracellular miRNAs were extracted from
supernatants of monocyte cultures using the miRNeasy
Mini Kit (Qiagen, CA, USA). e miRNAs from cells
or supernatants were reversely transcribed with miS-
cript Reverse Transcription Kit (Qiagen, CA, USA).
e real-time PCR for the miRNAs quantification was
carried out with miScript Primer Assays using miS-
cript SYBR Green PCR Kit from Qiagen as previously
described [32].
HIV GAG gene quantication
HIV GAG gene copy numbers in monocytes or mono-
cytes culture supernatants were determined by the
real-time PCR. RNAs from cells or the cell-free super-
natants were extracted with Tri-reagent (for tissues,
cells cultured in monolayer, or cell pellets) or Tri-rea-
gent (for whole blood, serum/plasma or cell culture
supernatant) according to the manufacturer’s instruc-
tions, respectively. HIV GAG standards with known
copy numbers were used to quantify viral GAG gene
expression in the culture supernatants.
Statistical analysis
Data were expressed as mean ± standard de viation
(mean ± SD) of three experiments using monocytes from
three different donors. Statistical significance was meas-
ured by Student’s t-test using GraphPad Prism Statistical
Software (GraphPad Software, La Jolla, USA). *P < 0.05
and **P < 0.01 indicate statistic difference between com-
pared groups.
Supplementary Information
The online version contains supplementary material available at https:// doi.
org/ 10. 1186/ s13578‑ 021‑ 00703‑4.
Table 1 Primers Pairs
Gene Orientation Sequence (5-3)
GAPDH Forward GGT GGT CTC CTC TGA CTT CAACA
Reverse GTT GCT GTA GCC AAA TTC GTTGT
HIV GAG Forward ATA ATC CAC CTA TCC CAG TAG GAG AAA
Reverse TTT GGT CCT TGT CTT ATG TCC AGA ATGC
OAS2 Forward CAG TCC TGG TGA GTT TGC AGT
Reverse ACA GCG AGG GTA AAT CCT TGA
GBP5 Forward CAG GAA CAA CAG ATG CAG GA
Reverse TCA TCG TTA TTA ACA GTC CTC TGG
ISG56 Forward TTC GGA GAA AGG CAT TAG A
Reverse TCC AGG GCT TCA TTC ATA T
Viperin Forward TGG GTG CTT ACA CCT GCT G
Reverse TGA AGT GAT AGT TGA CGC TGGT
ISG15 Forward GGC TGG GAG CTG ACG GTG AAG
Reverse GCT CCG CCC GCC AGG CTC TGT
IRF1 Forward TGA AGC TAC AAC AGA TGA GG
Reverse AGT AGG TAC CCC TTC CCA TC
IRF3 Forward ACC AGC CGT GGA CCA AGA G
Reverse TAC CAA GGC CCT GAG GCA C
IRF5 Forward AAG CCG ATC CGG CCAA
Reverse GGA AGT CCC GGC TCT TGT TAA
IRF7 Forward TGG TCC TGG TGA AGC TGG AA
Reverse GAT GTC GTC ATA GAG GCT GTTGG
STAT1 Forward CCG TGG CAC TGC ATA CAA TC
Reverse ACC ATG CCG AAT TCC CAA AG
STAT2 Forward CCC CAT CGA CCC CTC ATC
Reverse GAG TCT CAC CAG CAG CCT TGT
STAT3 Forward CTG CCC CAT ACC TGA AGA CC
Reverse TCC TCA CAT GGG GGA GGT AG
IFN‑α Forward TTT CTC CTG CCT GAA GAA CAG
Reverse GCT CAT GAT TTC TGC TCT GACA
IFN‑β Forward GCC GCA TTG ACC ATC TAT GAGA
Reverse GAG ATC TTC AGT TTC GGA GGT AAC
IFN‑λ1 Forward CTT CCA AGC CCA CCC CAA CT
Reverse GGC CTC CAG GAC CTT CAG C
CD4 Forward AGT CCC TTT TAG GCA CTT GC
Reverse GAT CAT TCA GCT TGG ATG G
CCR5 Forward CAA GTG TCA AGT CCA ATC TA
Reverse ACC AAA GAT GAA CAC CAG TG
Page 8 of 9
Liuetal. Cell Biosci (2021) 11:194
Additional le1: Fig. S1. Effect of METH on the cell viability of human
monocytes. Freshly isolated human monocytes were treated with
METH at the indicated concentrations for 96 hours. The cell viability was
assessed by MTS assay. Data are showed as the absorbance (490 nm)
relative to untreated control, which is defined as 1.0. The results shown
were obtained as mean ± SD from three independent experiments with
triplicate wells.
Additional le2: Fig. S2. Effect of METH on CD4 and CCR5. (A, B) Freshly
isolated human monocytes were treated with METH (150 μM) at the indi‑
cated time points. The cellular RNA was subjected to the real‑time PCR for
CD4 and CCR5 expression. (C, D) Freshly isolated monocytes were treated
with METH (150 μM) for 24 h and then collected for the flow cytometry
analysis of CD4 and CCR5 protein expression. Data are shown in A and B
as mean ± SD from three independent experiments with triplicate wells.
Flow cytometry data shown in C and D are the representative pictures of
three independent experiments.
Acknowledgements
Not applicable.
Authors’ contributions
YL, FZM, XW, WZH designed the research; YL, FZM, PW, JBL, WBY, and WHH
performed the experiments and analyzed the data; YL, FZM, XW, and WZH
interpreted the data and wrote the paper; YL, FZM, XW, JBL, WHH, WZH
discussed and edited the paper. All authors read and approved the final
manuscript.
Funding
This work is financially supported by the NIH (DA 51893 and DA 41302 for WZ
Ho).
Availability of data and materials
The datasets used and/or analyzed during the current study are available from
the corresponding author on reasonable request.
Declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
All authors agree to publish this paper.
Competing interests
No potential conflict of interest was reported by the authors.
Author details
1 Department of Pathology and Laboratory Medicine, Temple University
Lewis Katz School of Medicine, 3500 N Broad St., Philadelphia, PA 19140, USA.
2 Center for Substance Abuse Research, Temple University Lewis Katz School
of Medicine, Philadelphia, PA 19140, USA.
Received: 31 May 2021 Accepted: 27 October 2021
References
1. Marshall BD, Wood E, Shoveller JA, Patterson TL, Montaner JS, Kerr T.
Pathways to HIV risk and vulnerability among lesbian, gay, bisexual,
and transgendered methamphetamine users: a multi‑cohort gender‑
based analysis. BMC Public Health. 2011;11:20.
2. Pitpitan EV, Semple SJ, Zians J, Strathdee SA, Patterson TL. Mood, meth,
condom use, and gender: latent growth curve modeling results from a
randomized trial. AIDS Behav. 2018;22(9):2815–29.
3. Semple SJ, Patterson TL, Grant I. Motivations associated with meth
amphetamine use among HIV+ men who have sex with men. J Subst
Abuse Treat. 2002;22(3):149–56.
4. Nerlander LMC, Hoots BE, Bradley H, Broz D, Thorson A, Paz‑Bailey G,
NHBS Group. HIV infection among MSM who inject methamphetamine
in 8 US cities. Drug Alcohol Depend. 2018;190:216–23.
5. Vu NT, Maher L, Zablotska I. Amphetamine‑type stimulants and HIV
infection among men who have sex with men: implications on HIV
research and prevention from a systematic review and meta‑analysis. J
Int AIDS Soc. 2015;18:19273.
6. Morin SF, Steward W T, Charlebois ED, Remien RH, Pinkerton SD,
Johnson MO, Rotheram‑Borus MJ, Lightfoot M, Goldstein RB, Kittel L,
et al. Predicting HIV transmission risk among HIV‑infected men who
have sex with men: findings from the healthy living project. J Acquir
Immune Defic Syndr. 2005;40(2):226–35.
7. Plankey MW, Ostrow DG, Stall R, Cox C, Li X, Peck JA, Jacobson LP. The
relationship between methamphetamine and popper use and risk
of HIV seroconversion in the multicenter AIDS cohort study. J Acquir
Immune Defic Syndr. 2007;45(1):85–92.
8. Shoptaw S, Reback CJ. Associations between methamphetamine use
and HIV among men who have sex with men: a model for guiding
public policy. J Urban Health. 2006;83(6):1151–7.
9. Shoptaw S, Reback CJ. Methamphetamine use and infectious disease‑
related behaviors in men who have sex with men: implications for
interventions. Addiction. 2007;102(Suppl 1):130–5.
10. Urbina A, Jones K. Crystal methamphetamine, its analogues, and HIV
infection: medical and psychiatric aspects of a new epidemic. Clin Infect
Dis. 2004;38(6):890–4.
11. Moore RD, Keruly JC, Chaisson RE. Differences in HIV disease progression
by injecting drug use in HIV‑infected persons in care. J Acquir Immune
Defic Syndr. 2004;35(1):46–51.
12. Ellis RJ, Childers ME, Cherner M, Lazzaretto D, Letendre S, Grant I. Group
HIVNRC: increased human immunodeficiency virus loads in active
methamphetamine users are explained by reduced effectiveness of
antiretroviral therapy. J Infect Dis. 2003;188(12):1820–6.
13. Carrico AW, Johnson MO, Colfax GN, Moskowitz JT. Affective correlates
of stimulant use and adherence to anti‑retroviral therapy among HIV‑
positive methamphetamine users. AIDS Behav. 2010;14(4):769–77.
14. Colfax GN, Vittinghoff E, Grant R, Lum P, Spotts G, Hecht FM. Frequent
methamphetamine use is associated with primary non‑nucleoside
reverse transcriptase inhibitor resistance. AIDS. 2007;21(2):239–41.
15. Fairbairn N, Kerr T, Milloy MJ, Zhang R, Montaner J, Wood E. Crystal meth‑
amphetamine injection predicts slower HIV RNA suppression among
injection drug users. Addict Behav. 2011;36(7):762–3.
16. Kapadia F, Cook JA, Cohen MH, Sohler N, Kovacs A, Greenblatt RM,
Choudhary I, Vlahov D. The relationship between non‑injection drug use
behaviors on progression to AIDS and death in a cohort of HIV seroposi‑
tive women in the era of highly active antiretroviral therapy use. Addic‑
tion. 2005;100(7):990–1002.
17. Massanella M, Gianella S, Schrier R, Dan JM, Perez‑Santiago J, Oliveira MF,
Richman DD, Little SJ, Benson CA, Daar ES, et al. Methamphetamine use
in HIV‑infected individuals affects T‑cell function and viral outcome dur‑
ing suppressive antiretroviral therapy. Sci Rep. 2015;5:13179.
18. Jiang J, Wang M, Liang B, Shi Y, Su Q, Chen H, Huang J, Su J, Pan P, Li
Y, et al. In vivo effects of methamphetamine on HIV‑1 replication: a
population‑based study. Drug Alcohol Depend. 2016;159:246–54.
19. Kedzierska K, Crowe SM. The role of monocytes and macrophages in the
pathogenesis of HIV‑1 infection. Curr Med Chem. 2002;9(21):1893–903.
20. Zhu T. HIV‑1 in peripheral blood monocytes: an underrated viral source. J
Antimicrob Chemother. 2002;50(3):309–11.
21. Strazza M, Pirrone V, Wigdahl B, Nonnemacher MR. Breaking down
the barrier: the effects of HIV‑1 on the blood‑brain barrier. Brain Res.
2011;1399:96–115.
22. Williams DW, Eugenin EA, Calderon TM, Berman JW. Monocyte matura‑
tion, HIV susceptibility, and transmigration across the blood brain barrier
are critical in HIV neuropathogenesis. J Leukoc Biol. 2012;91(3):401–15.
23. Sanmarti M, Ibanez L, Huertas S, Badenes D, Dalmau D, Slevin M, Krupin‑
ski J, Popa‑Wagner A, Jaen A. HIV‑associated neurocognitive disorders. J.
Mol Psychiatry. 2014;2(1):2.
24. Zayyad Z, Spudich S. Neuropathogenesis of HIV: from initial neuroinva‑
sion to HIV‑associated neurocognitive disorder (HAND). Curr HIV/AIDS
Rep. 2015;12(1):16–24.
Page 9 of 9
Liuetal. Cell Biosci (2021) 11:194
25. Skowronska M, McDonald M, Velichkovska M, Leda AR, Park M, Toborek M.
Methamphetamine increases HIV infectivity in neural progenitor cells. J
Biol Chem. 2018;293(1):296–311.
26. Calderon TM, Williams DW, Lopez L, Eugenin EA, Cheney L, Gaskill PJ,
Veenstra M, Anastos K, Morgello S, Berman JW. Dopamine increases
CD14(+)CD16(+) monocyte transmigration across the blood brain
barrier: implications for substance abuse and HIV neuropathogenesis. J
Neuroimmune Pharmacol. 2017;12(2):353–70.
27. Gaskill PJ, Calderon TM, Coley JS, Berman JW. Drug induced increases in
CNS dopamine alter monocyte, macrophage and T cell functions: impli‑
cations for HAND. J Neuroimmune Pharmacol. 2013;8(3):621–42.
28. Honda K, Taniguchi T. IRFs: master regulators of signalling by Toll‑like
receptors and cytosolic pattern‑recognition receptors. Nat Rev Immunol.
2006;6(9):644–58.
29. Osterlund PI, Pietilä TE, Veckman V, Kotenko SV, Julkunen I. IFN regulatory
factor family members differentially regulate the expression of type III IFN
(IFN‑lambda) genes. J Immunol. 2007;179(6):3434–42.
30. Chen K, Liu J, Cao X. Regulation of type I interferon signaling in immunity
and inflammation: a comprehensive review. J Autoimmun. 2017;83:1–11.
31. Hillmer EJ, Zhang H, Li HS, Watowich SS. STAT3 signaling in immunity.
Cytokine Growth Factor Rev. 2016;31:1–15.
32. Wang X, Ye L, Hou W, Zhou Y, Wang YJ, Metzger DS, Ho WZ. Cellular
microRNA expression correlates with susceptibility of monocytes/mac‑
rophages to HIV‑1 infection. Blood. 2009;113(3):671–4.
33. Nair MP, Saiyed ZM, Nair N, Gandhi NH, Rodriguez JW, Boukli N,
Provencio‑Vasquez E, Malow RM, Miguez‑Burbano MJ. Methampheta‑
mine enhances HIV‑1 infectivity in monocyte derived dendritic cells.
J Neuroimmune pharmacology : the official journal of the Society on
NeuroImmune Pharmacology. 2009;4(1):129–39.
34. Liang H, Wang X, Chen H, Song L, Ye L, Wang SH, Wang YJ, Zhou L, Ho WZ.
Methamphetamine enhances HIV infection of macrophages. Am J Pathol.
2008;172(6):1617–24.
35. Wang X, Wang Y, Ye L, Li J, Zhou Y, Sakarcan S, Ho W. Modulation of intra‑
cellular restriction factors contributes to methamphetamine‑mediated
enhancement of acquired immune deficiency syndrome virus infection
of macrophages. Curr HIV Res. 2012;10(5):407–14.
36. Lawson KS, Prasad A, Groopman JE. Methamphetamine enhances HIV‑1
replication in CD4(+) T‑cells via a novel IL‑1beta auto‑regulatory loop.
Front Immunol. 2020;11:136.
37. Prasad A, Kulkarni R, Shrivastava A, Jiang S, Lawson K, Groopman JE.
Methamphetamine functions as a novel CD4(+) T‑cell activator via the
sigma‑1 receptor to enhance HIV‑1 infection. Sci Rep. 2019;9(1):958.
38. Wires ES, Alvarez D, Dobrowolski C, Wang Y, Morales M, Karn J, Harvey
BK. Methamphetamine activates nuclear factor kappa‑light‑chain‑
enhancer of activated B cells (NF‑κB) and induces human immunode‑
ficiency virus (HIV) transcription in human microglial cells. J Neurovirol.
2012;18(5):400–10.
39. Hou W, Wang X, Ye L, Zhou L, Yang ZQ, Riedel E, Ho WZ. Lambda
interferon inhibits human immunodeficiency virus type 1 infection of
macrophages. J Virol. 2009;83(8):3834–42.
40. Wang Y, Li J, Wang X, Zhou Y, Zhang T, Ho W. Comparison of antiviral
activity of lambda‑interferons against HIV replication in macrophages. J
Interferon Cytokine Res. 2015;35(3):213–21.
41. Fagone P, Nunnari G, Lazzara F, Longo A, Cambria D, Distefano G,
Palumbo M, Nicoletti F, Malaguarnera L, Di Rosa M. Induction of OAS
gene family in HIV monocyte infected patients with high and low viral
load. Antiviral Res. 2016;131:66–73.
42. Krapp C, Hotter D, Gawanbacht A, McLaren PJ, Kluge SF, Sturzel CM, Mack
K, Reith E, Engelhart S, Ciuffi A, et al. Guanylate binding protein (GBP) 5
is an interferon‑inducible inhibitor of HIV‑1 infectivity. Cell Host Microbe.
2016;19(4):504–14.
43. Morales DJ, Lenschow DJ. The antiviral activities of ISG15. J Mol Biol.
2013;425(24):4995–5008.
44. Nasr N, Alshehri AA, Wright TK, Shahid M, Heiner BM, Harman AN, Bot‑
ting RA, Helbig KJ, Beard MR, Suzuki K, et al. Mechanism of interferon‑
stimulated gene induction in HIV‑1‑infected macrophages. J Virol.
2017;91(20):e00744.
45. Nasr N, Maddocks S, Turville SG, Harman AN, Woolger N, Helbig KJ, Wilkin‑
son J, Bye CR, Wright TK, Rambukwelle D, et al. HIV‑1 infection of human
macrophages directly induces viperin which inhibits viral production.
Blood. 2012;120(4):778–88.
46. Okumura A, Lu G, Pitha‑Rowe I, Pitha PM. Innate antiviral response targets
HIV‑1 release by the induction of ubiquitin‑like protein ISG15. Proc Natl
Acad Sci USA. 2006;103(5):1440–5.
47. Ikushima H, Negishi H, Taniguchi T. The IRF family transcription factors
at the interface of innate and adaptive immune responses. Cold Spring
Harb Symp Quant Biol. 2013;78:105–16.
48. Brierley MM, Fish EN. Stats: multifaceted regulators of transcription. J
Interferon Cytokine Res. 2005;25(12):733–44.
49. Raftery N, Stevenson NJ. Advances in anti‑viral immune defence:
revealing the importance of the IFN JAK/STAT pathway. Cell Mol Life Sci.
2017;74(14):2525–35.
50. Huang J, Wang F, Argyris E, Chen K, Liang Z, Tian H, Huang W, Squires K,
Verlinghieri G, Zhang H. Cellular microRNAs contribute to HIV‑1 latency in
resting primary CD4+ T lymphocytes. Nat Med. 2007;13(10):1241–7.
51. Pilakka‑Kanthikeel S, Nair MP. Interaction of drugs of abuse and microRNA
with HIV: a brief review. Front Microbiol. 2015;6:967.
52. Swaminathan G, Navas‑Martin S, Martin‑Garcia J. MicroRNAs and HIV‑1
infection: antiviral activities and beyond. J Mol Biol. 2014;426(6):1178–97.
53. Whisnant AW, Bogerd HP, Flores O, Ho P, Powers JG, Sharova N, Stevenson
M, Chen CH, Cullen BR. In‑depth analysis of the interaction of HIV‑1
with cellular microRNA biogenesis and effector mechanisms. mBio.
2013;4(2):e000193.
54. Sandau US, Duggan E, Shi X, Smith SJ, Huckans M, Schutzer WE, Loftis JM,
Janowsky A, Nolan JP, Saugstad JA. Methamphetamine use alters human
plasma extracellular vesicles and their microRNA cargo: an exploratory
study. J Extracell Vesicles. 2020;10(1):e12028–e12028.
55. Zhu L, Zhu J, Liu Y, Chen Y, Li Y, Chen S, Li T, Dang Y, Chen T. Chronic
methamphetamine regulates the expression of MicroRNAs and puta‑
tive target genes in the nucleus accumbens of mice. J Neurosci Res.
2015;93(10):1600–10.
56. Nathans R, Chu CY, Serquina AK, Lu CC, Cao H, Rana TM. Cellular
microRNA and P bodies modulate host‑HIV‑1 interactions. Mol Cell.
2009;34(6):696–709.
57. Liu MQ, Zhao M, Kong WH, Peng JS, Wang F, Qiu HY, Zhu ZR, Tang L,
Sang M, Wu JG, et al. Antiretroviral therapy fails to restore levels of
HIV‑1 Restriction miRNAs in PBMCs of HIV‑1‑infected MSM. Medicine.
2015;94(46):e2116.
58. Yin C, Zhang T, Qu X, Zhang Y, Putatunda R, Xiao X, Li F, Xiao W, Zhao H,
Dai S, et al. In vivo excision of HIV‑1 provirus by saCas9 and multiplex
single‑guide RNAs in animal models. Mol Ther. 2017;25(5):1168–86.
59. Kalasinsky KS, Bosy TZ, Schmunk GA, Reiber G, Anthony RM, Furukawa
Y, Guttman M, Kish SJ. Regional distribution of methamphetamine in
autopsied brain of chronic human methamphetamine users. Forensic Sci
Int. 2001;116(2–3):163–9.
60. Klette KL, Kettle AR, Jamerson MH. Prevalence of use study for
amphetamine (AMP), methamphetamine (MAMP), 3,4‑methylenedioxy‑
amphetamine (MDA), 3,4‑methylenedioxy‑methamphetamine (MDMA),
and 3,4‑methylenedioxy‑ethylamphetamine (MDEA) in military entrance
processing stations (MEPS) specimens. J Anal Toxicol. 2006;30(5):319–22.
61. Schepers RJ, Oyler JM, Joseph RE Jr, Cone EJ, Moolchan ET, Huestis MA.
Methamphetamine and amphetamine pharmacokinetics in oral fluid and
plasma after controlled oral methamphetamine administration to human
volunteers. Clin Chem. 2003;49(1):121–32.
62. Takayasu T, Ohshima T, Nishigami J, Kondo T, Nagano T. Screening and
determination of methamphetamine and amphetamine in the blood,
urine and stomach contents in emergency medical care and autopsy
cases. J Clin Forensic Med. 1995;2(1):25–33.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in pub‑
lished maps and institutional affiliations.
... Similarly, the use of methamphetamine via IDU or "booty bumping" can result in HIV infection through direct inoculation of the bloodstream from a shared needle or damage the rectal mucosa, which increases biological susceptibility to HIV infection if exposed during condomless receptive anal sex (Cantrell et al., 2006). Methamphetamine also directly alters cellular processes, resulting in decreased intracellular immunity to HIV and greater HIV replication in human monocyte cells (Liu et al., 2021). ...
Article
Full-text available
Introduction: Substance use, including methamphetamine use, is a contributing factor in HIV acquisition and treatment. Stimulant use is linked to mental health yet there is limited data from youth in community-based settings. Design: One hundred marginally housed or homeless transitional age youth (TAY) were recruited at Larkin Street Youth Services and completed a survey on mental health and substance use. Methods: We conducted secondary data analysis using multivariable logistic regression models to identify the correlates of methamphetamine use among TAY. Results: The participants' mean age was 22. Of those who reported methamphetamine use in the past 3 months, 64% were Gay, Bisexual, or Pansexual. Factors independently associated with methamphetamine use were; living with HIV (adjusted odds ratio [aOR] = 3.18, 95% CI = 1.11-9.15), depressive symptoms (aOR = 6.02, 95% CI = 1.46-24.78), symptoms of PTSD (aOR = 13.38, 95% CI = 1.59-112.73), polysubstance use in the past 3 months (aOR = 50.02, 95% CI = 9.72-257.46) and a history of injection drug use (aOR = 8.38, 95% CI = 1.87-37.53). Conclusions: Results from this study suggest a need to develop, adapt, and rapidly implement comprehensive interventions that address the combined epidemics of substance use, HIV, and mental health among TAY. Clinical relevance: This article examines factors associated with methamphetamine use among transitional age youth (TAY) experiencing homelessness or housing instability. Several factors were associated with use, including depression, PTSD, HIV status, polysubstance use, and injection drug use. These findings highlight the need for nurses to assess for methamphetamine use among youth as well as associated mental health and physical health problems. Nurses should link TAY who are using methamphetamine to evidence-based treatment programs to address substance use and comorbid conditions.
... that reported increased HIV infection in other cell types including macrophages (46,47). ...
Article
Full-text available
HIV-associated neurocognitive impairment (HIV-NCI) persists in 15-40% of people with HIV (PWH) despite effective antiretroviral therapy. HIV-NCI significantly impacts quality of life, and there is currently no effective treatment for it. The development of HIV-NCI is complex and is mediated, in part, by the entry of HIV-infected mature monocytes into the central nervous system (CNS). Once in the CNS, these cells release inflammatory mediators that lead to neuroinflammation, and subsequent neuronal damage. Infected monocytes may infect other CNS cells as well as differentiate into macrophages, thus contributing to viral reservoirs and chronic neuroinflammation. Substance use disorders in PWH, including the use of methamphetamine (meth), can exacerbate HIV neuropathogenesis. We characterized the effects of meth on the transcriptional profile of HIV-infected mature monocytes using RNA-sequencing. We found that meth mediated an upregulation of gene transcripts related to viral infection, cell adhesion, cytoskeletal arrangement, and extracellular matrix remodeling. We also identified downregulation of several gene transcripts involved in pathogen recognition, antigen presentation, and oxidative phosphorylation pathways. These transcriptomic changes suggest that meth increases the infiltration of mature monocytes that have a migratory phenotype into the CNS, contributing to dysregulated inflammatory responses and viral reservoir establishment and persistence, both of which contribute to neuronal damage. Overall, our results highlight potential molecules that may be targeted for therapy to limit the effects of meth on HIV neuropathogenesis.
... A majority of studies on METHinduced immune responses in monocytes/macrophages focused on the process of HIV infection. Results suggested that METH promoted HIV replication in monocytes/macrophages [19,20]. Furthermore, as a weak base, METH led to acidic organelle injury, and disrupted the pH gradient, leading to immune cell dysfunction [21,22]. ...
Article
Full-text available
Background Methamphetamine (METH) abuse causes serious health problems, including injury to the immune system, leading to increased incidence of infections and even making withdrawal more difficult. Of course, immune cells, an important part of the immune system, are also injured in methamphetamine abuse. However, due to different research models and the lack of bioinformatics, the mechanism of METH injury to immune cells has not been clarified. Methods We examined the response of three common immune cell lines, namely Jurkat, NK-92 and THP-1 cell lines, to methamphetamine by cell viability and apoptosis assay in vitro, and examined their response patterns at the mRNA level by RNA-sequencing. Differential expression analysis of two conditions (control and METH treatment) in three types of immune cells was performed using the DESeq2 R package (1.20.0). And some of the differentially expressed genes were verified by qPCR. We performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis of differentially expressed genes by the clusterProfiler R package (3.14.3). And gene enrichment analysis was also performed using MetaScape (www.metascape.org). Results The viability of the three immune cells was differentially affected by methamphetamine, and the rate of NK-cell apoptosis was significantly increased. At the mRNA level, we found disorders of cholesterol metabolism in Jurkat cells, activation of ERK1 and ERK2 cascade in NK-92 cells, and disruption of calcium transport channels in THP-1 cells. In addition, all three cells showed changes in the phospholipid metabolic process. Conclusions The results suggest that both innate and adaptive immune cells are affected by METH abuse, and there may be commonalities between different immune cells at the transcriptome level. These results provide new insights into the potential effects by which METH injures the immune cells.
... Gene expression analyses of human macrophages treated with meth could determine the transcriptional pathways that meth alters to mediate differential protein expression. In vivo studies of gene expression in SIV-infected macaque brains exposed to meth found an increased proportion of infected macrophages, which is consistent with our mass spectrometry data and prior in vitro studies [92,93]. These analyses demonstrated the differential expression of genes in the complement pathway and electron transport chain function [93]. ...
Article
Full-text available
HIV-neurocognitive impairment (HIV-NCI) can be a debilitating condition for people with HIV (PWH), despite the success of antiretroviral therapy (ART). Substance use disorder is often a comorbidity with HIV infection. The use of methamphetamine (meth) increases systemic inflammation and CNS damage in PWH. Meth may also increase neuropathogenesis through the functional dysregulation of cells that harbor HIV. Perivascular macrophages are long-lived reservoirs for HIV in the CNS. The impaired clearance of extracellular debris and increased release of reactive oxygen species (ROS) by HIV-infected macrophages cause neurotoxicity. Macroautophagy is a vital intracellular pathway that can regulate, in part, these deleterious processes. We found in HIV-infected primary human macrophages that meth inhibits phagocytosis of aggregated amyloid-β, increases total ROS, and dysregulates autophagic processes. Treatment with widely prescribed ART drugs had minimal effects, although there may be an improvement in phagocytosis when co-administered with meth. Pharmacologically inhibited lysosomal degradation, but not induction of autophagy, further increased ROS in response to meth. Using mass spectrometry, we identified the differentially expressed proteins in meth-treated, HIV-infected macrophages that participate in phagocytosis, mitochondrial function, redox metabolism, and autophagy. Significantly altered proteins may be novel targets for interventional strategies that restore functional homeostasis in HIV-infected macrophages to improve neurocognition in people with HIV-NCI using meth.
Article
Full-text available
Methamphetamine use has become a rampant public health issue that not only causes devastating consequences to the user but also poses a burden to surrounding communities. A spectrum of ophthalmic sequelae is associated with methamphetamine use and includes episcleritis, scleritis, corneal ulceration, panophthalmitis, endophthalmitis, retinal vasculitis, and retinopathy. In many instances, prompt recognition of the condition and associated infectious process and early initiation of antimicrobial therapy are crucial steps to preventing vision loss. In this review, we summarize the reported ocular complications that may result from methamphetamine use in addition to several postulated mechanisms regarding the ocular toxicity of methamphetamine. The increasing prevalence of methamphetamine use as a public health threat highlights the need for continued investigation of this ophthalmologic issue.
Article
METH and HIV Tat treatment results in increased oxidative stress which affects cellular metabolism and causes DNA damage in the treated microglia. Both, METH ± HIV Tat impair mitochondrial respiration, leading to dysfunction in bioenergetics and increased ROS in microglial cells. Our data indicate that mitochondrial dysfunction may be key to the METH and/or HIV Tat-induced neuropathology. METH and/or HIV Tat induced changes in the protein, lipid and nucleotide concentration in microglial cells were measured by Raman Spectroscopy, and we speculate that these fundamental molecular-cellular changes in microglial cells contribute to the neuropathology that is associated with METH abuse in HIV patients.
Article
Full-text available
Methamphetamine (MA) is the largest drug threat across the globe, with health effects including neurotoxicity and cardiovascular disease. Recent studies have begun to link microRNAs (miRNAs) to the processes related to MA use and addiction. Our studies are the first to analyse plasma EVs and their miRNA cargo in humans actively using MA (MA‐ACT) and control participants (CTL). In this cohort we also assessed the effects of tobacco use on plasma EVs. We used vesicle flow cytometry to show that the MA‐ACT group had an increased abundance of EV tetraspanin markers (CD9, CD63, CD81), but not pro‐coagulant, platelet‐, and red blood cell‐derived EVs. We also found that of the 169 plasma EV miRNAs, eight were of interest in MA‐ACT based on multiple statistical criteria. In smokers, we identified 15 miRNAs of interest, two that overlapped with the eight MA‐ACT miRNAs. Three of the MA‐ACT miRNAs significantly correlated with clinical features of MA use and target prediction with these miRNAs identified pathways implicated in MA use, including cardiovascular disease and neuroinflammation. Together our findings indicate that MA use regulates EVs and their miRNA cargo, and support that further studies are warranted to investigate their mechanistic role in addiction, recovery, and recidivism.
Article
Full-text available
Methamphetamine (Meth) abuse is a worldwide public health problem and contributes to HIV-1 pathobiology and poor adherence to anti-retroviral therapies. Specifically, Meth is posited to alter molecular mechanisms to provide a more conducive environment for HIV-1 replication and spread. Enhanced expression of inflammatory cytokines, such as Interleukin-1β (IL-1β), has been shown to be important for HIV-1 pathobiology. In addition, microRNAs (miRNAs) play integral roles in fine-tuning the innate immune response. Notably, the effects of Meth abuse on miRNA expression are largely unknown. We studied the effects of Meth on IL-1β and miR-146a, a well-characterized member of the innate immune signaling network. We found that Meth induces miR-146a and triggers an IL-1β auto-regulatory loop to modulate innate immune signaling in CD4⁺ T-cells. We also found that Meth enhances HIV-1 replication via IL-1 signaling. Our results indicate that Meth activates an IL-1β feedback loop to alter innate immune pathways and favor HIV-1 replication. These observations offer a framework for designing targeted therapies in HIV-infected, Meth using hosts.
Article
Full-text available
Methamphetamine (Meth) exacerbates HIV-1 pathobiology by increasing virus transmission and replication and accelerating clinical progression to AIDS. Meth has been shown to alter the expression of HIV-1 co-receptors and impair intrinsic resistance mechanisms of immune cells. However, the exact molecular mechanisms involved in augmenting HIV-1 replication in T-cells are still not yet clear. Here, we demonstrate that pretreatment with Meth of CD4+ T-cells enhanced HIV-1 replication. We observed upregulation of CD4+ T-cell activation markers and enhanced expression of miR-34c-5p and miR-155 in these cells. Further, we noted activation of the sigma-1 receptor and enhanced intracellular Ca2+ concentration and cAMP release in CD4+ T-cells upon Meth treatment, which resulted in increased phosphorylation and nuclear translocation of transcription factors NFκB, CREB, and NFAT1. Increased gene expression of IL-4 and IL-10 was also observed in Meth treated CD4+ T-cells. Moreover, proteasomal degradation of Ago1 occurred upon Meth treatment, further substantiating the drug as an activator of T-cells. Taken together, these findings show a previously unreported mechanism whereby Meth functions as a novel T-cell activator via the sigma-1 signaling pathway, enhancing replication of HIV-1 with expression of miR-34c-5p, and transcriptional activation of NFκB, CREB and NFAT1.
Article
Full-text available
Methamphetamine use poses increased risk for HIV and other sexually transmitted infections. There is robust evidence that methamphetamine use increases sexual risk behavior, like condomless sex, primarily among men who have sex with men but also among heterosexual women and men. Gender differences have been found among women and men who use meth, and there is a high degree of interconnectedness between meth use, depression, and condomless sex. The aims of the current study are to evaluate the efficacy of a theory-based, tri-focal intervention designed to reduce depression, meth use, and condomless sex among women and men, and to examine gender as a moderator of efficacy. A total of 432 HIV-negative women and men who use meth participated in a two-arm randomized controlled trial and completed baseline and follow-up assessments at 4, 8, and 12 months. We used latent growth curve modeling techniques to analyze the data. Results showed that while all participants exhibited reductions in depression, meth use, and condomless sex, the intervention and comparison groups did not differ in changes over time. However, we did find a significant gender moderation effect, such that among men, those in the intervention arm reported greater reductions in meth use relative to those in the comparison group; reduced meth use was associated with reduced condomless sex, but not depression. In contrast, women in the intervention condition did not differ from women in the comparison condition in changes in any of the three outcome variables. Interventions targeting heterosexual women and men who use meth must be gender-specific, and take into account the unique vulnerabilities and experiences of women, including the perceived positive aspects of using meth, gendered power dynamics, higher depression, and violence.
Article
Full-text available
HIV-1 infection and methamphetamine (METH) abuse frequently occur simultaneously and may have synergistic pathological effects. Although HIV-positive/active METH users have been shown to have higher HIV viral loads and experience more severe neurological complications than non-users, the direct impact of METH on HIV infection and its link to the development of neurocognitive alternations are still poorly understood. In the present study, we hypothesized that METH impacts HIV infection of neural progenitor cells (NPC) by a mechanism encompassing NFκB/SP1-mediated HIV-LTR activation. Mouse and human NPC were infected with EcoHIV (modified HIV virus infectious to mice) and HIV, respectively, in the presence or absence of METH (50 or 100 µM). Pretreatment with METH, but not simultaneous exposure, significantly increased HIV production in both mouse and human NPC. To determine the mechanisms underlying these effects, cells were transfected with different variants of HIV-LTR promoters and then exposed to METH. METH treatment induced transcriptional activity of HIVLTR promotor, an effect that required both NFκB and SP1 signaling. Pretreatment with METH also decreased neuronal differentiation of HIVinfected NPC in both in vitro and in vivo settings. Importantly, NPC-derived daughter cells appeared to be latently infected with HIV. This study indicates that METH increases HIV infectivity of NPC, through the NFκB/SP1- dependent activation of HIV LTR, and with the subsequent alterations of NPC neurogenesis. Such events may underlie METH- exacerbated neurocognitive dysfunction in HIV-infected patients.
Article
Full-text available
Viruses manipulate the complex interferon and interferon-stimulated gene (ISG) system in different ways. We have previously shown that HIV inhibits type I and III interferons in its key target cells but directly stimulates a subset of >20 ISGs in macrophages and dendritic cells, many of which are antiviral. Here, we examine the mechanism of induction of ISGs and show this occurs in two phases. The first phase was transient (0 to 24 h postinfection [hpi]), induced mainly by extracellular vesicles and one of its component proteins, HSP90α, contained within the HIV inoculum. The second, dominant, and persistent phase (>48 hpi) was induced via newly transcribed HIV RNA and sensed via RIGI, as shown by the reduction in ISG expression after the knockdown of the RIGI adaptor, MAVS, by small interfering RNA (siRNA) and the inhibition of both the initiation and elongation of HIV transcription by short hairpin RNA (shRNA) transcriptional silencing. We further define the induction pathway, showing sequential HIV RNA stimulation via Tat, RIGI, MAVS, IRF1, and IRF7, also identified by siRNA knockdown. IRF1 also plays a key role in the first phase. We also show that the ISGs IFIT1 to -3 inhibit HIV production, measured as extracellular infectious virus. All induced antiviral ISGs probably lead to restriction of HIV replication in macrophages, contributing to a persistent, noncytopathic infection, while the inhibition of interferon facilitates spread to adjacent cells. Both may influence the size of macrophage HIV reservoirs in vivo. Elucidating the mechanisms of ISG induction may help in devising immunotherapeutic strategies to limit the size of these reservoirs.
Article
Full-text available
Interferon-alpha (IFN-α) is a potent anti-viral cytokine, critical to the host immune response against viruses. IFN-α is first produced upon viral detection by pathogen recognition receptors. Following its expression, IFN-α embarks upon a complex downstream signalling cascade called the JAK/STAT pathway. This signalling pathway results in the expression of hundreds of effector genes known as interferon stimulated genes (ISGs). These genes are the basis for an elaborate effector mechanism and ultimately, the clearance of viral infection. ISGs mark an elegant mechanism of anti-viral host defence that warrants renewed research focus in our global efforts to treat existing and emerging viruses. By understanding the mechanistic role of individual ISGs we anticipate the discovery of a new “treasure trove” of anti-viral mediators that may pave the way for more effective, targeted and less toxic anti-viral therapies. Therefore, with the aim of highlighting the value of the innate type 1 IFN response in our battle against viral infection, this review outlines both historic and recent advances in understanding the IFN-α JAK/STAT pathway, with a focus on new research discoveries relating to specific ISGs and their potential role in curing existing and future emergent viral infections.
Article
Full-text available
CRISPR-associated protein 9 (Cas9)-mediated genome editing provides a promising cure for HIV-1/AIDS; however, gene delivery efficiency in?vivo remains an obstacle to overcome. Here, we demonstrate the feasibility and efficiency of excising the HIV-1 provirus in three different animal models using an all-in-one adeno-associated virus (AAV) vector to deliver multiplex single-guide RNAs (sgRNAs) plus Staphylococcus aureus Cas9 (saCas9). The quadruplex sgRNAs/saCas9 vector outperformed the duplex vector in excising the integrated HIV-1 genome in cultured neural stem/progenitor cells from HIV-1 Tg26 transgenic mice. Intravenously injected quadruplex sgRNAs/saCas9 AAV-DJ/8 excised HIV-1 proviral DNA and significantly reduced viral RNA expression in several organs/tissues of Tg26 mice. In EcoHIV acutely infected mice, intravenously injected quadruplex sgRNAs/saCas9 AAV-DJ/8 reduced systemic EcoHIV infection, as determined by live bioluminescence imaging. Additionally, this quadruplex vector induced efficient proviral excision, as determined by PCR genotyping in the liver, lungs, brain, and spleen. Finally, in humanized bone marrow/liver/thymus (BLT) mice with chronic HIV-1 infection, successful proviral excision was detected by PCR genotyping in the spleen, lungs, heart, colon, and brain after a single intravenous injection of quadruplex sgRNAs/saCas9 AAV-DJ/8. In conclusion, in?vivo excision of HIV-1 proviral DNA by sgRNAs/saCas9 in solid tissues/organs can be achieved via AAV delivery, a significant step toward human clinical trials.
Article
Background and aims: Men who have sex with men (MSM) and inject drugs are at risk for HIV infection. Although research exists on non-injection methamphetamine (meth) use and sexual risk among MSM, less is known about meth injection and its association with HIV infection among MSM who inject drugs. Methods: We analyzed data from men aged ≥18 years who reported injecting drugs and male-to-male sexual contact. Men were recruited using respondent-driven sampling, interviewed, and tested for HIV during the 2012 and 2015 cycles of National HIV Behavioral Surveillance among persons who inject drugs. We included data from 8 cities where ≥10 MSM reported meth as the primary drug injected. We assessed differences in demographic characteristics, past 12 months risk behaviors, and HIV infection between MSM who primarily injected meth and those who primarily injected another drug. Results: Among 961 MSM, 33.7% reported meth as the drug they injected most often. Compared to MSM who primarily injected other drugs, MSM who primarily injected meth were more likely to have had ≥5 condomless anal sex partners, have been diagnosed with syphilis, and were less likely to report sharing syringes. In multivariable analysis, injecting meth was associated with being HIV-positive (adjusted prevalence ratio 1.48; 95% confidence interval 1.08-2.03). Including number of condomless anal sex partners in mediation analysis rendered this association no longer significant. Conclusions: HIV prevalence among MSM who primarily injected meth was almost 50% higher than among MSM who primarily injected other drugs, and this association was mediated by sexual risk.