Download Multiple Origins of Virus Persistence during Natural

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Men who have sex with men blood donor controversy wikipedia , lookup

Transcript
Article
Multiple Origins of Virus Persistence during Natural
Control of HIV Infection
Graphical Abstract
Authors
Eli A. Boritz, Samuel Darko,
Luke Swaszek, ..., Stephen H. Hughes,
Steven G. Deeks, Daniel C. Douek
Correspondence
[email protected]
In Brief
HIV persistence in people who can
spontaneously control the infection
involves different mechanisms within
distinct anatomic and functional
compartments.
Highlights
d
In HIV controllers, both TFH and non-TFH lymph node CD4
T cells contain HIV
d
Lymph node viruses in both TFH and non-TFH have attributes
of active replication
d
Rare, recently infected cells that produce virus upon
stimulation circulate in blood
d
Archival proviruses predominant in clonally expanded blood
cells can be inducible
Boritz et al., 2016, Cell 166, 1004–1015
August 11, 2016 Published by Elsevier Inc.
http://dx.doi.org/10.1016/j.cell.2016.06.039
Accession Numbers
KX390978
KX394124
GSE83482
Article
Multiple Origins of Virus Persistence
during Natural Control of HIV Infection
Eli A. Boritz,1 Samuel Darko,1 Luke Swaszek,1 Gideon Wolf,1 David Wells,2 Xiaolin Wu,2 Amy R. Henry,1 Farida Laboune,1
Jianfei Hu,1 David Ambrozak,3 Marybeth S. Hughes,4 Rebecca Hoh,5 Joseph P. Casazza,3 Alexander Vostal,3
Daniel Bunis,1 Krystelle Nganou-Makamdop,1 James S. Lee,1 Stephen A. Migueles,6 Richard A. Koup,3 Mark Connors,6
Susan Moir,6 Timothy Schacker,8 Frank Maldarelli,7 Stephen H. Hughes,7 Steven G. Deeks,5 and Daniel C. Douek1,*
1Human
Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
3Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
4Thoracic and Gastrointestinal Oncology Branch, NCI, NIH, Bethesda, MD 20892, USA
5Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
6Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD 20892, USA
7HIV Dynamics and Replication Program, NCI, NIH, Frederick, MD 21702, USA
8Program in HIV Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
*Correspondence: [email protected]
http://dx.doi.org/10.1016/j.cell.2016.06.039
2Leidos
SUMMARY
Targeted HIV cure strategies require definition of the
mechanisms that maintain the virus. Here, we tracked
HIV replication and the persistence of infected CD4
T cells in individuals with natural virologic control by
sequencing viruses, T cell receptor genes, HIV integration sites, and cellular transcriptomes. Our results
revealed three mechanisms of HIV persistence operating within distinct anatomic and functional compartments. In lymph node, we detected viruses with
genetic and transcriptional attributes of active replication in both T follicular helper (TFH) cells and nonTFH memory cells. In blood, we detected inducible
proviruses of archival origin among highly differentiated, clonally expanded cells. Linking the lymph
node and blood was a small population of circulating
cells harboring inducible proviruses of recent origin.
Thus, HIV replication in lymphoid tissue, clonal expansion of infected cells, and recirculation of recently infected cells act together to maintain the virus in HIV
controllers despite effective antiviral immunity.
INTRODUCTION
During chronic HIV infection, multiple mechanisms combine to
ensure the persistence of virus-infected CD4 T cells despite innate
and adaptive antiviral responses. Foremost among these is
ongoing virus replication, which by itself can maintain an infected
CD4 T cell pool in the absence of antiretroviral therapy (ART) (Ho
et al., 1995). Even under ART, however, HIV-infected CD4 T cells
remain detectable in blood and lymphoid tissue. This may partly
reflect the persistence of memory cells that harbor replicationcompetent proviruses for long periods without expressing them
(Chun et al., 1997a, 1997b; Finzi et al., 1997, 1999; Hermankova
et al., 2003; Wong et al., 1997). That such cells can show a resting
memory phenotype has led to their identification as a latent reservoir and has spurred development of ‘‘shock and kill’’ HIV cure
strategies (Archin et al., 2012; Rasmussen et al., 2014; Routy
et al., 2012; Søgaard et al., 2015; Spivak et al., 2014). Nevertheless,
recent studies have also demonstrated clonal expansion of HIV-infected CD4 T cells under ART (Cohn et al., 2015; Maldarelli et al.,
2014; Simonetti et al., 2016; Wagner et al., 2014), raising questions
about the intrinsic properties of infected cells in this setting (Kim
and Siliciano, 2016). The further characterization of mechanisms
by which HIV-infected CD4 T cells persist under different conditions in vivo has thus emerged as a key research goal.
Here, we investigated the mechanisms that maintain HIV
in vivo through a detailed genetic analysis of virus sequences
from CD4 T cell subsets in blood and lymphoid tissue. We chose
people with natural control of the virus for this study. These individuals, termed HIV controllers, represent a rare group whose
HIV-specific immune responses enable them to control the virus
without ART (Migueles and Connors 2015; Walker and Yu, 2013).
Despite evidence of ongoing virus replication in HIV controllers
not receiving ART (Boufassa et al., 2014; Chun et al., 2013; Fukazawa et al., 2015; Hatano et al., 2013; Mens et al., 2010;
O’Connell et al., 2010; Salgado et al., 2010), prior work has
shown fewer CD4 T cells containing HIV DNA (Julg et al., 2010)
and replication-competent HIV (Blankson et al., 2007) in HIV controllers than in non-controllers. We reasoned that this would
allow us to sample more of the total virus population in these individuals and therefore obtain a comprehensive view of the infected CD4 T cell pool. Thus, we used sequencing not only to
help infer mechanisms of HIV persistence during natural virologic
control, but also to elucidate cellular processes that may maintain the virus both in HIV controllers and in non-controllers.
RESULTS
Distribution of HIV among Blood CD4 T Cell Subsets in
HIV Controllers
We enrolled 14 HIV controllers, defined by plasma HIV RNA
levels <1,000 copies/ml during chronic infection without ART,
1004 Cell 166, 1004–1015, August 11, 2016 Published by Elsevier Inc.
105
0.313
V907
103
S1270
102
S1349
S1475
101
S1492
100
TCM TTM TEM
104
TN
TCM TTM TEM
S1495
S1541
0.001 0.079
0.688 0.844
S1548
0.009
0.563
S1788
LIR01
103
LIR02
102
LIR03
101
LIR04
100
Non-controllers
-1
10
TN
TCM TTM TEM
TN
TCM TTM TEM
102
101
100
V016
V257
V621
V667
V723
V965
Figure 1. HIV DNA and RNA Levels in
Circulating CD4 T Cell Subsets from HIV
Controllers
(A) HIV DNA copies detected by FCA per 106 TN,
TCM, TTM, or TEM cell equivalents. The participant
color code at right applies to all figures. Horizontal
bars indicate median values in all figures.
(B) Numbers of HIV-infected TN, TCM, TTM, and TEM
cells per milliliter of blood, calculated by adjusting
values in (A) for CD4 counts and proportions of
CD4 T cells in each subset.
(C) Copies of unspliced (circles) and spliced (diamonds) HIV RNAs in TCM, TTM, and TEM cells from
HIV controllers, measured by qRT-PCR and
normalized to values in (A). Undetectable values
are plotted at the assay’s limit of detection (LOD)
with open symbols. Wilcoxon signed-rank test
p values are shown.
In (A) and (B), all Wilcoxon signed-rank test
p values for comparisons between TN and memory
subsets in HIV controllers are <0.0001, and MannWhitney p values for comparisons between HIV
controllers and non-controllers are <0.001 for all
cell subsets. In (C), all Wilcoxon signed-rank test
p values for comparisons of unspliced or spliced
RNA between subsets are >0.05.
See also Figures S1 and S2.
infected cell, we also found that the TEM
and TTM subsets accounted for most of
the infected CD4 T cells in blood in HIV
controllers (Figure 1B), with a median
72.6% of total HIV DNA copies in TEM
cells and a median 21.2% in TTM cells.
Although we hypothesized that this characteristic distribution in HIV controllers
reflected greater HIV expression and
replication within TEM- and TTM-like cells
in vivo, qRT-PCR revealed low or undetectable levels of unspliced and spliced HIV RNA in all CD4
T cell subsets from these individuals (Figure 1C). We also found
no correlation across individual HIV controllers between the level
of HIV genomic DNA in each subset and the absolute count of
each subset in blood (TCM, Spearman r = 0.200, p = 0.492;
TTM, r = 0.477, p = 0.087; TEM, r = 0.033, p = 0.916), as might
have been expected with ongoing, active replication and resulting cell depletion.
Open symbols
indicate LOD
10-1
U
ns
pl
Sp .
lic
ed
10-2
TCM
TTM
U
ns
pl
Sp .
lic
ed
HIV DNA copies/mL
0.035
HIV controllers
V912
B
Copies/HIV DNA copy
0.438 0.219
104
TN
C
0.025 0.268
U
ns
pl
Sp .
lic
ed
HIV DNA copies/106 cells
A
TEM
as well as six non-controllers with plasma HIV RNA levels
>10,000 copies/ml off ART (Table S1). Participants had been
documented HIV seropositive for a median of 15.5 years, with
a median of 18 years in the controller group (range 4–30) and
6 years in the non-controller group (range 2–29; Mann-Whitney
p = 0.1040 for controllers versus non-controllers). Seven of 14
controllers and two of six non-controllers carried protective
class I major histocompatibility complex (MHC) alleles including
multiple HLA-B57 subtypes and HLA-B2703. Blood CD4 T cell
counts were higher in the controllers than in the non-controllers
(Mann-Whitney p = 0.0064).
We first characterized naive (TN), central memory (TCM), transitional memory (TTM), and effector memory (TEM) CD4 T cells in
blood as hosts for the virus in these individuals by quantifying
HIV nucleic acids in cell subsets sorted by fluorescence-activated cell sorting (FACS) (Figure S1). In HIV controllers, fluorescence-assisted clonal amplification (FCA; Figure S2) revealed
that TEM and TTM cells were more likely to contain HIV DNA
ex vivo than were TCM cells (Figure 1A). By adjusting for
absolute cell numbers and assuming one copy of HIV DNA per
HIV DNA Sequence Analysis in Blood CD4 T Cell Subsets
in HIV Controllers
To clarify the roles of circulating CD4 TCM, TTM, and TEM cells
as hosts for the virus in HIV controllers, we sequenced the
single-template PCR products derived by FCA from these cell
subsets and plasma virions. Compared to 608 HIV sequences
from the non-controllers, the 1,279 sequences from the
HIV controllers showed two striking patterns (compare Figures
S3 and S4). First, blood CD4 T cell-associated HIV sequences
in each HIV controller differed markedly from that individual’s
plasma virus sequences. HIV sequences in blood CD4 T cells
Cell 166, 1004–1015, August 11, 2016 1005
Figure 2. HIV DNA Sequence Analysis in Circulating CD4 T Cell
Subsets from HIV Controllers
1 plasma virus match
No plasma virus match
60
(A) Number of distinct HIV DNA sequences detected in blood CD4 T cells from
each HIV controller, with number of sequences matching one or more plasma
virus in red.
(B) Average genetic distances between plasma HIV RNA sequences and HIV
DNA sequences in TCM, TTM, or TEM cells in HIV controllers and non-controllers. Mann-Whitney p values are shown.
(C) Phylogenetic analysis of all sequences in the study, with labels colored by
participant. The arrow shows one clade in which G-to-A hypermutated sequences from multiple participants are intermingled.
(D) Genetic compartmentalization between HIV DNA sequences in TCM, TTM, or
TEM cells and plasma viruses in HIV controllers and non-controllers, as
determined by Slatkin-Maddison testing with Bonferroni correction. Only 13
participants are shown for TCM cells because participant S1270 had no
detectable HIV DNA in TCM cells. Fisher’s exact test p values for comparisons
between controllers and non-controllers are shown.
(E) Normalized Shannon diversities of plasma viruses and HIV DNA sequences
in TCM, TTM, and TEM cells from HIV controllers and non-controllers. MannWhitney p values for comparisons between controllers and non-controllers are
in black. Wilcoxon signed-rank test p values for comparisons between subsets
in HIV controllers are in green. All Wilcoxon signed-rank test p values for
comparisons between subsets in non-controllers are >0.05.
See also Figures S3 and S4.
40
20
0
V9
1
V9 2
S1 07
2
S1 70
3
S1 49
4
S1 75
4
S1 92
4
S1 95
5
S1 41
5
S1 48
7
LI 88
R0
LI 1
R
LI 02
R
LI 03
R0
4
Distinct HIV seqs.
A
Participant ID
HIV distance to plasma
B
0.7271
0.5704
0.2599
C
C
C
0.15
0.10
0.05
0.00
NC
TCM
NC
TTM
0.04
LI
LI
R0
LIR0
R01_
1_
32
33
31
1_9
30
22
21
20
19
18
17
13
12
11
3
1LI
LILIR R01
LI
SSS12
_15
R01
R
SSS1SSSS1111112
70
01
_4
LI
_5
_8
_7
_1
12 2222277770
_2
_6
_2
_3
001
_5
R801
_6
264879R
5LI
01
RLI
55 01234567841
SSS127S277770000____5
01
_1
_2
SSS1S1SSSS1S1S1S11111122021720_07270_4_7___13456891123340123456789013679012358946123456789
R_1
40016 LI
LILIRRLI LI
12122122217222172722777S00000350_14__427
LIR
_2
R
LI
01
7727070707007070070_010____711171150S
01
LI
RR01
01_5
R_3
3 01
R
LI
_6
01
_3
_409
_4
01
01
R
_5
_4
_3
01
_0__8_02_72___34567_81_1721_11276348771326712538049790458124569404151227
01
_4
_8
_5
9172654_4
_5
8_5
_5
3934586057
51438542417401901367851742352863152049_33 0_43
3 136
97
9
S13
1827
71392V
_
1
V912VV_991112225__1V
4
V912_82
39
1V912_62
_2
2V912_119
19
V912_128
V912_115
V912_92
V912_108
V912_54
V912_104
V912_109
V912_77
V912_121
V912_110
9
V912_111
V912_71
1
V912_114
2
V912_116
V912_118
_32
33
4
V912_12
37
35
_102
_90
_100
_88
60
8_52
_87
V912_136
V912_13
8_56
3
8_58
8_46
V912_1
8_10
S1548
45
9
43
8
4
S1548
V912_9
S154
S154
V912_1
S154
V912_
V912_
V912_7
54
57
61
62
64
73
5
38
77
82
2
36
29
8
48_
105
8_70
4
3
0
8_12
7 3S15
48_
48_
48_
8_11
S15
48_
S154
92
89
86
85
8_68
S154
84
8_10
8_10
80
S15
S15
53
48
47
24
45
48_
S154
44
16
41
15
75
S154
42
S154
48_
48_1
S15S15
48_23
_33
48_
S15
S15
S15
548
S1
9 S15
_66
_93
_79
_11
_78
548
_76
_71
51
_67
40
39
_55
34
31
548
8_49
S1
8_
548
548
S1
54
54
S1
8_
37060
S1
54
10
S1
S18_
54
8_
74
5197
43
12
8_
54
20
S1
35
54
21547
16
8_
10
63
8_
8_
54
8_
8_
S1
81
S1
69
8_
54
8_
54
37
50
54
8_
54
54
S1
21594304629
691
54
96
S1
23
54
92
S1
83
91
8_
28
89
8_
88
11
S1
54
87
18
S1
S1
9876543214001626
86
85
8_
S1
73
8_
11
96
59
S1
8_
10
9_
54
94
54
8_
S1
55___111
13
9_
54
21
9_
54
14
8_
8_
54
S1
_4
34
S1
9_
1
5_5
_4
9_
54
_4
27
_9
8_
S1
34
S1
54
54
34
25
8_
98
2489
S1
34
49921532976543109867654
S1
_3
34
S1S1
54
49
49
_3
8_
_2
12
15
_545235567901235614
17
S1
S1
8_
19
54
22
8_
24
26
S1
27
54
49
13
8_
13
S1
8_
95 S1
4949
54
S129511191__4_84_2292111110877665232102987543210832198765432109
S1
_1
54
_7
_4
S
8_
S
_1
S13
_2
_9
S1
54
54
8_99
S1
_2
S1
_3
5 _ 1 17
_3453789013579012345
54
09SS
49
S873S13
S1
9849
S1
49
49
913
S1
49
499___56676789012467890123 5214_S9995555____1105_7964857
13
13
404S_2
S154
_1
13
13
_3
SS
_3
1133344449999____7889494S59_111114444449999995555_592_15103
S
_2
_1
_3
249
SSS13
49
49
49
49
13
49
_1
SSS111111333333444499 1S41 SSSSSSSS111111144449199545_
13
13
S
S13
13
SSSS
13
49
SSS 11 S
SSS4S4
138
_5
S
SS11
49S
V912_8
14
06 V912_35
3
6
7
4
V9
91
12
2_
_3
30
3
2
V912_31
V
V912_29
V912_28
V912_7
V912_6
V912_5
V912_4
V912_2
V912_3
V912_67
V912_140
V912_94
V912_107
V912_99
V912_90
V912_88
V912_138
V912_89
V912_75
V912_58
V912_65
V912_95
V912_12
V912_1
V912_1
V912_5
V912_5
V912_1
V912_6
V912_1
V912_
01 244
902
36
V912_
13
146
155
V912_
157
162
168
V912_
170
171
176
V912
180
V912
181
_190
_196
V912_197
_85
_192
V912
_195
_185
_178
V912
_158
V912
_148
V912
_144
_78
_63
_142
_69
V91
2_61
V91
V91
2_76
V91
2_79
2_11
V91
2_12
V91
2_1
2_1
2_9
V91
2_5
7
3972
2_4
00
V91
V91
03
7
2_4
6
2
2_1
V91
2_3
1
2_6
V91
0
2_1
2
9
86
V91
2_9
2_8
23
2_7
41
3
V912_5
2_6
1
2_5
V9
2
0
V9
6
12_
5
2
V9
1
12_
49
0
48
V9
47
12_
26
25
V9
24
12
23
22
_2
V9V9
21
12
_1
0
12
9
_1
7
6
_7
V9
5
12
_1
1
V9
V9
0_1
12
_8
05
V9
12
7_1
12
V9
_1
12
V9
12
_1
V9
43
65
45
47
12
_1
V9
49
50
12
V9
51
_1
52
53
V9
12
_1
54
56
59
V9
12
_1
60
61
V9
63
12
_1
64
66
67
12_1
69
V912
72
_182
73
12
74
75
77
79
_1
86
87
88
_1
89
91
93
94
98
LIR
01_1
4
91921_21_8V41912_183
0
8V
V912_V
C
NC
TEM
55
87_
4_466
827629827V
7_7__163__14276
37 6666767667V7_7_
7_3 VVV6V6V66666
25184251310926_99603616362774__43563 VV
77_7__6_15_4V73454V
9536
73_6
6666666666V766776720_6_77_7_6546_4_V
VVVV
667_266V6_23766
678901234567 012345678901
VV6V
7V67_6V7_2986954291430825
____8_9911118901011234562347567120372689067890 V66V
3
8
13
7
81
0
4
00 _ 1
0_2
___7
_7
35_
54
6 785575
_8
_2
_6
_8
_4
_1
_9
_55_
_8
278346759_4
_9
_7
_1
6 47
_1
222277777770000000____1 VV7V_643614
75
947
775
93
75
_6
714
75
_6
_7
94
_7
_9
75
775
_4
_1
_2
_8
_2
92_95
_5
S1
14
_7
75
75
14
S14
75
_6
S1
SSSSS111111112222277 V_4676SSS1S
14
S1492_
75
114
SS14
90
75
91
92S14
89
S
88
87
75
86
14
14
84
2_
83
SSS 1 6174 SS
82
_7
81
14
80
2_
S75
64
63
62
49
14
61
2_
S14
60
S1492_
59
58
17
49
75
33
2_
57
36
34
56
67_
55
S1
SS14
54
2_
49
53
52
51
S1
50
5_
2_
49
49
48 32
21
44
S1
46
2__47
49
54
51 2
56
49
3_6V103127 S14
_46
_44
S1
_43
47
18
_42
19
20
_41
49
5_
25
_38
26
S1
_37
27
30
2792_
492
5_
_33
32
35
_29
37
38
39
S1
45
5_
48
70V6__7645 47
492
52
S1
55
10
S14
58
47
60
92_
61
S1
66
5_
47
68
70
80
35
3
5_
S1
5_5_
30
45
S14
6_V656 S1
5_
S1
47
68
67
78
S1
57
92_
5_
26
15
40
4747
92_
47
92_
65
13
92_
92_
73
75
23
S14
7
92_
64
47S1
85
S1
20
6V7
77
S14
S1
92_
21
1
S14
82_9
4
92_
S1
S14
S14
82_72
92_
S14
47
92_
5_
22
S15_
92_4
50
S14
2_31
92_1
42
S14
2_69
S14
2_76
S14
V6
2_39
2_79
S14
4731
2_28
S1
5_
2_34
S14
S149
S149
2_36
2_1
S149
S149
2_22
S149
S149
2_74
S149
2_16
475_
S1
_71
S149
2_10
2_6
_2
_3
S149
_5
_12
72
_17
S1492
111
S147
06
S1492
_24
10
_25
66
17
70
S1548_3
S1
S1548_
27
S1492
S1548_12
S1492_
S1548_1
92_19
S1548_1
809
LIR04_1
LIR01_
6
7 S1548_1
S1475_
S1270_13
S1270_13
LIR04_106
V965_116
LIR04_170
LIR04_164
S1495_145
S1475_69
S1475_94
S1475_78
LIR01_52
LIR04_25
LIR03_38
LIR04_168
LIR04_31
LIR03_182
LIR04_163
LIR03_180
LIR03_36
LIR03_69
LIR03_78
V016_74
V016_127
V016_52
V016_128
V016_89
V016_91
LIR02_145
LIR02_155
LIR02_110
LIR02_98
LIR02_78
LIR02_48
LIR02_47
LIR02_22
LIR02_111
LIR02_215
LIR02_118
LIR02_144
LIR02_70
LIR02_65
LIR02_136
LIR02_43
LIR02_44
LIR02_45
LIR02_91
LIR02_194
LIR02_53
LIR02_54
LIR02_94
LIR02_108
LIR02_80
LIR02_28
LIR02_93
LIR02_10
LIR02_21
LIR02_11
LIR02_85
LIR02_6
904
LIR02_2
631
LIR02_8
5
LIR02_1
LIR02_1
LIR02_1
LIR02_
LIR02_
792
60
LIR02_
48
152
75
LIR02_
161
LIR02
130
LIR02_
LIR02
LIR02
LIR02
127
_86
LIR02
31
_137
LIR02
_153
_96
_197
LIR02
LIR02
_141
LIR02
_101
LIR0
_19
_73
LIR0
_5
_7
2_14
_105
2_21
2_32
2_46
LIR0
2_88
2_11
LIR0
LIR0
2_15
LIR0
2_19
LIR0
LIR
2_21
LIR
LIR0
2_99
2_90
2
2_2
2_14
LIR
4
2_1
02_
9
02_
2_5
2_1
18
20
13
61
02_
63
56
95
74
02_11
7
04
84
114
95
119
140
LIR
196
LIR
LIR
LIR
LIR
02_
LIR
LIR
02_
02_
LIR
LIR
02_
02_
LIR
142
02_
LIR
LIR
02_
51
192
LIR
17
162
02_
LIR
71
02_
116
02_
11
02_
LIR
27
169
206
LIR
02
178
_1
LIR
23
12102_
128
LIR
LIR
02_
_7
212
97
_5
02
16
_2
LIR
02
LIR
24
LIR
02
_3
02_26
5
_6
02
9
02
LIR
0
LIR
LIR
LIR
_1
_1
02_
_7
4
02
6
_1
183
LIR
02
_7
74
2
LIR
_13
_1
106
02
02
_2
_2
02
02
80
_7
619
_6
LIR
LI
07
39
LIR
02
_1
07
_1
_6
_1
LI
7
LILILI
_8
R0
02
_1
147
03
00
LI
02
_2
2
R0
1
35
LI
3
22
R0
9
02
64
_1
LI
R0
2_
93
R0
R0
03
_1
14
_2
LIR LILI
2_
LI
R
LI
R0
_2
R
90
2_
17
2_
13
20
LI
2_
02
R0
02
2_
25
02
RR0
2_
30
2_
45
57
LI
58
1
59
2_
21
R
60
49
02LI
62
LI
12
_4
_1
64
10
_1
67
2_
02
_2
R
68
14
15
715
02
R
20
98
375
_1
8
_9
221
59
_8
_1
R
02
_3
8109
LILI
02
_4
57
_1
R81
LI
_1
RLILI
02
20987650 09
29
R_1
33
LI
76
84
85
02
86
LI
87
88
0291
_1
89
R
R
R02
VR
_1
56
02
_1
82
V62
02
02
25
66V
34
_3
_4
32
_5
_1
LI_6
VVV66VVV6V626VV
_1
21R
62121_2_149
43
1V
67
212626211_
68
902
70
77
6221VV
__66616_22_2161_11212_19_67_40915 _1
VVVV6VV6VV2616V61V
2V226_11711073411__2_7151426_32432 79
6V22621V
61221_16_61_6_212_2_311_1_2111185_4321401753128621723057
LIR
1V12_890112970__0479887172097
02LI
3
6
5
6
8
_1
0
1
V
0
R66
VVV6 VVVV6V66260819
02RLI
LI
_1
R38
VVVVV66626V2V6V6V22626221_16
02
02
_1_1
_2
VVVVV666V66V66V266V2V26221611221V116__16V_62_26321621121211_2_1_1_1___11723653452309
LIR
LI
72
13
6222211121216_V1_221V_912_6_41494120218__1_1_822_012865484
02
2602
_1
1V1____97V6_V_6161625_0311203194_127851563
_1
65
73
58
6_2821191041263262_261491_9 036
13_952 211_7_3120
110 _1623
68 52
SS1 SS
S14 SS11
SS14
SSSSSS1S11SSS4SSS11111S494S49511SSS11S441441999
14
S1S
S1141414149 444995_4149149455_
S14
95
S
SS
S_2
SSSS95
14
14
S
S95
14
14
1158
194SSSSSSSS1111144944499594459955_95_959_955_15555____49_215496955795__9_55__121_25043
495
14
_1
S
14
14
SSSS
95
14
495
S
14
14
95
SS
14
14
14
9_1
14
5_1
9_1151464S4S11444999955_1_2_1223_728_5011234556347890123456783415_268_721504986309
S
17
95
95
14
95
95
_1
5_1
95
_3
14
95
95
95
95
_90
_2
_1
_2
_1
_1
95
LIR
86
1432123_4456789911955415439__99555_____34789023435917234562712945689017 60171220558046
_2
460
_2
95
LIR
_2
_1
SS
_2
_1
57
03
05
10
88
S
18
30
14
_1
LIR
31
03_2
34
S1
42
85 23 14680
03
38
_8
44
S
LIR
53
37
S1
14
S
7
03
40
85
LIR
95
14
14
S1
S1
00
S14
03
49
S1
16 S1
LIR
15
_274
8 6_9259653_4_125346789034567890123890
S1
95
49
S1
14
S1
03
S1
13
14
12
LIR
11
_2
_1
95
49
95
10
S1
5_
95
49
S1
08
03
49
49
LIR
84
S1
49
_2
_1
5_
72
S1
49
49
69
49
95
LIR
03
68
S1
67
5_
_2
31
S1
_1
58
_2
18
49
_9
5_
5_
_1
LIR
49
57
5_
5_
03
51
18
62
49
49
55
5_
11
42
S1
_1
44
49
49
LIR
36
5_
5_
43
_9
49
39
03
25
38
22
19
05
03_
LIR
20
5_
2
37
_1
59
49
18
81
30
5_
28
24
93
27
72
LIR
90
71
26
70
5_
03_
25
5_
50
98
36
55_
24
289
6532492
35
19
LIR
119
23
22
756
117
22
20
11
23
11
114
5_
1
26
18
03_
110
17
52
23
109
LIR
24
107
03_
106
104
25
LIR
8613
102
24
37
0
100
1
03_
4124
3
LIR
5
99
6
8
97
0
96
9
03_
93
LIR
92
91
90
03_
LIR
89
88
87
85
LIR
03_
84
83
LIR0
82
81
03_
80
LIR0
79
76
72
3_6
68
LIR0
67
3_5
3_4
6
LIR0
3_45
5
3_41
4
3_37
1
3_31
LIR0
9
3_30
3_27
LIR0
LIR0
3_24
LIR0
3_19
3_18
LIR0
LIR03
LIR03
3_11
3_10
3_4
3_3
LIR03
3_63
3
15
_108
LIR03
_86
_171
LIR03
LIR03
_112
_209
LIR03
_155
_77
LIR03_
_152
_134
LIR03_
_136
LIR03_
_116
LIR03_
150
LIR03_
145
146
LIR03_
103
95
16
LIR03_1
132
140
LIR03_1
73
48
LIR03_15
LIR03_94
41
47
LIR03_73
35
LIR03_75
LIR03_50
LIR03_48
LIR03_42
6
LIR03_98
LIR03_133
LIR03_111
LIR03_142
LIR03_23
LIR03_207
LIR03_32
LIR03_71
LIR03_105
LIR03_74
LIR03_131
LIR03_170
LIR03_46
LIR03_43
LIR03_153
LIR03_28
LIR03_206
LIR03_186
LIR03_22
LIR03_33
LIR03_191
LIR03_165
LIR03_34
LIR03_15
LIR03_40
LIR03_5
LIR03_8
LIR03_11
LIR03_44
LIR03_52
LIR03_53
LIR03_54
LIR03_55
LIR03_59
LIR03_121
LIR03_122
LIR03_162
LIR03_166
LIR03_187
LIR03_189
LIR03_190
LIR03_192
LIR03_193
LIR03_194
LIR03_196
9 10
LIR03_197
LIR03_200
LIR03_203
LIR03_205
29
LIR03_204
LIR03_62
9
LIR03_15
LIR03_188
LIR03_70
18
LIR03_25
LIR03_2
13
LIR03_7
56
57
LIR03_6
60
LIR03_1
LIR03_
202
LIR03_
61
_163
_35
_179
_164
_12
_14
71
LIR03_
_195
_198
3_17
LIR03_
_201
3_20
LIR03_
_199
3_21
LIR03
0
3_26
LIR03
3_29
3_17
LIR03
LIR03
LIR03
3_39
_1
LIR0
3_47
3_58
LIR03
3_16
LIR0
LIR0
LIR03
LIR0
16
0.044
S17
S
SS1
S1
S
S
17S
S1
17
S178
78
17_5
88
S1
88
78
S1
88
78
8_
882356_3
_4
78
_3
S1
S1
_3
S178
_2
8_S1
_6
8_
S1
_7
78
8_
S1
33
788_
50
8_
78
78
S1
S1
S178
77
5789_2921
78
583416
8_
73
8_20
S1
78
8_
S1
78
8_
S1
78
S1
8_
8_
78
V9
60
21
S1
78
18
69
8_
78
S1
11
61
V9
8_
78
62
8_
58
V9
78
57
S113
8_
54
07
46
S1
44
78
8_
41
8_
40
V9
78
68
07
34
V9
30
8_
26
07
59
22
8_
_1
76
7878
19
V9
67
10
17
42
V9
15
49
8_
_1
07
51
47
07
38
_9
18
74
V907_
24
_5
16
V9
72
23
8_37
V9
07
71
12
V9
70
07
8_
65
22
_1
V9
43
66
V9
_1
468_
V9
3
_9
07
_1
07
0
07
_1
V9
V9
36
1
07
_4
07
00
07
_3
_9
01
_3
124
V9
_1
_8
V9
07_
4 4514
07_
V9
_2
607_
_7
_1
V9
5
V90
195
V9
19
V9
07_
07_
V90
V90
07_
21
90
98
07_
V90
07_
V907_71
V90
V90
7_1
993 HXB2
513
7_1
7_1
V907
_74
7_9
_73
615
7_4
7_1
2
20
23
_47
V907
02
17
V907_2
V907_23
V90
_45
_44
V90
7V90
_43
V907
V90
_42
_39
7_3
V90
_37
7_1
V90
_34
9 V90
7_7
7_6
V90
7_3
7_6
7_2
V907_40
7_3
7_4
866
V90
7_3
V90
7_7
7_1
23
4
V907
1003
V90
9
V90
7_1
072
7_1
V90
7_4
7_3
7_27
_19
7_28
7_25
06
7_18
8
V907_21
V907_
_20
V907_
22
26
51
52
V907_
53
V907_5
54
55
V907_6
56
57
58
9
V907_65
0
V907_66
1
V907_67
2
V907_75
3
4
V907_76
V907_77
V907_78
V907_79
V907_80
V907_81
V907_82
V907_83
V907_84
V907_85
V907_86
V907_87
V907_88
V907_89
V016_122
V907_91
V
V907_92
0110
1661
6__6
V907_115
_18_
1184
021
7
V907_107
8
3
0V
VV016_100
0V
V
01
16
6_
_6
48
0_
18
64
_4
79
0
2
VV
0V
1
6
S1
54
SS1S1_
54
1S_1
SSSS115S12885
5524S SSSSSS11111155555544414541S514S1_411155S1LIRL
0LIR4
11_5SS111115555544441111____11_780_41415 LLLIR
S1SS
S
4
4 IR
LLIR
SSS1S1S
IR
IR
0_0
15S151S
IR
S15155115SS553S1141925515554_44448111111_1_____5_566677347567890123456789039118_605_1L51IR
8_40003444041400_43_441_71
SS1S
1S5551414541551411_4514_4_144_114114_1S
_1151415571__333480280136012
15
SSS15
___11__130543281
15
S
441144_191293__115365__22971_12_924545184320_14_4762_1123641990134567
15
41
15
41
SSS15
15
41
_21_65187643
77609685542931746
_904
_5
4141
_8
41
_9
9
_1_9
6
_1
00987421098724
_1
01
02
03
04
11
12
13
17
14
S1
10
S7S11
5544
11__
1100
568
1_
54
S1
# of participants
D
94583866 731
1
___98543_21_6452234531019_7__18717656
_8 2
222111212_1_182911_6271_2121_11
57
2322
VVV6V66V66V226266V1612V_V62V166
_57_V
V
5725
VV6V
V2V
4
_5
71047247
57
V2
__57432_182_1226305 22
55577575_7_1671825_1_185913_1579763_421846_254039367641
45
VVV2V2V5275227527_752_75_4752_75_2775__7657_7686_71_70_5 5677_4_36
2VV
2725_724528_538 3V
V225V225V
5_532172_5
VVV2VVVV
2V725V25V7_25977V55_721397_05483293747_565_967_17436V
5_077_707623V
V7525V5_2275575763752_57_72_7_752V7_5_572_675_277V
527__27
V5V
222V
2VV
2252V
2 540916 7
V5_2V918653
13
40
39
V2VVVVV2VV
3
38
35
28
_97
24
37
22
47
1_865
17
80
7 _1
11
98
10
842
4531
45
14
15
10
7V9
655_
8 5210
43
96
5_
5_
5_
16
99
3 65
34
19
62
311
5_
90
V9
65
65
5_
7_7
36
292
_5
5_
5_
96
10
65_1
5_
_8
VVV9V
96
96
5_
V9
_2
V965
912
96
5_
5_
3_7
8
21
72
V9
_1
96
96
265
65
_1
85
_2
5_
6112
5_
_5
96
48V9
60
_43
87_1
96
65
_3
4
V96
65
5_
_9
_8
30
09
96
515
83 _7
96
8_8
16
5_1
VV
_4
V9
0
26_3
65
469
41 7 _8065
65
V9
5
VVV
_8
_4
767
_4
_7
7
_6
V9
35
_1
V9
_7
_6
9
V0
_2
65
_1
_2
_5
30
_1
65
_6
16_
_1
_2
_8
_3
46
65
_5
V96
VV96
106
V9
_9
_70_1
6_60
_8
7
96
65
V9
65
65
65
_6
6_9
V9
_5
82
65
65
_7
16_
6_4
V
_3
12
65
65
1
V0
65
65
65
16
65
65
65
V9
109
65
V9
6_2
64
16_
16_
65
V9
V01
6
V9
65
V0
65
79
V9
V9
V9
16_
65
65
4
25
V9
6_2
6_1
_5
V9
5V9
6V0
9
V9
V01
V9
V0
V0
3
V9
87
16_
6_1
16_
V9
V9
V9
V9V9
V9
V0
02
4
6_1
V01
6_2
_7
00
V9
9_6
0
V01
65
7
04
V9
05
3_7
V0
8
V0
6_2
6_7
V01
23
9_1
8
_1
01
_6
16_5V016_
V01
65
_4
6_3
65
_6
1
_1
V9
6_1
_6
V01
9
6_4
0021
_5
_5
65
65
_1
V01
6_6
1
V9
V9
65
65
14
6_1
65
V01
6_8
1 V0
65
65
V01
6_5
V9
6_73
V9
6_1
V01
V9
V9
V9
6_2
V01
V01
V9
6_11
V9
_42
654_9
6_32
V01
6_68
_58
_77
_3
_95
V016
_15
V96_5
V01
V01
_93
65
V016
_81
V016
V016
V016
65_2
_59
_65_108
V016
_40
V016
_69
_54
V9V9
V016
V016
111
V016
V016
V016
V016
_71
119
V016_
75
36
120
53
V016
101
118
V016_
17
V016_
28
9
V016_
V016_
17
V016_
V016_9
27
V016_1
V016_5
636
V016_9
V016_6
V016_7
V016_8
V016_10
V016_70
V016_90
V016_33
V016_63
V016_94
V016_55
V016_96
V016_8
V016_50
V016_115
V016_22
V016_34
V016_10
V016_31
V016_45
V016_102
V016_92
V016_116
V016_76
V016_30
V016_21
101616_6_6
VV016_113
41_1
8
0V
9
010
V0V0
55244
184
V
8
3
1061_6_3
3_94
0
3_91
2
3_87
3
3_85
V72
3_8
3_7
8
5
3_5
V72
1
3_4
0
3_3
8
V72
3
3_3
763
432
V72
3_2
V72
3_5
3_3
3_1
3_4
10
V72
1
2
V72
3_2
V72
7
3_7
3_6
9
3_8
3_1
3_2
3_1
33
56
V72
V72
V72
61
3_2
V72
62
V72
74
V72
95
23_
82
65
V72
66
23_
V7
45
6_7
_7
23_
_1
23_
_1
897
V7
_4
23
1
23_
69
_4
473
V7
V7
23
_2
90
_8
23
23
40
_7
V7
V7
_8
6
23
_2
5_6
23
_2
23
_4
V7
98_6
5_9
V7
_6
_1
23
31 V7V7
23
8783747
V7
_9
V7
23
_7
_9
_2
23
_1
_3
_1
23
23
V7
V7
_4
_1
_6
491
V7
23
2_7
23
23
_3
23
_8
_6
V7
_3
_4
V7
23
2
0_5
V7
23
_5
_8
7654985
V7
V7
23
23
23
99_1
9 0 V7
79
23
V7
23
_5
54
V7
V7
45
V7
2323
43
_6
2323
_1
23
42
23
41
_8
_6
V7
36
V7
_5
V7
_9
_3
12_4
31
_1
_9
8_4
29
0_9
23
V7
28
V7
V7
V7
23
V7
23
V7
16871421907854
_1
23
623
V7
23
23
_4
23
238_5
8V7
23
04
23
_1
V7
_8
V7
V7
_7
_8
23
04
_1_9
V7
R
_7
V7
V7
_9
V7
23
V7
07V7
23
V7_7
_3
04
23
R
_2
_5
23
LI
V7
_1
832127
0404
_6
78420956905
04
R
_5
V7V7
_1
23_5
_6
04
V7V7
RR
R
LI
23
67
V723 LI
30
_5
04
04
_9
_8
R
_2
LILI
_5
00 V7
LI
_6
04
_1
_7
04
20
R
3573854172
R
_5
_1
LI
_1
RR04
04
04
51
R
161411
LILI
04
LI
R
39
235_1
04
R
_1_0_8
04
04
82248_3864 5
04
R04
3
_1
RR
1204
4_4_1
LILI
V7_4
R
LI
LI
R
23
LI
040IR
41__0846173_440035685401 03 47_253125621 52932_7336104086
04
LI
LIR
LI
_8
RR
_3
LILI
V7
LI
IR
0IR
LLIR
4000_4_4_01_4_714_52763167064_640637118_23828590334206_044814__1161_937491784716489514892456170170_0810244740__404_41_
LI04
0L0L04IR
LIR
45310IR
0IR0 101_145_45380_52540__4_14_0_IR
LIR LLLIR
IR
L0IR
140IR
040
IR
0_440414000__440441064___480__1IR
IR
LIR
L
IR
IR
44L71LL16LIR
404_00__44_0441_00_024L4000IR
4LL__IR
L92IR
0IRIR
11L96IR
L0L4IR
IR
IR
LLLIR
L00IR
L004IR
692520IR
IRIR054044_4__L
IR
LIR
LIRLLLLLLIR
LIR
LIR
0LL40LIR
IR
IR4__9314L_IR
IR
LLLIR
LIR
06400
IR
LL_IR
9IR
14410 LLLIR
LIR
0IR
6
0
_L1IR
_
IR
044LL
2_0_6L5143
0LL144IR
IR0
IR
4L_0IR
LLIR
L0IR
7
LIR_L7
04
LIR
0.157
0.002
12
Compart. v. plas.
Not compart. v. plas.
8
4
0
C NC C NC C NC
TCM
TTM
TEM
0.0012
E
0.3396
<0.0001
0.0001
0.0015
0.0002
C
C
0.1786
HIV diversity
1.0
0.5
0.0
C
NC
TCM
NC
TTM
NC
TEM
1006 Cell 166, 1004–1015, August 11, 2016
C
NC
Plasma
from HIV controllers rarely matched plasma viruses (Figure 2A).
Although genetic distances among HIV sequences might be
expected to be greater in non-controllers than in controllers
due to higher levels of virus replication in non-controllers, the
genetic distances between cell-associated virus DNA sequences and plasma viruses in controllers were as great as
those in non-controllers (Figure 2B). The large genetic distances observed in controllers were not due to dual infections
because sequences from all study participants were genetically
clustered by participant (Figure 2C). Moreover, whereas HIV
DNA sequences in blood cells from non-controllers were genetically intermingled with plasma viruses, HIV DNA sequences in
blood cells from controllers were frequently compartmentalized
from plasma viruses (Figure 2D). This was especially true in the
TEM subset, where compartmentalization from plasma viruses
was seen in 13/14 controllers but only in one of six noncontrollers.
In addition to genetic divergence from plasma viruses, HIV
DNA sequences in blood cells from controllers showed large
clusters of identical sequences not commonly seen in non-controllers. As measured by normalized Shannon diversity, which increases with the proportion of unique sequences in a population,
HIV DNA sequences were less diverse in controllers than in noncontrollers within each CD4 T cell subset (Figure 2E). This was
true despite similar levels of diversity in plasma viruses from
the two groups (Figure 2E). Clusters of identical sequences
were particularly prominent in TEM cells from controllers, with
lower HIV diversity in TEM cells than in TTM or TCM cells (Figure 2E).
Diversity of HIV DNA sequences in non-controllers was similar
among TCM, TTM, and TEM subsets (Figure 2E). Thus, among
memory CD4 T cell subsets in HIV controllers, the TEM subset accounted for most of the infected cells in blood but generally
harbored proviruses that were transcriptionally quiescent, markedly distinct from plasma viruses, and rich in clusters of identical
sequences.
0.0001
A
Figure 3. Clonality of Cells and HIV DNA Sequences in Circulating CD4 T Cell Subsets
from HIV Controllers
E
0.0001 0.0001
TCR diversity
1.0
NSD1
(88.8%)
0.5
0.0
TCM
B
TTM
TEM
0.2188
0.0007
0.0001
0.6875
0.0001
0.2188
HIV distance to MRCA
0.14
0.12
0.10
MED26
(2.6%)
0.05
0.00
Plas. TCM TTM TEM
Controllers
Non-controllers
0.05
F
C
15
1 plasma virus match
No plasma virus match
POLM (95.5%)
10
5
0
V9
1
V9 2
S1 07
2
S1 70
3
S1 49
4
S1 75
4
S1 92
4
S1 95
5
S1 41
5
S1 48
7
LI 88
R0
LI 1
R
LI 02
R
LI 03
R0
4
HIV seq. clusters
Plas. TCM TTM TEM
Participant ID
HIV seq. cluster distance to plasma
D
0.15
r = 0.4190
P = <0.0001
(A) Normalized Shannon diversities of T cell receptor beta (TCRB) sequences from TCM, TTM, and
TEM cells in HIV controllers. Wilcoxon signed-rank
test p values are shown.
(B) Average genetic distances of plasma virus and
TCM, TTM, and TEM cell-associated HIV DNA sequences from most recent common ancestral
(MRCA) sequences in HIV controllers and noncontrollers. Wilcoxon signed-rank test p values are
shown.
(C) Number of recurrent HIV DNA sequences detected in circulating CD4 T cells of each HIV
controller, with number of distinct sequences
matching one or more plasma virus sequence
in red.
(D) Correlation between the abundance in blood of
each recurrent HIV DNA sequence in circulating
CD4 T cells from HIV controllers (x axis) and the
average genetic distance of that sequence to
plasma viruses (y axis). Each symbol represents
one distinct sequence and is colored by participant. Spearman r and p values are shown.
(E and F) HIV DNA sequences associated with
expanded cellular clones in HIV controllers V907
(E) and S1349 (F), illustrated using dashed borders
within phylogenetic trees. Gene locus names
corresponding to the HIV integration sites in these
expanded clones are shown. Each number in parentheses represents the percentage of all copies
of HIV DNA in blood CD4 T cells from the individual
deriving from the indicated HIV integrant. Sequences from participant V907 showing G-to-A
hypermutation and associated with expanded
cellular clones are shown as detached branches;
other hypermutated sequences are omitted for
clarity. The large sequence cluster in participant
S1349 is shown separated from the tree for ease of
viewing; an arrowhead shows the position of this
sequence on the tree.
See also Table S2.
0.10
0.02
0.05
0.00
10-1
100
101
102
103
HIV seq. cluster copies/mL blood
HIV Clonality in Blood CD4 T Cell Subsets from HIV
Controllers
Based on these results, we hypothesized that each cluster of
identical HIV DNA sequences represented one CD4 T cell clone
harboring a single HIV provirus amplified in vivo through cell
proliferation. We also hypothesized that the TEM subset might
be enriched for recurrent HIV sequences because memory
CD4 T cells undergoing clonal expansion in vivo tend to differentiate into TEM cells. To test this, we first compared the clonotypic
diversity of the three memory CD4 T cell subsets using T cell
receptor beta chain (TCRB) deep sequencing. We found that TCRB diversity
decreased progressively along the putative pathway of differentiation from TCM
to TTM to TEM (Figure 3A), consistent
with the hypothesis.
We evaluated the alternative explanation for clusters of identical HIV DNA sequences in blood cells, whereby sequences closely related to
the actively replicating virus pool and matching over the region
we sequenced might accumulate through bursts of virus replication. Using plasma viruses as a genetic surrogate for actively
replicating viruses, we observed the opposite pattern. In general,
HIV sequences from blood TCM, TTM, and TEM cells in HIV controllers were ancestral to plasma viruses (Figure 3B). Sequences
occurring at least twice rarely matched any plasma virus
sequence (Figure 3C). Moreover, recurrent sequences that accounted for higher copy numbers in blood (i.e., occurred in
Plasma
TCM
TTM
TEM
Cell 166, 1004–1015, August 11, 2016 1007
TEM
A
14
9
3
10
28
TTM
10
25
TCM
TTM
TCM
B
TEM
CXCR5 + + - - - - - - CCR6 - + - + - + - - +
CD57 - - - - - - + - V912_9
V912_25
V912_26
V912_47
V907_25
V907_16
S1475_18
S1492_23
S1492_42
S1495_30
S1495_14
S1495_32
S1495_43
S1548_44
S1548_24
S1548_31
S1548_28
S1548_34
S1548_22
S1548_48
S1548_43
HIV DNA copies/mL
C
0.5431
0.0002
0.0256
<00001
104
more cells) were genetically more distant from plasma viruses
(Figure 3D), with average genetic distances to plasma viruses
frequently >5%. This suggested greater expansion among older
HIV-infected CD4 T cell clones in HIV controllers, which was not
apparent in sequences from non-controllers (Spearman r =
0.2706, p = 0.2117; data not shown). Therefore, recurrent
HIV DNA sequences in blood CD4 T cells from HIV controllers
had genetic attributes of expanded cellular clones rather than virus replicative bursts.
We used HIV integration site analysis to prove the presence of
clonally expanded, HIV-infected cells in blood from HIV controllers. Although too few infected cells to permit this analysis were
available from some participants, we identified two HIV integration sites in clonally expanded cells for participant V907 and one
such site for participant S1349. We then linked recurrent env sequences with their integration sites by PCR from the forward env
primer to the integration site. Sequencing the env portions of
these amplicons confirmed that clusters of identical sequences
detected by FCA arose from expanded CD4 T cell clones (Figures 3E and 3F). These clones accounted for 91.3% and
95.5% of all amplifiable env DNA copies in blood CD4 T cells
from these participants. Therefore, integration site analysis in
HIV controllers confirmed both the presence of clonally
expanded, HIV-infected cells and the predominance of these
cells within the circulating, HIV-infected CD4 T cell pool.
We next characterized further the recurrent HIV DNA sequences detected in blood cells within the full HIV controller
cohort. Excluding the six of 105 such sequences with <1% genetic distance from plasma viruses as possible replicative
bursts, we analyzed the remaining 99 as coming from presumptive expanded clones (Table S2). These sequences were relatively abundant in blood, together accounting for between
32.7% and 96.8% of HIV DNA sequences in circulating CD4
T cells from each individual, at levels ranging between 0.32
and 1368.61 copies/ml for each sequence. Twenty-two of these
sequences (22.2%) contained one or more stop codon associated with G-to-A hypermutation, and an additional sequence
contained a frameshift in gp120. The remaining 76 sequences
(76.8%) contained neither stop codons nor frameshifts over
the region of env sequenced (HXB2 bases 7011–7502). We found
32 (32.3%) of these sequences in more than one memory CD4
T cell subset (Figure 4A) and many of these in multiple subsets
within TCM, TTM, and TEM defined by the markers CCR6,
CXCR5, and CD57 (Figure 4B). Therefore, confirmed and
103
102
1
10
100
C
NC
Repeat
HM
C
NC
Singlet
HM
C
NC
Repeat
no mut.
C
NC
Singlet
no mut.
Figure 4. Subset Distribution and Genetic Attributes of HIV DNA
Sequences in Circulating CD4 T Cells
(A) Subset distribution of HIV sequences from presumptive expanded CD4
T cell clones in HIV controllers.
1008 Cell 166, 1004–1015, August 11, 2016
(B) Distribution of HIV DNA sequences from presumptive expanded clones
across subsets within TCM, TTM, and TEM populations defined by CXCR5,
CCR6, and CD57. Each row represents one sequence, labeled by participant
and a unique number. Yellow indicates that the sequence was detected in the
given subset. Italics indicate G-to-A hypermutated sequences.
(C) Levels of HIV DNA sequences in circulating CD4 T cells from HIV controllers
and non-controllers categorized according to the number of occurrences
(Repeat, >1 occurrence; Singlet, one occurrence) and the presence or absence of
G-to-A hypermutation (HM, hypermutation detected; no mut., no lethal genetic
defect detected). To allow display of these wide-ranging values—including
several values of zero—on a logarithmic scale, each plotted value represents the
measured value + 1. Mann-Whitney p values are shown. Sequences with lethal
genetic defects other than hypermutation were rarely detected.
See also Table S2.
presumptive HIV-infected CD4 T cell clones in blood from HIV
controllers showed evidence of in vivo maturation and functional
differentiation. Finally, although repeated and hypermutated sequences accounted for a strikingly high proportion of all HIV DNA
copies in blood CD4 T cells from HIV controllers, equal or higher
absolute numbers of these sequences were detected in blood
CD4 T cells from non-controllers (Figure 4C).
Inducible Proviruses in Blood CD4 T Cells from HIV
Controllers
Because some expanded HIV DNA sequences may have been
lethally mutated outside the region we sequenced, we tested
whether CD4 T cells harboring these sequences could produce
virions by stimulating them through the T cell receptor and
then sequencing RNA from virions released in culture.
Although these experiments revealed no virion production
from most presumptive expanded clones (Figure S5, open
bars), we recovered virion RNA matching two recurrent DNA
sequences from TEM cells in participant S1270 (Figure S5, filled
bars; Figure 5A, x-y plots, filled circles). One of these DNA
sequences was highly expanded, divergent from plasma viruses,
and close to the most recent common ancestor (MRCA), consistent with an expanded clone carrying an archival provirus.
The other appeared to be more recent (arrowhead) but contained
a frameshift in gp120. Importantly, while abundant virion production was again detected from cells harboring the latter sequence
in a repeat experiment, this was not the case for cells harboring
the former sequence, even though this sequence occurred
within many more cells in the culture (Figure 5B). Therefore,
although virion production was occasionally inducible from proviruses bearing genetic signatures of expanded cellular clones,
the inducibility of a given provirus was not always uniform among
the cells that harbored it.
Importantly, we also detected a second class of inducible viruses with a distinct genetic signature in these experiments. In
participants S1495, V912, and S1349, we found multiple induced
viruses that had not been detected as recurrent DNA sequences
in separate aliquots of the same cell populations (stars in Figure 5A, x-y plots). All but one of these ‘‘unique induced’’ viruses
were closely related to plasma viruses and divergent from ancestral sequences, suggesting recent in vivo infection of the cells
producing them. Assuming that recurrent virion RNA copies of
unique induced sequences represented progeny virions from
one cell, we calculated that cells harboring these proviruses accounted for 0.19%–1.48% of all HIV-infected, circulating memory CD4 T cells from the individuals in whom they were detected
(Figure 5A, hemispheres). Of note, unique induced viruses were
detected in all circulating memory CD4 T cell subsets and were
found mainly in the two HIV controllers with the highest plasma
viremia. Overall, therefore, inducible proviruses in circulating
memory CD4 T cells from HIV controllers fell into two distinct categories: expanded, archival proviruses in TEM cells, and proviruses of recent origin in rare TCM, TTM, and TEM cells.
Lymphoid Tissue Viruses in HIV Controllers
To help determine the origin of circulating CD4 T cells harboring
unique induced viruses in HIV controllers, we characterized HIV
in lymph node (LN) from four study participants (Table S3). Three
of these four had detectable plasma viremia and showed high
levels of HIV DNA in LN CD4 T cells. In these viremic HIV controllers, the highest levels of HIV DNA were observed in germinal
center (GC) and non-germinal center (non-GC) TFH cells (Figure 6A). These TFH subsets accounted for 73.1%–86.3% of the
infected CD4 T cells in LN (Figure 6D). However, HIV-infected
cells were also detected in LN TCM, CD57+, and TEM subsets,
at levels that were higher than in TCM, TTM, and TEM cells from
blood (Figure 6A). HIV DNA in both follicular and non-follicular
LN memory CD4 T cells was also associated with higher HIV
RNA levels than was HIV DNA in blood cells (Figures 6B and
6C). Detection of cell-associated HIV RNA by in situ hybridization
using intact LN tissue samples confirmed that HIV RNA+ LN cells
were present both inside and outside follicles in these individuals
(Figure 6E).
To clarify the relationship of HIV in TFH and non-TFH LN cells to
viruses in blood cells and plasma, we analyzed HIV DNA
sequences in LN cell subsets. We found that sequences from
GC and non-GC TFH cells in the three viremic controllers
(n = 113 sequences) were more closely related to plasma viruses
than were HIV DNA sequences from blood cells (Figure 7A, left
panels). In two participants, sequences in TFH cells were also
significantly further from the MRCA than were blood cell-associated sequences. Although recurrent HIV DNA sequences were
observed in TFH cells, these sequences too were closely related
to plasma viruses and divergent from ancestral sequences, thus
distinguishing them from recurrent HIV DNA sequences in blood
cells. Importantly, analysis of 145 additional sequences from
non-TFH TCM and TEM LN cells in these individuals showed a
close relationship with viruses in TFH cells (Figure 7A, right
panels, green symbols). By contrast, the sequences from circulating TCM, TTM, and TEM cells were significantly more distant
from viruses in TFH cells (Figure 7A, right panels, red symbols).
Thus, among CD4 T cells of a given maturation phenotype,
we identified sharp distinctions in the genetic characteristics
and transcriptional activity of cell-associated HIV populations
detected in different anatomic sites. Whole-transcriptome
sequencing and principal component analysis (PCA) of cell subsets from these sites revealed that LN TEM cells had a transcriptional profile that was related to that of TFH cells and distinct from
that of circulating CD4 TEM cells (Figure 7B).
Finally, in one HIV controller with undetectable plasma viremia,
levels of LN cell-associated HIV DNA were nearly undetectable
(Table S3). In this individual, only a single, G-to-A-hypermutated
copy of HIV was detected among all LN CD4 T cells studied, with
no HIV DNA found in >2 3 104 TFH and GC TFH (Figure S6I). Flow
cytometry confirmed the expected distribution of LN CD4 T cells
among TN, TFH, and other memory subsets in this individual (Figures S6A–S6H). Furthermore, plasma virus sequences from this
individual showed a relatively high average intragroup pairwise
genetic distance (Figure S6J) and an average divergence from
MRCA that was the highest out of all 20 participants (Figure S6K).
Therefore, HIV-infected CD4 T cells were very rare in LN from
one HIV controller even though plasma viruses showed genetic
markers of diversification and ongoing evolution. This individual
also showed TCM cell-associated HIV DNA sequences in blood
that were more genetically distant from plasma viruses than
were TEM-associated sequences. Among all 14 HIV controllers
Cell 166, 1004–1015, August 11, 2016 1009
Dist. to plasma NN
Dist. to plasma NN
Dist. to plasma NN
Dist. to plasma NN
Dist. to plasma NN
A
0.08
Participant S1495
pVL 525 copies/mL
HIV DNA 53.2 copies/mL
Uniq. ind. 0.79 cell/mL
0.04
0.00
0.00
0.04
0.08
0.09
0.06
Participant V912
pVL 442 copies/mL
0.03
HIV DNA 93.5 copies/mL
Uniq. ind. 0.76 cell/mL
0.00
0.00
0.06
0.12
0.04
Participant S1788
pVL 41 copies/mL
HIV DNA 8.3 copies/mL
Uniq. ind. LOD 0.010 cell/mL
0.02
0.00
0.00
0.02
0.04
0.02
Participant S1349
pVL <40 copies/mL
HIV DNA 188.1 copies/mL
Uniq. ind. 0.36 cell/mL
0.01
0.00
0.00
0.01
0.02
0.06
Participant S1270
pVL <40 copies/mL
HIV DNA 83.7 copies/mL
Uniq. ind. LOD 0.006 cell/mL
0.03
0.00
0.00
0.03
0.06
Distance to MRCA
B
Plasma
TTM cell DNA
TEM cell DNA
Virion Expt. 1
Virion Expt. 2
Panels A, x-y plots
Type of virus sequence
Cell repeat, >102 copies/mL
Cell repeat, 10-102 copies/mL
Cell repeat, 1-10 copies/mL
Cell repeat, 10-1-1 copy/mL
Cell singlet
Unique induced virion RNA
Induced virion CD4 T cell subset
TCM
TTM
TEM
Panels A, hemispheres
Unique induced
All other HIV DNA
1010 Cell 166, 1004–1015, August 11, 2016
0.02
Figure 5. RNA Sequences of Virions Induced
from Circulating TCM, TTM, and TEM Cells in HIV
Controllers
(A) x-y plots: proximity of each HIV DNA sequence from
circulating CD4 T cells or virions induced from TCM, TTM,
and TEM cells to the participant’s MRCA (x axis) and
nearest genetic neighbor (NN) from plasma virus (y axis).
HIV DNA sequences detected once are shown as gray
dots; recurrent HIV DNA sequences are shown as black
circles scaled by the abundance of the sequence; and
sequences detected in induced virion RNA but not in DNA
from a second aliquot of cells (i.e., ‘‘unique induced’’ viruses) are shown as stars. Where an induced virion RNA
sequence matched a recurrent HIV DNA sequence from
blood cells, the circle corresponding to that DNA
sequence is filled. The arrow shows one sequence from
participant S1270 containing a lethal deletion within
gp120. For alignment production, this deletion was
filled with the participant’s consensus sequence; the
measured genetic distance of this sequence to the
plasma virus NN is therefore an underestimate. Hemispheres: quantities of unique induced proviruses and all
other HIV DNA sequences in circulating CD4 T cells from
HIV controllers. Plots are scaled to show relative levels of
HIV DNA in circulating CD4 T cells from the five participants. For participants with undetectable unique induced
viruses from blood cells, the LOD of this measurement is
shown.
(B) Sequences in virions induced from blood cells in
participant S1270. Cyan indicates sequences from an
initial experiment; magenta indicates sequences from a
repeat experiment. Sequences in which relative insertions were excised or deletions filled in alignment
production are shown with gray arrows.
See also Figure S5.
0.0012 0.0007
HIV DNA copies/
106 cells
105
104
0.0004
104
103
102
0.0004
3
10
102
Follic. Other Blood
LN
LN
101
100
Follic. Other Blood
LN
LN
0.5245 0.0079
C
104
0.0004
Panels A-C
TCM
TTM
TEM
CD57
Non-GC TFH
GC TFH
103
102
101
100
10-1
Follic. Other Blood
LN
LN
TCM
CD57
Non-GC TFH
GC TFH
TEM
50
0
LI
R0
2
LI
R0
3
LI
R0
4
% HIV DNA copies
D
100
Follicular
Extrafollicular
Participant LIR03
Participant LIR02
E
studied, the relative genetic proximity of TCM-associated HIV
DNA sequences to plasma viruses was directly associated with
plasma viremia (Figure S7).
DISCUSSION
10-1
101
Spliced RNA copies/
DNA copy
0.3566 0.0002
B
Unspl. RNA copies/
DNA copy
A
In this study, we investigated the mechanisms of virus persistence in HIV controllers using a combination of genetic analyses
to track HIV replication and the persistence of HIV-infected cells
in vivo. We found that the populations of HIV-infected CD4 T cells
in blood and lymphoid tissue from these individuals differed
markedly from one another. The infected cell pool in blood
largely comprised archival proviruses within highly differentiated
cells that appeared to be clonally expanded and that occasionally expressed virions when stimulated. In sharp contrast, the
lymphoid tissue was often rich in TFH and other memory CD4
T cells bearing HIV genetic markers of recent infection and containing abundant virus transcripts. Despite these differences,
however, we also detected rare circulating CD4 T cells that inducibly expressed HIV proviruses with the genetic signature of an
actively replicating virus pool. These cells link the infected populations from blood and lymphoid tissue and may thus reflect the
hematogenous dissemination of newly infected cells. Therefore,
our findings suggest a single model in which HIV persists despite
natural virologic control by three interrelated mechanisms:
(1) ongoing infection of cells in lymphoid tissue, (2) survival and
recirculation of some of these cells, and (3) long-term persistence of proviruses in clonally expanded cells.
Our results support this model through key advances in
several areas. Consistent with previous studies of SIV-infected
macaques and HIV-infected long-term nonprogressors (Fukazawa et al., 2015; Perreau et al., 2013), we detected viruses
with genetic and transcriptional markers of active replication
most abundantly within PD1high, TFH-enriched cell populations
in hosts who control the virus spontaneously. However, by using
single-copy sequence analysis to identify this virus population,
we were also able to detect its dissemination to extrafollicular
LN cells. This implies either HIV transmission across follicle
boundaries or differentiation of infected TFH cells into non-TFH
cells. Furthermore, the genetic similarity among plasma viruses,
viruses in LN cells, and unique induced viruses from blood cells
suggests that some infected LN cells survive their initial
encounter with infectious virus long enough to recirculate. We
also identified blood TEM cells that appeared to be clonally
expanded as a source of archival virus in HIV controllers. Our
finding that inducible proviruses of recent origin were present
but rare amid the excess of archival proviruses in highly differentiated, clonally expanded blood cells explains the large genetic
50 M
Figure 6. HIV DNA and RNA Levels in LN CD4 T Cell Subsets
(A) Levels of HIV DNA measured by FCA in LN non-GC TFH and GC TFH; nonfollicular LN subsets (CD57+ subset collected only for participant LIR02); and
blood TCM, TTM, and TEM subsets.
(B and C) Copies of unspliced (B) and spliced (C) HIV RNAs in LN
memory CD4 T cell subsets, measured by qRT-PCR and normalized to
values in (A). Each cell subset is shown with a unique shape and colored by
participant. Undetectable values are plotted at the assay LOD with open
symbols.
(A)–(C) include results from blood CD4 TCM, TTM, and TEM subsets that are also
shown in Figure 1. Mann-Whitney p values are shown.
(D) The percentage of all HIV DNA copies in LN memory CD4 T cells from each
participant detected in each subset.
(E) HIV RNA+ LN cells detected by in situ hybridization using 35S-labeled riboprobes in two study participants. White arrows indicate examples of HIV
RNA+ cells. Some such cells were associated with areas of diffusely increased
signal corresponding to the follicular dendritic cell network (follicular); others
were outside such areas (extrafollicular).
See also Figures S6 and S7 and Table S3.
Cell 166, 1004–1015, August 11, 2016 1011
0.02
EM
T
TM
T
CM
CM
0.00
0.06
T
0.03
0.04
T
0.03
0.00
0.00
0.06
EM
Plasma P <0.0001
MRCA P = 0.0035
All P < 0.05
T
0.06
Distance to TFH NN
Distance to plasma NN
Figure 7. Analysis of HIV DNA Sequences
and Host Gene Expression in LN CD4
T Cell Subsets from HIV Controllers
Participant LIR02
A
EM
TM
T
T
CM
0.00
0.10
T
0.05
0.03
CM
0.00
0.00
0.06
T
0.05
All P < 0.05
0.09
EM
Plasma P <0.0001
MRCA P = 0.1349
T
0.10
Distance to TFH NN
Distance to plasma NN
Participant LIR03
EM
T
TM
T
CM
0.00
0.08
T
0.04
0.02
CM
0.00
0.00
0.04
T
0.04
All P < 0.05
0.06
EM
Plasma P = 0.0008
MRCA P = 0.0036
T
0.08
Distance to TFH NN
Distance to plasma NN
Participant LIR04
Distance to MRCA
Panels A, left
B
LN/PB TN
0.2
Blood cell DNA sequences
Repeat, 10-102 copies/mL
Repeat, 10-1-1 copy/mL
Singlet
PC2 (0.014)
Repeat, 1-10 copies/mL
GC/non-GC TFH
0.1
TFH cell DNA sequences
(GC and non-GC)
Repeat, High rel. abund.
Repeat, Mod. rel. abund.
PB TTM
0.0
LN TEM
-0.1
PB TEM
-0.2
-0.134
-0.132
Panels A, right
-0.128
PC1 (0.939)
Repeat, Low rel. abund.
Singlet
-0.130
Panel B
PB TEM
PB TN
LN TEM
Lymph node
LN TN
Non-GC TFH
Blood
PB TTM
GC TFH
1012 Cell 166, 1004–1015, August 11, 2016
(A) Left: proximity of each HIV DNA sequence from
LN GC and non-GC TFH cells to each participant’s
MRCA (x axis) and plasma virus NN sequence
(y axis). HIV DNA sequences from blood cells are
shown as in Figure 5; HIV DNA sequences from GC
and non-GC TFH cells are shown as green-filled
circles scaled by their relative abundance in LN.
Sequences from GC and non-GC TFH cells were
compared to those from blood cells for genetic
distance to plasma virus NN and MRCA sequences. Mann-Whitney p values for these comparisons are shown. Right: proximity of each HIV
DNA sequence from non-TFH TCM and TEM LN cells
and TCM, TTM, and TEM blood cells to the NN
sequence from GC and non-GC TFH cells. All
Mann-Whitney p values for comparisons between
LN cell subsets and blood cell subsets are <0.05.
(B) PCA of transcriptomes from blood (PB) and LN
CD4 T cell subsets. Clusters of symbols representing samples of the same cell subset from
multiple study participants are demarcated with
dashed boundaries.
See also Figures S6 and S7 and Table S3.
distance between HIV sequences in blood cells and plasma
observed previously in controllers (Bailey et al., 2006; O’Connell
et al., 2010). Finally, although prior studies have found expanded
clones of HIV-infected cells ex vivo (Cohn et al., 2015; Maldarelli
et al., 2014; Wagner et al., 2014), virus production has been
shown previously from only one clone in one individual (Maldarelli et al., 2014; Simonetti et al., 2016) and never in an HIV
controller. Although CD4 T cell clonal proliferation may involve
transcriptional processes that elicit HIV expression and thus
select for lethal mutations among expanded proviruses, the
dissociation of virion production from cellular stimulation supported by our findings provides one mechanism for proliferative
self-renewal of HIV-producing CD4 T cells in vivo.
The heterogeneity that we observed among HIV-infected TEM
cells from HIV controllers was also noteworthy. Most HIV DNA
sequences in circulating TEM cells appeared to be associated
with expanded cellular clones. We propose that this reflects
the basic biology of TEM cells, which may also harbor infected
cells that have clonally expanded in ART-treated individuals
(von Stockenstrom et al., 2015). The relatively high levels of
HIV DNA we found in blood TEM cells distinguish HIV controllers
from ART-treated individuals (Chomont et al., 2009) and may
stem from higher levels of virus replication and immune activation (Hunt et al., 2008, 2011; Krishnan et al., 2014) stimulating
HIV-infected cells to proliferate and differentiate in HIV controllers. At the same time, by sampling lymphoid tissue or virions
induced from blood cells instead of blood cell DNA, we uncovered viruses of recent origin in rare TEM cells. Therefore, it appears that both archival and actively replicating virus populations
persist within TEM cells in HIV controllers, but by distinct mechanisms. Importantly, lack of CD27 expression represents an inclusive definition for the TEM subset, and finer distinctions among
subsets of CD27– cells may yield additional insights in future
studies. Furthermore, the lifespan of circulating CD27– cells carrying inducible, recently acquired proviruses remains to be
determined. Nonetheless, these considerations make clear
that targeting less differentiated TCM and TTM cells is unlikely to
eliminate all potentially infectious proviruses in HIV controllers.
Despite these diverse mechanisms of persistence, we detected profound imprints of effective antiviral defenses among
HIV DNA sequences from HIV controllers. In peripheral blood,
restricted replication of the virus was associated with a predominance of recurrent sequences within clonally expanded cells, a
scarcity of cell-associated virus transcripts, and a small number
of cells harboring unique induced viruses. While expanded
G-to-A hypermutant viruses were abundant in circulating infected cells from some HIV controllers, this most likely reflects
a predominance of non-replicating proviruses in these individuals, rather than excess activity of APOBEC3G (Abdel-Mohsen
et al., 2013). In lymphoid tissue, the enrichment for recently infected cells containing abundant HIV transcripts within TFH populations is consistent with a key role for virus-specific CD8+
T cells in suppressing HIV replication outside follicles (Fukazawa
et al., 2015). In fact, we documented this effect in individuals
lacking major protective HLA-B alleles, suggesting that the suppression of extrafollicular virus replication may occur widely
among HIV controllers. Finally, we propose that the relationship
between plasma viremia and the genetic proximity of blood TCM-
associated HIV DNA sequences to plasma viruses in HIV controllers reflects reduced dissemination of recently infected CD4
T cells in individuals with greater virologic control. When ongoing
virus replication is very limited, rare recently infected cells—
more readily detected by sequencing in the TCM subset than in
the more heavily expanded TTM and TEM subsets—may occur
too rarely for detection in a single blood sample. Taken to an
extreme, the limited dissemination of recently infected CD4
T cells in blood may prevent the virus from spreading throughout
the lymphoid tissue compartment, as for one participant in our
study. Thus, in rare cases, natural antiviral responses may
restrict the regional anatomic distribution of the virus.
Our findings reflect in vivo properties of HIV-infected CD4
T cell populations that may be generalizable to all infected individuals. Of particular interest is the uncertain significance of
expanded CD4 T cell clones as barriers to cure. On the one
hand, the rarity of virion production from expanded proviruses
in our study mirrors results in ART-treated individuals, calling
into question whether this mechanism alone could account for
the near certainty of rebound viremia upon ART interruption.
On the other hand, the recent temporal clustering of unique
induced proviruses in our study suggests that newly infected
cells recirculating from lymphoid tissue may disappear quickly
if not maintained by clonal expansion. Complicating attempts
to resolve these findings, however, is the evident difficulty of eliciting virus expression from some inducible proviruses in HIV controllers, as shown previously in ART-treated non-controllers (Ho
et al., 2013). In addition to preventing full characterization of
inducible proviruses ex vivo, this difficulty may reflect natural
heterogeneity in the cellular response to stimulation that serves
to protect some proviruses from antiviral defenses in vivo. Therefore, while our findings support the role of the B cell follicle in
ongoing HIV replication in HIV controllers, we also demonstrate
extrafollicular mechanisms that could maintain and disseminate
the virus even if its replication inside follicles were disrupted.
Were any approach to a functional or sterilizing cure to succeed
either in HIV controllers or in non-controllers, it would need to
target these multiple distinct cellular processes.
EXPERIMENTAL PROCEDURES
Peripheral Blood Mononuclear Cell and LN Samples
Participants were recruited from UCSF and NIAID and gave informed consent
for all procedures. Studies were approved by the UCSF and NIAID institutional
review boards. Peripheral blood mononuclear cells (PBMCs) were isolated
from blood or leukapheresis by density gradient centrifugation. LN excisional biopsy samples were sectioned and filtered to generate single-cell
suspensions.
Fluorescence-Activated Cell Sorting
Viable PBMCs were stained and sorted on a FACSAria (BD). Subset definitions
were as shown in Figure S1 for PBMCs and in Figure S6 for LN cells.
Nucleic Acid Extraction
RNA and DNA were extracted in separate fractions from sorted cells lysed in
RNAzol RT (MRC), according to the manufacturer’s instructions.
Recovery of HIV Virion RNA
Virion RNA was extracted from whole plasma or pelleted virions using the
QiaAmp vRNA mini kit (QIAGEN), according to the manufacturer’s instructions.
Cell 166, 1004–1015, August 11, 2016 1013
Fluorescence-Assisted Clonal Amplification for Single-Copy HIV
Quantification and Sequencing
Cell DNA or reverse-transcribed HIV virion RNA was amplified at limiting dilution
in replicate PCR wells using primers targeting HXB2 positions 6908–7517, with
SYBR green I as marker of double-stranded DNA (dsDNA). Amplification was detected by real-time fluorescence and confirmed by melt curve (Figure S2). Single
copies of HIV DNA were enumerated for each sample by limiting dilution calculations. Amplified env products were reamplified by nested PCR and Sanger
sequenced. Because all primers in these PCRs targeted regions within the HIV
genome, the resulting sequence data were considered to reflect all HIV DNA
copies in samples from cells, including both integrated and unintegrated forms.
Quantification of HIV RNAs
Cellular total RNA samples were tested for HIV RNA by qRT-PCR for unspliced
(gag) RNA or transcripts spliced between the SD1 and SA4 sites (Purcell and
Martin 1993). Copies of RNA were enumerated using standard curves generated from dilutions of synthetic RNAs.
HIV Sequence Analysis
Sanger sequence reads defining single HIV sequences spanning HXB2 base
positions 7011–7502 were edited, analyzed for diversity by subset within
each participant, aligned by participant, and subjected to phylogenetic analysis after identification and removal of G-to-A hypermutated sequences.
T Cell Receptor Beta Gene Deep Sequencing
Sequencing and annotation of expressed TCRB genes were performed as
described (Gros et al., 2014). The normalized Shannon diversity was calculated for the set of TCRB sequences from each CD4 T cell subset by the
same method used for populations of HIV sequences (see Supplemental
Experimental Procedures), determining the maximum Shannon diversity for
each sample based on the number of cells used in library preparation.
HIV Integration Site Deep Sequencing
Integration site analysis was performed as described (Maldarelli et al., 2014).
Sequencing of Viruses Induced In Vitro
Sorted TCM, TTM, and TEM cells were stimulated in culture through the T cell receptor in the presence of antiretrovirals, and virions were pelleted from culture
supernatants for RNA extraction after 4 and 8 days of culture. Virion RNA was
reverse transcribed and amplified for single-copy sequencing by FCA.
In Situ Hybridization
Hybridization of 35S-labeled riboprobes for the detection of HIV RNA+ cells in
LN tissue slices available from two study participants was performed as
described (Rothenberger et al., 2015).
Whole-Transcriptome Sequencing
Messenger RNA libraries were constructed as described (Sandler et al., 2014)
and sequenced in 2-3-75-base, paired-end runs on an Illumina HiSeq. Highquality reads were mapped to the Hg19 reference using Tophat (v.2.0.8) with
a reference annotation (Ensembl ‘‘Homo_sapiens.GRCh37.74.gtf’’). Samples
with low map rates were discarded. Transcript abundance was determined
with Cufflinks (v2.1.1). PCA was performed using ‘‘princomp’’ in the R ‘‘stats’’
package. The abundance of each transcript was log transformed before PCA.
To limit the influence of low transcript abundance levels, the level of each transcript was set to 1 if the measured level was <1.
ACCESSION NUMBERS
The accession numbers for the HIV Sanger sequencing data reported in this
paper are GenBank: KX390978-KX394124 The accession number for the
whole transcriptome sequencing data reported in this paper is GEO:
GSE83482. The accession number for the TCR deep sequencing data reported in this paper is NCBI Short Read Archive: SRP076794. The accession
number for the HIV integration site sequencing reported in this paper can be
found by searching the NCI HIV Dynamics and Replication program website
at https://rid.ncifcrf.gov/index.php using the paper’s PubMed ID.
1014 Cell 166, 1004–1015, August 11, 2016
SUPPLEMENTAL INFORMATION
Supplemental Information includes Supplemental Experimental Procedures,
seven figures, and four tables and can be found with this article online at
http://dx.doi.org/10.1016/j.cell.2016.06.039.
AUTHOR CONTRIBUTIONS
Conceptualization, E.A.B. and D.C.D.; Methodology, E.A.B., X.W., J.P.C., T.S.,
and D.C.D.; Investigation, E.A.B., S.D., L.S., G.W., D.W., X.W., A.R.H., F.L.,
D.A., A.V., D.B., and K.N.-M.; Formal Analysis, S.D. and J.H.; Resources,
M.H., R.H., S.A.M., M.C., S.M., and S.G.D.; Writing – Original Draft, E.A.B.
and D.C.D.; Writing–Review and Editing, E.A.B., S.H.H., and D.C.D.; Supervision, R.A.K., F.M., S.H.H., S.G.D., T.S., and D.C.D.
ACKNOWLEDGMENTS
We thank the study participants for their involvement in the study. We thank
S. Kosakovsky-Pond for help with phylogenetic analysis and C. Petrovas for helpful discussions. D.C.D. and E.A.B. are funded by the NIH Intramural Research
Program. D.C.D. is also funded by the NIAID Division of AIDS and the NIH Office
of AIDS Research. Additional funding came from AIDS Vaccine Discovery grant
OPP1032325 from the Bill and Melinda Gates Foundation (to R.A.K.), the Delaney
AIDS Research Enterprise (AI096109 to S.G.D.), NIAID K24 (AI069994 to S.G.D.),
the UCSF/Gladstone Institute of Virology & Immunology CFAR (P30 AI027763 to
S.G.D.), and federal funds from the NCI (to F.M. and S.H.H.).
Received: February 25, 2016
Revised: May 9, 2016
Accepted: June 20, 2016
Published: July 21, 2016
REFERENCES
Abdel-Mohsen, M., Raposo, R.A., Deng, X., Li, M., Liegler, T., Sinclair, E.,
Salama, M.S., Ghanem, Hel.-D., Hoh, R., Wong, J.K., et al. (2013). Expression profile of host restriction factors in HIV-1 elite controllers. Retrovirology
10, 106.
Archin, N.M., Liberty, A.L., Kashuba, A.D., Choudhary, S.K., Kuruc, J.D.,
Crooks, A.M., Parker, D.C., Anderson, E.M., Kearney, M.F., Strain, M.C.,
et al. (2012). Administration of vorinostat disrupts HIV-1 latency in patients
on antiretroviral therapy. Nature 487, 482–485.
Bailey, J.R., Williams, T.M., Siliciano, R.F., and Blankson, J.N. (2006). Maintenance of viral suppression in HIV-1-infected HLA-B*57+ elite suppressors
despite CTL escape mutations. J. Exp. Med. 203, 1357–1369.
Blankson, J.N., Bailey, J.R., Thayil, S., Yang, H.C., Lassen, K., Lai, J., Gandhi,
S.K., Siliciano, J.D., Williams, T.M., and Siliciano, R.F. (2007). Isolation and
characterization of replication-competent human immunodeficiency virus
type 1 from a subset of elite suppressors. J. Virol. 81, 2508–2518.
Boufassa, F., Lechenadec, J., Meyer, L., Costagliola, D., Hunt, P.W., Pereyra,
F., Deeks, S., Pancino, G., Taulera, O., Lichterfeld, M., et al.; ANRS CO18 HIV
Controllers Cohort; Cascade Collaboration in Eurocoord; SCOPE Cohort;
International HIV Controllers Study (2014). Blunted response to combination
antiretroviral therapy in HIV elite controllers: an international HIV controller
collaboration. PLoS ONE 9, e85516.
Chomont, N., El-Far, M., Ancuta, P., Trautmann, L., Procopio, F.A., YassineDiab, B., Boucher, G., Boulassel, M.R., Ghattas, G., Brenchley, J.M., et al.
(2009). HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat. Med. 15, 893–900.
Chun, T.W., Carruth, L., Finzi, D., Shen, X., DiGiuseppe, J.A., Taylor, H., Hermankova, M., Chadwick, K., Margolick, J., Quinn, T.C., et al. (1997a). Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection.
Nature 387, 183–188.
Chun, T.W., Stuyver, L., Mizell, S.B., Ehler, L.A., Mican, J.A., Baseler, M.,
Lloyd, A.L., Nowak, M.A., and Fauci, A.S. (1997b). Presence of an inducible
HIV-1 latent reservoir during highly active antiretroviral therapy. Proc. Natl.
Acad. Sci. USA 94, 13193–13197.
Chun, T.W., Shawn Justement, J., Murray, D., Kim, C.J., Blazkova, J., Hallahan, C.W., Benko, E., Costiniuk, C.T., Kandel, G., Ostrowski, M., et al.
(2013). Effect of antiretroviral therapy on HIV reservoirs in elite controllers.
J. Infect. Dis. 208, 1443–1447.
Mens, H., Kearney, M., Wiegand, A., Shao, W., Schønning, K., Gerstoft, J.,
Obel, N., Maldarelli, F., Mellors, J.W., Benfield, T., and Coffin, J.M. (2010).
HIV-1 continues to replicate and evolve in patients with natural control of
HIV infection. J. Virol. 84, 12971–12981.
Migueles, S.A., and Connors, M. (2015). Success and failure of the cellular immune response against HIV-1. Nat. Immunol. 16, 563–570.
Cohn, L.B., Silva, I.T., Oliveira, T.Y., Rosales, R.A., Parrish, E.H., Learn, G.H.,
Hahn, B.H., Czartoski, J.L., McElrath, M.J., Lehmann, C., et al. (2015). HIV-1
integration landscape during latent and active infection. Cell 160, 420–432.
O’Connell, K.A., Brennan, T.P., Bailey, J.R., Ray, S.C., Siliciano, R.F., and
Blankson, J.N. (2010). Control of HIV-1 in elite suppressors despite ongoing
replication and evolution in plasma virus. J. Virol. 84, 7018–7028.
Finzi, D., Hermankova, M., Pierson, T., Carruth, L.M., Buck, C., Chaisson, R.E.,
Quinn, T.C., Chadwick, K., Margolick, J., Brookmeyer, R., et al. (1997). Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 278, 1295–1300.
Perreau, M., Savoye, A.L., De Crignis, E., Corpataux, J.M., Cubas, R., Haddad,
E.K., De Leval, L., Graziosi, C., and Pantaleo, G. (2013). Follicular helper T cells
serve as the major CD4 T cell compartment for HIV-1 infection, replication, and
production. J. Exp. Med. 210, 143–156.
Finzi, D., Blankson, J., Siliciano, J.D., Margolick, J.B., Chadwick, K., Pierson,
T., Smith, K., Lisziewicz, J., Lori, F., Flexner, C., et al. (1999). Latent infection of
CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in
patients on effective combination therapy. Nat. Med. 5, 512–517.
Purcell, D.F., and Martin, M.A. (1993). Alternative splicing of human immunodeficiency virus type 1 mRNA modulates viral protein expression, replication,
and infectivity. J. Virol. 67, 6365–6378.
Fukazawa, Y., Lum, R., Okoye, A.A., Park, H., Matsuda, K., Bae, J.Y., Hagen,
S.I., Shoemaker, R., Deleage, C., Lucero, C., et al. (2015). B cell follicle sanctuary permits persistent productive simian immunodeficiency virus infection in
elite controllers. Nat. Med. 21, 132–139.
Gros, A., Robbins, P.F., Yao, X., Li, Y.F., Turcotte, S., Tran, E., Wunderlich,
J.R., Mixon, A., Farid, S., Dudley, M.E., et al. (2014). PD-1 identifies the patient-specific CD8+ tumor-reactive repertoire infiltrating human tumors.
J. Clin. Invest. 124, 2246–2259.
Hatano, H., Yukl, S.A., Ferre, A.L., Graf, E.H., Somsouk, M., Sinclair, E., AbdelMohsen, M., Liegler, T., Harvill, K., Hoh, R., et al. (2013). Prospective antiretroviral treatment of asymptomatic, HIV-1 infected controllers. PLoS Pathog. 9,
e1003691.
Hermankova, M., Siliciano, J.D., Zhou, Y., Monie, D., Chadwick, K., Margolick,
J.B., Quinn, T.C., and Siliciano, R.F. (2003). Analysis of human immunodeficiency virus type 1 gene expression in latently infected resting CD4+ T lymphocytes in vivo. J. Virol. 77, 7383–7392.
Ho, D.D., Neumann, A.U., Perelson, A.S., Chen, W., Leonard, J.M., and Markowitz, M. (1995). Rapid turnover of plasma virions and CD4 lymphocytes in
HIV-1 infection. Nature 373, 123–126.
Ho, Y.C., Shan, L., Hosmane, N.N., Wang, J., Laskey, S.B., Rosenbloom, D.I.,
Lai, J., Blankson, J.N., Siliciano, J.D., and Siliciano, R.F. (2013). Replicationcompetent noninduced proviruses in the latent reservoir increase barrier to
HIV-1 cure. Cell 155, 540–551.
Hunt, P.W., Brenchley, J., Sinclair, E., McCune, J.M., Roland, M., Page-Shafer, K., Hsue, P., Emu, B., Krone, M., Lampiris, H., et al. (2008). Relationship
between T cell activation and CD4+ T cell count in HIV-seropositive individuals
with undetectable plasma HIV RNA levels in the absence of therapy. J. Infect.
Dis. 197, 126–133.
Hunt, P.W., Landay, A.L., Sinclair, E., Martinson, J.A., Hatano, H., Emu, B.,
Norris, P.J., Busch, M.P., Martin, J.N., Brooks, C., et al. (2011). A low T regulatory cell response may contribute to both viral control and generalized immune activation in HIV controllers. PLoS ONE 6, e15924.
Julg, B., Pereyra, F., Buzón, M.J., Piechocka-Trocha, A., Clark, M.J., Baker,
B.M., Lian, J., Miura, T., Martinez-Picado, J., Addo, M.M., and Walker, B.D.
(2010). Infrequent recovery of HIV from but robust exogenous infection of activated CD4(+) T cells in HIV elite controllers. Clin. Infect. Dis. 51, 233–238.
Kim, M., and Siliciano, R.F. (2016). Reservoir expansion by T-cell proliferation
may be another barrier to curing HIV infection. Proc. Natl. Acad. Sci. USA 113,
1692–1694.
Krishnan, S., Wilson, E.M., Sheikh, V., Rupert, A., Mendoza, D., Yang, J., Lempicki, R., Migueles, S.A., and Sereti, I. (2014). Evidence for innate immune system
activation in HIV type 1-infected elite controllers. J. Infect. Dis. 209, 931–939.
Maldarelli, F., Wu, X., Su, L., Simonetti, F.R., Shao, W., Hill, S., Spindler, J., Ferris, A.L., Mellors, J.W., Kearney, M.F., et al. (2014). HIV latency. Specific HIV
integration sites are linked to clonal expansion and persistence of infected
cells. Science 345, 179–183.
Rasmussen, T.A., Tolstrup, M., Brinkmann, C.R., Olesen, R., Erikstrup, C., Solomon, A., Winckelmann, A., Palmer, S., Dinarello, C., Buzon, M., et al. (2014).
Panobinostat, a histone deacetylase inhibitor, for latent-virus reactivation in
HIV-infected patients on suppressive antiretroviral therapy: a phase 1/2, single
group, clinical trial. Lancet HIV 1, e13–e21.
Rothenberger, M.K., Keele, B.F., Wietgrefe, S.W., Fletcher, C.V., Beilman,
G.J., Chipman, J.G., Khoruts, A., Estes, J.D., Anderson, J., Callisto, S.P.,
et al. (2015). Large number of rebounding/founder HIV variants emerge from
multifocal infection in lymphatic tissues after treatment interruption. Proc.
Natl. Acad. Sci. USA 112, E1126–E1134.
Routy, J.P., Tremblay, C.L., Angel, J.B., Trottier, B., Rouleau, D., Baril, J.G.,
Harris, M., Trottier, S., Singer, J., Chomont, N., et al. (2012). Valproic acid in
association with highly active antiretroviral therapy for reducing systemic
HIV-1 reservoirs: results from a multicentre randomized clinical study. HIV
Med. 13, 291–296.
Salgado, M., Brennan, T.P., O’Connell, K.A., Bailey, J.R., Ray, S.C., Siliciano,
R.F., and Blankson, J.N. (2010). Evolution of the HIV-1 nef gene in HLA-B*57
positive elite suppressors. Retrovirology 7, 94.
Sandler, N.G., Bosinger, S.E., Estes, J.D., Zhu, R.T., Tharp, G.K., Boritz, E.,
Levin, D., Wijeyesinghe, S., Makamdop, K.N., del Prete, G.Q., et al. (2014).
Type I interferon responses in rhesus macaques prevent SIV infection and
slow disease progression. Nature 511, 601–605.
Simonetti, F.R., Sobolewski, M.D., Fyne, E., Shao, W., Spindler, J., Hattori, J.,
Anderson, E.M., Watters, S.A., Hill, S., Wu, X., et al. (2016). Clonally expanded
CD4+ T cells can produce infectious HIV-1 in vivo. Proc. Natl. Acad. Sci. USA
113, 1883–1888.
Søgaard, O.S., Graversen, M.E., Leth, S., Olesen, R., Brinkmann, C.R., Nissen,
S.K., Kjaer, A.S., Schleimann, M.H., Denton, P.W., Hey-Cunningham, W.J.,
et al. (2015). The depsipeptide romidepsin reverses HIV-1 latency in vivo.
PLoS Pathog. 11, e1005142.
Spivak, A.M., Andrade, A., Eisele, E., Hoh, R., Bacchetti, P., Bumpus, N.N.,
Emad, F., Buckheit, R., 3rd, McCance-Katz, E.F., Lai, J., et al. (2014). A pilot
study assessing the safety and latency-reversing activity of disulfiram in
HIV-1-infected adults on antiretroviral therapy. Clin. Infect. Dis. 58, 883–890.
von Stockenstrom, S., Odevall, L., Lee, E., Sinclair, E., Bacchetti, P., Killian, M.,
Epling, L., Shao, W., Hoh, R., Ho, T., et al. (2015). Longitudinal genetic characterization reveals that cell proliferation maintains a persistent HIV type 1 DNA
pool during effective HIV therapy. J. Infect. Dis. 212, 596–607.
Wagner, T.A., McLaughlin, S., Garg, K., Cheung, C.Y., Larsen, B.B., Styrchak,
S., Huang, H.C., Edlefsen, P.T., Mullins, J.I., and Frenkel, L.M. (2014). HIV latency. Proliferation of cells with HIV integrated into cancer genes contributes
to persistent infection. Science 345, 570–573.
Walker, B.D., and Yu, X.G. (2013). Unravelling the mechanisms of durable control of HIV-1. Nat. Rev. Immunol. 13, 487–498.
Wong, J.K., Hezareh, M., Günthard, H.F., Havlir, D.V., Ignacio, C.C., Spina,
C.A., and Richman, D.D. (1997). Recovery of replication-competent HIV
despite prolonged suppression of plasma viremia. Science 278, 1291–1295.
Cell 166, 1004–1015, August 11, 2016 1015
Supplemental Figures
A
E
24.1
CD4
SSC-A
71.0
FSC-A
CD8
B
F
85.5
FSC-H
SSC-A
99.9
FSC-A
Lineage dump
C
G
TN
23.7
CD3
CD27
74.5
Viability dump
DP
16.0
TEM
43.2
CD45RO
D
H
TCM
47.9
CCR7
CD14/CD11c
99.4
CD4
TTM
41.3
SSC-A
Figure S1. Gating for Fluorescence-Activated Cell Sorting of CD4 T Cell Subsets from Blood, Related to Figure 1
(A–F) Leukocytes (A) not part of multi-cell conjugates (B) that were viable and stained with the T cell marker CD3 (C) but not myeloid cell markers CD14 and CD11c
(D), the cytotoxic T cell marker CD8 (E), or lineage dump markers including CD20, CD56, and TCR-gd (F) were divided by CD27 and CD45RO staining and
collected as TN (G, top-left gate) or TEM (G, bottom gate) subsets.
(H) Cells that were double-positive (DP; G, top-right gate) for CD27 and CD45RO were further divided by CCR7 staining (H) into TCM (top) and TTM (bottom)
subsets. Both CD4high and CD4low cells (E) were included in sorted subsets to ensure collection of any cells with CD4 downregulation due to active HIV infection
and Nef activity. Numbers on plots represent percentages of plotted cells falling within the gates shown.
Cell 166, 1004–1015, August 11, 2016 S1
Fluorescence
A
Cycle #
Deriv. Fluorescence
B
Temperature
C
M- - + - - + - - - - - - - - - - + + - - ++ - +
M- + - - - - - + -+ - ++ - - - - - + - - + - M+ - - + - - - ++ - ++ - - + - - - - - - - + M+ -+ - - - - - - - - - - - + - - + - - - + - -
D
HIV DNA copies detected
50
40
30
20
10
0
FCA
gag
Figure S2. Fluorescence-Assisted Clonal Amplification, Related to Figure 1
Cell-associated HIV DNA or plasma virion cDNA was plated in 384-well plates at an estimated 0.3 copies/well and amplified by PCR using primers flanking a
fragment of the env gene including HXB2 base positions 6908-7517, in the presence of SYBR Green I as a marker of dsDNA.
(A and B) Wells showing fluorescence amplification (A) and amplicon melting temperatures between 76.0-82.0 C (B) were considered to be positive.
(legend continued on next page)
S2 Cell 166, 1004–1015, August 11, 2016
(C) Specificity of FCA analyzed by agarose gel electrophoresis. Lanes corresponding to FCA wells from (A) and (B) that contained amplification products with
melting temperatures in the target range are indicated with ‘‘+’’ symbols. M, molecular weight marker. Gel images were cropped to include only the relevant size
range. Results are shown for a sample consisting of linearized HIV plasmid diluted into HIV-negative PBMC DNA at 1 copy per 1,000 cell equivalents.
(D) The sensitivity of FCA was similar to the sensitivity of HIV gag taqman PCR for single copies of HIV in equal aliquots of this sample run in 96 PCR wells for each
test (D).
Cell 166, 1004–1015, August 11, 2016 S3
D
A
J
G
M
0.02
0.02
0.02
0.02
0.02
B
E
H
K
N
0.02
0.05
0.02
0.05
C
F
I
L
0.02
Legend
0.02
0.02
0.05
0.02
Plasma
TCM
TTM
TEM
Figure S3. Phylogenetic Trees Representing Single-Copy HIV Sequence Results Obtained by FCA from Plasma Virions and from CellAssociated DNA of CD4 TCM, TTM, and TEM in HIV Controllers, Related to Figure 2
Each tree represents results from a single study participant, rooted on the HXB2 sequence. Sequences from each participant showing G-to-A hypermutation are
indicated as detached branches in the box associated with that participant’s tree. Sequences in which relative insertions were excised or deletions filled in
alignment production are shown with gray arrows.
(A–N) Participant identifiers are as follows: (A) S1492, (B) S1270, (C) S1788, (D) S1548, (E) LIR04, (F) V907, (G) S1475, (H) S1349, (I) S1495, (J) S1541, (K) LIR02, (L)
LIR01, (M) V912, and (N) LIR03.
S4 Cell 166, 1004–1015, August 11, 2016
A
C
E
0.05
0.02
0.05
B
D
F
0.02
0.02
0.05
Legend
Plasma
TCM
TTM
TEM
Figure S4. Phylogenetic Trees Representing Single-Copy HIV Sequence Results Obtained by FCA from Plasma Virions and from CellAssociated DNA of CD4 TCM, TTM, and TEM in Non-controllers, Related to Figure 2
Each tree represents results from a single study participant, rooted on the HXB2 sequence. Sequences from each participant showing G-to-A hypermutation are
indicated as detached branches in the box associated with that participant’s tree. Sequences in which relative insertions were excised or deletions filled in
alignment production are shown with gray arrows.
(A–F) Participant identifiers are as follows: (A) V016, (B) V257, (C) V621, (D) V667, (E) V965, and (F) V723.
Cell 166, 1004–1015, August 11, 2016 S5
00
10
60
0
0
20
40
30
20
10
0
Recurrent HIV DNA sequence
S1495_14
S1495_30
S1495_32
S1495_34
S1495_43
S1495_55
S1495_58
V912_14
V912_16
V912_25
V912_26
V912_29
V912_30
V912_41
V912_47
V912_8
V912_9
S1788_29
S1788_3
S1349_10
S1349_17
S1349_6
S1349_7
S1349_9
S1270_1
S1270_2
S1270_5
S1270_6
S1270_7
S1270_8
S1270_9
Cells in stimulation culture
Figure S5. Calculated Numbers of Cells from Presumptive Expanded CD4 T Cell Clones Included in Virion Induction Cultures of CD4 TCM,
TTM, and TEM Cells from HIV Controllers, Related to Figure 5
Each presumptive expanded clone is defined by a distinct HIV DNA sequence (y axis labels). Blue bars indicate TCM cultures, green bars indicate TTM cultures, and
red bars indicate TEM cultures. Filled bars indicate sequences also detected in virions induced from the given CD4 T cell subset.
S6 Cell 166, 1004–1015, August 11, 2016
E
I
10-2
10-3
10-4
CD8
29.8
G
37.8 57.1
0.59 4.08
EM
T
T
FH
FSC-A
C
0.08
Avg. pairwise dist. plas.
CD4
FSC-H
J
CM
10-5
TM
83.2
10-1
T
F
Blood
100
T
CM
N
on CD
-G 57
C
T
G FH
C
T
90.9
Myeloid dump
Lymph Node
101
T
CD4
SSC-A
FSC-A
B
HIV DNA copies/100 cells
99.6
75.8
EM
A
0.06
0.04
0.02
Viability dump
D
C
N
C
1.50 0.37
96.8 1.31
CD57
99.6
CD4
K
CD45RO
H
Avg. plas. dist. to MRCA
CD3
CD27
0.00
0.15
0.10
0.05
C
N
PD1
C
0.00
CD20
Figure S6. Study Participant LIR01, Related to Figures 6 and 7
(A–H) Gating for fluorescence-activated cell sorting (FACS) of CD4 T cell subsets from lymph node. Leukocytes (A) not part of multi-cell conjugates (B) that were
viable and stained with the T cell marker CD3 (C) but not B cell marker CD20 (D), myeloid cell markers CD14 and CD11c (E), or the cytotoxic T cell marker CD8 (F)
were divided by CD27 and CD45RO staining and collected as TN (G, top-left quadrant) or TEM (G, bottom quadrants) subsets. Cells that were double-positive for
CD27 and CD45RO (G, top-right quadrant) were further divided by CD57 and PD1 staining (H) into CD57+ TCM (top-left quadrant), GC TFH (top-right quadrant) and
non-GC TFH (bottom-right quadrant) subsets. Both CD4high and CD4low cells (F) were included in sorted subsets to ensure collection of any cells with CD4
downregulation due to active HIV infection and Nef activity. Numbers on plots represent percentages of plotted cells falling within the gates or quadrants shown.
(I) Ex vivo quantification of HIV DNA in memory CD4 T cell subsets from lymph node and blood. Undetectable values are plotted at the assay LOD with open bars.
(J) Average pairwise genetic distances among plasma virus sequences from all study donors, with the value for participant LIR01 shown in dark blue.
(K) Average genetic distances to MRCA of plasma virus sequences from all study donors, with the value for participant LIR01 shown in dark blue. C, HIV
controllers; NC, non-controllers.
(I) includes results from blood CD4 TCM, TTM, and TEM subsets that are also shown in Figure 1. (K) shows results that are also shown in Figure 3.
Cell 166, 1004–1015, August 11, 2016 S7
Plasma HIV RNA (copies/mL)
1500
r = 0.5453
P = 0.0469
1000
500
0
0.5
1.0
1.5
2.0
TCM relative proximity to plasma
Figure S7. Correlation between the Relative Genetic Proximity of Blood TCM Cell-Associated HIV DNA Sequences to the Plasma Virus and
Plasma HIV RNA Levels in HIV Controllers, Related to Figures 6 and 7
Genetic proximity to the plasma virus was defined as the reciprocal of the average genetic distance between HIV DNA sequences in a given subset and plasma
virus RNA sequences. The genetic proximity to the plasma virus of HIV DNA sequences from the TCM subset was normalized to the corresponding value for HIV
DNA sequences from the TEM subset in the same individual to determine the relative genetic proximity of TCM cell-associated HIV DNA sequences to the plasma
virus for that individual. For participant S1270, who had no detectable HIV DNA in blood TCM cells, the relative genetic proximity of blood TCM cell-associated HIV
DNA sequences to the plasma virus was set equal to the lowest value among the remaining participants. This was done to reflect the absence of HIV DNA
sequences closely related to the plasma virus within the blood TCM cell subset in this individual. Spearman r and p values are shown.
S8 Cell 166, 1004–1015, August 11, 2016
Cell, Volume 166
Supplemental Information
Multiple Origins of Virus Persistence
during Natural Control of HIV Infection
Eli A. Boritz, Samuel Darko, Luke Swaszek, Gideon Wolf, David Wells, Xiaolin Wu, Amy R.
Henry, Farida Laboune, Jianfei Hu, David Ambrozak, Marybeth S. Hughes, Rebecca
Hoh,
Joseph
P.
Casazza,
Alexander
Vostal,
Daniel
Bunis,
Krystelle
Nganou-Makamdop, James S. Lee, Stephen A. Migueles, Richard A. Koup, Mark
Connors, Susan Moir, Timothy Schacker, Frank Maldarelli, Stephen H.
Hughes, Steven G. Deeks, and Daniel C. Douek
SUPPLEMENTAL EXPERIMENTAL PROCEDURES
Extraction and quality analysis of cell-associated RNA and DNA. CD4 T cell subsets were sorted by FACS into heatinactivated fetal calf serum and kept on ice until further processing. Cells were then sedimented by centrifugation to 420 x g
for 7 minutes at 4°C, lysed in RNAzol RT at <5 x 106 cells/mL, homogenized by pipetting, and stored at -80°C until
extraction. For dual RNA and DNA extraction, 0.4 volume of sterile H2O was added to each lysate to allow aqueous and
organic phase separation. Total RNA was extracted from the aqueous phase according to the manufacturer’s instructions. The
organic phase of each lysate was solubilized in DNAzol (Molecular Research Centers) to allow DNA extraction according to
the manufacturer’s instructions. RNA yield and integrity were verified by microelectrophoresis using the RNA 6000 Pico
Bioanalyzer Kit (Agilent). DNA yield was determined on a Nanodrop spectrophotometer (Thermo Scientific). For DNA
samples from memory CD4 T cell subsets yielding no amplifiable HIV DNA by fluorescence-assisted clonal amplification
(FCA), PCR inhibition was ruled out by quantitative PCR for the albumin gene in serial sample dilutions.
Cell staining for FACS. Cells were labeled with LIVE/DEAD Aqua stain (Molecular Probes) and the following antibodies:
CXCR5-Alexa Fluor 488 (Pharmingen); CD27-Cy5PE (Coulter); CD45RO-PE-Texas Red (Coulter); CD14-PE (BD);
CD11c-PE (BD); CD3-H7APC (BD); CCR7-Alexa Fluor 700 (Pharmingen); CD20-APC (BD); CD56-APC (BD); T cell
receptor gamma delta (TCR-γδ)-APC (Pharmingen); CD57-QDot 800 (Invitrogen); PD1-Brilliant Violet 711 (BioLegend);
CD4-Briliant Violet 785 (BioLegend); CD8-QDot 655 (Invitrogen); and CCR6-Brilliant Violet 421. LN cells were stained
with the same antibodies, except for the inclusion of CD20-Brilliant Violet 570 (BioLegend) in place of CD20-APC and the
exclusion of CD56-APC and TCR-γδ-APC. Due to sample availability constraints, cells were only sorted according to the
markers CXCR5, CCR6, and CD57 for participants V912, V907, S1475, S1492, S1495, and S1548.
Reverse transcription of HIV virion RNA. Virion RNA samples for FCA and sequencing were denatured at 65°C for 10
minutes, annealed to env reverse transcription primer (either LSr1 or envB3out) at a primer concentration of 100 nM, and
reverse transcribed using SuperScript III (Life Technologies) at 50°C for 50 minutes followed by incubation at 85°C for 10
minutes to inactivate the reverse transcriptase.
Fluorescence-assisted clonal amplification (FCA). FCA for single-copy quantification and sequencing of CD4 T cellassociated HIV DNA molecules or single-copy sequencing of HIV virion cDNA molecules was performed in replicate PCR
wells at an estimated 0.3 HIV copies/well using primer mixes ESf2 and LSr1, which were prepared as equimolar
combinations of related oligonucleotides accounting for degenerate sequences at several base positions (see Table S4). These
primer mixes were added to a final concentration 150 nM for each oligonucleotide. Amplification was performed with
Platinum Taq DNA Polymerase High Fidelity (Life Technologies) in the presence of a 1:250,000-fold dilution of SYBR
Green I Gel Stain (Molecular Probes) as a double-stranded DNA marker. To inhibit primer amplification artifacts, yeast
transfer RNA was included in each reaction at a concentration inversely related to the quantity of CD4 T cell DNA template
in the reaction. Up to 5,000 cell-equivalents of template DNA were loaded per reaction well. Reactions were run in 384-well
standard PCR plates (ThermoFisher catalog # AB1384) under the following cycling conditions: 94°C for 2 minutes; then 50
cycles of 94°C for 15 seconds, 63°C for 15 seconds, and 68°C for 45 seconds; and then melt curve analysis. Positive wells
were defined as those showing exponential amplification of SYBR green I fluorescence and a discrete melt curve peak
between 76.0°C and 82.0°C. For each sample analyzed by FCA, a small sample aliquot was initially run in enough replicate
wells to allow rough quantification of HIV DNA copy number, calculated as follows: HIV DNA copy number = (number of
replicate wells) x (-ln(fraction of negative replicate wells)). The quantity of each sample used in this quantification FCA was
chosen based on plasma virus load (for plasma virion cDNA samples) or the proportion of CD4 T cells in the sample
predicted to contain HIV DNA (generally 10-4-10-3), with the goal of achieving 50% positive wells. Quantification FCAs
yielding non-quantifiable results (i.e., 0% or 100% positive wells) were repeated after adjustment of sample input quantity.
Based on quantification FCA results, a volume of sample predicted to contain approximately 30 HIV DNA copies (where
available) was loaded into enough replicate wells to ensure limiting dilution. Preliminary quantifications of HIV DNA copy
number were determined from these larger-scale FCA runs. Positive wells from FCA runs where <35% of wells were
positive (i.e., samples run at limiting dilution) were reamplified with primers FCAf3 and FCArB using Platinum Taq DNA
Polymerase High Fidelity under the following cycling conditions: 94°C for 2 minutes; then 35 cycles of 94°C for 15
seconds, 60°C for 15 seconds, and 68°C for 45 seconds. Reamplified products were then Sanger sequenced. Products not
yielding the expected HIV env sequence were considered to reflect poor amplification due to possible mismatch between
nested primer sequences and the original FCA reaction products, and were thus “rescued” by reamplification of original FCA
reaction products with the outer primers ESf2 and LSrB instead of the nested primers. FCA wells that were originally called
positive but that did not yield the expected HIV env sequence upon rescue reamplification were considered to be false
positives. Final quantifications of HIV DNA copy number were determined after excluding these false positive wells. To
determine the percentage of cells from each sorted CD4 T cell subset containing HIV DNA, each HIV DNA copy number
value was normalized to the sorted cell number.
Quantitative reverse transcription PCR (qRTPCR) for HIV RNAs. Total RNA samples from CD4 T cell subsets
extracted as described above were treated with DNAse I for 20 minutes at 37°C and then 15 minutes at 70°C to inactivate the
DNAse. Unspliced and spliced HIV transcripts were quantified using the RNA UltraSense One-Step Quantitative RT-PCR
System (ThermoFisher). For unspliced (gag) RNA detection, the forward primer was gag F, the reverse primer was gag R,
and the probe was gag P (see Table S4). For spliced RNA detection, the forward primer was rev F, the reverse primer was rev
R, and the probe was rev P. Cycling conditions were 45°C for 30 minutes, 95°C for 2 minutes, and then 45 cycles of 95°C for
15 seconds and 60°C for 60 seconds. Template RNA was loaded at between 4,000 and 5,500 cell-equivalents per reaction.
Sanger sequence editing. Base calls for Sanger sequence reads were initially made in Sequencher after ends trimming with
default settings. Each sequence read with a high-quality base call at every position within the range defined by HIV HXB2
coordinates 7011-7502 was included in later analysis without further editing. Each sequence read with a single ambiguous
base call (defined by the presence of a secondary, in-phase chromatogram peak with a magnitude at least 25% of that of the
primary peak) over this same env region was considered a possible PCR error and included in later analysis with the
ambiguity included in the final sequence. Each sequence read with ≥2 ambiguous base calls separated by unambiguous base
calls was considered to be non-clonal and was thus excluded from later analysis. Reads with widespread ambiguities
occurring after long poly(dA) runs were attributed to polymerase slipping during PCR and were manually overwritten
assuming this mechanism. Downstream analysis of these sequences universally showed G-to-A hypermutation. In rare
examples of this pattern, base call ambiguities were too severe to be overwritten manually. Such sequence reads were
trimmed and used for virus DNA quantification and diversity analyses, but not for any phylogenetic tree-based analysis.
HIV sequence analysis. Edited sequences from all study participants were aligned using GeneCutter at www.hiv.lanl.gov.
The alignment was improved manually and used for maximum-likelihood phylogenetic tree construction in MEGA, allowing
identification and removal of rare contaminants. The remaining sequences from each participant were compared to one
another in pairwise fashion without alignment. Each group of matching sequences thus identified was collapsed to generate a
uniquely identified cluster tagged with the number of corresponding reads from each virion or cell subset. Normalized
Shannon diversity was determined by using the R package, Vegan, to calculate the absolute Shannon diversity within the
collapsed, counted data from each virion or cell subset and then dividing this by the maximum Shannon diversity for that
subset determined by the number of sequence reads analyzed. Sequences were then realigned by participant and used to
create maximum-likelihood trees in DIVEIN (https://indra.mullins.microbiol.washington.edu/DIVEIN/) (Deng et al. 2010).
Sequences showing G-to-A hypermutation were tentatively identified within each participant’s alignment using Hypermut at
www.hiv.lanl.gov and confirmed by their genetic distance from the group of non-hypermutated sequences in that
participant’s tree. Hypermutated sequences were removed from each participant’s alignment before realignment and
construction of a final phylogenetic tree in DIVEIN, rooted on the HXB2 sequence. Genetic characteristics of individual HIV
sequences or sequence clusters were determined using pairwise distances calculated in this analysis. Copy numbers of
repeated CD4 T cell-associated HIV DNA sequences per volume of blood were calculated for each CD4 T cell subset as
follows: copy number from subset/mL = (participant’s CD4 count in cells/mL) x (proportion of CD4 T cells from subset in
participant) x (copies HIV DNA per cell in subset in participant) x (proportion of HIV DNA copies corresponding to repeated
sequence in subset in participant). The total copy number of each repeated sequence per volume of blood was then
determined by summing the results for that sequence from all subsets. Relative abundance values of repeated CD4 T cellassociated HIV DNA sequences from LN were calculated in similar fashion, but without adjustment for absolute CD4 T cell
numbers across different participants. Repeated sequences in LN cells were classified as occurring at low, medium, or high
relative abundance after stratifying all such sequences by their relative abundance levels into three groups of equal size.
Genetic compartmentalization of HIV sequences was assessed by Slatkin-Maddison testing using HyPhy (Pond, Frost, and
Muse 2005).
Analysis of G-to-A hypermutation. The Original Hypermut algorithm on the LANL HIV sequence database website was
used to identify sequences with G-to-A hypermutation. Because the use of the HXB2 sequence as a reference in this
algorithm caused an apparent decrease in sensitivity in our initial trials, we chose a reference sequence for each participant
from the set of cell- and virion-associated sequences determined for that participant. This was accomplished by determining a
“least hypermutated” sequence from each participant’s alignment after scoring all possible pairwise comparisons between
sequences for evidence of APOBEC3G-induced hypermutation using Hyperpack (Kijak et al. 2007). Sequences with
significant evidence of hypermutation (P < 0.05) using the least hypermutated sequence as a reference in the Hypermut
algorithm were considered to be hypermutated if they did not cluster with non-hypermutated sequences in the phylogenetic
tree for the participant.
TCRB gene deep sequencing. Total RNA from sorted CD4 T cell subsets was reverse transcribed using a modified version
of the SMARTer cDNA Synthesis kit (Clontech). TCRB sequences were then amplified using the SMARTer 5’ primer and
TCRB constant region-specific primer Hu/Rh MBC2. The amplicons were amplified further to incorporate the Illumina flow
cell binding sequences, Illumina read 1 priming sequences, and unique identifiers. Libraries were sequenced in 2 x 150 bp
paired-end runs to obtain V region, J region, and CDR3 sequences. TCRB annotation was performed by combining a custom
Java program written for these analyses and the National Center for Biotechnology Information’s BLAST program. Germline
V and J genes of a TCRB read were identified first, and the CDR3 was determined by finding the conserved cysteine at the 5’
end of the CDR and the conserved phenylalanine at the 3’ end of the CDR. Unique TCRB combinations (V-CDR3-J) were
collapsed to determine counts.
HIV integration site library preparation and confirmatory PCR. Genomic DNA was fragmented by random shearing
into 300- to 500-bp fragments. Linker-mediated nested PCR was performed to amplify the human genomic regions and the
linked virus sequences from both the 5’ and 3’ long terminal repeats (LTRs). Paired-end sequencing was carried out using the
MiSeq 2 x 150-bp paired end kit (Illumina). The sequences of the host-virus junctions and the host DNA breakpoints were
determined. The host DNA sequences were then mapped to human genome (hg19) with BLAT. A stringent filter was used
to check quality of recovered integration sites. Sequences with exactly the same integration site but different host DNA
breakpoints came from different cells; this test thus identifies proviruses in clonally amplified cells. Integration sites in
clonally expanded cells identified by this method were confirmed by PCR using a modified version of the FCA forward
primer (ESf2Short, see Table S4) and a reverse primer targeting the integration site. In two cases, 45 PCR cycles generated
~3.2Kb products that were purified after excision from agarose gels and Sanger sequenced to determine the associated env
region sequences. In a third case, 35 cycles of nested PCR using the first-round PCR product were necessary before gel
purification and Sanger sequencing of the confirmatory ~3.2Kb product. These PCR reactions were performed using
Platinum Taq Polymerase (Life Technologies) and additional reagents supplied by the manufacturer.
Virion induction in vitro. Sorted CD4 TCM, TTM, and TEM cell subsets in RPMI 1640 + 10% fetal calf serum +
penicillin/streptomycin/L-glutamine supplemented with Abacavir (5 nM) and Efavirenz (40 nM) were stimulated using T cell
activation/expansion beads (Miltenyi) according to the manufacturer’s instructions. T cell activation was confirmed by flow
cytometric analysis for CD25 and CD69 expression after 24-48 hours. After 4 days of culture, 75% of volume of medium
from each culture was withdrawn for virion isolation and replaced with fresh medium. Each culture was then incubated for an
additional 4-5 days, and culture medium was again withdrawn for virion isolation. Virions were isolated by high-speed
centrifugation as described (Yukl et al. 2011) after an initial pre-clarification spin at 2,000 x g for 10 minutes and after
spiking with known quantities of RCAS virions as a recovery control (Palmer et al. 2003). Virion RNA extracted from
resulting pellets was reverse transcribed for FCA or subjected to FCA without reverse transcription to exclude HIV DNA
contamination. Two sequences detected from induced virions were determined potentially to have come from contaminating
DNA and were therefore excluded from further analysis.
SUPPLEMENTAL TABLES
Table S1. Related to Figure 1. Characteristics of study participants. C, HIV controller; NC, HIV-infected non-controller.
ID
Sex
V912
V907
S1270
S1349
S1475
S1492
S1495
S1541
S1548
S1788
LIR01
LIR02
LIR03
LIR04
V016
V257
V621
V667
V723
V965
M
M
M
M
M
M
M
M
F
M
M
F
M
M
M
M
M
M
M
M
Clinical
Group
C
C
C
C
C
C
C
C
C
C
C
C
C
C
NC
NC
NC
NC
NC
NC
HIV Diagnosis
Year
1989
2007
1986
2005
1992
2004
1992
1996
1991
2000
1984
2005
1994
2003
2003
2003
1993
1980
2008
2008
Sample
Date
4/2011
4/2011
4/2014
2/2014
6/2010
6/2010
4/2014
1/2014
5/2010
3/2014
4/2014
6/2014
7/2014
11/2014
7/2010
4/2008
10/2010
10/2009
7/2010
3/2011
HLA-B
Alleles
B44/53
B07/45
B*0705/*5701
B*2705/*3501
B*3501/*5101
B*3501/*5703
B*2705/*4402
B*0702/*1402
B*4901/*5201
B*2705/*5701
B*1401/*5701
B14/15
B57/8101
B15/40:12
B*5701
B*5701/*0801
B*0801/*4801
B*4403/*5301
B35/39
Not performed
CD4 count
(cells/µL)
604
772
578
620
1216
746
293
933
846
503
934
551
494
530
230
373
412
564
501
467
Plasma HIV RNA
(copies/mL)
442
854
<40
<40
66
486
525
988
118
41
<40
267
159
132
22240
25300
21381
13500
17728
23492
Table S2. Related to Figures 3 and 4. Characteristics of HIV DNA sequences in presumptive expanded CD4 T cell clones
from HIV controllers.
Participant ID
Sequence IDa
Defectb
Copies/mL
Average Distance
to Plasma HIVc
% Blood CD4 T Cell
HIV DNA
V912
V912
V912
V912
V912
V912
V912
V912
V912
V912
V907
V907
V907
V907
V907
S1270
S1270
S1270
S1270
S1270
S1270
S1270
S1349
S1349
S1349
S1349
S1349
S1475
S1475
S1475
S1475
S1475
S1475
S1475
S1475
S1492
S1492
S1492
S1492
S1495
S1495
S1495
S1495
S1495
S1495
S1495
S1541
S1541
S1541
S1541
S1541
V912_26
V912_47
V912_30
V912_41
V912_29
V912_14
V912_16
V912_25
V912_9
V912_8
V907_16
V907_25
V907_15
V907_19
V907_13
S12702014_1
S12702014_2
S12702014_6
S12702014_9
S12702014_8
S12702014_7
S12702014_5
S1349_7
S1349_9
S1349_17
S1349_6
S1349_10
S1475_18
S1475_17
S1475_19
S1475_12
S1475_8
S1475_35
S1475_24
S1475_20
S1492_23
S1492_42
S1492_27
S1492_22
S1495_14
S1495_43
S1495_55
S1495_32
S1495_58
S1495_30
S1495_34
S15412014_1
S15412014_4
S15412014_9
S15412014_13
S15412014_3
HM
HM
Del
HM
HM
HM
HM
HM
HM
HM
HM
HM
HM
39.03
10.88
2.84
1.89
1.89
1.89
1.21
0.80
0.53
0.53
1368.61
39.64
4.95
2.70
2.70
64.66
9.04
1.87
1.25
1.25
1.25
1.25
180.35
0.92
0.71
0.37
0.37
15.62
2.87
2.85
2.40
1.60
1.60
1.19
0.95
97.84
7.58
2.33
2.33
30.55
4.97
1.29
1.29
0.65
0.65
0.32
172.32
10.99
4.01
4.01
2.67
10.37%
12.11%
6.49%
3.68%
9.72%
4.01%
4.03%
4.71%
3.51%
3.51%
na
na
4.78%
4.78%
5.33%
5.26%
1.08%
4.86%
na
4.40%
na
na
1.46%
na
1.68%
1.68%
1.90%
7.97%
8.57%
8.25%
2.91%
2.45%
na
4.46%
7.27%
4.04%
na
1.72%
1.53%
5.62%
5.90%
2.06%
5.65%
na
3.71%
2.58%
5.19%
na
2.28%
na
na
41.7%
11.6%
3.0%
2.0%
2.0%
2.0%
1.3%
0.9%
0.6%
0.6%
88.8%
2.6%
0.3%
0.2%
0.2%
76.8%
10.7%
2.2%
1.5%
1.5%
1.5%
1.5%
95.5%
0.5%
0.4%
0.2%
0.2%
35.1%
6.4%
6.4%
5.4%
3.6%
3.6%
2.7%
2.1%
77.4%
6.0%
1.8%
1.8%
57.2%
9.3%
2.4%
2.4%
1.2%
1.2%
0.6%
80.4%
5.1%
1.9%
1.9%
1.2%
Gened
NSD1
MED26
POLM
S1541
S1541
S1541
S1548
S1548
S1548
S1548
S1548
S1548
S1548
S1548
S1548
S1548
S1788
S1788
S1788
S1788
S1788
S1788
LIR01
LIR01
LIR01
LIR01
LIR01
LIR02
LIR02
LIR02
LIR02
LIR02
LIR02
LIR02
LIR03
LIR03
LIR03
LIR03
LIR04
LIR04
LIR04
LIR04
LIR04
LIR04
LIR04
LIR04
LIR04
LIR04
LIR04
LIR04
LIR04
a
S15412014_10
S15412014_6
S15412014_27
S1548_24
S1548_31
S1548_22
S1548_43
S1548_34
S1548_44
S1548_48
S1548_28
S1548_33
S1548_38
S1788_3
S1788_29
S1788_1
S1788_2
S1788_6
S1788_16
LIR01_3
LIR01_4
LIR01_6
LIR01_2
LIR01_5
LIR02_8
LIR02_31
LIR02_1
LIR02_23
LIR02_19
LIR02_4
LIR02_18
LIR03_6
LIR03_7
LIR03_8
LIR03_18
LIR04_19
LIR04_26
LIR04_31
LIR04_96
LIR04_92
LIR04_66
LIR04_6
LIR04_32
LIR04_60
LIR04_24
LIR04_16
LIR04_13
LIR04_100
HM
HM
HM
HM
HM
HM
HM
HM
HM
HM
-
2.67
1.46
1.46
147.24
126.61
75.90
51.83
43.20
34.67
27.68
27.13
21.10
7.06
1.38
1.16
0.55
0.54
0.38
0.38
4.20
1.97
0.84
0.52
0.39
3.72
2.26
2.05
1.22
1.12
1.02
1.02
9.82
6.26
4.43
1.26
1.67
1.64
1.55
1.04
1.04
1.04
1.01
0.60
0.50
0.50
0.50
0.50
0.40
4.30%
na
na
4.89%
8.33%
9.73%
na
9.65%
2.79%
na
9.48%
3.60%
8.51%
1.37%
1.52%
1.16%
na
na
na
13.50%
9.50%
2.72%
10.66%
4.81%
3.66%
5.62%
na
3.86%
2.16%
3.47%
1.76%
4.96%
5.64%
5.75%
3.95%
6.37%
4.04%
4.34%
na
na
2.44%
1.80%
6.92%
na
1.92%
3.19%
3.01%
6.63%
1.2%
0.7%
0.7%
18.8%
16.1%
9.7%
6.6%
5.5%
4.4%
3.5%
3.5%
2.7%
0.9%
15.9%
13.3%
6.3%
6.2%
4.4%
4.4%
35.39%
16.59%
7.06%
4.36%
3.26%
9.8%
6.0%
5.4%
3.2%
3.0%
2.7%
2.7%
23.5%
15.0%
10.6%
3.0%
6.3%
6.2%
5.9%
3.9%
3.9%
3.9%
3.9%
2.3%
1.9%
1.9%
1.9%
1.9%
1.5%
Each unique HIV env sequence was given a unique identifier, indicated as “Participant ID_sequence number.”
HM, G-to-A hypermutation pattern; Del, internal deletion and frameshift; -, no defect detected over the region sequenced.
c
Calculated as the average pairwise genetic distance between the indicated sequence and all plasma virus sequences. na,
calculation not attempted for sequences showing G-to-A hypermutation.
d
Integration site as demonstrated by long-range PCR from the env forward primer to an integration site-specific reverse
primer, confirmed by sequencing of the env portion of the PCR product.
b
Table S3. Related to Figures 6 and 7. HIV controllers who underwent LN biopsy.
LIR01
CD4 count
(cells/µL)
934
Plasma HIV RNA
(copies/mL)
<40
HLA-B
Alleles
B*1401/*5701
Biopsy
Site
Inguinal
Cell
Yield
3.74 x 107
CD4 T Cell
HIV DNA Copies
[47]a
LIR02
551
267
B14/15
Inguinal
1.37 x 108
27859
LIR03
494
159
B57/8101
Axillary
5.50 x 108
90152
Inguinal
7
26189
ID
LIR04
530
132
B15/40:12
9.50 x 10
a
Bracketed value represents total HIV DNA copy number calculated based on detection of a single HIV DNA copy in all
sorted material. This single detected copy showed G-to-A hypermutation.
SUPPLEMENTAL REFERENCES
Deng, W., B. S. Maust, D. C. Nickle, G. H. Learn, Y. Liu, L. Heath, S. L. Kosakovsky Pond, and J. I. Mullins. 2010.
'DIVEIN: a web server to analyze phylogenies, sequence divergence, diversity, and informative sites',
Biotechniques, 48: 405-8.
Kijak, G. H., M. Janini, S. Tovanabutra, E. E. Sanders-Buell, D. L. Birx, M. L. Robb, N. L. Michael, and F. E. McCutchan.
2007. 'HyperPack: a software package for the study of levels, contexts, and patterns of APOBEC-mediated
hypermutation in HIV', AIDS Res Hum Retroviruses, 23: 554-7.
Palmer, S., A. P. Wiegand, F. Maldarelli, H. Bazmi, J. M. Mican, M. Polis, R. L. Dewar, A. Planta, S. Liu, J. A. Metcalf, J.
W. Mellors, and J. M. Coffin. 2003. 'New real-time reverse transcriptase-initiated PCR assay with single-copy
sensitivity for human immunodeficiency virus type 1 RNA in plasma', J Clin Microbiol, 41: 4531-6.
Pond, S. L., S. D. Frost, and S. V. Muse. 2005. 'HyPhy: hypothesis testing using phylogenies', Bioinformatics, 21: 676-9.
Yukl, S. A., P. Li, K. Fujimoto, H. Lampiris, C. M. Lu, C. B. Hare, S. G. Deeks, T. Liegler, M. Pandori, D. V. Havlir, and J.
K. Wong. 2011. 'Modification of the Abbott RealTime assay for detection of HIV-1 plasma RNA viral loads less
than one copy per milliliter', J Virol Methods, 175: 261-5.