Вы находитесь на странице: 1из 18

Articles

DOI: 10.1038/s41564-017-0006-5

Gut-homing Δ42PD1+Vδ2 T cells promote


innate mucosal damage via TLR4 during acute
HIV type 1 infection
Allen Ka Loon Cheung1, Hau-yee Kwok1, Yiru Huang1, Min Chen1,2, Yufei Mo1, Xilin Wu1, Ka-shing Lam1,
Hoi-Kuan Kong3, Terrence Chi Kong Lau   3, Jingying Zhou1, Jingjing Li1, Lin Cheng1,4, Boon Kiat Lee1,
Qiaoli Peng4, Xiaofan Lu5, Minghui An6, Hui Wang4, Hong Shang6, Boping Zhou4, Hao Wu5, Aimin Xu7,
Kwok-Yung Yuen1 and Zhiwei Chen   1,4*

The innate immune cells underlying mucosal inflammatory responses and damage during acute HIV-1 infection remain incom-
pletely understood. Here, we report a Vδ​2 subset of gut-homing γ​δ​T cells with significantly upregulated Δ​42PD1 (a PD1 iso-
form) in acute (~20%) HIV-1 patients compared to chronic HIV-1 patients (~11%) and healthy controls (~2%). The frequency of
Δ​42PD1+Vδ​2 cells correlates positively with plasma levels of pro-inflammatory cytokines and fatty-acid-binding protein before
detectable lipopolysaccharide in acute patients. The expression of Δ​42PD1 can be induced by in vitro HIV-1 infection and is
accompanied by high co-expression of gut-homing receptors CCR9/CD103. To investigate the role of Δ​42PD1+Vδ​2 cells in vivo,
they were adoptively transferred into autologous humanized mice, resulting in small intestinal inflammatory damage, probably
due to the interaction of Δ​42PD1 with its cognate receptor Toll-like receptor 4 (TLR4). In addition, blockade of Δ​42PD1 or TLR4
successfully reduced the cytokine effect induced by Δ​42PD1+Vδ​2 cells in vitro, as well as the mucosal pathological effect in
humanized mice. Our findings have therefore uncovered a Δ​42PD1–TLR4 pathway exhibited by virus-induced gut-homing Vδ​2
cells that may contribute to innate immune activation and intestinal pathogenesis during acute HIV-1 infection. Δ​42PD1+Vδ​2
cells may serve as a target for the investigation of diseases with mucosal inflammation.

H
uman Vγ​9Vδ​2 T cells (Vδ​2) are unconventional gamma-delta ‘HIV enteropathy’ characterized by diarrhoea, increased gastroin-
T lymphocytes (γ​δ​-T) that are associated with gut infiltration testinal inflammation, enhanced intestinal permeability and mal-
and inflammation. γ​δ​T cells express T cell receptor rearranged absorption5. By histological analysis, the intestinal villi are often
with the T cell receptor gamma (TCRG) and delta (TCRD) genes and seen with atrophy and blunting, epithelial damage and infiltrating
comprise 3–10% of human peripheral blood lymphocytes, with the Vδ​ lymphocytes in both patients and acute simian immunodeficiency
2 subset dominant over Vδ​1 in healthy individuals. Vδ​1 cells reside in virus (SIV)-infected macaques, and this can occur in the absence
the gut epithelium and maintain homeostasis and surveillance, while of microbial translocation6,7. Inflammatory mediators such as infil-
Vδ​2 cells have a critical role in important innate and adaptive immune tration and activation of CD8+ T cells, pro-inflammatory cytokines
responses against pathogens and in sterile inflammation, includ- and CD4 T cell depletion are believed to contribute to these drastic
ing cytolysis of infected cells and rapid production of cytokines1. In changes in the small intestine following local immune activation8–11.
vivo activation of Vδ​2 cells in macaques led to accumulation in the While the Vδ​1 subset of γ​δ​T cells undergoes rapid expansion in
gut and the onset of antimicrobial responses. This is also reported in mucosal tissues due to immune activation12, the circulating Vδ​2
Crohn’s disease and inflammatory bowel disease, where Vδ​2 cells were cells also increases within days to a few weeks after infection but
found to be gut-homing and might be responsible for the pathology begin to diminish by 6 weeks post-infection in an SIV model or
observed in small intestinal and colonic tissues2,3. Apart from the rec- in human intestines13,14. Interestingly, during HIV-1 infection, cir-
ognition of phosphoantigen that leads to their activation, Vδ​2 cells can culating Vδ​2 cells appear to have potential gut-homing properties,
crosstalk with B cells or dendritic cells (DCs), migrate to mucosal sites, as demonstrated by high CCR5 expression and upregulated CCR9
and serve as antigen presenting cells (APCs) to α​β​-T cells1,2,4. and CD10314,15. The depletion of Vδ​2 cells in the gut, however, may
During acute human immunodeficiency virus (HIV)-1 infec- be mediated through gp120, which becomes abundant in the intes-
tion, it is well documented that patients’ small intestines undergo tines due to replicating virus and infected CD4 T cell death in acute

1
​AIDS Institute, Research Center for Infection and Immunity, Department of Microbiology, Li Ka Shing Faculty of Medicine, University of Hong Kong,
999097 Hong Kong SAR, China. 2​Yunnan Center for Disease Control and Prevention, 650000 Kunming, Yunnan Province, China. 3​Department of
Biomedical Sciences, City University of Hong Kong, 999097 Hong Kong SAR, China. 4​HKU-AIDS Institute Shenzhen Research Laboratory and AIDS Clinical
Research Laboratory, Guangdong Key Laboratory of Emerging Infectious Diseases, Shenzhen Key Laboratory of Infection and Immunity, Shenzhen Third
People’s Hospital, 518000 Shenzhen, China. 5​STD/HIV Research Laboratory and Department of Infectious Diseases, Beijing Key Laboratory of HIV/AIDS
Research, Beijing You-An Hospital, Capital Medical University, 100000 Beijing, China. 6​Key Laboratory of AIDS Immunology of National Health and Family
Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, China Medical University, 110000
Shenyang, China. 7​Department of Pharmacology & Pharmacy and Department of Medicine, Research Centre of Heart, Brain, Hormone and Healthy Aging,
Li Ka Shing Faculty of Medicine, University of Hong Kong, 999097 Hong Kong SAR, China. *e-mail: zchenai@hku.hk

Nature Microbiology | VOL 2 | OCTOBER 2017 | 1389–1402 | www.nature.com/naturemicrobiology 1389


© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Articles NaTuRe MICRObIOLOgy

HIV-1 infection, and subsequently acts on CCR5 expressed by these However, a higher frequency of CD3+Vδ​1 cells than CD3+Vδ​2 was
cells14,16. As a result, an inversion of the Vδ​2:Vδ​1 subsets ratio occurs observed in acute patients, which contrasts with the predominance
shortly after HIV-1 infection in peripheral blood, and is not readily of Vδ​2 over Vδ​1 in healthy individuals (Fig. 1d). This is consistent
restored with highly active antiretroviral treatment (HAART)12,14,17. with a disproportionate change in Vδ​2 and Vδ​1 cells in acute and
The functions of Vδ​2 cells are dysregulated during this time, when chronic HIV-1 patients, as reported previously12,14,17. This difference
they have an increased ability to produce IL-17 but have impaired was more dramatic in chronic patients, where an expansion of Vδ​1
cytotoxicity and IFN-γ​ response18–21. In particular, the ability of Vδ​2 cells was observed (Fig. 1d). Increased CD8+ T cell activation was
cells to crosstalk with DCs is significantly impeded as a result of evident in acute and chronic HIV-1 patients on measuring CD38 and
HIV-1 infection and aberrant cytokines being produced22. However, HLA-DR expression (Fig. 1e). In addition, a comparison between
apart from phenotypic or functional changes, little is known of the two stages of acute HIV-1 infection based on Fiebig status showed
mechanism by which Vδ​2 cells perform their immune stimulatory significantly higher Δ​42PD1+Vδ​ 2 cells in seronegative patients
role once they migrate to the gut during early HIV-1 infection2,23. (Fiebig I–II) than in seropositive (Fiebig III–V) patients, indicating
Interactions with other cell types, such as DCs, follicular T helper that the rapid onset of Δ​42PD1 expression probably occurred early
and conventional T cells that may contribute to HIV pathogenesis, (<​20 days) after HIV-1 infection (Fig. 1f). We also performed cor-
are proposed2,4, but defining the interacting receptor/ligand respon- relation analyses between the frequency of Δ​42PD1+Vδ​2 cells from
sible for mediating pathogenic effects is critical in understanding acute and chronic patients with viral load, CD4:CD8 ratio, CD4
the role of Vδ​2 cells during acute HIV-1 infection. count, CD8 count, frequency of CD38+HLA-DR+ CD4+ and CD8+
We recently identified an alternatively spliced isoform of human T cells, but only the frequency of Δ​42PD1+Vδ​2 cells and viral load
programmed death 1 (PD-1) containing a 42-nucleotide deletion— in acute patients had a statistically significant positive correlation
Δ​42PD1— which is capable of inducing pro-inflammatory cyto- (Supplementary Fig. 3).
kines from human peripheral blood mononuclear cells (PBMCs) Due to the increased CD8+ T cell activation observed in acute
when expressed as a recombinant soluble protein24. Using in-house patients, we then analysed their plasma cytokine levels, because cyto-
anti-Δ​42PD1 monoclonal antibodies25, we demonstrate here that Δ​ kine storm is one of the landmark characteristics of early HIV-1 infec-
42PD1 is significantly expressed on up to 20% of peripheral Vδ​2 tion26. Surprisingly, we found that the frequency of Δ​42PD1+Vδ​2
cells in acute HIV-1 patients, which correlates with the level of cells was positively correlated with plasma levels of TNF-α​, IL-6,
plasma pro-inflammatory cytokines (tumour-necrosis factor IL-1β​and IFN-α​ (Fig. 1g), suggesting a potential role of Δ​42PD1 in
(TNF)-α​, interleukin (IL)-6, IL-1β​, interferon (IFN)-α​). Further innate immune activation. The endotoxin lipopolysaccharide (LPS)
analysis showed that one-third of Δ​42PD1+Vδ​2 cells co-express level in acute patient plasma (0.84 ±​ 1.15 endotoxin unit (EU) ml–1),
the gut-homing receptors CCR9/CD103 in patient samples. Thus, however, was similar to healthy controls (0.79 ±​ 0.69 EU ml–1), consis-
we hypothesized that Δ​42PD1 plays a role in early immune activa- tent with the lack of microbial translocation during the acute phase
tion during acute HIV-1 infection in the gut. To test this, we used of infection27. We therefore concluded that Δ​42PD1+Vδ​2 cells might
a humanized mouse model where HIV-1-induced Δ​42PD1+Vδ​2 play a role in immune activation soon after HIV-1 primary infection,
cells transmigrate to the intestines, promoting inflammation dam- probably before seroconversion and microbial translocation.
age and increased IL-6 expression. Importantly, we identified that
the innate Toll-like receptor (TLR)4/MD2 complex is probably the Δ42PD1 expression is regulated by cytokines induced by HIV-1
cognate receptor for Δ​42PD1 by which pro-inflammatory cytokines infection. To determine if HIV-1 infection can stimulate the expres-
are induced in monocyte-derived DCs. This study thus discovers sion of Δ​42PD1 on Vδ​2 cells, we performed in vitro viral infec-
a previously unrecognized mechanism exhibited by gut-homing tion of healthy human PBMCs. Fresh PBMCs were infected with
Δ​42PD1+Vδ​2 cells that may contribute to innate immune activation X4-tropic HIV-1NL4-3, R5-tropic HIV-1Henan or mock. Detection of
and mucosal damage during acute HIV-1 infection. proviral DNA by qPCR, long terminal repeat (LTR) transcripts by
quantitative reverse-transcription real-time PCR (qRT–PCR) and
Results intracellular p24 signals in CD4+ T cells by flow cytometry indi-
Increased Δ42PD1 expression on γδ T cells in acute and chronic cated successful infection (Supplementary Fig.  4). By 3 days after
HIV-1-infected patients. To study the biological role of Δ​42PD1, HIV-1 infection of PBMCs, ~10% of Vδ​2 cells expressed Δ​42PD1,
we recently generated two anti-Δ​42PD1 monoclonal antibodies for of which ~50% of cells were CD69+, indicating activation (Fig. 2a).
immune assays25. Lymphocytes freshly isolated from healthy indi- Other viruses, including human cytomegalovirus (CMV), HIV
viduals were tested by multicolour flow cytometry and gating on pseudoviruses with vesicular stomatitis virus glycoprotein (VSV-
specific cell markers, CD4+ T, CD8+ T, B, NK, NKT and γ​δ​T cells G) or ADA envelope, could also induce Δ​42PD1 expression on Vδ​
(Supplementary Fig. 1a). Δ​42PD1 was found to be expressed mainly 2 cells, suggesting a generalized response (Supplementary Fig. 5a).
on ~6% of γ​δ​T cells among these cell types (Supplementary Fig. 1b). In a time-course experiment, induction of Δ​42PD1 expression on
We also found that Δ​42PD1 is exclusively expressed on the Vδ​2 but Vδ​2 cells reached the highest level by day 3 post-infection (p.i.) as
not the Vδ​1 subset of γ​δ​T cells (Supplementary Fig. 1c). compared to mock, and remained so at day 6 p.i. for both HIV-1NL4-3
To determine the role of Δ​42PD1 in HIV/AIDS, we examined and HIV-1Henan (Fig. 2b). Moreover, cell numbers of CD3+Vδ​2+ and
two cohorts of HIV-1 acutely infected patients (n =​ 57) and one Δ​42PD1+ CD3+Vδ​2+ cells were increased by an average of two- and
cohort of chronic HIV-1 patients (n =​ 50) for the expression of ninefold, respectively, after HIV-1 infection compared to mock
Δ​42PD1 on Vδ​2 cells from frozen PBMC samples and compared (Supplementary Fig. 5b,c). However, direct application of HIV-1 to
with healthy controls (n =​ 22) (Supplementary Table  1). By flow purified γ​δ​T cells resulted in neither induction of Δ​42PD1 nor suc-
cytometry, the frequency of CD3+Vδ​2 cells in the PBMCs of the cessful infection, due mainly to undetectable intracellular p24. In
acute patients was found to be 2.62 ±​  1.99% (mean  ±​  s.d.), which contrast, HIV-1 induction resulted in the upregulation of CD4 on
is significantly higher than in chronic patients (1.61 ±​  1.73%, Δ​42PD1+CD3+Vδ​2+ cells (Supplementary Fig.  6a,b) but not CD8
P <​  0.001) but comparable to healthy controls (2.65  ±​  1.49%, (Supplementary Fig. 6c,d). Therefore, it is possible that certain sol-
P =​  0.5225) (Fig.  1a,b). When we further examined the expres- uble factors resulting from infected PBMCs probably upregulated
sion of Δ​42PD1, the proportions of Δ​42PD1+Vδ​2 cells were sig- Δ​42PD1 expression on Vδ​2 cells.
nificantly higher in acute HIV-1 patients (18.53 ±​  10.26%) than Previous studies have shown that γ​δ​T cells can be activated by
in chronic patients (11.72 ±​  9.95%, P <​ 0.001) and healthy con- cytokines such as IL-12 and IL-1528. Our multiplex cytokine analysis
trols (2.18 ±​  1.41%, P <​  0.001) (Fig. 1c and Supplementary Fig. 2). also revealed IL-12 release in supernatants after HIV-1NL4-3 infection

1390 Nature Microbiology | VOL 2 | OCTOBER 2017 | 1389–1402 | www.nature.com/naturemicrobiology

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
NaTuRe MICRObIOLOgy Articles
a b c
15 P = 0.5225 80 ***

∆42PD1+ CD3+ Vδ2+ (%)


1.05
*** *** *** ***

CD3+ Vδ2+ (%)


60
10

Vδ2–PE
SSC

40
5
20
88.6
0 0
FSC CD3-PB Acute Chronic Healthy Acute Chronic Healthy

0.313 7.53 d e
∆42PD1-AF647
***

CD38+HLA–DR+ CD8+ (%)


40 80
*** *** ***
Isotype

Vδ1
30 Vδ2 60

CD3+ (%)
**
67.2 20 40
***
CD3-PB CD3-PB 10 20

0 0
Acute Chronic Healthy Acute Chronic Healthy
g
50,000 800
R2 = 0.5251 R2 = 0.2343
40,000 P < 0.0001 P = 0.0122
TNF-α (pg ml–1)

600

IL-6 (pg ml–1)


f 30,000
50 *** 400
20,000
∆42PD1+Vδ2+ (%)

40
10,000 200
30
0 0
20 0 10 20 30 0 10 20 30

10
4,000 6,000
R2 = 0.4714 R2 = 0.1672
0 P = 0.0001 P = 0.0381
3,000

IFN-α (pg ml–1)


IL-1β (pg ml–1)

I-II III-V 4,000


Fiebig status
2,000
2,000
1,000

0 0
0 10 20 30 0 10 20 30
∆42PD1+Vδ2+ (%) ∆42PD1+Vδ2+ (%)

Fig. 1 | Δ42PD1+Vδ2 cells are found in early HIV-1 infection and correlate with immune activation. a, Representative plots for the detection of
Δ​42PD1-expressing CD3+Vδ​2+ cells in PBMCs of acute HIV-1-infected patients by flow cytometry based on isotype control. Numbers on scatter plots
represent percentages. b,c, Acute (n =​ 57, triangles) and chronic (n =​ 50, squares) HIV-1 patients and healthy controls (n =​ 29, circles) were assessed for
frequency of CD3+Vδ​2+ cells (b) and Δ​42PD1-expressing CD3+Vδ​2+ cells (c). d, Proportions of Vδ​1 and Vδ​2 cells among CD3+ cells analysed in HIV-1
acute and chronic patients and healthy controls (n =​ 12 for each group). e, Frequency of CD38+HLA-DR+ CD8 T cells for acute (n =​ 47), chronic (n =​ 17)
and healthy (n =​ 14) individuals. f, Δ​42PD1+Vδ​2+ cells among acute HIV-1 patients who are seronegative (Fiebig I–II, n =​ 17) and seropositive (Fiebig III–V,
n =​ 31) were assessed. Data represent mean ±​ s.e.m. and statistics were calculated based on the Mann–Whitney U-test; *P <​ 0.05, **P <​ 0.01, ***P <​ 0.001.
(a–f). g, Acute HIV-1 patient plasma cytokines were measured in a multiplex assay. Levels of cytokines were correlated with frequency of Δ​42PD1-
expressing CD3+Vδ​2+ cells by linear regression (95% confidence intervals shown as dotted lines) to calculate statistics. Each symbol represents the data
of one individual patient (n =​ 19); n, number of individual patient samples.

of PBMCs (Supplementary Fig.  7). We therefore sought to deter- day 3 p.i. Interestingly, only Vγ​9/Jγ​1.2 transcript was detected in
mine the specific effects of IL-12 and IL-15 on inducing Δ​42PD1 both mock- and HIV-1-induced Vδ​2+ cells (Supplementary Fig. 8a).
expression with or without neutralizing antibodies in PBMC cul- Furthermore, Vγ​9/Jγ​1.2 can be detected in both HIV-1 stimulated
tures infected with HIV-1NL4-3 or HIV-1Henan for 3 days. Compared Δ​42PD1− or Δ​42PD1+ CD3+Vδ​2+ cells (Supplementary Fig. 8b). To
to control, anti-IL-12 and/or anti-IL-15 abrogated the expression of verify if there are differences in TCR arrangement in the Vγ​9/Jγ​1.2
Δ​42PD1 on Vδ​2 cells (Fig. 2c). These data demonstrated that IL-12 transcripts, we performed sequence alignment analysis on TRGV9
or IL-15 cytokines arising from HIV-1 infection could activate Vδ​2 and TRGJP reference regions (Supplementary Fig.  8c,d). In the
cells to express Δ​42PD1. junction CDR3 region, we found one nucleotide change at position
The majority of Vδ​ 2 cells in peripheral blood express the 350 that led to an amino acid change from methionine (M) to lysine
TCR-Vγ​9 chain. Previous reports have shown that phosphoanti- (K) for Δ​42PD1− CD3+Vδ​2+ cells, which might differ in response
gen-responsive Vδ​2 cells employ the Jγ​P (or Jγ​1.2) gene segment to HIV-1. Nonetheless, our analysis shows that Δ​42PD1 may not be
from the junction region and these cells are depleted in HIV-1 related to phosphoantigen recognition for Vδ​2 cells.
patients29–31. To investigate if Δ​42PD1+Vδ​2 cells express TCR-Vγ​9/ The gut-homing property of Δ​42PD1+Vδ​2 cells has been shown
Jγ​1.2 during HIV-1 infection, we performed RT–PCR analysis on in a previous study where CCR9 and CD103 were upregulated
CD3+Vδ​2+ cells isolated from mock or HIV-1-treated PBMCs at during HIV-1 infection, and this might facilitate their migration to the

Nature Microbiology | VOL 2 | OCTOBER 2017 | 1389–1402 | www.nature.com/naturemicrobiology 1391


© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Articles NaTuRe MICRObIOLOgy

∆42PD1+ ∆42PD1+
a b
28.1
20
47.3

∆42PD1+Vδ2+ (%)
9.52 15 * *
* HIV–1NL4-3

∆42PD1-AF647
10 HIV–1Henan
*
** * *
Vδ2–PE

– –
∆42PD1 ∆42PD1 5
78.9 6.45 Mock

CD103-AF488
0
CD3-PB CD3-PB 0 1 2 3 4 5 6
0.331
Day p.i.

SSC
CD69-PE/Cy7 CCR9-PerCP/
Cy5.5
c d e
80 80 80 ***
40 ** ∆42PD1+
CCR9+CD103+ Vδ2+ (%)

Mock
** ** ** ∆42PD1–

CCR9+CD103+ (%)
∆42PD1+Vδ2+ (%)

30
HIV-1NL4-3 60 *** 60 60
* HIV-1Henan ** ***
20 40 *** 40 ** 40

10 ** *** 20 20 20
** **
* *
0 0 0 0
3 4 5 6 3 4 5 6 ∆42PD1+ ∆42PD1–
pe

5
-1

-1

-1
ty

IL

IL

IL

Day p.i. (HIV-1NL4-3) Day p.i. (HIV-1Henan)


Iso

ti-

ti-

ti-
An

An

an
2/
-1
IL
ti-
An

Fig. 2 | HIV-1 infection induces Δ42PD1 expression on Vδ2 cells. a, Flow cytometry plots for assessing the frequency of Δ​42PD1 expression on
CD3+Vδ​2+ cells from human PBMCs infected with X4-tropic HIV-1NL4-3 at day 3 p.i., representative of four independent experiments. Expression of
activation marker CD69 and gut-homing receptors CCR9/CD103 among Δ​42PD1+ and Δ​42PD1− subpopulations is shown. b, Data from mock (black
circles), HIV-1NL4-3 (white squares) or R5-tropic HIV-1Henan (grey triangles) infected PBMCs in a 6 day time-course experiment (four independent
experiments). c, PBMCs infected with mock, HIV-1NL4-3 or HIV-1Henan were examined at day 3 p.i. for expression of Δ​42PD1 on Vδ​2+ cells in the presence
of neutralizing antibodies against IL-12 and/or IL-15 (four independent experiments). d,e, Expression of CCR9/CD103 on Δ​42PD1+ and Δ​42PD1− CD3+Vδ​
2+ cells, shown in a time-course experiment (four independent experiments) (d) or acute cohorts (n =​ 57) (e). Data were acquired by flow cytometry and
represent mean ±​ s.e.m.. Statistics were calculated based on Student’s paired t-test (b–d) or Mann–Whitney U-test (e); *P <​ 0.05, **P <​ 0.01, ***P <​ 0.001.

intestines in patients14. We therefore examined this in a time-course propria (LP) and intraepithelial lymphocytes (IEL) of the small
experiment of PBMCs infected with either HIV-1NL4-3 or HIV-1Henan intestine in mice receiving HIV-Vδ​2 cells as compared to mock-Vδ​2
by flow cytometry (Fig. 2a). We found that CCR9 and CD103 expres- by flow cytometry among other tissues (Fig. 3b and Supplementary
sion is mainly found in the Δ​42PD1+ subpopulation of Vδ​2 cells Fig.  9b). In comparison, we also found that NSG-HuPBL mice
following either viral infection (Fig. 2d). In contrast, Δ​42PD1−Vδ​2 without adoptive transfer harboured minimal CD3+Vδ​2+
cells had significantly less coexpression of CCR9 and CD103. To cells (Supplementary Fig.  10a). To confirm that transferred Vδ​2
determine the biological relevance of CCR9/CD103 expression on cells migrate to the small intestines, we injected carboxyfluorescein
Δ​42PD1+Vδ​2 cells, we examined the two acute cohorts of HIV- succinimidyl ester (CFSE)-labelled Vδ​2 cells isolated from mock
infected patients and found that Δ​42PD1+Vδ​2 cells significantly and HIV-1 PBMCs and, by day 3 post-adoptive transfer, the highest
(~30%) harboured CCR9/CD103 coexpression compared with number of CFSE+ cells were found in the small intestines (detected
Δ​42PD1−Vδ​2 cells (Fig. 2e). by flow cytometry; Supplementary Fig. 10b). Using a more sensitive
and stable dye, we performed another set of experiment with Qdot
Δ42PD1+Vδ2 cells migrates to the small intestines in a human- nanocrystal-labelled Vδ​2 cells, which also showed migration of
ized mouse model. To determine the in vivo relevance of transferred cells to the small intestine (detected by flow cytometry
Δ​42PD1+Vδ​2 cells during HIV-1 acute infection, we used NOD. and confocal microscopy; Fig. 3e,f and Supplementary Fig. 10c).
Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice humanized with freshly iso- Small intestinal pathology was then examined in mice receiving
lated human PBMCs (NSG-HuPBL), as we recently described32. HIV-Vδ​2 cells (haematoxylin and eosin (H&E) staining, Fig.  3c).
Three weeks after transfusion of 1 ×​  107 PBMCs, immune cells were In contrast to the Mock-Vδ​2 control, which showed intact villi
reconstituted in the NSG mice with detectable human CD45+ cells structures, HIV-Vδ​2 mice demonstrated the apparent development
and subpopulations of CD3+, CD4+ and CD8+ T cells in peripheral of Gruenhagen’s space and vacuolization (black arrows, Fig.  3c)
blood (Supplementary Fig. 9a). Next, to determine if Δ​42PD1+Vδ​2 and progression to detachment of epithelium (asterisk), mucosal
cells are capable of homing to mucosal tissues in vivo, autologous ulceration and disintegration of the LP (red arrow)33–35 (Fig.  3c).
CD3+Vδ​2+ cells were sorted from mock (Mock-Vδ​2) or HIV-1NL4-3- Furthermore, fluorescent immunohistochemistry (IHC) stain-
infected (HIV-Vδ​2) PBMC cultures at day 3 p.i. and intravenously ing of tissue sections showed infiltration of Δ​42PD1+Vδ​2+ cells in
(i.v.) transfused into NSG-HuPBL mice (Fig. 3a). After 3 days, cell the LP (white arrow, Fig.  3d). We therefore concluded that puri-
preparations from different tissues were analysed. We found that a fied HIV-induced Δ​42PD1+Vδ​2+ cells preferentially migrated to
higher frequency of Δ​42PD1+Vδ​2+ cells was detected in both lamina the small intestines after adoptive transfer, leading to inflamma­tory

1392 Nature Microbiology | VOL 2 | OCTOBER 2017 | 1389–1402 | www.nature.com/naturemicrobiology

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
NaTuRe MICRObIOLOgy Articles
a b 60
1 × 107 Mock-Vδ2
2 × 105 CD3+Vδ2+ (i.v.)

Vδ2+CD3+ (%)
PBMCs i.p. Endpoint HIV-Vδ2 *
40

Day –21 Day 0 Day 3 20 *


0
Mock-Vδ2 (n = 4) HIV-Vδ2 (n = 5)
NSG-HuPBL (n = 9)

en
Sp C

LN
SI LP)
LI L)
LI P)
L)
M

(I E

(I E
le

(L
(
PB

SI
c Mock-Vδ2 HIV-Vδ2 d Vδ2 Δ42PD1 Merge
*

Mock-Vδ2
HIV-Vδ2
e f
1,500 Mock-Vδ2 HIV-Vδ2 No transfer
*
/104 lymphocytes
No. Qdot+CD3+

1,000

500
Qdot
Hoechst
0
Mock-Vδ2 HIV-Vδ2

Fig. 3 | Preferential migration of HIV-induced CD3+Vδ2+ cells to the intestines in humanized mice. a, Schematic diagram of humanized mice experiment
using human PBMC reconstituted NSG mice (HuPBL). b, Flow cytometry data showing the detection of CD3+Vδ​2+ cells in different tissue compartments,
at 3 days post-adoptive transfer (i.v.) of FACSorted CD3+Vδ​2+ cells from PBMCs infected with mock (Mock-Vδ​2; n =​ 4) or HIV-1 (HIV-Vδ​2;
n =​ 5) into mice. LN, lymph nodes; LP, lamina propria; IEL, intraepithelial lymphocytes of small intestine (SI) and large intestine (LI); n, number of
individual mice per group. c, Microscopic analysis of small intestine tissue sections 3 days post-transfer, by H&E, representative of ten images taken
per mice. Intestinal pathology show Gruenhagen’s space and vacuolization (black arrows), epithelium detachment (asterisk) and mucosal ulceration
and disintegration of the LP (red arrow). Yellow scale bars, 20 μ​m. d, Fluorescent IHC images for the detection of cells expressing Vδ​2 and Δ​42PD1,
counterstained with Hoechst 33258, and co-localization of Vδ​2 and Δ​42PD1-positive cells (merged), are shown for small intestine tissue sections of
mice receiving HIV-Vδ​2 cells (enlarged inset) and absent in mock. Images are representative of ten images taken per sample. e, Qdot-labelled Vδ​2+ cells
purified from mock or HIV-1 infected PBMCs were i.v. transferred into NSG-HuPBL mice (n =​ 4 each) for 3 days before assessment by flow cytometry.
CD3+Qdot+ cell numbers in the small intestine homogenates are plotted. f, Representative confocal microscopy images showing Qdot+ cells (white
arrows) found in the villi of small intestines. At least ten images were taken per mouse small intestine tissue sample. Two mice without transfer served as
control. White scale bars in d and f, 20 μ​m. Data represent mean ±​ s.e.m. and statistics were calculated based on Mann–Whitney U-test (b,e); *P <​ 0.05.

pathology and tissue damage there. To determine the clinical rel- to endogenous intracellular or soluble proteins36. To test this
evance of this phenomenon, we performed correlation analysis of hypothesis, monocyte-derived dendritic cells (MoDCs) pre-treated
HIV-1 patient plasma of markers associated with intestinal muco- with blocking antibody against TLR2, TLR4 and TLR6 were used
sal damage, that is, soluble (s)CD14, fatty acid binding protein 2 to monitor cytokine signals after sΔ​42PD1fc protein or LPS treat-
(FABP2) and haptoglobin to the frequency of Δ​42PD1+Vδ​2+ cells ment. Purified proteins were assessed for endotoxin by the limulous
(Supplementary Fig. 11a). Negative and positive correlations were amoebocyte lysate (LAL) assay or oxidized phospholipid contami-
found for sCD14 and FABP2, respectively. However, the only sig- nants by measuring oxidized low-density lipoproteins (oxLDL), but
nificant difference for FABP2 was found between acute and chronic no positive results were found. By qRT–PCR analysis, TNF-α​, IL-6
patients (Supplementary Fig.  11b). These findings suggest that and IL-1β​transcriptional levels were found to be reduced when
Δ​42PD1+Vδ​2+ cells may infiltrate to the intestines and, in part, play TLR4 was inhibited, but not TLR2 or TLR6 (Fig. 4a). Consistently,
a role in causing damage to the mucosal barrier. MoDCs pretreated with the TLR4-specific inhibitor CLI-095 abro-
gated TNF-α​, IL-6 and IL-1β​release by LPS and sΔ​42PD1fc treat-
Δ42PD1 induction of cytokines from DCs is dependent on TLR4. ment on MoDCs (Fig.  4b). Furthermore, when we used vectors
To determine the molecular mechanism of possible Δ​42PD1+Vδ​2+ encoding short hairpin RNA (shRNA) against TLR4, TNF-α​ and
-mediated production of pro-inflammatory cytokines, we sought to IL-6 production levels were significantly decreased compared to
identify the cognate receptor of Δ​42PD1 on DCs. Because extra- control MoDCs following sΔ​42PD1fc protein treatment (Fig.  4c).
cellular treatment of Δ​42PD1 mainly induces pro-inflammatory The effect of shRNA in downregulating TLR4 and CD14 proteins
cytokines in human PBMCs24, we suspect that its receptor is one of was shown in MoDCs using western blotting and flow cytom-
the surface innate membrane activators, such as TLR2, TLR4 and etry (Supplementary Fig.  12a,b). In contrast, shRNA against
TLR6, although they have been shown previously only to respond another component of the TLR4 complex, CD14, resulted in an

Nature Microbiology | VOL 2 | OCTOBER 2017 | 1389–1402 | www.nature.com/naturemicrobiology 1393


© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Articles NaTuRe MICRObIOLOgy

a tnfa il6 il1b d *


25 200 100 Isotype 1.0
Anti-TLR2 * * Media

Absorbance (620 nm)


0.8 LPS
Relative expression

20 150 80 Anti-TLR4
Anti-TLR6 sΔ42PD1fc
15 60 0.6
* 100
10 40 0.4
50
5 20 0.2

0 0 0 0.0
a

a
fc

fc
a
S

ly

4
trl
fc
D1

1
i

i
i
LP

LP

LP
PD
ed

LR

D1
ed

PD
ed

on

-C
2P
M

-T

-C
M
M

42

42

NA
lls
4

NA

NA

Ce

iR

iR

iR
ps

ps

ps
b e
150 TLR4 *
* *
TLR4/MD2-wt

Normalized luciferase
8,000 4,000 DMSO
600
CLI-095 * TLR4/MD2-F126A
100 TLR4/MD2-C95Y
TNF-α (pg ml–1)

IL-1β (pg ml–1)


6,000 3,000
IL-6 (pg ml–1)

400
4,000 2,000
* *
* 50
200 **
2,000 1,000
ND ND ND ND
0 0 0 0
ia

ia

ia
c
fc

α
sΔ PS

sΔ PS

fc

ag
c
1

1
1f

1f

LP

F-
LP

PB
ed

PD
PD

PD
ed

ed

1fl
PD
L

PD

TN
M

M
42
42

42

42

42


c f
**
*
** ** *
psiRNA-Ctrl 60 *
2,500 6,000 psiRNA-TLR4
*
Normalized luciferase
psiRNA-CD14
2,000
TNF-α (pg ml–1)

* 40
IL-6 (pg ml–1)

4,000
1,500
*
1,000 * 20
2,000
500
0 0 0
c
ia
ia

D1
g

Ab

D1

Ab

D1
S

1f
1f

PD
PD

Ne
LP

ed
ed

PD

LP

2P

2P

2P
iso

iso
M
M

42
42

Δ4

Δ4

Δ4
+

+

ti-

ti-
ia

5
-1
293A cells
ed

an

an
IL
M

2/
+

+
ia

5
-1

-1
ed

TLR4 IL

IL
M

TLR4/MD2-WT 2/
-1
IL

TLR4/MD2-F126A γδ T cells
TLR4/MD2-C95Y

Fig. 4 | Δ42PD1 functions via TLR4 for the induction of cytokine production. a,b, Human MoDCs were pre-treated with antibody blockers against
TLR2, TLR4, TLR6 or isotype antibody control (n =​ 4) (a), or TLR4-specific inhibitor CLI-095 or DMSO (n =​ 4) (b) before addition of LPS (100 ng ml–1)
or recombinant purified sΔ​42PD1fc protein (2 μ​g ml–1). MoDCs in media only serve as negative control. TNF-α​, IL-6 and IL-1β​mRNA expression at 6 h
post-treatment or supernatant cytokine were analysed by qRT–PCR and multiplex cytokine bead assay, respectively. ND, not detected. c, MoDCs were
transfected with control vector (Ctrl) or vector encoding shRNA against TLR4 (psiRNA-TLR4) or CD14 (psiRNA-CD14) and analysed for TNF-α​ and
IL-6 production following 24 h treatment with LPS or sΔ​42PD1fc (n =​ 3). d, HEKBlue-hTLR4 cell line was transfected with psiRNA-Ctrl, psiRNA-TLR4 or
psiRNA-CD14 before treatment with LPS or sΔ​42PD1fc. e,f, Transient co-transfection of 293A cells with NF-κ​B-luc, TLR4 and/or MD2-wt, MD2-F126A
or MD2-C95Y, was performed, followed by treatment with sΔ​42PD1fc, sΔ​42PD1flag, LPS or TNF-α​ (100 ng ml–1) (n =​ 3) (e); or co-cultured at 1:1 with cells
293A-Neg, -PD1, -Δ​42PD1; or media or IL-12/IL-15-stimulated γ​δ​T cells that were pre-treated with isotype or α​-Δ​42PD1 antibody (n =​ 3) (f). Data are
presented as mean ±​ s.e.m. of independent experiments (n) and statistics were calculated using Student’s paired t-test (a–f); *P <​ 0.05, **P <​ 0.01.

increased level of cytokines induced by sΔ​42PD1fc, suggesting that experiments was performed. First, HEKBlue-hTLR4 cells were
CD14 probably hinders Δ​ 42PD1–TLR4 signalling (Fig.  4c). In downregulated for CD14 using psiRNA-CD14 and subsequently
addition, when we performed experiments in the TLR4 signalling MD2 using two commercial siRNAs. The effect of downregula-
reporter cell line (HEKBlue-hTLR4), we found that LPS-induced tion was assessed by flow cytometry (Supplementary Fig. 13a). sΔ​
signalling was decreased when using shRNA against TLR4 or CD14. 42PD1fc and sΔ​42PD1flag proteins or LPS stimulation resulted in
However, sΔ​42PD1fc only induced reporter signalling when CD14 reporter activity. We used two different tagged proteins to show
was downregulated (Fig. 4d). These results demonstrate that Δ​42PD1 that the tag did not affect the function of sΔ​42PD1. CD14 down-
can induce cytokines through TLR4 in the absence of CD14. regulation resulted in a higher response by sΔ​42PD1, in contrast
To further illustrate that Δ​ 42PD1 can induce TLR4 down- to LPS, which was decreased (Supplementary Fig. 13b), consistent
stream signalling and to eliminate the possibility of endotoxin or with the above results. Knockdown of MD2 by siRNA resulted
oxidized phospholipids that require MD2, a series of reporter cell in a further reduction in LPS response, but had no significant

1394 Nature Microbiology | VOL 2 | OCTOBER 2017 | 1389–1402 | www.nature.com/naturemicrobiology

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
NaTuRe MICRObIOLOgy Articles
a b
sΔ42PD1fc coated sPD1fc coated
10 10
KD = 6.82 µM
Time
8 0
TLR4/MD2 (µM)

Response unit

Response unit
6 TLR4/MD2 (µM) –10
0.49
44.1
4 –20 1.64
14.7
2 –30 4.9
4.9
1.64 14.7
0 –40 44.1
0.49
Time

sΔ42PD1fc protein

TLR4-His protein
IP: anti-rabbit Fc
TLR4 – + – + + – –
TLR4/MD2 – – + – – + +

Marker
LBP – – – + – + –
HMGB1 – – – – + – +

IB: anti-His
- 70 kDa

IB: anti-rabbit Fc - 70 kDa

d Pre-bleach → Post-bleach FRET 0.3 **


CFP channel

FRET efficiency
0.2
sΔ42PD1-CFP

0.1
YFP channel

130 0

sΔ42PD1-CFP/TLR4-YFP

sΔ42PD1-CFP mixed
110
Grey value

with TLR4-YFP
90

co-transfected
YFP channel
70
TLR4-YFP

50
0 100 200 300 400
Distance (pixels)

Fig. 5 | Direct interaction between Δ42PD1 and TLR4. a,b, SPR of flowing concentrations (0.49 to 44.1 μ​M) of TLR4/MD2 purified protein against
coated sΔ​42PD1fc (a) or sPD1fc (b). c, Co-IP of purified sΔ​42PD1fc and TLR4 or TLR4/MD2 proteins with or without LBP and HMGB1. Detection of
immunoprecipitate (IP) pulldown by anti-rabbit Fc agarose beads and immunoblotting (IB) by western blotting using anti-His or anti-rabbit Fc antibodies.
Dotted line: the same immunoblot from two exposure times to avoid over-developed TLR4 protein control signals. d, FRET analysis of co-transfecting sΔ​
42PD1-CFP and TLR4-YFP in 293T cells. Confocal microscopy was used to detect CFP and YFP channels, and FRET analysis was performed using PixFRET
(grey to white signals, blue background) with values measured against distance. FRET efficiency was calculated by subtracting background regions
from signal regions, and statistics were calculated by Student’s paired t-test of at least 20 fields from two independent experiments. A representative
fluorescence image is shown. Bar graph data represent mean ±​ s.e.m; **P <​ 0.01.

effect on sΔ​ 42PD1 proteins. We also performed co-culture expression plasmids encoding NF-κ​B-luc and TLR4, with or without
experiments using previously described stably expressing PD1 or MD2-WT (wild-type), MD2-F126A or MD2-C95Y (Supplementary
Δ​42PD1 293A cells24,25. 293A-Δ​42PD1 showed a significant signal- Fig.  15b), cells were treated with sΔ​42PD1fc and sΔ​42PD1flag pro-
ling response only when CD14 was downregulated, but was unaf- teins, LPS or TNF-α​. The results show that, unlike TNF-α​, LPS could
fected by a reduction in MD2 (Supplementary Fig. 13c). Furthermore, induce NF-κ​B activity with TLR4/MD2-WT transfected cells, but
when we used IL-12/IL-15-stimulated γ​δ​T cells (with upregulated not TLR4 alone, TLR4/MD2-F126A or TLR4/MD2-C95Y (Fig. 4e).
Δ​42PD1) for co-culture, similar results were observed. Compared In contrast, sΔ​42PD1fc and sΔ​42PD1flag proteins could trigger NF-κ​
to media-treated cells, IL-12/IL-15-γ​δ​T cells could induce reporter B activity with only TLR4 being expressed, despite the presence of
activity on CD14-downregulated cells (Supplementary Fig.  13d), wild-type or MD2 mutants (Fig.  4e). Similar findings were found
but this was reduced when Δ​42PD1 was blocked using an in-house for 293A-Δ​42PD1 or IL-12/IL-15-γ​δ​T cells co-cultures with these
anti-Δ​42PD1 antibody (clone CH101, Supplementary Fig.  14). To reporter cells (Fig. 4f). Together, our data illustrate that Δ​42PD1 can
verify whether Δ​42PD1 requires MD2 to trigger TLR4 signalling, trigger TLR4 downstream signalling independent of MD2, which
site-directed mutagenesis was performed to generate F126A and eliminates the possibility of contaminants.
C95Y mutations of the pEFBOS-humanMD2 expression plasmid
(provided by K. Miyake) (Supplementary Fig. 15a). These mutations Δ42PD1 directly interacts with its cognate receptor TLR4.
would lead to MD2 unresponsiveness to endotoxin and TLR4 sig- To determine if direct interaction occurs between Δ​42PD1 and
nalling37–40. Following transient co-transfection of 293A cells with TLR4, several experiments were performed. A surface plasmon

Nature Microbiology | VOL 2 | OCTOBER 2017 | 1389–1402 | www.nature.com/naturemicrobiology 1395


© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Articles NaTuRe MICRObIOLOgy

resonance (SPR) experiment was first carried out by flowing dif- HIV-induced Δ​42PD1+Vδ​2 cells abrogated cytokine induction. As
ferent concentrations of TLR4/MD2 to immobilized sΔ​42PD1fc controls, MoDCs treated with sΔ​42PD1fc or LPS led to the expres-
protein. The results in Fig.  5a show positive binding kinetics of sion of tnfa, il1b and il6. To confirm these findings, an enzyme-linked
TLR4/MD2 to sΔ​42PD1fc and the equilibrium dissociation con- immunosorbent assay (ELISA) was performed for IL-6 secretion in
stant (KD) value of the interaction is calculated to be 6.82 μ​M the co-culture experiment between sorted HIV-Vδ​2 and autologous
(Fig.  5a). In contrast, TLR4/MD2 had no interaction with MoDCs (Fig.  6b), and this correlated with the qRT–PCR results
sPD1fc proteins (Fig.  5b) and LPS showed no binding activity to (Fig.  6a). Also, blocking the Δ​42PD1–TLR4 pathway with TLR4-
sΔ​42PD1fc (Supplementary Fig. 16a). Furthermore, we confirmed that specific inhibitor CLI-095 or antibodies significantly decreased the
no binding exists between sΔ​42PD1fc and PD-L1 (Supplementary level of IL-6 release following co-culture (Fig. 6b).
Fig.  16b), while a strong interaction was observed for sPD1fc and To determine the in vivo relevance of these findings, we inves-
PD-L1 (Supplementary Fig.  16c). In another set of experiments tigated if Δ​42PD1+Vδ​2 cell transmigration could lead to increased
based on fluorescence binding, the interaction between Δ​42PD1 pro-inflammatory cytokines in the gut using our humanized mouse
and TLR4/MD2 was confirmed with a KD (4 μ​M) similar to that model. We found that in the small intestines of NSG-HuPBL mice
of the SPR analysis (Supplementary Fig.  16d). Also, no signifi- with adoptively transferred HIV-Vδ​2, Δ​42PD1+ cells were found in
cant interaction of Δ​42PD1 and PD-L1 was observed in this assay close proximity to TLR4+ cells in the LP of intestinal villi, which
(Supplementary Fig. 16e). Second, co-immunoprecipitation (co-IP) appear to show pathological detachment of the epithelium (Fig. 6c).
experiments were conducted. Purified sΔ​42PD1fc protein was Furthermore, Vδ​2+ cells appear to be located in cellular regions of
mixed with TLR4-His or TLR4/MD2-His complex proteins in a 1:1 increased IL-6 expression, and this may suggest that their inter-
ratio. Because LPS binding protein (LBP) and high-mobility growth action with TLR4-expressing cells induced IL-6, mediating the
box 1 (HMGB1) could interact with TLR441, they were added to the inflamed pathology observed in the gut tissue (Fig. 6d).
mixture in equal amounts to determine if the interaction between
sΔ​42PD1fc-TLR4 was affected. Following western blotting, the anti- Inhibition of Δ42PD1–TLR4 alleviates gut pathology in human-
rabbit Fc detected sΔ​42PD1fc and indicated successful pulldown ized mice. Because the Δ​42PD1–TLR4 pathway exhibited by HIV-
despite different combinations of proteins being mixed (Fig.  5c). induced Vδ​2 cells may confer pathological effects in the gut, we
Anti-His detection demonstrated co-precipitated TLR4-His after next sought to determine if blocking this pathway would reduce
sΔ​42PD1fc was mixed with TLR4/MD2 or TLR4, confirming their inflammation in the small intestine. In another set of NSG-HuPBL
interaction. Interestingly, the presence of HMGB1, but not LBP, mice experiments, anti-Δ​42PD1 (clone CH101), isotype antibody
decreased the interaction between sΔ​42PD1fc and TLR4 or TLR4/ or CLI-095 were administered i.p. 2 h before adoptive transfer of
MD2 (Fig.  5c), suggesting that HMGB1 might have outcompeted mock- or HIV-induced Vδ​2 cells and by another injection 2 days
sΔ​42PD1fc in its interaction with TLR4. Moreover, in a dose-depen- after transfer (Fig. 6e). H&E staining of the small intestines showed
dent experiment, decreasing the amount of sΔ​42PD1fc reduced the that mice receiving Mock-Vδ​2 cells did not present with gut inflam-
detection of TLR4 in the co-IP experiment but appeared to saturate mation, whereas transfer of HIV-Vδ​2 into mice treated with isotype
at a ratio of more than 1:1 (Supplementary Fig. 17). Finally, to fur- antibody resulted in significant pathological effects (Fig. 6f), consis-
ther demonstrate that sΔ​42PD1fc interacts with TLR4, fluorescence tent with the above results (Fig. 3c). In contrast, mice that received
resonance energy transfer (FRET) experiment was performed by anti-Δ​42PD1 antibody had markedly reduced gut pathology, similar
co-transfecting plasmids encoding sΔ​42PD1−CFP and TLR4-YFP to CLI-095-treated mice (Fig. 6f). Flow cytometry analysis showed
into 293T cells. After one day, confocal microscopy was performed that the frequency of CD3+Vδ​2+ cells among human CD45+ cells was
to assess if FRET had occurred. By comparing pre- and post- significantly reduced in the small intestines of anti-Δ​42PD1 treated
photobleaching of the donor TLR4-YFP fluorescence, an increase mice compared to isotype, but not CLI-095 treatment (Fig.  6g).
in sΔ​42PD1−CFP fluorescence was observed in the YFP channel Therefore, the Δ​42PD1–TLR4 pathway may be an important con-
and by software analysis using PixFRET (Fig.  5d, white regions). tributor to gut inflammation following HIV-1 infection, and this
In contrast, the negative control pair sΔ​42PD1-CFP and TLR9- effect can be reduced using the anti-Δ​42PD1 antibody.
YFP showed no significant fluorescence transfer (Supplementary
Fig.  18). Overall, these experiments demonstrate that sΔ​42PD1fc Discussion
binds to TLR4 as its cognate cell surface receptor; this has not been In this study, to our knowledge, we describe for the first time that
demonstrated previously. an endogenously surface-expressed protein, Δ​42PD1, uses TLR4/
MD2 as an innate receptor to perform its immune stimulatory func-
HIV-induced Δ42PD1+Vδ2 cells with gut-homing capabilities tion (Fig. 5). One critical role of this innate Δ​42PD1–TLR4 pathway
can crosstalk with DCs via TLR4 to induce robust cytokine pro- was found to be regulating HIV-1 infection through the induction
duction. Because Δ​42PD1 interacts directly with the TLR4/MD2 of Δ​42PD1+Vδ​2 cells, which co-expressed the gut-homing recep-
receptor complex, we sought to determine if Vδ​2 cells engage this tors CCR9 and CD103 (Fig. 2a,d). Once these HIV-1-induced Vδ​2
Δ​42PD1–TLR4 pathway for cytokine induction. To test this hypoth- cells transmigrated into the small intestines, they promoted mucosal
esis, we used HIV-induced Δ​42PD1+Vδ​2+ cells to co-culture with inflammation and pathology in a humanized mouse model system by
autologous MoDCs at a 1:1 ratio for 24 h before performing qRT– triggering pro-inflammatory cytokine responses, probably through
PCR for cytokine expression. Vδ​2 cells from mock infection that TLR4-expressing DCs (Figs.  3 and 6c,d,f). In addition, a high fre-
express minimal levels of Δ​42PD1 were also sorted and used for quency of gut-homing Δ​42PD1+Vδ​2 cells was readily found among
comparison. Antibody blockers against TLR4 and Δ​42PD1 were acutely HIV-1-infected individuals, especially before seroconversion
included to show the specificity of the Δ​42PD1–TLR4 pathway. (Fig. 1f), and was correlated positively with plasma cytokine levels
Collectively, there were significantly increased expressions of tnfa, (Fig.  1g). The elevated cytokines may contribute to the polyclonal
il1b, il6 and ifna after co-culture of MoDCs with HIV-induced activation of CD38+HLA-DR+ CD8 T cells in the early phase of HIV
Δ​42PD1+Vδ​2 cells as compared to Mock-Vδ​2 (Fig.  6a). Blocking infection and mucosal damage (Figs. 1e and 6f). Our findings there-
TLR4 or Δ​42PD1 significantly reduced the level of cytokine induc- fore reveal a previously unrecognized Δ​42PD1–TLR4 pathway in
tion during co-culture (Fig. 6a), demonstrating a role of cell surface regulating immune activation in acute HIV-1 pathogenesis.
Δ​42PD1 as an inducer of cytokines in the context of HIV-1 infec- Δ​42PD1 is the first endogenous surface protein identified on
tion via TLR4 that was previously unrecognized. Moreover, cell–cell human cells that interacts with TLR4. Previous studies have only
interaction was required, because transwell separating MoDCs and identified endogenously secreted molecules that could signal through

1396 Nature Microbiology | VOL 2 | OCTOBER 2017 | 1389–1402 | www.nature.com/naturemicrobiology

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
NaTuRe MICRObIOLOgy Articles
a 40
tnfa
50
il6 b
30 *** 40 ***
20 30 * 4,000 *
5 *** 15 ***
4 *** * **
10 3,000
Relative gene expression
3

IL-6 (pg ml–1)


2 5
1 2,000
0 0

il1b il1b 1,000


80 80
70 ** 70 ** + ND
60 60 0
50 * 50 *
*** ***

IsoAb

Untreated
DMSO
CLI-095

sΔ42PD1fc
Anti-TLR4

γδ-T only

LPS
Anti-Δ42PD1
30 30
20 20
10 10
ND ND ND ND
0 0
Co-DC DC
IsoAb
Anti-Δ42PD1
Anti-TLR4
IsoAb
Anti-Δ42PD1
Anti-TLR4
Transwell
Untreated
sΔ42PD1fc
LPS

IsoAb
Anti-Δ42PD1
Anti-TLR4
IsoAb
Anti-Δ42PD1
Anti-TLR4
Transwell
Untreated
sΔ42PD1fc
LPS
Mock-Vδ2 HIV-Vδ2 DC Mock-Vδ2 HIV-Vδ2 DC
Co-DC Co-DC

c Δ42PD1 d Vδ2
e 1 × 107
TLR4 IL-6 Hoescht
PBMCs i.p. 1 × 105 CD3+Vδ2+ (i.v.) Endpoint

Day –21 Day 0 Day 3


–2 h Day 2

NSG-HuPBL (n = 1 5) Ab or CLI-095 (i.p.) Ab or CLI-095 (i.p.)

1. Mock-Vδ2 (n = 2) ( )
2. IsoAb + HIV-Vδ2 (n = 5) ( )
Groups -
3. Anti-Δ42PD1 + HIV-Vδ2 (n = 5) ( )
f Mock-Vδ2 IsoAb + HIV-Vδ2 4. CLI-095 + HIV-Vδ2 (n = 3) ( )

** g
4 20
Per cent CD3+Vδ2+ of CD45+

*
3 15
pathology grade
Small intestine

2 10

1 5

0 0
Ab

95
Ab

D1
2

2
1

95
PD

Iso

I-0
2P
Iso

I-0
2
k-

k-

Δ4
Δ4

CL
CL
oc

oc

ti-
ti-
M

An
An

Anti-D42PD1 CLI-095
+HIV-Vδ2 +HIV-Vδ2

Fig. 6 | HIV-induced Δ42PD1-expressing γδ T cells can induce robust cytokines from autologous DCs via Δ42PD1–TLR4. a, CD3+Vδ​2+ cells were sorted
from mock or HIV-1 infected PBMCs at day 3 p.i. and co-cultured 1:1 with autologous MoDCs (Co-DC) for 8 h (four independent replicates).
Anti-TLR4 or anti-Δ​42PD1, or isotype antibodies or transwell were used. qRT–PCR for the expression of tnfa, il6, il1b and ifna was performed on sorted
CD11c+ MoDCs. LPS or sΔ​42PD1fc protein served as control. ND, not detected. b, ELISA for IL-6 in co-culture experiments between HIV-1-induced sorted
Vδ​2+ cells and autologous MoDCs after 24 h with anti-TLR4 or anti-Δ​42PD1 antibodies, CLI-095 or respective controls (four independent replicates). c,d,
Fluorescent IHC staining of small intestine tissue sections from humanized mice that received sorted CD3+Vδ​2+ cells from mock (Mock-Vδ​2) or HIV-1-
infected PBMC cultures (HIV-Vδ​2) for 3 days. Immunostaining for human Δ​42PD1 and TLR4 (c) or IL-6 and Vδ​2 and counterstained for nucleus (d) was
performed to show close proximity between cells of interest (inset, white arrows). Images represent more than ten images taken per mouse from at least
four mice (per group). White scale bars, 20 μm. e, Schematic representation of NSG-HuPBL mice receiving Mock-Vδ​2 or HIV-Vδ​2 cells with anti-Δ​42PD1
Ab or CLI-095 administered (i.p.) 2 h prior and 2 days after cell transfer. (Iso)type antibody was used as control. f, Representative H&E images of small
intestine showing gut pathology from each group of mice, with grading shown as a graph. Images from at least 20 fields were taken and assessed. Yellow
scale bars, 20 μ​m. g, Flow cytometry detection of CD3+Vδ​2+ cells gated on human CD45+ cells. Graph data represent mean ±​ s.e.m. and statistics were
calculated based on Student’s paired t-test (a,b) or Mann-Whitney U-test (f,g); *P <​ 0.05, **P <​ 0.01, ***P <​ 0.001.

TLR4, including danger-associated molecular patterns (DAMPs) such which are produced as a result of cell death or pathological condi-
as HMGB1, and liver secretory glycoprotein Fetuin-A; but others have tions, we found that cell-surface-expressed Δ​42PD1 can drive a rapid
shown Ni2+ can genuinely trigger TLR441–44. Unlike the danger signals, inflammatory response via TLR4 on DCs (Fig. 4). Other studies have

Nature Microbiology | VOL 2 | OCTOBER 2017 | 1389–1402 | www.nature.com/naturemicrobiology 1397


© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Articles NaTuRe MICRObIOLOgy

shown that γ​δ​T cells and DCs can crosstalk to achieve dual activa- model (Fig. 3e,f). Once reaching the gut, they may engage in TLR4
tion of both cell types45,46, which is consistent with our finding that signalling, which is abundantly expressed by DCs and other muco-
Δ​42PD1+Vδ​2 cells can act on DCs through TLR4 signalling to stimu- sal cell types, including macrophages, myofibroblasts, endothelial
late a pro-inflammatory response. cells and circulating polymorphonuclear neutrophils (PMNs) to
To present Δ​42PD1 as a genuine TLR4 agonist, three postu- induce pro-inflammatory cytokines33. This may be the underlying
lates should be met, as proposed by Mancek-Keber and Jerala47 and mechanism behind the increased IL-6 expression in the small intes-
should address the possibility of contaminants such as endotoxin tines in humanized mice and/or gut pathology (Fig.  6d,f). While
and oxidized phospholipids in experiments. These postulates are as TLR4 signalling is an important regulator in intestinal homeosta-
follows: (1) binding to TLR4 or TLR4/MD2, (2) activation of the sis49, the onset of CCR9+CD103+Δ​42PD1+Vδ​2 cells in natural acute
receptor signalling by in situ produced agonist, and (3) structural HIV-1 infection can stimulate an inflammatory response through
identification for molecular interactions with TLR4 or TLR4/MD2. TLR4 that may contribute to gut CD4+ and Th17 T cell deple-
Because endotoxin and oxidized phospholipids engage MD2 to tion once migrated11,50–52. Furthermore, positive correlation of the
achieve TLR4 signalling, in our reporter assays the downregulation frequency of Δ​42PD1+Vδ​2 cells with FABP2 but not haptoglobin in
of MD2 or the use of the F126A and C95Y mutants demonstrates acute HIV-1 patients suggest that intestinal epithelial cell apoptosis
that Δ​42PD1 could still function, which indicates the absence of occurred rather than tight junction defects. The resulting muco-
such contaminants from these purified proteins. Also, no signifi- sal integrity damage caused by Δ​42PD1–TLR4 and possibly sub-
cant differences in downstream signalling between TLR4 or TLR4/ sequent microbial translocation may account for systemic chronic
MD2 were found with Δ​42PD1 protein stimulation or Δ​42PD1- immune activation—a hallmark of HIV-1 pathogenesis. However,
expressing cells, in consistency with the co-IP results, suggesting other factors such as oxidative stress may contribute to acute HIV-
that Δ​42PD1 may interact with TLR4 at sites distinguished from associated inflammatory responses (Supplementary Fig. 20), as has
those identified for LPS48. Together with SPR analysis, interaction of been suggested for chronic HIV-153,54.
Δ​42PD1 with TLR4/MD2 was confirmed, and Δ​42PD1 is capable HIV/AIDS is mainly a sexually transmitted disease, for which
of triggering TLR4 signalling. Furthermore, blocking of Δ​42PD1 early innate immunity at mucosal surfaces remains incompletely
in IL-12/IL-15-induced Δ​42PD1+γ​δ​ -T cell co-culture experi- characterized. As mounting evidence points to a role of the acute
ments with reporter cell lines or DCs demonstrates that naturally innate immune response in the control or development of HIV-1
expressed Δ​42PD1–TLR4 does indeed play a role in the induction disease, our findings may provide new insights into the understand-
of cellular responses. Determination of the crystal structure of Δ​ ing of a new pathway that underlies acute HIV-1 infection. Vδ​2 cells
42PD1 is currently under way, but we have attempted to gain some are characterized for other functions such as antigen presentation
idea as to where the interaction between Δ​42PD1 and TLR4 may and cytotoxicity1,55,56, whereas TLR4 has been implicated to affect
occur by docking wild-type PD-1 (known structure) to TLR4/MD2 T cell stimulation and function36,57,58, and future work to define the
in silico (Supplementary Fig.  19). It appears that PD-1 does not role of Δ​42PD1–TLR4 in the functional aspects of these cell types
interact with MD2, and the 42-nucleotide deletion may result in a may identify new immune mechanisms. Because Vδ​2 cells are
different structure for Δ​42PD1 to bind to the central N-terminal important players in other mucosal infections and disease mod-
region of TLR4. Without the structure of the TLR4/MD2/Δ​42PD1 els59,60, studies should examine the role of the Δ​42PD1–TLR4 path-
complex, we cannot, at this moment, definitively draw a conclusion way in settings besides HIV/AIDS.
about how Δ​42PD1 interacts with TLR4/MD2. However, we dem-
onstrate that Δ​42PD1 is an agonist for TLR4. Methods
The observation of upregulated Δ​42PD1 in response to acute Study subjects. All study subjects gave informed consent, and procedures
HIV-1 infection suggests that this protein can be an important reg- were reviewed and approved by the Beijing You-An Hospital Research Ethics
Committee, The First Hospital of China Medical University Ethics Committee
ulator of early innate immune responses. Given the rapid-response and the Ethics Review Committee of Shenzhen Third People’s Hospital. A cohort
property of the γ​δ​T cells and their function as a bridge between of members of the high-risk MSM (men who have sex with men) group were
innate and adaptive immune responses, we speculate that Δ​42PD1 enrolled in Beijing You-An Hospital, Beijing, China, and was screened every two
might play an essential role in modulating these functions. Indeed, months for HIV-1 infection. A total of 45 acute HIV-1-infected individuals were
enrolled in this study at Beijing You-An Hospital, Capital Medical University,
we found that the highest proportion of these cells arose at the earli-
Beijing, China. Another cohort of 12 acute HIV-1-infected individuals (MSM)
est stages of acute HIV-1 infection and reduced when the patients were enrolled from The First Hospital of China Medical University, Shenyang,
seroconverted, with a further decrease when the chronic state was China. They were subjected to voluntary HIV-1 testing and counselling, given
reached (Fig. 1c). However, HIV-1 infection induces Δ​42PD1+Vδ​2 necessary healthcare, and monitored regularly for HIV PCR testing at 6 week
cells, which were found to express the Vγ​9/Jγ​1.2 TCR, which may to 3 month intervals. Apart from signed informed consent forms for the
collection of samples, all subjects received subsequent analyses and completed an
suggest that these cells are depleted after early infection14,16. Our administered epidemiological questionnaire. Once identified as infected, patients
data therefore uncover a previously unrecognized cellular mecha- were followed up and blood samples were obtained in 1 or 2 week intervals.
nism by which early immune activation following HIV-1 infec- All new infections were classified based on the detection of HIV-1-specific
tion can occur through TLR4 signalling, leading to induction of RNA, antigen and antibody in plasma, according to the Fiebig system. Acute
pro-inflammatory cytokines and polyclonal activation of CD8+ T HIV positivity was achieved when real-time PCR showed positive detection
of HIV RNA, with or without western blot detection of all three Gag, Env and
cells, as well as subsequent intestinal inflammation and damage. Pol antigens, no antibody against HIV was detected by ELISA, but with HIV
To this end, the co-upregulation of gut-homing receptors CCR9/ seroconversion occurring within 6 months of follow-up. A further 17 chronic
CD103 on Δ​42PD1+Vδ​2 cells suggests their potential function in patients infected for more than 2 years were also recruited from Beijing You-An
modulating gut mucosal integrity. Previous studies show that the Hospital and 33 from Shenzhen Third People’s Hospital. None of the acute or
frequency of Vδ​2 cells is significantly reduced both in peripheral chronic patients received antiretroviral treatment. The patient characteristics are
shown in Supplementary Table 1.
blood and in the gut despite HAART during HIV-1 infection when
compared with healthy people14,29,31. In our study, up to 20% of Vδ​2 PBMC, MoDC and transfection. Fresh PBMCs were collected following gradient
cells expressed Δ​42PD1 in acute HIV-1-infected individuals, of centrifugation (Ficoll-Paque, GE Healthcare) of healthy human blood donor buffy
which a majority were positive for gut-homing receptors CCR9 and coats. Use of buffy coats received ethics approval from the Institutional Review
CD103 (Fig. 2a,d). This is in line with previous studies that showed Board of the University of Hong Kong/Hospital Authority Hong Kong West Cluster
#UW13-476. MoDCs were generated by 5 or 7 day culture of freshly purified
high α​4β​7 and CCR5 expression on Vδ​2 cells following in vitro CD14+ cells from healthy human PBMCs using microbeads (>​95% purity, Miltenyi
HIV-1 infection of human PBMCs16. The translocation ability of Biotec) in RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS,
Δ​42PD1+Vδ​2 cells was further demonstrated in a humanized mouse GIBCO), 2 mM l-glutamine (GIBCO), 1% streptomycin/penicillin (GIBCO),

1398 Nature Microbiology | VOL 2 | OCTOBER 2017 | 1389–1402 | www.nature.com/naturemicrobiology

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
NaTuRe MICRObIOLOgy Articles
10 mM HEPES (GIBCO), 2 mM β​-mercaptoethanol (GIBCO), 20 ng ml–1 each of Lyse) and flow cytometry. Three weeks after reconstitution, mice were injected i.v.
recombinant human GM-CSF and IL-4 (Sigma-Aldrich). A 50% medium change with 1 ×​  105 CD3+Vδ​2+ cells FACSorted from the same donor PBMCs following
was carried out every 3 days. MoDCs were transfected with 1 μ​g of plasmids 3 days of in vitro HIV-1 infection. For tracking experiments, FACSorted CD3+Vδ​
encoding shRNA (InvivoGen) against TLR4 (psiRNA-TLR4) or CD14 (psiRNA- 2+ cells were labelled with CFSE using the CellTrace CFSE proliferation kit or
CD14), or a control plasmid (psiRNA-Ctrl), using Lipofectamine 2000 (Invitrogen) Qdot 655 nanocrystals according to the manufacturer’s instructions (Thermo
in Opti-MEM medium (Invitrogen) for 48 h with Zeocin selection before treatment Fisher Scientific), before i.v. injection. For the in vivo blocking experiment,
with stimulating agents. Success of downregulation was observed by western anti-Δ​42PD1 (CH101 clone) antibody or TLR4 inhibitor CLI-095 (InvivoGen)
blotting. The HEKBlue-TLR4 cell line was used according to the manufacturer’s were administered i.p. at 2 mg kg–1 and 3 mg kg–1, respectively, 2 h before and
instructions (InvivoGen) and expression of human TLR4, CD14 and MD2 was 2 days after CD3+Vδ​2+ cell transfer. Three days after transfer, mice were killed
monitored by flow cytometry. for blood and the following tissues collected for analysis: spleen, lymph nodes,
small intestine and large intestine. LP cells and IELs were isolated according to
Antibodies and flow cytometry. The following antibodies were used for western a published protocol66. After formalin fixation and paraffin embedding, tissue
blotting: mouse anti-His (R&D Systems #MAB050), anti-mouse IgG HRP (GE sections and H&E staining were performed at the Department of Pathology,
Healthcare #NA931V) and anti-rabbit IgG HRP (GE Healthcare #NA934V). For HKU LKS Faculty of Medicine, Queen Mary Hospital, Pokfulam, Hong Kong.
flow cytometry, the following antibodies against human surface proteins were Gut pathology was graded in at least ten fields of vision per tissue sample slide
used: anti-CD3 Pacific Blue (PB; clone UCHT1, BD Biosciences #558117), under light microscopy according to refs. 67,68, and the median was used for
anti-γ​δ​-TCR APC (clone B1, BD Biosciences #555718), anti-Vδ​2 PE (clone analysis. Briefly, as shown in Fig. 3c, the small intestinal pathology was graded as
B6, BD Biosciences #555739), anti-CD4 PerCP-Cy5.5 (clone OKT4, Biolegend follows: Gruenhagen’s space and vacuolization, grade 1 (black arrows); epithelium
#317428), anti-CD8a PE/Cy7 (clone SK1, eBioscience #25-0087-42), anti-CD8 detachment, grade 2 (asterisk); mucosal ulceration, grade 3; disintegration of the
Alexa Fluor 700 (clone RPA-T8, BD Biosciences #561453) anti-CD11c APC (clone LP, grade 4 (red arrow). Intact epithelial villi are graded 0 (as seen in mock).
3.9, eBioscience #17-0116-42), anti-CD19 PE (clone 1D3, eBioscience #12-0193-
82), anti-CD14 PE/Cy7 (eBioscience), anti-CD56 APC (clone HCD56, Biolegend Fluorescent IHC. Tissue sections of small intestines from humanized mice
#318310), anti-CD69 APC/Cy7 (clone FN50, Biolegend #310914), anti-CD103 were immunostained as previously described32, with the primary antibodies
Alexa Fluor 488 (clone Ber-ACT8, Biolegend), anti-CCR9 PerCP/Cy5.5 anti-human Vδ​2-TCR (clone 15D, Thermo Fisher Scientific), anti-Δ​42PD1 (clone
(clone L053E8, Biolegend) and anti-HLA-DR PerCP/Cy5.5 (clone LN3, CH34 or CH101), anti-TLR4 (Abcam) and anti-IL-6 (clone M-19, Santa Cruz),
eBioscience #25-9956-42). Relevant matching isotype controls were used. followed by the secondary antibodies anti-mouse IgG1-Alexa Fluor 488, mouse
Two mouse monoclonal antibodies (clones CH34 and CH101) against Δ​42PD1 IgG1-Alexa Fluor 568, anti-mouse IgG2a-Alexa Fluor 488 and anti-mouse IgG2b-
were generated as described in ref. 25 (by using a Δ​42PD1 expression plasmid DNA Alexa Fluor 568 (Invitrogen), counterstained using Hoechst 33258, and mounted
to prime BALB/c mice followed by a boost with soluble Δ​42PD1 protein). Briefly, using fluorescence mounting medium (DAKO). Signals were captured using
serum samples were collected at week 10 post-vaccination and analysed for anti-sΔ​ a Carl Zeiss LSM700 confocal microscope (×​20 and ×​40 objective lenses at 1.3
42PD1 antibodies by ELISA. Splenocytes from mice showing high antibody titre aperture) and acquired using ZEN software at room temperature. ImageJ software
were used to fuse with SP2/0-Ag14 myeloma cells. The hybridoma cells, which (http://imagej.nih.gov/ij/) was used for analysis and to merge images.
produced Δ​42PD1-specific monoclonal antibody (no cross reaction with wild-
type PD-1), were purified twice using a limited-dilution subcloning technique. Purification of recombinant proteins. HEK-293F cells (Freestyle 293-F cells, Life
The positive clones (CH34 and CH101) were confirmed by flow cytometry using Technologies, mycoplasma negative) were transfected with pVAX-sPD1-Fc, pVAX-sΔ​
Δ​42PD1-expressing 293T cells. These antibodies are capable of distinguishing 42PD1-Fc or pVAX-sΔ​42PD1-Flag plasmids using polyethylenimine (PEI; Sigma).
between wild-type PD-1 and Δ​42PD1 by flow cytometry25 and the CH101 After 24 h, cells were 1:1 diluted with pre-warmed medium supplemented with
clone has a blocking effect against Δ​42PD1-induced signalling (Supplementary valproic acid (adjusted to a final concentration of 2.2 mM in the cell culture). Half
Fig. 14). Anti-Δ​42PD1 antibodies were labelled using an Antibody Labeling of the cell suspension was collected on day 5 post-transfection and an equivalent
Kit (Invitrogen). Clone CH34 was labelled with Alexa Fluor (AF) 488 and clone volume of pre-warmed medium was supplemented to the culture. Four days after the
CH101 with Alexa Fluor 647. first collection, cells in the suspension were collected. The harvested cell suspension
was centrifuged at 3,220g for 10 min at 4 °C. Supernatant was then aspirated and
Viruses. HIV-1NL4-3 (X4-tropic) and HIV-1Henan (R5-tropic) virus stocks and 0.2 μ​m-filtered. Supernatant containing sPD1fc or sΔ​42PD1fc proteins was purified
pseudoviruses ADA and VSV-G were prepared by human PBMC expansion and by passing through a column with protein G agarose beads (Sigma), while that
used for infection at 50 ng p24 (determined using ZeptoMatrix p24 ELISA kit) for containing sΔ​42PD1flag was passed through a column with anti-FLAG M2 affinity
5 ×​  106 cells per ml RPMI 1640 +​ 10% FBS for 2 h, as previously described61. Human gel (Sigma). The columns were appropriately washed and the purified protein was
cytomegalovirus strain AD169 was prepared as previously described62. Freshly then eluted, neutralized and concentrated using Amicon Ultra-4 Centrifugal Filter
isolated PBMCs without PHA or IL-2 stimulation were used for infection. Units (50K for sΔ​42PD1fc and 10K for sΔ​42PD1flag, Merck Millipore). An endotoxin
removal step with EndoTrap red (Hyglos) was then performed for the proteins
RT–PCR and TCR-γ analysis. PBMCs were infected with HIV-1NL4-3 for 3 days according to the manufacturer’s instructions. Protein was eluted and concentrated
before sorted for Vδ​2+ cells using anti-phycoerythrin (PE) microbeads kit after using the above centrifugal filter units, its concentration determined by BCA protein
staining with anti-Vδ​2 PE antibody, or further stained with anti-Δ​42PD1 antibody assay kit (Thermo Fisher Scientific) and then stored at –80 °C in multiple aliquots.
and FACSorted for Δ​42PD1+ and Δ​42PD1− cells using a BD AriaIII instrument.
RNA was then extracted with RNAiso Plus (Takara) and cDNA generated by Analysis of cytokine expression. Endotoxin contamination was not detected
PrimeScript RT kit (Takara). PCR was performed using the following primers63,64 in all protein preparations tested using an E-TOXATE kit (<​0.03 endotoxin
with PrimeStar HS DNA Polymerase (Takara): Vγ​I, 5′​-GAAGATCTAGACAGGC units ml–1, Sigma-Aldrich) or Pierce LAL Chromogenic Endotoxin Quantitation
CGACTGGGTCATCTGC-3′​; Vγ​II, 5′​-GAAGATCTAGACAGCCCGCCTG Kit (Thermo Fisher Scientific). LPS (100 ng ml–1) were also used for treatment.
GAATGTGTGG-3′​; Vγ​III, 5′​-AGCATGGGTAAGACAAGCAAA-3′​; Jγ​1.1/2.1, MoDCs were treated with LPS or recombinant human sΔ​42PD1fc proteins as
5′​-GAAGATCTAGACTTACCAGGTGAAGTTACTATAAGC-3′​; Jγ​1.2, AAGAA described previously24. For inhibition experiments, MoDCs were pre-treated with
AACTTACCTGTAATGATAAGC-3′​; Jγ​1.3/2.3, 5′​-CTAGTCTAGACCGTA polyclonal blocking antibodies against TLR2, TLR4 and TLR6 (InvivoGen) for
TATGCACAAAGCCAGATC-3′​; hu-β​-actin forward, 5′​-GGATGATGATATC 30 min, or the TLR4-specific inhibitor CLI-095 (InvivoGen) for 1 h. After 6 h,
GCCGCG-3′​, reverse, 5′​-GGATAGCAACGTACATGGCTGGG-3′​. PCR products qRT–PCR was performed for the human genes tnfa, il6, il1b and ifna using specific
were electrophoresed on 1% (wt/vol) agarose gel, extracted using a QIAquick primers. HIV-1 infection of freshly unstimulated human PBMCs was performed
gel extraction kit (Qiagen) or MEGA-spin(TM) Agarose Gel DNA Extraction for 24 h before supernatants were collected for cytokine analysis. Cytokine release
Kit (Intron) and sent for Sanger sequencing. The original gel photos are provided was measured using the Human Th1/Th2/Th9/Th17/Th22 13plex, Human
in Supplementary Fig. 22. For TCR-Vγ​II/Jγ​1.2 sequence analysis, sequences Inflammation 20plex Ready-to-Use FlowCytomix Multiplex kit and analysed using
were aligned to the best-matched germline V and J gene (TRGV9*01: FlowCytomixPro software (version 3) or Human IL-6 Quantikine ELISA Kit (R&D
accession no. X07205; and TRGJP*01: accession no. M12950) on the Systems) according to the manufacturer’s instructions (eBioscience).
IMGT/V-QUEST database65 (www.imgt.org).
Co-IP and western blotting. Recombinant proteins sΔ​42PD1fc and sPD1fc were
Humanized mice experiments. NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice (NSG, generated as described above. Purified sΔ​42PD1fc (0.5 μ​g) was mixed with 0.5 μ​g
Jackson Laboratories) were bred and housed in the minimal disease area in of commercial carrier-free purified TLR4-His or TLR4/MD2-His proteins
HKU Laboratory Animal Unit. All animal experiments were approved by (R&D Systems), or with equal amounts of LBP and HMGB1 (R&D Systems)
the Committee on the Use of Live Animals in Teaching and Research at the in 500 μ​l IP buffer (25 mM Tris, 150 mM NaCl, pH 7.2) with gentle rocking
Laboratory Animal Unit of the University of Hong Kong. Mice were randomly overnight at 4 °C. Following addition of anti-rabbit IgG agarose beads for 2 h,
chosen with no blinding, and gender was chosen between groups randomly. Four- protein mixture was centrifuged at 5,000 r.p.m. for 10 min before being washed
to six-week-old mice were reconstituted with 1 ×​  107 fresh healthy human PBMCs in IP buffer and centrifuged again. Bead-bound proteins were eluted using IgG
by intraperitoneal (i.p.) injection in 0.5 ml RPMI. At day 10, blood was drawn elution buffer and neutralized with 1 M Tris (pH 9.0) buffer (10 μ​l per 100 μ​l
to assess the existence of human lymphocytes by red blood cell lysis (BD Pharm protein eluate). Proteins with loading dye were then boiled for 5 min and loaded

Nature Microbiology | VOL 2 | OCTOBER 2017 | 1389–1402 | www.nature.com/naturemicrobiology 1399


© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Articles NaTuRe MICRObIOLOgy
onto 4–10% SDS–PAGE gel for electrophoresis and transferred to polyvinylidene FRET. 293T cells were co-transfected with plasmids encoding sΔ​42PD1-CFP
difluoride (PVDF) membrane (Millipore) using a Trans-Blot SD Semi-Dry (acceptor) and TLR4-YFP (donor) or TLR9-YFP (CFP and YFP encoding plasmids
Transfer Cell (Bio-Rad). Membrane was blocked using PBS-T with 0.5% bovine were provided by K. H. Kok, University of Hong Kong). The 293T cells were
serum albumin (BSA) and 5% blotting-grade blocker (Bio-Rad) for 1 h, before mycosplasma negative. Settings were assisted by single transfections. Photobleaching
incubation with primary mouse anti-His (R&D Systems) or anti-Δ​42PD1 (clone was carried out using an LSM510 confocal microscope and detection of FRET in
101) antibody, followed by anti-mouse HRP-conjugated secondary antibody (GE the YFP channel of the acceptor with the following settings: bleach for 5 s, repeat 5 s,
Healthcare). Membrane blot was developed using Superbright Femto enhanced 20 iterations. FRET analysis was performed as previously described69, and regions
chemiluminescence (ECL) substrate (Thermo Fisher Scientific) and signals were of interest were identified in multiple areas without cells and areas inside the cells
captured by an Amersham Hyperfilm enhanced chemiluminescence (ECL) (GE without photobleaching and from areas with photobleaching to calculate the FRET
Healthcare) and Developer/Fixative solutions (Kodak) or scanned with a LICOR efficiency. ImageJ and the PixFRET plug-in were used to analyse regions of FRET
C-DiGit scanner. Negative data for sPD1fc are not shown. The original photos are and increase in pixel intensity70.
provided in Supplementary Fig. 21.
SPR. Recombinant purified sΔ​42PD1fc or sPD1fc protein was immobilized
DC co-culture experiments. MoDCs were generated as above (see section ‘PBMC, on a CM5 chip by amine coupling for SPR in a Biacore X100 instrument (GE
MoDC and transfection’) and used at 5 days after culture with GM-CSF/IL-4 and Healthcare). Commercial TLR4/MD2-His complex or PD-L1 purified proteins
co-cultured with CD3+Vδ​2+ cells obtained from FACSorting of HIV-1-infected (R&D Systems) was used to flow over the chip at different concentrations. LPS
PBMC cultures using the BDAriaIII instrument. After sorting, cells were used (Sigma) was also examined against coated sΔ​42PD1fc. Analysis was performed on
immediately in RPMI +​10% FBS medium. Cells were pre-treated with blocking Biacore X100 evaluation software (GE Healthcare).
antibodies against TLR4 (pAb, InvivoGen) or Δ​42PD1 (CH101) or isotype IgG1
(DAKO) for 30 min before co-culture at a 1:1 ratio in a U-bottomed 96-well culture Fluorescence binding assay. Recombinant proteins of soluble domains of ∆​42PD1
plate (Greiner Bio-One), or separated in a Transwell-96 culture plate (Corning). were expressed in Escherichia coli and purified as described previously71. Proteins
Cytokine production was assessed by qRT–PCR and IL-6 in the supernatant by were allowed to refold overnight and purified by size-exclusion chromatography.
ELISA (Biolegend) after 24 h of co-culture. Purified recombinant TLR4/MD2 and PD-L1 were purchased from R&D Systems
and conjugated with Alexa Fluor 594 succinimidyl ester (Thermo Fisher Scientific)
Generation of MD2 mutants. MD2-F126A or -C95Y mutation was introduced according to the manufacturer’s manual. The labelled protein was then purified
into pEFBOS/humanMD2-FlagHis expression plasmid (provided by K. Miyake, from unlabelled dyes using a spin column. The binding experiment was performed
University of Toyko) by site-directed mutagenesis using the QuikChange II Site- by titrating increasing concentrations of soluble ∆​42PD1 with 10 nM labelled
Directed Mutagenesis Kit (Agilent Technologies) according to the manufacturer’s TLR4/MD2 or PD-L1 protein in PBS buffer. The change in fluorescence was
protocol. The primer pair for mutagenesis of human MD2-F126A mutant measured and the binding isotherm was fitted with the Hill equation using
comprised 5­′​-­CA­AC­AC­AT­TT­GT­AT­TT­TC­CC­TT­AG­GC­AA­TT­TT­AT­TC­CC­TT­ IgorPro (WaveMetrics).
GAAGGAGAATGATATTGTT-3′​and 5′​-­AACAATATCATTCTCCTTCAAGG
GAATAAAATTGCCTAAGGGAAAATACAAATGTGTTG-3′​, both of which Molecular docking of PD1 to TLR4/MD2. Protein–protein docking was
were designed via the web-based QuikChange Primer Design Program available performed using the ClusPro web server with default parameters72. Briefly, the
online at http://www.genomics.agilent.com/primerDesignProgram.jsp. structure of PD1 (5GGS) was docked with the TLR4/MD2 complex (3FXI) and
The primer pair for mutagenesis of human MD2-C95Y mutant comprised the docking model with the best cluster scores calculated by balanced coefficients
5′​-GCAAAGAAGTTATTTACCGAGGATCTGATGACGATTAC-3′​ and was selected. The model was visualized and analysed with Discovery Studio
5′​-GTAATCGTCATCAGATCCTCGGTAAATAACTTCTTTGC-3′​, as described visualizer (Accelrys).
previously39. Introduction of the mutation was confirmed by DNA sequencing
using the forward primer of MD2 (5′​-GCATTTGTAAAGCTTTGGAGA-3′​) at
Statistics. All statistical analyses were performed using a paired two-tailed
Beijing Genomics Institute (Hong Kong) (Supplementary Fig. 15a). Detection
Student’s t-test to calculate P values for equal numbers between groups. P <​  0.05
of the expression of wild-type or mutant MD2 by flow cytometry in transfected
was considered statistically significant. Data are presented as mean ±​ s.e.m. of at
cells was performed using the anti-Flag antibody (clone M2, Sigma).
least three independent experiments unless indicated in the figure legends. Linear
regression was performed for correlation analysis with data from acute HIV
Reporter cell line experiments. HEKBlue-hTLR4 cells (InvivoGen) were obtained patients with R2 and P values depicted in the graphs. Data comparisons between
commercially and tested by LPS as a positive control to authenticate the cell line. acute HIV and chronic HIV patient samples, and healthy controls, or groups of
HEKBlue-hTLR4 cells were first transfected with psiRNA-hCD14 (InvivoGen) sub- mice were analysed using the Mann–Whitney U-test.
cultured in Zeocin-selection medium, then transiently transfected with MD2 siRNAs
(ID s24262 and s24264, Silencer Select pre-designed siRNA; Thermo Fisher Scientific)
Data availability. The data that support the findings of this study are available
or Silencer Select negative control #1 siRNA (Thermo Fisher Scientific). After 48 h,
from the corresponding author upon reasonable request.
the effect of downregulation was determined by flow cytometry using anti-Flag (for
MD2) and anti-CD14 antibodies. The 293A cells were co-transfected with NF-kB
Luciferase reporter expression vector (Clontech, provided by K. H. Kok, University Ethical approvals. Informed consent was obtained for all participants and
of Hong Kong), pUNO1-hTLR4-HA3x (InvivoGen), pEFBOS/humanMD2 and its reviewed and approved by the Beijing You-An Hospital Research Ethics
F126A mutant. The 293A cells are mycoplasma negative. Expression of TLR4 and Committee and Ethics Review Committee, Shenzhen Third People’s Hospital. All
MD2 was determined on 293A cells by flow cytometry. For co-culture experiments, animal experiments were approved by the Committee on the Use of Live Animals
~2.5 ×​ 104 transfected HEKBlue-hTLR4 cells or ~5 ×​ 104 transiently transfected 293A in Teaching and Research at the Laboratory Animal Unit of the University of
cells were co-cultured 1:1 with 293A cells (negative or stably transfected with PD1 Hong Kong. Use of healthy human PBMCs was approved by the Institutional
and Δ​42PD1) or γ​δ​T cells (untreated or treated with IL-12 (4 ng ml–1) and IL-15 Review Board of the University of Hong Kong/Healthy Authority Hong Kong
(20 ng ml–1) (Peprotech) for 5 days, then pre-treated with anti-Δ​42PD1 antibody West Cluster.
(clone CH101) or IgG1 isotype (DAKO)), or treated with sΔ​42PD1fc or sΔ​42PD1flag
purified protein (2 μ​g ml–1), LPS (100 ng ml–1) or TNF-α​ (10 ng ml–1). Results were Received: 24 October 2016; Accepted: 5 July 2017;
determined using the VICTOR3 1420 Multilabel Counter (PerkinElmer) after 12 h Published online: 14 August 2017
incubation for SEAP reporter response (absorbance at 620 nm) from HEKBlue-
hTLR4 cells, or after 24 h for 293A cells for luciferase activity. For luciferase, cells
were washed with PBS and then lysed. The lysates were used in a Luciferase Assay References
System (Promega) and CellTiter-Glo Luminescent Cell Viability Assay (Promega) to 1. Vantourout, P. & Hayday, A. Six-of-the-best: unique contributions
normalize for cell number differences. of γ​δ​T cells to immunology. Nat. Rev. Immunol. 13, 88–100 (2013).
2. McCarthy, N. E. et al. Proinflammatory Vδ​2+​T cells populate the human
Plasma endotoxin, oxidized phospholipid, and biomarker ELISA. Healthy human intestinal mucosa and enhance IFN-γ​production by colonic α​β​T cells.
plasma or HIV-1 patient plasma was assessed for endotoxin level using a Pierce LAL J. Immunol. 191, 2752–2763 (2013).
Chromogenic Endotoxin Quantification kit (Thermo Fisher Scientific). Plasma 3. McCarthy, N. E. et al. Azathioprine therapy selectively ablates human Vδ​2(+​)
was centrifuged at 16,000g for 5 min, then 5 μ​l was diluted 1:10 with endotoxin- T cells in Crohn’s disease. J. Clin. Invest. 125, 3215–3225 (2015).
free water and incubated at 75 °C for 15 min, then assayed according to the 4. Brandes, M., Willimann, K. & Moser, B. Professional antigen-presentation
manufacturer’s instructions. All recombinant proteins used contained <​0.1 EU ml–1. function by human γ​δ​T cells. Science 309, 264–268 (2005).
Acute and chronic HIV-1 patient plasma was assessed for the biomarkers soluble 5. Bjarnason, I. et al. Intestinal inflammation, ileal structure and function in
CD14, FABP2 and haptoglobin using Quantikine ELISA kits (R&D Systems), as well HIV. AIDS 10, 1385–1391 (1996).
as oxidized phospholipid using the Human Oxidized LDL ELISA kit (oxLDL, Cell 6. Batman, P. A. et al. Jejunal enteropathy associated with human
Biolabs) according to the manufacturer’s instructions. Recombinant proteins showed immunodeficiency virus infection: quantitative histology. J. Clin. Pathol. 42,
negative results for oxLDL (below the detection limit). 275–281 (1989).

1400 Nature Microbiology | VOL 2 | OCTOBER 2017 | 1389–1402 | www.nature.com/naturemicrobiology

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
NaTuRe MICRObIOLOgy Articles
7. Heise, C. et al. Primary acute simian immunodeficiency virus infection of 36. Erridge, C. Endogenous ligands of TLR2 and TLR4: agonists or assistants?
intestinal lymphoid tissue is associated with gastrointestinal dysfunction. J. Leukoc. Biol. 87, 989–999 (2010).
J. Infect. Dis. 169, 1116–1120 (1994). 37. Teghanemt, A. et al. Novel roles in human MD-2 of phenylalanines 121
8. Kotler, D. P., Reka, S. & Clayton, F. Intestinal mucosal inflammation and 126 and tyrosine 131 in activation of Toll-like receptor 4 by endotoxin.
associated with human immunodeficiency virus infection. Dig. Dis. Sci. 38, J. Biol. Chem. 283, 1257–1266 (2008).
1119–1127 (1993). 38. Yu, L. et al. NMR studies of hexaacylated endotoxin bound to wild-type and
9. McGowan, I. et al. Increased HIV-1 mucosal replication is associated with F126A mutant MD-2 and MD-2.TLR4 ectodomain complexes. J. Biol. Chem.
generalized mucosal cytokine activation. J. Acquir. Immune Defic. Syndr. 37, 287, 16346–16355 (2012).
1228–1236 (2004). 39. Schromm, A. B. et al. Molecular genetic analysis of an endotoxin
10. Olsson, J. et al. Human immunodeficiency virus type 1 infection is associated nonresponder mutant cell line: a point mutation in a conserved region
with significant mucosal inflammation characterized by increased expression of MD-2 abolishes endotoxin-induced signaling. J. Exp. Med. 194,
of CCR5, CXCR4, and β​-chemokines. J. Infect. Dis. 182, 1625–1635 (2000). 79–88 (2001).
11. Harouse, J. M. et al. Distinct pathogenic sequela in rhesus macaques infected 40. Gruber, A. et al. Structural model of MD-2 and functional role of its basic
with CCR5 or CXCR4 utilizing SHIVs. Science 284, 816–819 (1999). amino acid clusters involved in cellular lipopolysaccharide recognition.
12. Autran, B. et al. T cell receptor γ​/δ​+ lymphocyte subsets during HIV J. Biol. Chem. 279, 28475–28482 (2004).
infection. Clin. Exp. Immunol. 75, 206–210 (1989). 41. Apetoh, L. et al. Toll-like receptor 4-dependent contribution of the immune
13. Gan, Y. H., Pauza, C. D. & Malkovsky, M. γ​δ​T cells in rhesus monkeys system to anticancer chemotherapy and radiotherapy. Nat. Med. 13,
and their response to simian immunodeficiency virus (SIV) infection. 1050–1059 (2007).
Clin. Exp. Immunol. 102, 251–255 (1995). 42. Andersson, U. & Tracey, K. J. HMGB1 is a therapeutic target for sterile
14. Poles, M. A. et al. Human immunodeficiency virus type 1 induces persistent inflammation and infection. Annu. Rev. Immunol. 29, 139–162 (2011).
changes in mucosal and blood T cells despite suppressive therapy. J. Virol. 77, 43. Pal, D. et al. Fetuin-A acts as an endogenous ligand of TLR4 to promote
10456–10467 (2003). lipid-induced insulin resistance. Nat. Med. 18, 1279–1285 (2012).
15. Glatzel, A. et al. Patterns of chemokine receptor expression on peripheral 44. Schmidt, M. et al. Crucial role for human Toll-like receptor 4 in the
blood γ​δ​T lymphocytes: strong expression of CCR5 is a selective feature of development of contact allergy to nickel. Nat. Immunol. 11, 814–819 (2010).
Vδ​2/Vγ​9 γ​δ​T cells. J. Immunol. 168, 4920–4929 (2002). 45. Conti, L. et al. Reciprocal activating interaction between dendritic cells and
16. Li, H. & Pauza, C. D. HIV envelope-mediated, CCR5/α​4β​7-dependent killing pamidronate-stimulated γ​δ​T cells: role of CD86 and inflammatory cytokines.
of CD4-negative γ​δ​T cells which are lost during progression to AIDS. Blood J. Immunol. 174, 252–260 (2005).
118, 5824–5831 (2011). 46. Scotet, E. et al. Bridging innate and adaptive immunity through γ​δ​
17. Hermier, F. et al. Decreased blood TcR γ​δ​+ lymphocytes in AIDS and T-dendritic cell crosstalk. Front. Biosci. 13, 6872–6885 (2008).
p24-antigenemic HIV-1-infected patients. Clin. Immunol. Immunopathol. 69, 47. Mancek-Keber, M. & Jerala, R. Postulates for validating TLR4 agonists.
248–250 (1993). Eur. J. Immunol. 45, 356–370 (2015).
18. Kosub, D. A. et al. γ​δ​T-cell functional responses differ after pathogenic 48. Park, B. S. et al. The structural basis of lipopolysaccharide recognition by the
human immunodeficiency virus and nonpathogenic simian TLR4-MD-2 complex. Nature 458, 1191–1195 (2009).
immunodeficiency virus infections. J. Virol. 82, 1155–1165 (2008). 49. Rakoff-Nahoum, S. et al. Recognition of commensal microflora by Toll-like
19. Li, Z. et al. γ​δ​T cells are involved in acute HIV infection and associated with receptors is required for intestinal homeostasis. Cell 118, 229–241 (2004).
AIDS progression. PLoS ONE 9, e106064 (2014). 50. Chen, Z. et al. CD4+ lymphocytopenia in acute infection of Asian macaques
20. Poccia, F. et al. Peripheral Vγ​9/Vδ​2 T cell deletion and anergy to nonpeptidic by a vaginally transmissible subtype-C, CCR5-tropic simian/human
mycobacterial antigens in asymptomatic HIV-1-infected persons. J. Immunol. immunodeficiency virus (SHIV). J. Acquir. Immune Defic. Syndr. 30,
157, 449–461 (1996). 133–145 (2002).
21. Wallace, M. et al. Functional γ​δ​T-lymphocyte defect associated with human 51. Mattapallil, J. J. et al. Massive infection and loss of memory CD4+ T cells in
immunodeficiency virus infections. Mol. Med. 3, 60–71 (1997). multiple tissues during acute SIV infection. Nature 434, 1093–1097 (2005).
22. Cardone, M. et al. HIV-1-induced impairment of dendritic cell cross-talk 52. Raffatellu, M. et al. Simian immunodeficiency virus-induced mucosal
with γ​δ​T lymphocytes. J. Virol. 89, 4798–4808 (2015). interleukin-17 deficiency promotes Salmonella dissemination from the gut.
23. Cimini, E. et al. Primary and chronic HIV infection differently modulates Nat. Med. 14, 421–428 (2008).
mucosal Vδ​1 and Vδ​2 T-cells differentiation profile and effector functions. 53. Mancek-Keber, M. et al. Toll-like receptor 4 senses oxidative stress mediated
PLoS ONE 10, e0129771 (2015). by the oxidation of phospholipids in extracellular vesicles. Sci. Signal. 8,
24. Zhou, J. et al. Potentiating functional antigen-specific CD8+ T cell immunity ra60 (2015).
by a novel PD1 isoform-based fusion DNA vaccine. Mol. Ther. 21, 54. Zidar, D. A. et al. Oxidized LDL levels are increased in HIV infection and
1445–1455 (2013). may drive monocyte activation. J. Acquir. Immune Defic. Syndr. 69,
25. Cheng, L. et al. Monoclonal antibodies specific to human Δ​42PD1: a novel 154–160 (2015).
immunoregulator potentially involved in HIV-1 and tumor pathogenesis. 55. Brandes, M. et al. Cross-presenting human γ​δ​T cells induce robust CD8+ α​β​
MAbs 7, 620–629 (2015). T cell responses. Proc. Natl Acad. Sci. USA 106, 2307–2312 (2009).
26. Stacey, A. R. et al. Induction of a striking systemic cytokine cascade prior to 56. Poonia, B. et al. γ​δ​T cells are ADCC effectors in elite HIV controllers.
peak viremia in acute human immunodeficiency virus type 1 infection, in Retrovirology 7(Suppl 1), O7 (2010).
contrast to more modest and delayed responses in acute hepatitis B and C 57. González-Navajas, J. M. et al. TLR4 signaling in effector CD4+ T cells
virus infections. J. Virol. 83, 3719–3733 (2009). regulates TCR activation and experimental colitis in mice. J. Clin. Invest. 120,
27. Brenchley, J. M. et al. Microbial translocation is a cause of systemic 570–581 (2010).
immune activation in chronic HIV infection. Nat. Med. 12, 58. Jin, B. et al. The effects of TLR activation on T-cell development and
1365–1371 (2006). differentiation. Clin. Dev. Immunol. 2012, 836485 (2012).
28. García, V. E. et al. IL-15 enhances the response of human γ​δ​T cells to 59. Tu, W. et al. The aminobisphosphonate pamidronate controls influenza
nonpetide microbial antigens. J. Immunol. 160, 4322–4329 (1998). pathogenesis by expanding a γ​δ​T cell population in humanized mice.
29. Enders, P. J. et al. HIV-mediated γ​δ​T cell depletion is specific for Vγ​2+ cells J. Exp. Med. 208, 1511–1522 (2011).
expressing the Jγ​1.2 segment. AIDS Res. Hum. Retroviruses 19, 21–29 (2003). 60. Xiang, Z. et al. Targeted activation of human Vγ​9Vδ​2-T cells controls
30. Evans, P. S. et al. In vitro stimulation with a non-peptidic alkylphosphate Epstein–Barr virus-induced B cell lymphoproliferative disease. Cancer Cell 26,
expands cells expressing Vγ​2-Jγ​1.2/Vδ​2 T-cell receptors. Immunology 104, 565–576 (2014).
19–27 (2001). 61. Kang, Y. et al. CCR5 antagonist TD-0680 uses a novel mechanism for
31. Hebbeler, A. M. et al. Failure to restore the Vγ​2-Jγ​1.2 repertoire in enhanced potency against HIV-1 entry, cell-mediated infection, and a
HIV-infected men receiving highly active antiretroviral therapy (HAART). resistant variant. J. Biol. Chem. 287, 16499–16509 (2012).
Clin. Immunol. 128, 349–357 (2008). 62. Cheung, A. K. L. et al. The role of the human cytomegalovirus UL111A gene in
32. Wu, X. et al. Brain invasion by CD4+ T cells infected with a transmitted/ down-regulating CD4+ T-cell recognition of latently infected cells: implications
founder HIV-1 during acute stage in humanized mice. J. Neuroimmune for virus elimination during latency. Blood 114, 4128–4137 (2009).
Pharmacol. 11, 572–583 (2016). 63. Ghosh, J. K., Romanow, W. J. & Murre, C. Induction of a diverse T cell
33. Abreu, M. T., Fukata, M. & Arditi, M. TLR signaling in the gut in health and receptor γ​/δ​repertoire by the helix-loop-helix proteins E2A and HEB in
disease. J. Immunol. 174, 4453–4460 (2005). nonlymphoid cells. J. Exp. Med. 193, 769–776 (2001).
34. Balazs, A. B. et al. Vectored immunoprophylaxis protects humanized mice 64. van Dongen, J. J. et al. Design and standardization of PCR primers and
from mucosal HIV transmission. Nat. Med. 20, 296–300 (2014). protocols for detection of clonal immunoglobulin and T-cell receptor
35. Im, E. et al. Elevated lipopolysaccharide in the colon evokes intestinal gene recombinations in suspect lymphoproliferations: report of the
inflammation, aggravated in immune modulator-impaired mice. Am. J. BIOMED-2 Concerted Action BMH4-CT98-3936. Leukemia 17,
Physiol. Gastrointest. Liver Physiol. 303, G490–G497 (2012). 2257–2317 (2003).

Nature Microbiology | VOL 2 | OCTOBER 2017 | 1389–1402 | www.nature.com/naturemicrobiology 1401


© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Articles NaTuRe MICRObIOLOgy
65. Lefranc, M. P. et al. IMGT®, the international ImMunoGeneTics information by research grants from the Hong Kong Research Grant Council (RGC: HKU5/
system® 25 years on. Nucleic Acids Res. 43, D413–D422 (2015). CRF/13G, RGC17103514, RGC17122915 and A-HKU709/14 to Z.C.); the Health and
66. Weigmann, B. et al. Isolation and subsequent analysis of murine lamina propria Medical Research Fund (HMRF: 14130582 to Z.C., 15140372 to A.K.L.C.); the San-
mononuclear cells from colonic tissue. Nat. Protoc. 2, 2307–2311 (2007). Ming Project of Medicine in Shenzhen (to Z.C. and H.Wa.); the National Science and
67. Erben, U. et al. A guide to histomorphological evaluation of intestinal Technology Major Project (2012ZX10001-009-001-001 to Z.C., 2012ZX1000-1006-
inflammation in mouse models. Int. J. Clin. Exp. Pathol. 7, 001-009 to H.S.) Beijing Key Laboratory of HIV/AIDS Research (BZ0089 to H.Wu)
4557–4576 (2014). and Beijing Municipal of Science and Technology Major Project (D161100000416003
68. Feinman, R. et al. HIF-1 mediates pathogenic inflammatory responses to to H.Wu) and the University Development Fund of the University of Hong Kong and
intestinal ischemia–reperfusion injury. Am. J. Physiol. Gastrointest. Liver Li Ka Shing Faculty of Medicine Matching Fund to AIDS Institute.
Physiol. 299, G833–G843 (2010).
69. Karpova, T. & McNally, J. G. Detecting protein–protein interactions with
CFP-YFP FRET by acceptor photobleaching. Curr. Protoc. Cytom. 12,
Author contributions
A.K.L.C. and Z.C. designed experiments, analysed data and wrote the manuscript.
12.7.1–12.7.11 (2007).
A.K.L.C., Y.H., H.-y.K., M.C., Y.M., X.W., K.-s.L., H.-k.K, T.C.K.L., J.Z. and B.K.L.
70. Feige, J. N. et al. PixFRET, an ImageJ plug-in for FRET calculation that can
performed experiments. J.L. and L.C. generated the Δ​42PD1-specific antibodies. Q.P.,
accommodate variations in spectral bleed-throughs. Microsc. Res. Tech. 68, X.L., M.A., H.Wa., H.S., B.Z. and H.Wu provided HIV patient samples. A.X. and K.-Y.Y.
51–58 (2005). provided critical comments and materials.
71. Nicoludis, J. M. et al. Structure and sequence analyses of clustered
protocadherins reveal antiparallel interactions that mediate homophilic
specificity. Structure 23, 2087–2098 (2015). Competing interests
72. Kozakov, D. et al. The ClusPro web server for protein–protein docking. Nat. The authors declare no competing financial interests.
Protoc. 12, 255–278 (2017).
Additional information
Acknowledgements Supplementary information is available for this paper at doi:10.1038/s41564-017-0006-5.
The authors thank K. Miyake for providing the MD2 plasmid, K.H. Kok for CFP, Reprints and permissions information is available at www.nature.com/reprints .
YFP and NF-κ​B-luciferase plasmids and C. Cheng-Mayer for critical discussions.
The authors thank L. Liu for technical advice with immunohistochemical staining. Correspondence and requests for materials should be addressed to Z.C.
The authors acknowledge the Faculty Core Facility of the LKS Faculty of Medicine, Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in
HKU, for technical assistance with confocal microscopy. This work was supported published maps and institutional affiliations.

1402 Nature Microbiology | VOL 2 | OCTOBER 2017 | 1389–1402 | www.nature.com/naturemicrobiology

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
nature research | life sciences reporting summary
Corresponding Author: Zhiwei Chen

Date: Jun 27, 2017

Life Sciences Reporting Summary


Nature Research wishes to improve the reproducibility of the work we publish. This form is published with all life science papers and is intended to
promote consistency and transparency in reporting. All life sciences submissions use this form; while some list items might not apply to an individual
manuscript, all fields must be completed for clarity.
For further information on the points included in this form, see Reporting Life Sciences Research. For further information on Nature Research policies,
including our data availability policy, see Authors & Referees and the Editorial Policy Checklist.

` Experimental design
1. Sample size
Describe how sample size was determined. At least 3 replicate experiments using individual donor cells were
performed to achieve Student t-test statistics. Acute patient cases are rare
and we were able to obtained 57 individual samples. With at least 16
samples comparing between acute and chronic patients, a minimum
statistical power of 80% can be achieved. Mann-Whitney test was
performed for the patient data. Linear regression was performed for
patient plasma data. As indicated in Statistics of the Materials and
Methods.
2. Data exclusions
Describe any data exclusions. The criteria for HIV patients categorized as acute patients is reported in
the Materials and Methods section, under Study Subjects, page 29. There
were no inclusion/exclusion criteria for animals.
3. Replication
Describe whether the experimental findings were reliably reproduced. At least 3 independent replicate experiments were performed. Statistical
analyses were done to illustrate significance.
4. Randomization
Describe how samples/organisms/participants were allocated into Animals chosen for different groups, sequence of adoptive transfer and
experimental groups. sacrificed were randomized.
5. Blinding
Describe whether the investigators were blinded to group allocation No blinding was performed.
during data collection and/or analysis.
Note: all studies involving animals and/or human research participants must disclose whether blinding and randomization were used.

May 2017

1
6. Statistical parameters

nature research | life sciences reporting summary


For all figures and tables that use statistical methods, confirm that the following items are present in relevant figure legends (or the Methods
section if additional space is needed).

n/a Confirmed

The exact sample size (n) for each experimental group/condition, given as a discrete number and unit of measurement (animals, litters, cultures, etc.)
A description of how samples were collected, noting whether measurements were taken from distinct samples or whether the same sample
was measured repeatedly.

A statement indicating how many times each experiment was replicated

The statistical test(s) used and whether they are one- or two-sided (note: only common tests should be described solely by name; more
complex techniques should be described in the Methods section)
A description of any assumptions or corrections, such as an adjustment for multiple comparisons

The test results (e.g. p values) given as exact values whenever possible and with confidence intervals noted

A summary of the descriptive statistics, including central tendency (e.g. median, mean) and variation (e.g. standard deviation, interquartile range)

Clearly defined error bars

See the web collection on statistics for biologists for further resources and guidance.

` Software
Policy information about availability of computer code
7. Software
Describe the software used to analyze the data in this study. Flowjo, ImageJ with relevant plug-ins, Graphpad Prism, Excel, Biacore X100
software, FlowCytomixPro, ZEN.
For all studies, we encourage code deposition in a community repository (e.g. GitHub). Authors must make computer code available to editors and reviewers upon
request. The Nature Methods guidance for providing algorithms and software for publication may be useful for any submission.

` Materials and reagents


Policy information about availability of materials
8. Materials availability
Indicate whether there are restrictions on availability of unique Materials commercially available are indicated, in-house antibodies are
materials or if these materials are only available for distribution by a used where appropriate.
for-profit company.
9. Antibodies
Describe the antibodies used and how they were validated for use in Materials and Methods section, page 31.
the system under study (i.e. assay and species).
10. Eukaryotic cell lines
a. State the source of each eukaryotic cell line used. Cell line sources are found in Materials and Methods. Materials and
methods, under Cells and transfection, page 30, for HEKBlue-hTLR4; page
31 and 38, for FreeStyle 293F cells page 38. HEK-293A page 39

b. Describe the method of cell line authentication used. Cell lines were authenticated using the relevant stimulation and is
mentioned in the Methods section.

c. Report whether the cell lines were tested for mycoplasma The cell lines were ordered directly from the company and used. Some lab
contamination. cells were mycoplasma previously.

d. If any of the cell lines used in the paper are listed in the database Two cell lines was used in this study: HEKBlue-hTLR4 from Invivogen
of commonly misidentified cell lines maintained by ICLAC, (Materials and methods, page 31 and 38), not listed in the misidentified
provide a scientific rationale for their use. cell line database; FreeStyle 293-F cells (Thermo Fischer, previously
Invitrogen) were used for protein production (page 35); HEK293T (from
ATCC) were used for FRET assay (page 40); 293A cells were used for
transfection.
May 2017

2
` Animals and human research participants

nature research | life sciences reporting summary


Policy information about studies involving animals; when reporting animal research, follow the ARRIVE guidelines
11. Description of research animals
Provide details on animals and/or animal-derived materials used in Materials and methods, section Humanized mice experiments, page 34 -
the study. Strain and age are reported but not the gender. Equal number of male/
female were randomized into each group where possible.

Policy information about studies involving human research participants


12. Description of human research participants
Describe the covariate-relevant population characteristics of the Materials and methods, Study subjects, page 29-30; Cells and transfection,
human research participants. page 30. Consent was obtained for each patient as stated on page 29-30.

May 2017

3
nature research | flow cytometry reporting summary
Corresponding Author: Zhiwei Chen

Date: Jun 27, 2017

Flow Cytometry Reporting Summary


Form fields will expand as needed. Please do not leave fields blank.

` Data presentation
For all flow cytometry data, confirm that:
1. The axis labels state the marker and fluorochrome used (e.g. CD4-FITC).
2. The axis scales are clearly visible. Include numbers along axes only for bottom left plot of group (a 'group' is an analysis of identical markers).
3. All plots are contour plots with outliers or pseudocolor plots.
4. A numerical value for number of cells or percentage (with statistics) is provided.

` Methodological details
5. Describe the sample preparation. In vitro experiment PBMCs or subsequent purified using
microbeads, were obtained from healthy individuals,
immunostained with relevant antibodies and fixed with 2% PFA.
Patient PBMCs were resuscitated, immunostained, and fixed with
2% PFA.

6. Identify the instrument used for data collection. BD Aria III and BD FACSCalibur

7. Describe the software used to collect and analyze the flow Acquiring data using BD Diva, analysis performed with Flowjo.
cytometry data.

8. Describe the abundance of the relevant cell populations Purity was determined by relevant staining using flow cytometry,
within post-sort fractions. these are stated in the manuscript where appropriate.

9. Describe the gating strategy used. All samples are FSC-A and SSC-A gated, followed by FSC-A/FSC-H
gating to select singlet cells. Subsequent relevant gating was
conducted, shown in the main figures or supplementary.

Tick this box to confirm that a figure exemplifying the gating strategy is provided in the Supplementary Information.

June 2017

Вам также может понравиться