Download Immunomodulatory Effects of Human Immunodeficiency

Document related concepts

Phagocyte wikipedia , lookup

Immune system wikipedia , lookup

Lymphopoiesis wikipedia , lookup

T cell wikipedia , lookup

Psychoneuroimmunology wikipedia , lookup

Adaptive immune system wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Molecular mimicry wikipedia , lookup

Innate immune system wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Immunomics wikipedia , lookup

Transcript
Linköping University Medical Dissertations No.1256
Immunomodulatory Effects of Human Immunodeficiency
Virus (HIV-1) on Dendritic Cell and T cell Responses
Studies of HIV-1 effects on Dendritic cell functionality reflected in primed T cells
Karlhans Fru Che
Division of Molecular Virology,
Department of Clinical and Experimental Medicine,
Linköping University SE-58185
Linköping 2011
Copyright © Karlhans Fru Che, 2011
Division of Molecular Virology
Department of Clinical and experimental medicine
Linköping University
SE-58185 Linköping
Cover: An HIV-1 exposed Dendritic cell priming naïve T cells
The cover picture and illustrations in this thesis were performed by Rada Ellegård.
Published articles have been reprinted with permission of the copyright holder.
Printed in Sweden by Liu-Tryck, Linköping, Sweden, 2011
ISBN: 978-91-7393-093-2
ISSN 0345-0082
“A determined hunter is not afraid of the jungle”
Nelson Mandela
“Our deepest fear is not that we are inadequate. Our deepest fear is that we are powerful
beyond measure. It is our light not our darkness that most frightens us. We ask ourselves,
“Who am I to be brilliant, gorgeous, talented and fabulous?” Actually, who are you not
to be? You are a child of God. Your playing small does not serve the world. There is
nothing enlightened about shrinking so that other people won’t feel insecure around you.
We are born to make manifest the glory of God that is within us. It is not just in some of
us. It’s in everyone. And as we let our light shine we give other people permission to do
the same. As we are liberated from our own fear, our presence automatically liberates
others”.
Marianne Williamson
“With the rapid developments in today’s life, the sky is now my spring board and no
longer my limit”.
Karlhans Fru Che
Supervisor
Committee Board
Professor Marie Larsson
Professor Sven Hammarström
Division of Molecular Virology
Division of Cell Biology
Department of Clinical and
Experimental Medicine
Department of Clinical and
Experimental Medicine
Linköping University
Linköping University
Linköping, Sweden
Linköping, Sweden
Co supervisors
Professor Jorma Hinkula
Professor Markus Maeurer
Division of Molecular Virology
Department of Microbiology Tumor and
Cell Biology (MTC)
Department of Clinical and
Experimental Medicine
Karolinska Institute
Linköping University
Stockholm
Linköping, Sweden
Sweden
Professor Karl-Eric Magnusson
Associate professor Maria Jenmalm
Division of Medical Microbiology
Division of Pediatrics
Department of Clinical and
Experimental Medicine
Department of Clinical and
Experimental Medicine
Linköping University
Linköping University
Linköping, Sweden
Linköping, Sweden
Faculty opponent
Professor Douglas Nixon
Professor Jan-Ingvar Jönsson
Division of Experimental Medicine
Division of Experimental Hematology
Department of Medicine
University of California San Francisco
Department of Clinical and
Experimental Medicine,
San Francisco, California, USA
Linköping University,
Linköping, Sweden
This work is dedicated to my entire Family
CONTENTS
LIST OF PAPERS ........................................................................................................................ 1
ABSTRACT ................................................................................................................................ 3
INTRODUCTION ....................................................................................................................... 5
Human Immunodeficiency Virus (HIV) ........................................................................................ 5
Discovery and Epidemiology ................................................................................................... 5
Etiology .................................................................................................................................... 5
Virology.................................................................................................................................... 5
Pathogenesis ........................................................................................................................... 7
HIV-1 Disease Progression ....................................................................................................... 9
Treatment .............................................................................................................................. 10
Dendritic Cell and Functions...................................................................................................... 11
DC subsets ............................................................................................................................. 11
In vitro generated DCs ........................................................................................................... 13
T cells ......................................................................................................................................... 13
T cell subsets.......................................................................................................................... 13
DC T cell priming .................................................................................................................... 15
DC-T cell-HIV-1 Interaction .................................................................................................... 17
HIV-1 Specific responses ........................................................................................................... 18
Immunomodulatory Potentials of HIV-1 ................................................................................... 19
Immunostimulatory and Inhibitory Molecules at a DC-T cell Encounter .................................. 20
Conclusions ................................................................................................................................ 23
AIMS AND OBJECTIVES .......................................................................................................... 25
Specific Aims .............................................................................................................................. 25
METHODS .............................................................................................................................. 27
Dendritic cell preparation.......................................................................................................... 27
Naïve T cell Separation .............................................................................................................. 28
Virus preparation....................................................................................................................... 28
DC T cell cocultures ................................................................................................................... 28
ELISPOT: ..................................................................................................................................... 29
Measurement of secretory factors............................................................................................ 29
ELISA: ..................................................................................................................................... 29
Intracellular cytokine staining ............................................................................................... 30
T cell proliferation analysis ........................................................................................................ 30
Fluorescence activated cell sorter (FACS) ................................................................................. 31
Gene expression by real time PCR (SYBR® Green) .................................................................... 32
Statistical analysis ...................................................................................................................... 32
FINDINGS AND DISCUSSION ................................................................................................... 33
Paper 1....................................................................................................................................... 33
Paper II....................................................................................................................................... 35
Paper III...................................................................................................................................... 37
Paper IV ..................................................................................................................................... 38
OVERALL FINDINGS/CONCLUSIONS ........................................................................................ 41
RECOMMENDATIONS AND FUTURE CHALLENGES .................................................................. 43
LAY LANGUAGE SUMMARY .................................................................................................... 45
ACKWOLEDGEMENTS............................................................................................................. 47
REFERENCES .......................................................................................................................... 51
LIST OF ABBREVIATIONS
Ab
Antibody
AIDS
Acquired immunodeficiency syndrome
ANOVA
Analysis of variance
APC
Antigen presenting cell
ART
Antiretroviral therapy
AT-2
Aldrithiol-2
BLIMP-1
B-lymphocyte-induced maturation protein-1
BTLA
B- and T-lymphocyte attenuator
DCs
Dendritic cells
CCR
Chemokine receptor
CD
Cluster of differentiation
CFSE
Carboxyfluorescein succinimidyl ester
CTLA-4
Cytotoxic T lymphocyte antigen-4
CXCR4
C-X-C chemokine receptor type
DNA
Deoxyribonucleic acid
DTX1
Deltex-1
EC
Elite controllers
EPG
Epithelial growth factor
FOXP3
Forkhead box P3
GM-CFS
Granulocyte-macrophage colony stimulating factor
gp
Glycoprotein
HAART
Highly active antiretroviral therapy
HIV-1
Human immunodeficiency virus-1
HVEM
Herpes virus entry mediator
IDCs
Immature dendritic cells
IDO
Indoleamine 2, 3 dioxygenase
IFN
Interferon
IL
Interleukin
JAK
Janus kinase
LAG-3
Lymphocyte-activation gene-3
LCs
Langerhans cells
LTNPs
Long term non-progressors
MAPK
Mitogen activated protein kinases
mDCs
Myeloid dendritic cells
MDC
Mature dendritic cells
MDDC
Monocyte derived dendritic cells
MHC
Major Histocompatibility complex
MLR
Mixed lymphocyte reaction
NFAT
Nuclear factor of activated T cells
NFkB
Nuclear factor kappa-light-chain enhancer of activated B cells
OIs
Opportunistic infections
PBMCs
Peripheral blood mononuclear cells
PCR
Polymerase chain reaction
pDCs
Plasmacytoid dendritic cells
PDGF
Platelet derived growth factor
PD-1
Programmed death-1
PD-1L
Programmed death-1 ligand
PHS
Pooled human serum
PI
protease inhibitor
PRR
Pattern recognition receptors
PAMP
Pathogen associated molecular patterns
PVL
Plasma viral load
RANTES
Regulated upon activation, normal T cell expressed and secreted
RNA
Ribonucleic acid
RT
Reverse transcriptase
SEB
Staphylococcal enterotoxin B
SIV
Simian immunodeficiency virus
STAT
Signal transducer and activator of transcription
SIV
Semian Immunodeficiency Virus
TCR
T cell receptor
TCM
Central memory T cells
TEM
Effector memory T cells
TGF-β
Transforming growth factor-β
TH1
T helper type 1
TH2
T helper type 2
TIM-3
T cell immunoglobulin domain and mucin domain-3
TLR
Toll like receptor
TNF
Tumor necrosis factor
TNFR
Tumor necrosis factor receptor
TRAIL
TNF-related apoptosis inducing ligand
Tregs
Regulatory T cells
Tr1
Regulatory T cells type 1
Tr3
Regulatory T cells type 3
VEGF
Vascular endothelial growth factor
VC
Viremic controllers
List of Papers
LIST OF PAPERS
1
In vitro priming recapitulates in vivo HIV-1 specific T cell responses,
revealing rapid loss of virus reactive CD4 T cells in acute HIV-1 infection.
Lubong Sabado R, Kavanagh DG, Kaufmann DE, Fru K, Babcock E, Rosenberg
E, Walker B, Lifson J, Bhardwaj N, Larsson M. PLoS One. 2009; 4(1): e4256.
2
HIV-1 impairs in vitro priming of naïve T cells and gives rise to contactdependent suppressor T cells.
Che KF, Sabado RL, Shankar EM, Tjomsland V, Messmer D, Bhardwaj N, Lifson
JD, Larsson M. European Journal of Immunology. 2010; 40(8): 2248-58.
3
Expression of a broad array of negative costimulatory molecules and Blimp-1
in T cells following priming by HIV-1 pulsed dendritic cells.
Shankar EM, Che KF, Messmer D, Lifson JD, Larsson M. Molecular Medicine.
2011;17(3-4): 229-40.
4
Cross-talk between P38MAPK and STAT3 regulates expression of negative
costimulatory molecules and transcription factors in HIV-1 primed T cells.
Che KF, Shankar EM, Sundaram M, Zandi S, Sigvardsson M, Hinkula J, Messmer
D, Lifson JD, and Larsson M (Submitted)
1
List of Papers
2
Abstract
ABSTRACT
The human immunodeficiency virus (HIV)-1 is the causative agent of acquired immune
deficiency syndrome (AIDS) worldwide. Till date there are no vaccines or cure for this
infection as the virus has adapted myriad ways to remain persistent in the host where it
causes severe damage to the immune system. Both humoral and cellular immune
responses are mounted against HIV-1 during the initial phase of infection but fail to
control viral replication as these responses are severely depleted during disease
progression. Of great importance in HIV-1 research today is the in depth understanding
of the types of immune responses elicited, the mechanisms behind their decline and how
these responses can be maintained overtime.
The focus of this thesis was to examine the possibility of priming HIV-1 specific T cell
responses in vitro from whole viral particles and in detail, scrutinize the type of T cell
responses and epitope specificities generated. Next was to investigate in vitro the factors
responsible for impaired immune responses in HIV-1 infected individuals. We were also
interested in understanding the underlying mechanisms through which HIV-1 initiate
suppression of T cell functionality.
Results showed that using HIV-1 pulsed monocyte derived dendritic cells (DCs), we
were able to prime HIV-1 specific CD4+ and CD8+ T cells from naïve T cells in vitro.
The epitopes primed in vitro were located within the HIV-1 envelope, gag, and pol
proteins and were confirmed ex vivo to exist in acute and chronically infected
individuals. We established that many of the novel CD4+ T cell epitopes primed in vitro
also existed in vivo in HIV-1 infected individuals during acute infection. These responses
declined/disappeared early on, which is in line with HIV-1 preferential infection of HIV1 specific CD4+ T cells.
Besides declining HIV-1 specific T cell responses, many HIV-1 infected individuals also
have impaired T cell functionality. We established that one reason behind the decline and
impairment in immune responses was the increased expression of inhibitory molecules
PD-1, CTLA-4, and TRAIL on HIV-1 primed T cells. These T cells had the capacity to
suppress new responses in a cell-cell contact dependent manner. The ability of the HIV-1
primed T cells to proliferate was severely impaired and this condition was reversed after
a combined blockade of PD-1, CTLA-4 and TRAIL. Furthermore, more inhibitory
molecules TIM-3, LAG-3, CD160, BLIMP-1, and FOXP3 were also found increased at
both gene and protein levels on HIV-1 primed T cells. Additionally, we showed
decreased levels of functional cytokines IL-2, IFN-γ and TNF-α, and the cytolytic
proteins perforin and granzyme in DC T cell priming cocultures containing HIV-1. This
could be as a result of the decreased T cell activation or impaired production by T cells.
The mechanisms responsible for the elevated levels of inhibitory molecules emanated
3
Abstract
mainly from the P38MAPK/STAT3 pathways. Blockade of these pathways in both
allogeneic and autologous DC-T cell assays significantly suppressed expression of
inhibitory molecules and subsequently rescued T cell proliferation.
In conclusion, HIV-1 pulsed DCs have the capacity to prime HIV-1 specific responses in
vitro that do exist in HIV-1 infected individuals and we found evidence that many of
these responses were eliminated rapidly in HIV-1 infected individuals. HIV-1 triggers
through P38MAPK/STAT3 pathway the synthesis of inhibitory molecules, namely
CTLA-4, PD-1, TRAIL, TIM-3, LAG-3, CD160, and suppression associated
transcription factors FOXP3, BLIMP-1 and DTX1. This is followed by decreased T cell
proliferation and functionality which are much needed to control viral replication.
4
Introduction
INTRODUCTION
Human Immunodeficiency Virus (HIV)
Discovery and Epidemiology
HIV-1, the primary cause of acquired immunodeficiency syndrome (AIDS) was initially
discovered by Luc Montagnier at the Pasteur Institute in 1983 (1) and was later
characterized in 1984 by Robert Gallo in Washington and Jay Levy in San Francisco.
HIV-2, a milder form of HIV, was later isolated from a West African patient in 1986 (2).
Current data suggests that HIV-1 comprises four groups, namely M, N, O, and P. The M
(major) group is predominant (90%) and constitutes 11 subtypes/clades (denoted by AK), and is cosmopolitan in distribution. The O (outliers) group is restricted to western and
Central Africa, whereas group N (Non-M and O), reported from Cameroon in 1998, is
extremely rare (3). The presumable fourth (P group) of HIV-1 was discovered in 2009 in
a Cameroonian woman and was closely associated with the gorilla simian
immunodeficiency virus (SIV) (4). HIV-2 subtypes are mainly restricted to West Africa
and comprise seven distinct phylogenetic lineages ranging from A-G. HIV-2 may be
categorized as epidemic subtypes (A and B) and non-epidemic subtypes (C-G) (5, 6). As
of 2009, over 33 million people are reportedly living with HIV-1 and among these, over
22 million were from sub-Saharan Africa. In 2009, 2.6 million new cases (~7000 new
cases/day) and 1.8 million deaths were registered. Therefore HIV-1 ranks highest
amongst the leading causes of death worldwide (www.uniaids.org).
Etiology
HIV-1 is a lentivirus similar to those found in primates called SIV believed to have
existed for more than 32,000 years (7). Available literature suggests that human
infections occurred through multiple zoonotic events (8). The human encounter with SIV
through primates led to crossing, and via a series of reinfections and mutations, HIV-1
originated (9, 10). Phylogenetic investigations have traced the origin of HIV-1 in humans
from chimpanzees, and HIV-2 from sooty mangabeys (10). Because these primate
species are indigenous to Central and West Africa, it is believed that HIV originated from
these African areas (9, 11).
Virology
HIV-1 is a positive, single-stranded (ss) RNA virus with a genome of 9.7 kilobases and is
classified under family Retroviridae, sub-family Lentiviridae, and genus Lentivirus. The
two ssRNA strands are surrounded by a capsid that encompasses ~2000 copies of the
core protein p24 (12, 13). The capsid is enclosed in a lipid envelope formed from host
cell membrane during viral budding (13)(see figure 1 for HIV-1 structure).
5
Introduction
Figure 1: HIV-1 structure.
HIV-1 life cycle involves a series of events within the host cell. Briefly, HIV-1 infects a
susceptible CD4+ target cell via binding its gp120 receptors to corresponding host CD4
receptors and coreceptors; chemokine receptors 5 (CCR5) or 4 (CXCR4) depending on
the viral tropism (i.e. R5 or X4 virus). This is followed by a conformational change in the
gp120 trimer leading to membrane fusion and delivery of the viral RNA into the
cytoplasm. Within the cytoplasm, viral RNA is transcribed by HIV-1 viral reverse
transcriptase (RT) into ssDNA, and subsequently into double stranded (ds) DNA. With
the help of viral integrase, the dsDNA is integrated into the host genome, and this
integrated DNA is called a provirus. Proviral DNA is transcribed alongside host DNA
during normal cell division (13) or remains latent in cells, which are called viral
reservoirs ensuring bust of infections especially after the interruption of antiretroviral
therapy (ART) (14-21). The RNA transcripts are spliced and translated into viral proteins
or remain unspliced and exported to the cytoplasm, where they are assembled as new
virions. Before viral assembly, immature viral polypeptides are processed into their
functional forms by viral protease and packaged with two full-length ssRNA transcripts
to make a mature viral particle. HIV-1 particles bud off from the cell membrane with an
envelope of host cell plasma membrane ready to infect other cells (13, 22). (Figure 2)
6
Introduction
Figure 2: HIV-1 replication cycle.
HIV-1 binds and fuses to target cells by the help of CD4 receptor and CCR5/CXCR4 coreceptors. The virus uncoats and the viral
ssRNA are released in the cytoplasm. Viral ssRNA is transcribed to dsDNA by the help of viral enzyme reverse transcriptase. The
DNA is incorporated in the human genome by help of integrase. New viral transcripts (proviruses) are synthesized during cell division
and assembled with translated protease-modified proteins at the cell membrane. New virions are formed and bud off from the host cell
plasma membrane
Pathogenesis
The hallmark of HIV-1 infection is destruction of CD4+ T cells and eventual collapse of
the host immune system. HIV-1 infects CD4+ immune cells including CD4+ T cells,
dendritic cells (DCs), macrophages, monocytes, thymocytes, and microglial cells (13,
22). New receptors against HIV-1 albeit the main receptors CD4, CCR5, and CXCR4 are
constantly emerging (23-25). HIV-1 transmission requires direct exposure of the body to
infected blood or secretions via needles, sharps, or abrasions in the mucosa during sexual
intercourse (26). The evolution of coreceptor use defines different phases of viral
pathogenesis involving a change from CCR5 use (R5 phenotype) to CXCR4 use (X4) or
in combination (R5X4) (27-31). The use of CXCR4 is characterized by rapid CD4+ T cell
loss and onset of opportunistic infections (OIs) (32). Studies by Grivel et al (1999)
7
Introduction
revealed that R5 HIV-1 is also highly cytopathic, but only for CCR5+/CD4+ T cells and
because these cells constitute only a small fraction of CD4+ T cells, their depletion does
not substantially change the total CD4+ T cell count. On the other hand, the effects of X4
HIV-1 is readily appreciated due to the extensive depletion of more CXCR4+CD4+ T
cells within the T cell pool (33). Within 3 weeks of infection (acute phase) the viral load
increases rapidly followed by increase in both cell-mediated and humoral responses (34,
35). The appearance of HIV-1 specific antibodies occurs approximately 3 weeks after the
onset of infection and during this time the subject may potentially transmit HIV-1
(window period) as infection is not tested without detectable antibody levels (36, 37).
Thereafter, the generated immune responses drastically curtail HIV-1 spread and keep
viral replication under control for a considerable duration (5-10 years) strictly depending
on both viral and host factors (38). During acute infection, subjects present flu-like
symptoms like fever, rashes, oral ulcers, lymphadenopathy, arthralgia, pharyngitis,
malaise, and weight loss (39). HIV-1 invades the gut, deplete CD4+ T cells and cause
severe damage in body organs, especially the lymphoid tissues (13, 40-42). HIV-1
continues to replicate evading host anti-viral responses, directly or indirectly targeting
HIV-1 specific T cells for elimination. This results to an eventual decline in immune
responses, a steady establishment of viral dominance and the onset of OIs. Subjects with
CD4+ T cell count ≤200cells/mm3 progress rapidly towards terminal AIDS and develop
OIs owing to viral, bacterial, fungal, parasitic infections, and neoplasms (43). (Figure 3)
Figure 3: HIV-1 pathogenesis.
During acute infection, viral load increases rapidly with a corresponding drop in CD4+ T cell counts for both typical and long term non
progressors. CD4+ T cells are significantly reestablished in LTNPs and maintained for over 10 years and viral replication is kept low
or below the limit of detection as in elite controllers. In typical progressors, the CD4+ T cell counts slightly increase after the initial
drop but then steadily declines as viral replication steadily rises.
8
Introduction
Following onset of infection, HIV-1 establishes a latent infection in certain cell types
namely resting memory CD4+ T cells (19, 44), dendritic cells (DCs) (20, 45, 46),
monocytes (21, 47-50), and macrophages (51-53). Naïve CD4+ T cells, pluripotent
progenitors in the bone marrow, and CD4+ T cells and macrophages in seminal fluid have
also been reported reservoirs for latent infection in subjects undergoing ART (17, 54-56).
Latent infection in cells located in the central nervous system (CNS), such as microglial
cells and macrophages migrating into the CNS, are critical in protecting HIV-1 from
antiretroviral drugs (18). Interruption of ART in patients with undetectable viremia
results in the reappearance of viral plasma, a phenomenon brought about by viral release
from the latently infected cells (44).
HIV-1 Disease Progression
The progression from HIV-1 infection to AIDS varies from person to person and depends
on both host and viral factors. For the host dependent factors, individuals with mutation
in the HIV-1 coreceptor CCR5Δ32 allele express a non-functional and truncated protein
that is unfit for viral binding and fusion and this offers a strong, but not complete
protection from infection in homozygous individuals (57), and a delayed disease
progression in heterozygous individuals (57, 58). About 10-15% of Caucasians are
heterozygous and 1% homozygous for the Δ32mutation (57). Another key host factor that
determines disease progression is HLA haplotypes (59, 60). HLA molecules are amongst
the most polymorphic gene products and therefore, depending on the composition of the
HLA haplotypes, individuals respond differently in immune strength to the same
pathogen (61). In humans, HLA B is the most diverse and polymorphic, rapidly evolving
and has been connected to a majority of HLA class I related diseases and disease
progression (62-64). In Chimpanzees as in humans, expression of the HLA B*27/B*57
haplotypes target conserved areas of SIV/HIV and minimize the chances of viral evasion
(65). In a study by Carrignton et al (66), Caucasians with HLA B*35 and HLA Cw*04
alleles were consistently associated with rapid progression to AIDS, whereas the
haplotypes HLA B*27, HLA B*57, and HLA B*5801 are associated with lower viral
loads and slower disease progression (67, 68). HLA class II haplotypes have also been
linked to disease progression. For instance, one study reported the association of HLA
DRB1*1303 alleles with decreased viral loads and slower disease progression (69). HLA
DRB1*01 was more common in HIV-1 negative as compared to positive individuals and
the expression of HLA DRB1*08 corresponded to lower median viral loads (70).
Interestingly, the age when acquiring the HIV-1 infection has also been shown to
influence disease progression as rapid disease progression seems to be more common in
older as compared to younger subjects (71, 72). Host habits and factors like smoking(73),
malnutrition(74) and depression (75) have also been shown to affect HIV-1 disease
progression, albeit not all studies confirm these findings.
9
Introduction
Viral factors, especially evolutionary changes in HIV-1 have been directly associated
with disease progression (30). Different HIV-1 strains have different replicating fitness,
which in turn can affect disease progression (76-78). Individuals with slow disease
progression are thought to harbor attenuated strains of HIV-1 or mutant forms of
important viral genes, e.g. nef, vpr, vif, or rev, which makes the virus less virulent (79).
The ability or inability to control disease progression has led to the categorization of
various stages of HIV-1 infection. Individuals with stable CD4+ T cell counts, lack of
disease progression, and symptoms of HIV-1 infection, and low or intermediate plasma
viral loads (PVLs) for >10 years of infection are classified as long term non-progressors
(LTNPs). LTNPs constitute 5-15% of HIV-1 infected individuals worldwide (80-85).
Elite controllers (ECs) make up <1% of all infected subjects and are characterized by
their ability to control their viral load (<50 copies/mL) below the limit of detection for
longer periods of time (64, 86, 87), whereas viremic controllers (VCs) display a lesser
degree of viremic control (low but detectable HIV-1 RNA levels (50-2000 copies/mL)) as
compared to ECs (88, 89). The central characteristic of ECs and VCs is their ability to
control viral load even if some of these subjects experience severe CD4+ T cell depletion
and advancement to AIDS. Likewise, LTNPs may remain clinically healthy with stable
CD4+ T cell counts, but consistently have uncontrolled PVL (90, 91).
Treatment
There is no effective vaccine available against HIV/AIDS, but ART regimens are widely
available to improve the life span and quality of life for infected individuals. Following
the advent of ART regimens, there has been a dramatic reduction in AIDS related
morbidity and mortality (92-94). According to recent treatment guidelines, patients who
show symptoms of AIDS regardless of CD4+ T cell counts or PVL, as well as
asymptomatic patients with PVLs ≥100,000copies/mm3 and CD4+ T cell counts
≤350cells/mm3 are eligible to initiate ART (95, 96).
There are four categories of antiretroviral drugs; (1) nucleoside reverse transcriptase
inhibitors that interfere with the viral RT, e.g. Zidovudine, Didanosine, and Stavudine,
(2) Nucleotide reverse transcriptase inhibitors that terminates chain elongation during
replication, e.g. Tenofovir and Adefovir, (3) Non-nucleoside reverse transcriptase
inhibitors that act via non-competitive binding to a hydrophobic pocket close to the RT
enzyme, e.g. Nevirapine, Efavirenz, and Delavirdine, and (4) Protease inhibitors that
block HIV-1 proteases preventing assembly of new viral particles, e.g. Nelfinavir and
Saquinavir (97, 98). Recently, several alternative approaches have gained acceptance as
treatment options and include fusion inhibitors that prevent viral entry into the cell e.g.,
Maraviroc and Enfuvirtide (99-102). In addition, integrase inhibitors that block the viral
integrase are being tested as potential regimens (103), and Raltegravir has recently been
approved by FDA (104). Viral maturation inhibitors are being widely investigated and
10
Introduction
they work by inhibiting formation of infectious viral particles and interferon alpha (IFNα) is the currently available agent in this class (105, 106). The CCR5 receptor
antagonists, e.g., Maraviroc, are the first drugs against HIV-1 infection that do not target
the virus directly (104) seeing that they shield viral attachment to the cell by blocking
CCR5 (102, 107, 108). Efficient adherence to treatment is important and maintains PVLs
at a constantly low and manageable level (109, 110). This also help prevent the
emergence of drug resistant strains (111, 112).
Dendritic Cell and Functions
DCs are defined as professional APCs that capture, process and present antigens to T
cells and are unique in their capacity to activate primary T cell responses (113). DCs
upon encountering pathogens capture and migrate to regional lymph nodes during which
the DCs mature, process, and load antigenic peptides on MHC class I or II molecules.
During DC migration, DC functionality is modulated by decreased phagocytosis,
enhanced antigen presenting abilities, and increased expression of MHC I/II molecules,
and costimulatory molecules (114-116). DC-T cell encounter in lymph nodes result either
in immune activation or suppression (117), the later function being the maintenance of
immune tolerance by DCs. Central tolerance occurs in the thymus for T cells and bone
marrow for B cells and acts to prevent the immune system from responding to self
antigens. The responsibilities of DCs in the thymus cannot be over emphasized as they
educate newly produced T cells and negatively selects or eliminates T cells reacting to
self MHC/peptide complexes (118, 119). DCs can also maintain peripheral tolerance by
active suppression of T cell responses deleterious to the host either through induction of
Tregs, or direct cell-to-cell contact mediated killing and induction of anergy (120).
DC subsets
There are multiple subsets of DCs depending on phenotype, functions, and location in the
body (Figure 4). Enormous knowledge exist regarding DC subsets found in mice (121).
The functional subsets of classical DCs in mice include CD8α+ DCs that induce T helper
type I (TH1) responses and CD8α- DCs that induce TH2 responses (122, 123). These
subsets have also been shown to have differential antigen processing outcomes whereby
CD8α+ DCs prime for CD8+ specific T cells and CD8α- DCs for CD4+ specific T cells
(124). Human DCs can be found either in blood or tissues in several distinct subsets
(121).
11
Introduction
Figure 4: Anatomical locations of human dendritic cells.
Human myeloid DCs (mDCs) and plasmacytoid DCs (pDCs) are bone marrow derived
cells and are distinct from follicular DCs (FDC), which are of stromal origin having no
role in T cell, NK cell, or NKT cell immunity (125). FDC will henceforth not be
discussed further in this thesis. Present in the body internal organs such as gut, lungs,
liver and kidney are interstitial tissue mDCs with the capacity to maintain surveillance,
bind antigen, and stimulate T cell responses in a similar manner to blood and cutaneous
DCs (126). Upon antigen encounter in these organs, the mDCs migrate to adjacent lymph
nodes where they present processed antigens to T cells thereby generating adaptive
responses against the pathogen of interest.
Cutaneous DCs: Different layers of human skin play host to different DC subsets. The
epidermis play host to Langerhans cells (LCs) and dermis to CD1a+ and CD14+ DCs,
distinguished by their phenotypic expression of surface molecules (127). LCs express
Langerin, a specific C-type lectin that makes up 2-3% of epidermal cells (128, 129). LCs
distinctively secrete large amounts of IL-15, whereas CD14+ DCs are unique in
producing a wide range of secretory factors, including IL-1β, IL-6, IL-8, IL-10, IL-12,
GM-CSF, MCP, and TGF-β (130). Some functional differences between CD14+ DCs and
LCs are the ability by CD14+ DCs to induce naïve CD4+ T cells to differentiate to T
follicular helper cells (131, 132), and induce naive B cells to switch isotype and become
plasma cells (133), whereas LCs effectively polarize naïve CD4+ T cells towards TH2
responses (133, 134). However comparative studies between LCs and CD14-CD1c+ DCs
12
Introduction
showed that LCs are significantly more efficient than CD14-CD1c+ DCs at secreting both
TH1 (IFN-γ and TNF-α) and TH2(IL-4 and IL-15) cytokines (135). LCs are also efficient
at priming and crosspriming naive CD8+ T cells that are highly efficient in tumor cell
killing as compared CD14+ DCs (130, 133, 135, 136) owing to their ability to secret IL15 (133, 137). The third group of cutaneous DC population CD1a+ DC is also good at
activating CD8+ T cells better than CD14+ T cells but less efficiently than LCs (133,
135).
Blood DCs: Based on their differential expression of surface molecules, three subsets of
blood DCs have been categorized. CD1c+ mDCs express a wide range of toll like
receptors (TLRs) including TLR 1-6, 8, and 10, and secrete proinflammatory cytokines
upon stimulation (138, 139). Secondly is the CD141+ mDC subset representing a minute
population that uniquely express CLEC9A, and is thought to serve as the human
accomplice for mice CD8+ DCs with an efficient ability to induce CD8+ T cell responses
through cross presentation and have also been report in non lymphoid tissues (140, 141).
The third subset of blood DCs are the pDCs expressing high levels of IL-3Rα chain
(CD123), BDCA2, and ILT7 (142). pDCs are unique in their ability to secret enormous
type 1 IFNs upon recognition of viral components through TLR7 and 9 (143). pDCs can
be functionally subdivided into CD2low and CD2high with the CD2high pDCs being more
potent than CD2 low pDCs in their ability to induce allogeneic T cell proliferation (144)
In vitro generated DCs
The low percentage of DCs in the body (1% of PBMC) has made DC studies
cumbersome and has prompted the establishment of ex vivo generated DCs (145, 146).
Several studies have established cytokine-driven methods for expanding and
differentiating DCs ex vivo creating sufficient numbers of DCs suitable for large scale
studies. According to a summary by Rossi et al (2005) (147), four categories of cytokine
ex vivo generated DCs were highlighted. The interstitial or dermal mDCs (1)
differentiated from the CD14+ precursor by GM-CSF, TNF-α, and IL-4; LCs (2)
differentiated from the CD14+ precursor by GM-CSF, TNF-α, and TGF-β; monocyte
derived DCs (MDDCs) (3) differentiated from CD14+ monocytes by GM-CSF, and IL4.The fourth group are the pDCs (4) which are sustained by IL-3 in cultures. In this
thesis, we have consistently used MDDCs in view to understand HIV-1 interaction with
DCs and T cells and their ability to prime T cell responses.
T cells
T cell subsets
T cell development and editing occur in the thymus and are selected according to MHC
class restrictions. Thymocytes responding to MHC II are positively selected as CD4+ T
13
Introduction
cells destined to provide helper functions (T helper cells), whereas thymocytes
responding to MHC I are selected as CD8+ T cells destined for cytotoxic activities
(cytotoxic T cells). On the other hand, T cells with receptors that strongly react to selfantigens on DCs are negatively selected, i.e. depleted (148). Positively selected naïve T
cells (TN) constantly circulate between lymph nodes and peripheral blood ready to be
primed in to effector T cells by APCs in lymph nodes. Effector T cells migrate out to
peripheral tissues to execute their functions as instructed by APCs (149). After clearance
of antigens, a majority of the effector cells undergo apoptosis and are cleared by
scavenger cells, i.e. macrophages and DCs. Sustained effector cells differentiate into
memory cells. The memory T cell pool is the hallmark of the acquired immune system
and persists for life (150). Memory T cells can be effector memory (TEM) or central
memory (TCM). The TEM circulate in the peripheral tissue and will mount an immediate
effector function, e.g. direct or cytokine-mediated cell killing after encountering a recall
pathogen. In contrast, TCM home to the T cell areas of secondary lymphoid tissues and
will expand to TEM upon antigenic stimulation to a recall pathogen (150, 151). Effector T
cell functions are either helper (TH) or cytotoxic (Tc). Helper effector T cells are
subdivided according to functionality. TH1 cells express transcription factor T-bet and
produce mostly IL-2 and IFN-γ. On the other hand, TH2 cells confer humoral immune
responses and possess transcription factor GATA-3 producing mostly IL-4, IL-5, and IL13 (152, 153). Regulatory T cells (Tregs) make up the third group of polarized TH cells
and are unique in their coexpression of high CD25, CD4, and expression of transcription
factor FOXP3. Induced Tregs have two functional subsets, type 1 (Tr1) and type 3 (Tr3),
which depends on IL-10 and TGF-β, respectively (154, 155). Tregs function as key
modulators of adaptive immunity and mediate tolerance to self. The fourth arm of
polarization is the TH17 subset commonly associated with transcriptional factors retinoid
acid receptor related orphan receptor (ROR) and signal transducer and activator of
transcription 3 (STAT3). TH17 cells are key producers of proinflammatory cytokines IL17, IL-6, IL-1, and IL-23 (152, 156, 157) (Figure 5).
14
Introduction
Figure 5: Dendritic cell polarized CD4+ T cells.
Different pathogens condition dendritic cells through their pathogen associated molecular patterns (PAMP) to activate naïve CD4+ T
cells and promote their development into TH1, TH2, TH17, or Tregs. This is strictly dependent on the type of cytokines generated
during the initial interaction hardwiring the generation of the different polarized T cell subsets.
Cytotoxic T cells are potent destructors of viral or intracellular pathogen infected cells
via direct killing of target cells by apoptosis or necrosis mediated mechanisms. The
common use of granzyme and perforin for cell killing is an important tool used by Tc to
execute this function (158). Tc cells are further subdivided into two subsets depending on
their cytokine profile with the Tc1 subset producing high amounts of IFN-γ and the Tc2
subset producing IL-4, IL-5, IL-10, IL-13 and low amounts of IFN-γ (159, 160).
DC T cell priming
The fate of DC T cell interaction is either activation or suppression of the immune
responses and this is a highly regulated process. The initiation of an effective immune
response involves 4 signals (Figure 6).
15
Introduction
Figure 6: Dendritic cell stimulatory signals to T cells.
Signal 1 is the antigen specific signal between the TCR and peptide/MHC molecule. Signal 2 is the costimulatory signal and it gives
either a stimulatory or inhibitory programming of the T cells. Signal 3 is the polarizing signal responsible for the promotion of TH1,
TH2, TH17, or Tregs subsets. Signal 4 (not indicated in the figure), also known as the homing signal, equips the polarized T cells with
homing molecules so they can migrate to their specific destination in the body, e.g. peripheral blood, gut, or skin.
Signal 1; an antigen specific signal that consists of the cognate interaction of T cell
receptor (TCR) complex with the DC peptide/MHC complex (161). To achieve full
activation, a costimulatory signal is required (signal 2) and without this signal the T cells
become anergic, which is characterized by decreased proliferation and IL-2 production
(162). Signal 3; also called the polarizing signal, is responsible for polarizing the T cells
to elicit a TH1, TH2, Treg, or TH17 response. The polarization depend on the prevailing
circumstances in the microenvironment and how the DCs have been activated i.e. the
type of PRR engagement and the cytokine profile elicited after pathogen encounter and
the quality of subsequent DC-T cell crosstalk (156, 157). For example, IL-12, IL-23, IL18, IL-27, type 1 IFNs, and the cell surface expressed intracellular adhesion molecule 1
(ICAM-1) polarizes T cells towards TH1 response, whereas the IL-4, CCL2, and OX40
ligand (OX40L) polarizes towards TH2. Tregs are polarized by IL-10 and transforming
growth factor-β (TGF-β) into either type 1 Tregs (Tr1) or type Tregs 3 (Tr3), respectively
(156, 157, 163). TH17 polarization is guided by the mixture of TGF-β, IL-6, and IL-23
differentiating from Treg (164) (refer to figure 5). After T cell stimulation, costimulation,
and polarization, effector T cells will home to their various organ destinations, e.g. skin,
mucosa, and brain, to exert their functions. Primed T cells are equipped with specific sets
of chemokine and adhesion molecules to guide them towards their final destinations (165,
166). For instance, increased expression of α4β7 integrins and chemokine receptor 9
(CCR9) directs effector T cells to the gut (167, 168), expression of α4β1 integrins direct
these cells to the brain (168), whereas T cells with high levels of cutaneous lymphocyte
associated antigen (CLA+) preferentially home to cutaneous inflammatory sites (169,
16
Introduction
170). To sum it all, during a DC-T cell interaction, the DC activates (signal 1),
costimulates (signal 2), polarizes (signal 3), and directs homing of primed cells (signal 4).
DC-T cell-HIV-1 Interaction
Mucosal DCs are amongst the first targets of infections during sexual transmission of
HIV-1 (129). At the site of infection, DCs pick up HIV-1, migrate to peripheral lymph
nodes, and effectively present HIV-1 peptides to naïve T cells and prime specific T cell
responses (171-175). Nonetheless, DCs play the role of a double-edged sword by
transferring whole viral particles across the immunological synapses, and in this case it is
called a virological synapse (129, 176-179). This leads to an efficient viral spread to
CD4+ T cells. The DC-induced T cell activation and subsequent proliferation triggers a
vicious cycle of viral dissemination (129, 179). (See figure 7). HIV-1 exposed DCs and
their ability to prime T cell responses will be the basis of this thesis.
Figure 7: HIV-1 dissemination from mucosal surfaces to lymph nodes.
During sexual transmission, HIV-1can establish contact with the intraepithelial DCs/LCs in the luminal surfaces as they extend their
dendrites to sample for luminal content or penetrate through lesions caused by venereal diseases, breaks in the mucosa caused by the
sexual intercourse, or through direct capture and transfer of virus by epithelia cells to the DCs located underneath. Within the DCs,
HIV-1 is trapped in specialized compartments that communicate with the external medium ready for transfer to T cells (178). DCs
17
Introduction
migrate to the regional lymph nodes where they form clusters with T cells and transfer infectious viral particles through the infectious
synapse to the T cells.
So far, in vitro studies by Granelli-Piperno et al (2004) have demonstrated that
supernatants derived from cocultures of HIV-1 pulsed MDDCs and autologous T cells
from either uninfected or infected individuals suppressed T cell proliferation in an IL-10
dependent manner (180). Of note, the HIV-1 infection did not induce DC maturation nor
were maturation stimuli able to induce maturation in already infected DCs (180).
Kawamura et al (2003) found that HIV-1 infected DCs significantly downregulated CD4
expression on DCs, after which stimulation with CD40L induced increased levels of IL12p70 and decreased levels of IL-10. Allogeneic cocultures of CD4+ T cells with HIV-1
infected DCs impaired T cell proliferation and IL-2 production, which was reversible by
addition of sCD4 (181). On the contrary, findings by Smed-Sorensen et al (2004)
revealed that HIV-1 infected DCs failed to produce IL-12p70 in response to CD40L
stimulation and had increased production of TNF-α (182). The formation of
immunological synapses are important for adequate TCR stimulation and immune
responses are impaired by HIV-1 as HIV-1 infected T cells poorly conjugate with APCs
and the synapses are abnormal due to the accumulation of TCR and Lck in the recycling
endosomal compartments. This was shown to be nef dependent as nef severely affected
their intracellular trafficking and signal transduction. Therefore, alteration of endocytic
and signaling networks at the immunological synapse likely impacts the function and fate
of HIV-1 infected cells (183). This important research area is still lacking many facts and
numerous studies are giving opposing findings. We have therefore conducted these
studies to gain better understanding regarding the immunological responses generated
from an HIV-1-DC-T cell interaction.
HIV-1 Specific responses
As discussed above, rapid or slow disease progression, exemplified in LTNPs, ECs, and
VCs, is dependent on both the host and viral properties/factors (29-31, 57-85). The
importance of HIV-1 specific immune responses in these individuals has been thoroughly
investigated and found to play critical roles in controlling viral replication and disease
progression (171-174, 179, 184, 185). In rapid progressors, HIV-1 specific CD4+ T cell
responses are generally weak or absent at all stages of the disease, whereas HIV-1
specific CD8+ T cell responses are more noticed but also wane as the disease progresses
(175, 185). However, increasing evidence suggest that early on in infection, HIV-1
specific CD4+ T cell responses are available but declines two - three weeks after infection
(171, 186) probably due to lack of CD127 expression (187). Longitudinal studies
revealed gag specific cytotoxic responses, very low numbers of infected CD4+ T cells,
and higher CD4+ T cell numbers for over 8 years in LTNPs as compared to rapid
progressors. Of note, gag specific responses were also noted during acute infection in
rapid progressors, but eventually disappeared with the onset of AIDS (173). Findings by
18
Introduction
Chinis et al (2003) (172) in pediatric patients showed highest HIV-specific CD8+
responses to gag, followed by gp120, gp41, and the V3 loop. Our in vitro studies
confirmed that HIV-1 exposed DCs could prime gag, env, and pol-specific CD4+ and
CD8+ T cell responses as well. We identified new CD4+ T cell specific epitopes and
follow up studies in acutely infected individuals revealed that these epitopes were
amongst the earliest recognized in vivo, but were lost presumably through activation
induced CD4+ T cell depletion (171). Rosenberg et al (1997) (174) showed that
individuals controlling viremia in the absence of ART, mount polyclonal, persistent, and
vigorous HIV-1 specific CD4+ T cell proliferative responses with increased production of
IFN-γ and antiviral chemokines. Therefore, the presence of protective cellular immunity
in LTNPs offers hope for a possible control of HIV-1 infection, if effective vaccines can
be created with the ability to elicit similar responses. However, the major hurdle to
achieve this vaccine is the discovery that HIV-1 specifically infects HIV-1 specific T
cells (184). The phenomenon that HIV-1 specifically infect the very cells that are
supposed to respond to it (184) adds skepticism to the fact that vaccines generating HIVspecific T cells may simply create potential targets for efficient HIV-1 replication.
Immunomodulatory Potentials of HIV-1
HIV-1 has adapted myriad ways to evade and hijack the immune system and to establish
persistent infection in humans (188). The hallmark of HIV-1 infection is the progressive
loss of immune cells ultimately leading to increased susceptibility to OIs and eventually
AIDS (189). HIV-1 affects the immune system in several ways that range from general
activation of the immune system, induction of general immune suppressive mechanisms,
and to a greater extent initiates direct and indirect killing of infected and uninfected cells
(189). The CD4+ T cells, known to regulate the adaptive arm of the immune system, are
the same cells preferentially infected by HIV-1. CD4+ T cell depletion during HIV-1
infection occurs via accelerated destruction, relocation of cells to special compartments
due to changes in homing receptors, chronic activation and T cell death, and impaired
regeneration of new T cells (190). APCs including DCs, monocytes, and macrophages
are also main targets of HIV-1 infection and contribute considerably to viral pathogenesis
(15). The levels of macrophages are not significantly affected in HIV-1 infected
individuals as oppose to monocytes and DCs. Their role as reservoirs for HIV-1 infection
provides sources of virions, which can infect more cells (16, 51). DCs contribute even
further to HIV-1 pathogenesis as they besides their ability to serve as reservoirs are
reduced in number in peripheral blood (191, 192), exhibit changes in their phenotype and
have impaired functionality (180, 193). HIV-1 proteins have been shown to alter the
expression of costimulatory molecules as well as chemokine receptor expression on DCs
(193-195). HIV-1 infected DCs in contact with T cells fail to give optimal feedback
signaling to T cells, due partly to impaired IL-12 production, which in turn fail to provide
optimal signals for DCs survival owing to decreased CD40L expression by the T cells
19
Introduction
(182). Furthermore, sustenance of T cell proliferation is impaired and this is partly due to
decreased IL-2 secretion (181, 196).
Immunostimulatory and Inhibitory Molecules at a DC-T cell
Encounter
Stimulation via positive costimulatory molecules during a DC-T cell encounter enhances
initial activation, provides additional signals for cell division, and favors survival or
induction of effector functions, such as cytokine secretion and cytotoxicity (197). The
most important costimulatory molecules belong to the B7-1 (CD80), B7-2 (CD86), CD28
or CTLA-4 superfamilies and upon ligation, depending on the prevailing circumstances,
they will positively or negatively regulate immune responses. Positive costimulatory
molecules are generally members of the immunoglobulin superfamily, e.g. CD28, and the
inducible T cell costimulator (ICOS), or the tumor necrosis factor receptor (TNFR)
family, e.g. CD27, OX40 (CD134), 4-IBB (CD137), and CD30 (198-200). In addition to
inducing costimulatory signals to T cells, DCs can also be activated via the ligation of
CD40 on its surface with CD40L on the T cells. Ligation of CD40 license DCs to
increase expression of B7 molecules, secretion of IL-12 needed for TH1 responses, and
also to drive effector Tc responses (201, 202) enhancing both positive costimulation and
survival of the DCs and T cells. On the contrary, negative costimulatory molecules
inhibit TCR mediated responses, impair T cell division, functional maturation, and
induces T cell tolerance or anergy (197). Normal physiological immunosuppression
makes sure to prevent responses to self antigens and will suppress excessive immune
responses deleterious to the host and involve for instance Tregs. Tregs suppress myriad
responses against autologous tumor cells (203), allergens (204), pathogenic or
commensal microbes (205), allogeneic organ transplants (206), and fetus during
pregnancy (207). Tregs may be naturally developed in the thymus and delivered to the
periphery called natural Tregs (nTregs) or induced from circulating lymphocytes called
adaptive Tregs (aTregs). aTregs have two functional subsets, type 1 Tregs (Tr1) and Tr3
Tregs depending on IL-10 and TGF-β, respectively (208, 209). Absence of Tregs leads to
autoimmune disease as shown in both mice and humans (210-212). Some pathogens
(HIV, HCV, and parasites) exploit the normal physiological immunosuppressive
responses to subvert protective immune responses to the host enabling the pathogens to
persist and establish chronic infections (205). The mechanisms of action of Tregs vary
according to type. Once activated by a particular antigen, Tregs can suppress responder T
cells irrespective of whether they share antigen specificity with the Tregs or not (213).
Tregs function by secretion of immunosuppressive cytokines, e.g. IL-10 and TGF-β,
establishing cell contact with the unwanted cell, or by functional modification or killing
of APCs (214). Contact-dependent suppression by Tregs (Figure 8) may involve
granzyme and perforin mediated killing, downregulation of costimulatory molecules on
APCs, or stimulation of DCs to express indoleamine 2, 3 dioxygenase (IDO), an enzyme
20
Introduction
that catabolizes the essential amino acid tryptophan to kynurenines that are toxic to T
cells. Tregs suppressive mechanisms appear to also depend on expression of the negative
costimulatory molecules, cytotoxic T lymphocyte antigen-4 (CTLA-4) and partially on
lymphocyte-activation gene 3 (LAG-3) on their surfaces (214).
Figure8: Contact dependent mechanisms of regulatory T cell functions
Regulatory T cells use different mechanisms of action and they include out competing effector T cells for binding on DCs due to their
increased affinity, modulation of DC functions by dampening the expression of costimulatory molecules and or direct cell killing of
DCs or effector cells
Thus far, several negative costimulatory molecules, viz., programmed death-1 (PD-1), T
cell immunoglobulin and mucin domain 3 (TIM-3), TNF-related apoptosis inducing
ligand (TRAIL), and CD160 together with suppression associated transcription factors
like B-lymphocyte-induced maturation protein-1 (BLIMP-1), FOXP3 and deltex-1
(DTX1) have been reported on activated T cells exerting suppressive responses (215221). Inhibitory costimulatory molecules regulate cell activity through diverse
mechanisms. For instance, the engagement of CTLA-4 to CD80/86 or PD-1 to PD-ligand
1/2 induce signaling cascades leading to impaired TCR mediated IL-2 production and T
cell proliferation (221-223). HIV-1 has been shown to exploit the immune modulatory
phenomena to their advantage by converting more naïve T cells to Tregs thereby
suppressing HIV-1 specific CD4+ and CD8+ T cells (224, 225). Assessment of T cells ex
vivo from HIV-1 infected patients and in vitro allogeneic mixed lymphocyte reactions
(MLRs) showed increased expression of PD-1 and CTLA-4 in the presence of HIV-1 and
blockade of these molecules significantly improved T cell functionalities (219, 221). The
mechanisms behind impaired T cell activity in HIV-1 infection have been reported, and
are largely attributed to PD-1 and CTLA-4. PD-1 expression on HIV-specific CD8+ T
cells correlated with impaired HIV-1 specific CD8+ T cell functions as well as predictors
of disease progression. Increased expression of PD-1 on CD4+ and CD8+ T cells
correlated positively with viral load and inversely with CD4+ T cell counts (226).
CTLA-4 expression was also upregulated on HIV-1 specific T cells and was positively
correlated with disease progression and negatively with the capacity of CD4+ T cells to
21
Introduction
produce IL-2 in response to viral antigens (221, 227). Blockade of these molecules
significantly restored T cell functions and their ability to respond to viral antigens (221,
226, 227). We also demonstrated in vitro, the expression of PD-1 and CTLA-4 on HIV-1
primed T cells (219, 220). Recent studies suggest a role of TIM-3 in mediating T cell
impairment and primarily acts following its ligation with galactin-9 and/or
phosphatidylserine ligands, triggering dysfunctional T cells and eventual cell death (215,
228-230). The levels of TIM-3 expression on T cells from HIV-1 subjects have correlated
positively with HIV-1 viral load and inversely with CD4+ T cell count. Progressively
infected patients upregulated TIM-3 expression on HIV-1 specific CD8+ T cells resulting
in failure to proliferate and secret cytokines in response to antigenic stimuli (230).
CD160, another inhibitory molecule impairs T cell activation through its ligand HVEM
by interfering with tyrosine phosphorylation proteins of CD3/CD28 T cell activation
pathways (231). The expression of CD160 has been reported in both acute and chronic
HIV-1 infections. Yamamoto et al (2011) (232) showed expression of multiple inhibitory
molecules, e.g. PD-1, CD160, and 2B4, on HIV-1 specific CD8+ T cells, which correlated
both with viral load and dysfunctional cytokine production (232, 233). In addition, LAG3 expression on T cells in HIV-1 subjects has been disputed seeing that comparative
studies between CMV and HIV-1 showed increased expression PD-1, CD160, 2B4, and
LAG-3 on CMV specific CD8+ T cells but not LAG-3 on the HIV-1 specific CD8+ T
cells (232). However, other reports have shown enhancement of LAG-3 on T cells in
HIV-1 infected subjects with unrestrained HIV-1 replication (234, 235). Our in vitro
studies showed that HIV-1 exposed DCs primed naive T cells with elevated gene and
protein expression levels of LAG-3 (220). LAG-3 binds with a higher affinity to MHC
class II molecules compared to the CD4 receptor and facilitates immunosuppression,
especially via Tregs (236), and has been linked to functional exhaustion of CD8+ T cells
in persistent viral infections (PVI) (237).
The role of TRAIL as a negative inhibitory molecule was brought to light when
Lunemann et al (2001) (238) demonstrated that TRAIL could inhibit antigen specific T
cell activation without necessarily inducing apoptosis. TRAIL also expanded
CD4+CD25+ Tregs in mice to control experimental autoimmune thyroiditis, another
possibility that TRAIL is not just an apoptosis inducing molecule, but could also serve as
a regulator of a hyperactivated immune system (239). We confirmed this by showing that
HIV-1 exposed DCs gave an increased expression of TRAIL on T cells, which
suppressed immune activation without inducing apoptosis (219, 220). We further showed
that only a synergistic effect of a combined blockade of TRAIL, CTLA-4, and PD-1
could significantly restore T cell proliferation, projecting TRAIL as a potential candidate
in the ranks of inhibitory molecules (219).
Notably, the transcription factor DTX1 has been linked to T cell anergy and suppression
of T cell activation through NFATc by both ubiquitin E3-dependent and independent
22
Introduction
mechanisms. Deletion of DTX1 promoted T cell activation, conferred resistance to
tolerance and led to increased inflammation and autoimmunity (218). On the other hand,
NFATc1 signaling has been shown to regulate PD-1 (240) and CTLA-4 (241) expression
upon T cell activation. BLIMP-1 has been termed a transcriptional repressor and
regulates T cell homeostasis and function (242) likewise regulating T cell exhaustion in
chronic viral infections (217). Elevated expression of BLIMP-1 in virus specific CD8+ T
cells repressed key aspects of normal memory CD8+ T cell differentiation and promoted
high expression of inhibitory receptors, PD-1, LAG-3, CD160, and 2B4, during chronic
LCMV infections (217). Both IL-2 and IL-2 activator Fos were directly repressed by
BLIMP-1, which attenuated T cell proliferation and survival (243).
The mechanisms whereby HIV-1 induces the upregulation of inhibitory molecules
remains unclear and therefore efforts to uncover the molecular mechanisms underlying
their upregulation will open up newer avenues for preventing viral suppression of the
host immune system.
Conclusions
In the acute phase of HIV-1 replication humoral and cellular immune responses are
generated and control this chronic disease for a considerable time period. Due to several
factors in the HIV-1 pathogenesis, viral immunity starts declining including loss of CD4+
T cells, DCs, and other immune cells, resulting in gradual increase of the PVL. The
individual eventually becomes vulnerable to opportunistic infections and malignancies,
which leads to death unless treated with ART. Humans can manage chronic viral
infections, e.g. CMV, EBV, and HSV, by generating immune responses that control
infection throughout life but has failed to do so against HIV-1. Therefore, knowledge
about the type of responses generated by the HIV-1 infection and subsequently, the
mechanisms whereby these immune responses gradually decline over time is of
paramount importance. Furthermore, in depth studies are needed to unveil the underlying
mechanisms contributing to HIV-immunomodulation, a critical aim in our strides which
can help design an effective anti-HIV vaccine.
23
Introduction
24
Aims and Objectives
AIMS AND OBJECTIVES
The studies were aimed to investigate the HIV-1 specific T cell responses generated by
DCs pulsed with HIV-1, and to understand the molecular mechanisms and underlying
signaling pathways associated with the onset of immune impairment in HIV-1 infection
in vitro.
Specific Aims
Paper 1: To decipher the type of HIV-1 specific T cell responses primed from naïve T
cells by HIV-1 pulsed DCs in vitro and to compare these responses with those existing in
HIV-1 infected individuals in vivo.
Paper 2: To investigate the effects HIV-1 exert on DCs’ ability to prime naïve allogeneic
T cell responses.
Paper 3: To investigate the array of inhibitory molecules upregulated by the presence of
HIV-1 during DC priming of T cell responses.
Paper 4: To unveil the underlying mechanisms through which HIV-1 give rise to
immunosuppressive T cells when exposed and primed with DCs.
25
Aims and Objectives
26
Methods
METHODS
Dendritic cell preparation
DCs were differentiated from monocytes in peripheral blood mononuclear cells
(PBMCs). Fresh blood samples from HIV-1 infected individuals or healthy donors were
diluted (50%) with RPMI1640 supplemented with 2mM EDTA and layered on FicollHypaque. The Buffy coat was separated by density gradient centrifugation at 2200rpm
for 20minutes with no brakes. Buffy coat was carefully collected and washed 4 times in
RPMI1640 at 1800rpm, 1500rpm, 1100 rpm, and 900rpm every 10minutes. Thirty to
forty million PBMCs were plated in tissue culture dishes in 1% plasma at 37°C in 5%
CO2 for 1-2 hours for monocytes (CD14+) to adhere. The non-adherent cells (NAC) were
washed off with RPMI1640. Adherent cells (DC progenitors) were differentiated in 1%
plasma supplemented with 100IU/mL recombinant granulocyte macrophage–colony
stimulating factor (rhGM-CSF) 300U/mL and recombinant human IL-4 (rhIL-4) (Figure
9).
Figure 9: Propagation of monocyte derived dendrite cells from blood progenitor
Cytokines were replenished every 2 days to facilitate the differentiation of progenitors
into MDDCs. Immature MDDCs were harvested on day 6. DC maturation was induced
by adding 30ng/mL of polyinosinic acid (Poly I:C).
27
Methods
Naïve T cell Separation
Naïve bulk T cells were prepared from NACs derived from PBMCs by magnetic bead
separation following manufacturer’s instructions (Miltenyi Biotech). In brief, naive T
cells were negatively selected using anti-CD19 microbeads for B cells, anti-CD56 for NK
cells, anti-CD45RO for memory T cells and anti-CD14 for monocytes. NACs were
incubated for 15 minutes with bead antibodies, washed once in MAC buffer and loaded
on a magnetic column. Labeled cells were trapped in the column while the unlabelled
(negatively selected naïve bulk T cells) cells were collected as the effluent.
Virus preparation
Virions of HIV-1MN (X4-tropic), clade B were produced by infection of CL.4/CEMX174
(T1) cells and purified by sucrose gradient ultracentrifugation as described previously
(244). Samples were titrated for the presence of infectious virus using AA2CL.1 cells and
HIV-1 p24 antigen capture kits (AVP, NCI). Aldrithiol-2 (AT-2)-inactivated HIV-1 (AT2 HIV) was prepared as described previously (245). In paper 1, infectious HIV-1MN (Lots
3807, 3966, and 3941: AVP, SAIC Frederick, Inc., NCI Frederick) and AT-2 HIV-1MN
(Lots 3808, 3965, 3937, and 3934) were used.
HIV-1 BaL/SUPT1-CCR5 CL.30 (Lot P4143) was produced using chronically infected
cultures of ACVP/BCP Cell line (No. 204), originally derived by infecting SUPT1-CCR5
CL.30 cells (graciously provided by Dr. J. Hoxie, University of Pennsylvania) with an
infectious stock of HIV-1 BaL (NIH AIDS Research and Reference Reagent Program,
Cat. No. 416, Lot 59155). Virus was purified by continuous flow centrifugation using a
Beckman CF32Ti rotor at 30,000 rpm (~90,000 x g) at a flow rate of 6 liters/hour
followed by banding for 30 minutes after sample loading. Sucrose density-gradient
fractions were collected and virus-containing fractions were pooled and diluted to less
than 20% sucrose, and virus pelleted at ~100,000 x g for 1 hour. The virus pellet was
resuspended at a concentration of 1,000 x g relative to the cell culture filtrate and aliquots
frozen in liquid N2 vapor. 1500ng/106 p24 equivalents of HIV-1 were pulsed with mature
or immature DCs for 24 hours before culturing with naïve T cells.
DC T cell cocultures
HIV-1-overnight-exposed immature(I) and mature(M) DCs were harvested, washed
twice, and cocultured with CFSE labeled naïve T cells in 5% pooled human serum (PHS)
in RPMI1640 at a ratio of 1:10 in 96-well plates. Autologous cocultures as in paper 1
were restimulated at day 7, 14, 21 and 28 by adding autologous HIV-pulsed DCs,
whereas allogeneic cocultures as in papers II, III and IV were restimulated once on day 7
by adding the same groups of DCs utilized to initiate the priming event. Expanded T cells
were analyzed for numerous parameters, e.g. antigen-specific epitopes on primed T cells
by ELISPOT assays, cytokine analysis by ELISA, Luminex, and intracellular staining. T
28
Methods
cell proliferation was measured by flow cytometry or 3H-Thymidine incorporation using
a β-counter. Gene expressions in primed T cells was by microarray and qRT-PCR,
whereas expression of cell surface markers was by flow cytometry
ELISPOT:
The ELISPOT assay is a widely used technique primarily developed to identify antibody
secreting cells (246). Today ELISPOT has been used to measure specific cytokineproducing cells as well as measuring epitope responses to particular peptides. ELISPOT
assays employ the sandwich enzyme-linked immunosorbent assay (ELISA) technique. In
brief, a polyvinylidene difluoride (PVDF) microplate is pre-coated with a monoclonal or
polyclonal antibody specific for the chosen analyte overnight at 4oC or 2-3 hours at 37oC.
The cells under investigation are then pipetted into the wells together with the stimulant
and the microplate incubated. During incubation (usually overnight), secreted antibodies
or cytokines bind to the immobilized antibody, in the immediate surroundings. Unbound
cells are washed and a biotinylated polyclonal antibody specific for the chosen analyte is
added to the wells for about 2 hours, washed and further incubated with an alkalinephosphatase conjugated to streptavidin. The unbound enzyme is subsequently removed
by washing and a substrate solution (usually BCIP/NBT) is added. Blue-black colored
precipitates are formed and appear as spots at the sites of analyte localization, with each
individual spot representing an individual analyte-secreting cell. The spots can be
counted with an automated ELISPOT reader system or manually, using a
stereomicroscope. In our system, expanded T cells were harvested and added to IFN-γ
ELISPOT plates at 50,000 T cells/well and restimulated with DCs infected with
Vaccinia-vectors (MOI = 2) (V-ctr, V-env, V-gag, V-pol, V-nef) to measure class 1
responses or pulsed with gp160, nef, p66, or p24 proteins (5 μg/ml; Protein Sciences
Corporation, Meriden, CT) for class II responses. The ratio of DCs to T cells was 1:10
and the assays were carried out as described above. Epitope mapping was by ELISPOT
using pools of overlapping peptides to initiate the responses. Positive wells were
reconfirmed with the single peptides by intracellular staining.
Measurement of secretory factors
ELISA: Cell secretions in supernatants from cocultures were measured by ELISA,
Luminex or intracellular cytokine staining. The principle of ELISA is much same as
ELISPOT with the only difference being the impossibility in estimating the number of
cells actually secreting the analyst of interest. Levels of p24, IL-10 and TGF-β in
supernatants from our cocultures were measured using ELISA kits as instructed by the
manufacturer (eBioscience). Supernatants from day 8 of cocultures were also analyzed
using the BioPlex Cytokine Luminex assay for IL-10, IL-2, IL-4, IL-5, IL-13, IL-17,
TNF-α, G-CSF, TGF-β, IFN-γ, RANTES, PDGF-β, MCP-1, IP-10, bFGF, eotaxia, IL-9,
IL-8, IL-6, IL-1R-α, VEGF, and IL-1Rβ following manufacturer’s instructions.
29
Methods
Intracellular cytokine staining: The benefit of intracellular cytokine staining is
the simultaneous analysis of surface molecules and intracellular antigens or proteins at a
cell to cell level. The cells are first stained for surface expression, fixed with a fixative
(e.g. formaldehyde) and permeabilized with a detergent (e.g. saponin) to allow antiantigen or cytokine antibodies to stain intracellularly. To measure cytokine levels in in
vitro stimulated cells, blockade of cytokine secretion with protein transport inhibitors e.g.
monensin or Brefeldin A (BFA) solution at the final hours of stimulation is important. In
our experiments, intracellular cytokine staining for the assessment of cytokine/chemokine
production was performed on primed T cells. Primed CD4+ and CD8+ T cells were
initially stimulated with HIV-1 MDDCs for 6 hours and in the presence of 10 μg/mL of
BFA. Fluorochrome-conjugated antibodies against IL-2, TNF-α, MIP-1β, and IFN-γ were
used. Intracellular cytokine staining for FOXP3 was performed according to
manufacturer’s instruction (eBioscience) and granzyme/perforin expression measured
from the allogeneic cocultures after day 7 restimulation (day 8). Samples were acquired
on a FACSCalibur or LSRII and data was analyzed using Cell Quest or FlowJo software.
T cell proliferation analysis
T cell proliferation is an important measurement for T cell response to particular stimuli.
Various methods are available but herein in this thesis have we described two methods
consistently. T cell proliferation was measured by either monitoring the degree of
dilution of the dye carboxyfluorescein succinimidyl ester (CFSE) by flow cytometry or
monitoring the levels of radioactive 3H-Thymidine incorporated in the proliferated cells
within the last 16 hours of coculture. CFSE is a dye that will spontaneously and
irreversibly binds to the amino acid lysine in both intracellular and cell surface proteins
of a cell. When the T cells divide, CFSE is equality distributed between the daughter cells
and as a result, the fluorescence intensity of the dye will be halved as the number of cell
cycles doubles. Halving of cellular fluorescence intensity marks each successive
generation in a population of proliferating cells and is readily measured by flow
cytometry (247) (Figure 10). In this thesis, naïve T cells (pellet) were resuspended in
10μM CFSE in 0.1%BSA for 10 minutes at 37oC. Cells were washed and cocultured with
mock or HIV-1 exposed DCs. At the end of culture, CFSE dilution was measured in
expanded T cells. The measurement of T cell proliferation by 3H-Thymidine was an
alternative method used in this thesis. During cell division, cell DNA replicates and thus
their precursors adenines, cytosine, guanine and thymidine are added.
30
Methods
Figure 10: CFSE dilution, a measurement of T cell proliferation.
The parent cells are coupled with CFSE dye and each cell division decreases the intensity to half as it is divided up between the parent
and the daughter cells. (A) Represents a histogram for allogeneic T cell activation with two peaks. The peak to right represents
parent/undivided cells with the strongest intensity and the peak to the left represent the daughter cells with weaker intensity. (B)
Shows a histogram for PHA activated T cells with clear peaks and each peak represents a successive cycle which is not visible in
allogeneic reactions. Measurement of the fluorescence intensity from parent to daughter cells gives a measurement of T cell
proliferation.
Radioactive thymidine (3H-Thymidine), will therefore be incorporated into the DNA as
the cells proliferate and levels will be measured with a scintillation counter. In this thesis,
4μCi of 3H-Thymidine was added to the cocultures during the last 16 hours. Cells were
harvested to a membrane, using a cell harvester, and membrane washed 5 times with
water and kept to dry. The membrane was then laminating in 5ml β-plate scintillation
fluid and loaded to the beta counter in a cassette. The level of radioactivity was measured
and shown to correlate positively with T cell proliferation.
Fluorescence activated cell sorter (FACS)
The principle guiding FACS is the rapid separation of cells in suspension based on the
size, granularity and color of their fluorescence. Antibodies towards a target(s) in
suspension are conjugated with a fluorescence dye. A cell suspension is acquired through
a nozzle permitting a stream of single cells to pass through. A laser beam directed
towards the stream excites the targets as each labeled cell passes through resulting in
fluorescence, which is detected by a detector. The cells are first separated depending on
light scatter (forward scatter measures cell size and side scatter measures cell
granularity). Different fluorochromes on different cell types or molecules on the same
cell will be recorded at different wavelength and depicted as quantitative measurements
for cell types, numbers or percentage of protein expressed on the cell. In our experiments,
31
Methods
antibody-cell-suspensions were incubated for about 30 minutes in FACS buffer (0.5%
BSA), washed and collected in FACS tubes for acquisition. Flow cytometric acquisition
was performed on a four color FACSCalibur or LSRII. Data were analyzed using Cell
Quest or FlowJo software.
Gene expression by real time PCR (SYBR® Green)
Real-time PCR allows the detection of amplified DNA in real time and therefore,
measures the kinetics of the reaction as compared to traditional PCR that measures only
the final phase of the reaction. The SYBR® Green technique is an alternative real time
method whereby the measurement is based on the intensity of fluorescence emission of a
dye (SYBR® Green). During amplification, dsDNAs are formed within which SYBR®
Green intercalates resulting in fluorescence. The intensity of fluorescence emission is
proportional to the amount of dsDNA formed, which depends on initial starting material.
A house keeping gene is required to normalize the final values thereby checking for
differences in starting concentrations. In our assays, mRNA was isolated using a
commercial Spin technology according to the manufacturer’s protocol (Qiagen). First
strand cDNA was made from 1 μg of mRNA in a 20 μL reaction volume. Twenty micro
liters of reaction mix containing 1 μL of oligo dT (50 μM), dNTP (10 mM),
SuperScriptTM III (200 U/mL), DTT (0.1 M), RNaseOUTTM (40 U/mL), 4 μL of 5 x
First-Strand buffer and 1 μg of mRNA in 11 μL of nuclease-free water. β-actin was used
as our endogenous control and qRT-PCRs was performed using Prism 7900HT. Samples
were run in triplicate each, a 10 μL reaction mixture that contained 5-10nM (1 μL each)
of forward and reverse primers, 5 μL Fast SYBR® Green Master Mix, 4 nM (2 μL)
template DNA and 1 μL of water. Results were expressed as ΔΔCt and the values
normalized.
Statistical analysis
Statistical significance was measured using GraphPad Prism 4.0 software (GraphPad, La
Jolla, CA, USA). The Wilcoxon non-parametric matched pairs test was used to compare
between paired groups showing decline in specific HIV-1 responses over time as in paper
I, whereas the unpaired t test and One way ANOVA-Tukey’s test was used to compare
between the groups as of paper II, III and IV. Of note, due to considerable degree of
variability within the experiments, we obtained statistical values after
normalizing/transforming the data whereby each data set was divided by the sum total of
all values in the data set. Each value within a group is presented as percentage.
Differences were considered significant when *P<0.05, **P<0.005 and ***P<0.001.
32
Findings and Discussion
FINDINGS AND DISCUSSION
Paper 1
Background
The progression to AIDS is eminent in most cases of HIV-1 infected subjects worldwide
if not treated with ART (90). This is owed to insufficient immune responses generated
and loss of CD4+ T cells. HIV-1 specific cellular immune responses have been reported
in rapid progressors and shown to wane as the disease progresses (175, 185). In
connection to this, HIV-1 favorably infect HIV-1 specific CD4+ T cells partially
explaining why the cellular responses rapidly disappeared over time (184). LTNPs
control viral replication and delay disease progression owing in part to efficient priming
and persistent HIV-1 specific T cells (172-174). These subjects have prompted intense
interest to understand the mechanisms under which sustained immune responses are
generated (90). DCs are professional APCs and even though they are responsible for viral
dissemination to T cells (129, 179) they also prime HIV-1 specific cellular responses
(172, 173). Drawing from a vaccine study by Lu et al (248), using autologous DCs pulsed
ex vivo with aldrithiol-2 (AT-2) inactivated HIV-1, they showed increase levels of HIV-1
specific T cells and decreased viral load and an eventual control of viral load in
chronically infected subjects. Different HIV-1 antigenic constructs have been used to
pulse DCs and shown to prime HIV-1 specific responses in vitro (249-253). However, the
capacity of DCs pulsed in vitro with whole virions; the most physiological source of
HIV-1 antigens, to prime T cells in vitro had not been fully investigated. Nevertheless,
animal studies using whole HIV-1 to prime HIV-1 specific responses had been conducted
(254, 255). The aim of this study was to examine whether whole HIV-1 could serve as an
efficient source of antigens for DCs to prime HIV-1 specific T cells from naïve cells in
vitro. Of note, we used both infectious and noninfectious AT-2 HIV-1 to allow us to
compare the nature of responses generated between these antigen sources, findings which
could be of great interest for vaccine design.
Principal Findings
The study investigated the effects exerted by HIV-1 on DC phenotype and functionality
and showed that HIV-1 did not affect the phenotypic profile of mature MDDCs, which is
an important component needed for naïve T cell stimulation. In addition, MDDCs
retained their ability to process, present, and activate HIV-1 specific CD4+ and CD8+ T
cells. Further to investigate if MDDCs could prime HIV-1 specific T cell responses from
naïve T cells, DCs were loaded with AT-2 or infectious HIV-1 and cultured with naïve T
cells in vitro. Priming with noninfectious AT-2 HIV-1 yielded a slightly higher response
compared to infectious HIV-1. MDDCs pulsed with HIV-1 activated significantly more
33
Findings and Discussion
HIV-1 specific CD4+ than CD8+ T cell responses. Assessment of the primed responses by
ELISPOT assays using a matrix of HIV-1 overlapping peptides showed that the epitopes
recognized by the primed T cells were from the env-gp41, env-gp120, gag-p17, gag-p24,
pol-INT, and pol-RT. The in vitro primed specific T cells secreted high levels of effector
cytokines (IL-2, IFN-γ, MIP, and TNF-α) and were comparable with responses existing
in infected individuals (256-259). We showed that five out of the ten CD4+ T cell
responses primed in vitro matched the responses seen ex vivo in HIV-1 infected
individuals at different stages of disease (256-259). The unmatched responses
(ELKKIIGQVRDQAEHLK pol-IN157-173, MTKILEPFRKQNPDIVIY pol-RT163-181,
AVLSIVNRVRQGYSPLSF env-gp41700-717, LELDKWASLWNWFNITNW envgp41601-678, and RPVVSTQLLLNGSLA env-gp120252-266 were considered novel as these
epitopes had not been reported before. We were able to detect three out of five of these
novel responses, i.e. pol-IN157-173, env-gp120252-266 and env-gp41700-717, in acutely infected
HIV-1 individuals suggesting that these responses arise very early in infection and
disappear even before they are detected. Although less frequent, HIV-1 specific CD8+ T
cell responses were also observed and they all matched the responses seen ex vivo in
HIV-1 infected individuals at different stages of disease (256-259). Improved priming of
specific CD8+ T cell responses were achieved after depletion of CD4+ T cells indicating
the possible dominance of CD4+ T cells over CD8+ T cells within the priming cultures.
Discussion/Conclusions
DCs help disseminate whole viral particles to T cells (129, 177-179), but can also prime
HIV-1 specific T cell responses in HIV-1 infected individuals (172-174, 179, 184, 185,
256-259). The ability of MDDCs to prime broad HIV-1 specific T cell responses in vitro
can be used in vivo to prime and also to activate HIV-1 specific memory T cells towards
the control of infection. Our study confirmed this notion showing that DCs pulsed with
infectious or AT-2 inactivated HIV-1 could prime T cell responses that arise naturally in
vivo in HIV-1 infected individuals and demonstrates that AT-2 HIV-1 was an excellent
source of antigens, a finding that may be of great importance in the quest for vaccine
development (248). The differences in responses between infectious and AT-2 HIV-1
remains unclear but could be attributed to direct effects of viral replication in the cells or
negative immunoregulatory effects exerted by HIV-1 on both DCs and T cells (180, 260).
All in vitro primed CD8+ T cell responses were confirmed in vivo in chronically infected
patients as compared to CD4+ responses where only five out of ten were confirmed. Of
interest, our studies in acutely infected HIV-1 individuals revealed that three out the five
of the novel epitopes existed initially in them but declined overtime, a finding that
confirmed the theory of rapid decline of CD4+ specific T cell responses during the course
of infection and the importance this may play in HIV-1 pathogenesis (175, 184, 185, 187,
261). Long-term studies in these infected subjects revealed both loss in gag, pol, env and
nef CD4+ specific T cell responses as well as IL-2 and IFN-γ secretion. This is true as
34
Findings and Discussion
HIV-1 specific T cell responses are preferential targets for elimination by HIV-1 (184)
and also as a consequence of chronic immune activation due partly to bacterial
translocation (41, 262). The overwhelming CD4+ T cell responses seen in vitro consisted
with the strong and broad CD4+ T cell responses we observed in individuals with acute
HIV-1 infection in vivo, which were greatly reduced or disappeared within three months
after onset of infection. Studies in LTNP have revealed the importance of HIV-1 specific
responses as they have increased and persistent levels for many years of infection (79, 90,
91, 172-174). Focus on enveloped based vaccines aimed at inducing neutralizing
antibody responses for sterilizing immunity (263, 264) has gradually shifted towards cellmediated responses (265-267), which have been shown to control viral load in
asymptomatic infected individuals (80-84, 86-89). Therefore, in depth understanding of
mechanisms for efficient priming of HIV-1 specific responses can be of paramount
benefit to foster the development of cell mediated therapeutic vaccines. On the other
hand, detailed studies to investigate the mechanisms behind the rapid decline in the CD4+
and CD8+ specific T cell responses in rapid progressors is crucial for the maintenance of
the eventual vaccine induced responses and which will be prime focus of our subsequent
investigations.
Paper II
Background
HIV-1 dissemination has been linked to DCs and T cells. DCs pick up HIV-1 at the sites
of infection, and migrate to the lymph nodes where they present viral peptides to T cells
thereby triggering specific immune responses (172-175) but at the same time transfer
whole viral particles to the CD4+ T cells (177-179). HIV-1 has adapted myriad ways of
evading and hijacking the immune system to establish a persistent infection in humans
(188). This includes viral disguise, general activation of the immune system, induction of
general immune suppressive mechanisms, and direct or indirect killing of infected and
uninfected cells (189). The majority of infected individuals with high viral loads have
both diminished levels and functionally impaired DC and CD4+ T cells (191, 192), which
reveals that the presence of high viral burden exerts negative and deleterious effects on
the host’s immune cells. Individual HIV-1 proteins, such as nef, vpr, and tat exert
negative effects on immune cells. Nef is associated with decreased surface expression of
MHC class I, CD80, and CD86 molecules on infected cells (194). In addition, nef
upregulates TNF-α production and Fas ligand (FasL) expression on DCs, resulting in
cytotoxic DCs with an impaired ability to activate CD8+ T cells (268). Vpr down
regulates the expression of costimulatory molecules on DCs (195). Given the myriad
effects observed when studying individual HIV-1 proteins, we wanted to study the effects
of the whole virion, i.e. the physiological source of HIV-1 antigens, on immune cell
functionality. Our aim therefore was to study the effects of whole HIV-1, infectious and
35
Findings and Discussion
noninfectious AT-2 HIV-1 BaL, on DCs ability to prime T cell responses. We were also
interested in pinpointing the main factors responsible for the immunomodulation.
Principal Findings
The results showed that in vitro exposure of DCs to both infectious HIV-1 and
noninfectious AT-2 HIV-1 impaired DC’s ability to prime naïve T cells leading to
decreased T cell proliferation. The involvement of T regulatory cell cytokines, i.e. IL-10
and TGF-β, was eliminated as there were no significant differences in their levels
between mock and HIV-1 exposed cocultures. We therefore postulated a contact
dependent suppressive mechanism as shown for naturally occurring regulatory T cells
(NTregs) (210, 214). Noteworthy, our findings showed that the addition of HIV-1 primed
T cells into new DC-T cell priming assays suppressed T cell proliferation. This indicated
that NTregs could be involved in T cell impairment but when the NTregs were depleted
from the naïve T cell pool, the impaired T cell proliferation persisted. The failure to
reverse T cell impairment in the absence of NTregs prompted an inquiry for inducible
suppressor T cells. To understand this concept, HIV-1 primed T cells were
immunophenotyped for inducible suppressor markers PD-1, CTLA-4, and TRAIL (221,
226, 227, 269). We showed increased expression of TRAIL, PD-1, and CTLA-4 on HIV1 primed T cells compared to mock T cells. Blocking TRAIL, PD-1, and CTLA-4
individually demonstrated a slight but insignificant restoration in T cell proliferation but
when simultaneously blocked did the restoration of T cell proliferation reach significant
levels.
Discussion/Conclusion
HIV-1 exposed DCs do not only infect T cells interacting with them (177, 178, 184) but
also, as we have shown in this study, induce immune impairment by giving rise to
increased expression of inhibitory molecules CTLA-4, TRAIL, and PD-1 on the primed T
cells. The T cells primed in the presence of HIV-1 induced impairment of other T cells in
a contact dependent manner. The mechanism of immune suppression by Tregs/suppressor
T cells varies depending on the microenvironment. Suppressor T cells are recruited to DC
areas to regulate antigen specific naïve T cell activation and are more efficient in their
interaction with DCs than naïve T cells due to their high expression of adhesion
molecules such as LFA-1 (210, 214). Suppressor T cells may as well induce suppression
by downregulating CD80/86 expression on DCs via a CTLA-4 dependent mechanism.
Certain suppressor T cells can directly kill or inactivate responder T cells by secreting
granzyme/perforin or immunosuppressive cytokine such as IL-10 and TGF-β (210, 214).
In our system, secretion of IL-10 and TGF-β was not responsible for T cell impairment,
rather a contact dependent interaction between cells. HIV-1 therefore has profound
immunomodulatory effects on DCs using them as vehicles of transport to the lymph
36
Findings and Discussion
nodes and infection of T cells (129, 177-179) as well as modulating DCs to prime
contact-dependent suppressor T cells.
Paper III
Background
Chronic viral infections, including HIV-1, are characterized by impaired immune
responses. HIV-1 affects the immune system in myriad ways but general immune
suppression is of critical importance for viral pathogenesis (189). Immune suppressive
mechanisms include the induction of immunosuppressive molecules on activated T cells
and expansion of Tregs (219, 221, 226, 269). Tregs dependent suppression may function
through the secretion of immunosuppressive cytokines, e.g. IL-10, and TGF-β (208), or
via cell-to-cell contact mechanisms such as granzyme and perforin (270, 271) killing
and/or through the delivery of negative signals to responder cells (272). Tregs may also
function by direct functional modification or killing of APCs, (214) downregulation of
costimulatory molecules on APCs in a CTLA-4 dependent mechanism and/or stimulate
DCs to express indoleamine 2, 3 dioxygenase (IDO), an enzyme catabolizing the
essential amino acid tryptophan to kynurenines and this substance is toxic to T cells
(214). In addition to expression of CTLA-4 on Tregs, HIV-1 primed T cells expressing
PD-1 and TRAIL (219, 221, 226, 227). We showed previously that TRAIL in
combination with CTLA-4, and PD-1 significantly impaired T cell proliferation in the
presence of HIV-1 and their simultaneous blockade drastically restored the T cell
proliferation. In this study we postulated other possible inhibitory molecules on T cells
primed by HIV-1 exposed DCs. LAG-3 and B and T lymphocyte attenuator (BTLA) have
been shown to be involved in regulation of cell cycle activity (216, 236) and functionality
of regulatory T cells (236). In addition, TIM-3 inhibited TH1 responses and promoted
immunological tolerance (229) and has been reported to be overexpressed in HCV
infected individuals (273). In addition to PD-1, CTLA-4, TRAIL, we therefore examined
the expression of LAG-3, BTLA, and TIM-3 on HIV-1 primed T cells. We also examined
the expression of CD160, as this is a potent immune regulator in disease conditions (274).
Furthermore, we studied the transcriptional factor BLIMP-1, which has also been
reported as a transcriptional repressor in CD8+ T cell exhaustion (217, 275) and T cell
homeostasis (242) and compared the molecular expression with mock primed T cells.
Principal Findings
Our findings demonstrated that presence of HIV-1 during DC priming of allogeneic naïve
T cells significantly increased expression of CTLA-4, PD-1, TRAIL, LAG-3, TIM-3, and
CD160 on the T cells primed by HIV-1 pulsed DCs. The gene expression levels of the
transcription repressors BLIMP-1 and FOXP3 were also significantly upregulated and
correlated with expression of inhibitory molecules. We further showed that the
37
Findings and Discussion
suppressor T cells coexpressed TIM-3, LAG-3, TRAIL, and/or CTLA-4 with PD-1. The
functional attributes of the HIV-1 primed T cell i.e; cytolitic proteins like perforin, and
granzyme, and cytokines; TNF-α, IFN-γ, and IL-2 were decreased compared to mock
primed T cells. This could be due to reduced T cell activation in the HIV-1 primed T cells
or impared abilty of production. Of note, increased expression of inhibitory molecules
correlated with BLIMP-1 expression and decreased T cell proliferation.
Discussion/Conclusions
We demonstrated expression of a broad array of negative costimulatory molecules and
elevated gene levels of transcription repressors BLIMP-1 and FOXP3 in HIV-1 primed T
cells and a correspondent decrease in functional attributes. These results correlated with
in vivo studies that showed positive correlations of TIM-3 (230), CD160 (232, 233),
LAG-3 (234, 235), CTLA-4 (227) PD-1 (221, 226, 227), and TRAIL (269) with viral
load and CD4+ T cell numbers. Suppressor T cells are known to induce suppression in a
contact dependent manner partly due to secretion of perforin and granzyme (210, 214,
270, 271). At the early phase of infection, HIV-1 specific CD4+CD25+ effector cytotoxic
T cells are generated that express increased levels of granzyme and perforin to control
viral replication (261) as well as IFN-γ and IL-2 (171), but the IFN-γ and IL-2 responses
declined overtime. Our suppressor T cells had decreased secretion of granzyme and
perforin which could be in part a reason behind the declining immunological functions in
HIV-1. In the same light, total levels of IFN-γ, TNF-α and IL-2 were also reduced which
could be a consequence of the decreased T cell activation or actually be due to impaired
production and immune activity induced by HIV-1. The secretion level per cell is
therefore important and needs further investigation for these cytokines. The
transcriptional repressor BLIMP-1 has been linked to regulate CD8+ T cell exhaustion
and correlates positively with expression of PD-1, LAG-3, CD160, and 2B4 during
chronic LCMV infections (217). We found that the inhibitory molecules we described
above correlated positively with BLIMP-1 concurring with the findings in LCMV
infections. Therefore, priming of T cells with HIV-1 exposed DCs in vitro leads to
expansion of suppressor T cells armed with numerous inhibitory molecules, which could
be part of the immune deficiencies seen in HIV-1 infected individuals.
Paper IV
Background
HIV-1 infections modulate the immune system to their advantage partly via induction of
negative inhibitory molecules on the T cell surfaces (221, 226, 227, 230, 232, 233, 269).
In vitro studies show that allogeneic HIV-1/DC-T cell cocultures lead to increase
expression PD-1, CTLA-4, TRAIL, LAG-3, TIM-3, CD160, and transcriptional factors
BLIMP-1, and FOXP3 (219, 220). The mechanisms under which HIV-1 initiates the
38
Findings and Discussion
prolific expression of these inhibitory molecules are not understood. Hence, the
understanding of these pathways could be a milestone in deciphering the path through
which vaccine studies could embark on. Owing to the role of DTX1 in T cell anergy
(218), NFAT in regulation of PD-1 (240) and CTLA-4 (241) or CD8+ T cell exhaustion
(276), MAPK in PD-1 induction (277), and the role of STAT3 in the downmodulation of
TH1 responses, induction of immunosuppressive genes (278-281), and generation of T
cells beneficial for tumor survival (281), we set out to study these pathways and their
involvement in immune modulation in HIV-1 primed T cells.
Principal Findings
We have shown previously that HIV-1 impairs DCs ability to prime naïve T cell
responses (219, 220) and further established that similar impairment occurred in memory
T cells but to a lesser extent. Viral access to the DC cytosol was necessary to elicit the
impaired T cell proliferation and increased gene and protein expression of inhibitory
molecules. We subsequently investigated the role of P38MAPK, STAT3, and NFATc
signaling pathways in the induction of impaired T cell proliferation and enhanced
expression of inhibitory molecules. The findings showed that HIV-1 utilized the STAT3
pathway to induce inhibitory molecules as blockade of this pathway significantly reduced
gene and protein expression of inhibitory molecules as well as rescued T cell
proliferation. The mechanism of STAT3 involvement was unclear as we did not detect
increased IL-6 and IL-10 secretion, which are known stimulators of the STAT3 pathway
(282, 283). Neither were the levels of other known extracellular STAT3 activators, e.g.
PDGF, G-CSF, EGF, and VEGF (284-286), increased in supernatants. Therefore we
suggested the involvement of the intracellular STAT3 regulator P38MAPK (287, 288).
Of interest, blockade of P38MAPK drastically curtailed expression of inhibitory
molecules and rescued T cell proliferation showing a mechanism of induction of
suppressor T cell by STAT3 through the P38MAPK route. We were able to show that
PD-1 expression was also regulated via NFATc (240) as well as the DTX1 (218) as
blockade reduced their expression but in the contrary gave slight increases in expression
of other inhibitory molecules. To confirm that these findings could be translated into an
autologous system did we use DCs pulsed with the superantigen Staphylococcus
enterotoxin B (SEB) to activate T cells in the presence or absence of HIV-1. This
autologous system gave similar increased expression of inhibitory molecules and
decreased proliferation of the primed T cells when HIV-1 was present as seen in our
allogeneic system and thus confirming the immunomodulatory effect of HIV-1. Blockade
of STAT3 and P38MAPK in the SEB activated autologous system also increased T cell
proliferation significantly.
39
Findings and Discussion
Discussion/Conclusions
Increased expression of PD-1, CTLA-4, TRAIL, CD160, TIM-3, and LAG-3 have been
documented in HIV-1 infected individuals (221, 227, 230, 233, 269, 289). Furthermore,
TN cells primed in the presence of HIV-1 in vitro expressed elevated levels of these
negative costimulatory molecules (220) and blockade of PD-1 and CTLA-4 dramatically
improved T cell functions in vitro (145, 221, 226, 227). The mechanisms and molecular
events underlying the upregulation of these molecules both in vivo and in vitro remained
elusive. Herein, we established that HIV-1 entry into the DCs cytosol is the first step
towards the induction of negative costimulatory molecules in addition to well known
transcriptional repressors BLIMP-1 (217, 220, 242, 243), DTX1 (218), and FOXP3
(290). We showed in this study for the first time that these inhibitory molecules are
regulated by P38MAPK/STAT3 pathways as blockade of these signaling cascades
significantly reduced expression of inhibitory molecules and rescued T cell proliferation.
STAT3 may be a potential target for HIV-1 to modulate the immune system seeing that
STAT3 has been reported in cancer as a potent inducer of immunosuppressive genes
(133, 279, 281). We eliminated the involvement of IL-6, IL-10, G-CFS, EGF, VEGF, and
PDGF, which are all known to induce STAT3 signaling (282-286, 291) and postulated an
intracellular mechanism through which HIV-1 mediated events (292, 293) that activated
STAT3 through the P38MAPK pathway (287, 288). The interruption of trafic through the
P38MAPK pathway significantly suppressed the expression of inhibitory molecules and
this proved to be the pathway HIV-1 utilized to induce inhibitory molecules in vitro and
possibly in vivo. We confirmed these findings in an autologous system whereby DC-T
cell cultures were activated with the superantigen SEB. We showed a significant rescue
in T cell proliferation when the P38MAPK/STAT3 pathway was interrupted. However,
we maintained the allogeneic system throughout our studies since this system best
mimics the mechanism of a precise T cell activation. T cell activation with superantigen
has been shown to circumvent the normal peptide/MHC-TCR interaction mechanism for
T cell activation (294, 295). In conclusion, presence of HIV-1 in the DCs cytosol
condition these cells to induce signaling via P38MAPK and subsequent activation of
STAT3 leading to transcriptional activation of inhibitory genes and proteins, which
render the primed T cells immunosuppressive.
40
Overall Findings/Conclusions
OVERALL FINDINGS/CONCLUSIONS
The immune system responds to HIV-1 by mounting both humoral and cellular specific
responses, which control viral replication. These responses are highly dependent on both
viral and host factors. These responses decline as the disease progresses due to massive
loss of CD4+ T cells and leads to reestablishment of viremia. Hence, the generation of
HIV-1 specific responses and persistence of these responses is a fundamental requisite to
control HIV-1 infections. We have shown that HIV-1 specific T cell responses can be
generated in vitro by pulsing MDDCs with infectious or non infectious HIV-1 and
coculturing them with autologous naïve T cells. The in vitro primed responses were
confirmed in vivo in HIV-1 infected individuals and it was clear that these responses
declined fast as the disease progressed. We next established in vitro a possible
mechanism through which immune responses to HIV-1 could be impaired as the disease
progressed. HIV-1 impaired DCs ability to activate functional allogeneic naïve T cells.
The T cells primed in the presence of HIV-1 had upregulated inhibitory molecules
CTLA-4, PD-1, TRAIL, and FOXP3 and had the ability to impair other naïve T cell
responses. The neutralization of the inhibitory molecules CTLA-4, PD-1, and TRAIL
restored T cell proliferation. The next point of focus was the possible involvement and
expression of other inhibitory molecules and was consequently screened on the HIV-1
primed T cells for well established inhibitory molecules, LAG-3, TIM-3, BTLA and
CD160, and transcription repressor BLIMP-1. We reported increased expression of all
these molecules except for BTLA on HIV-1 primed T cells. HIV-1 primed T cells also
produced reduced levels of granzyme and perforin as well as TNF-α, IL2 and IFN-γ.
These finding reaffirmed the decline in immune responses that occur in vivo in HIV1infected as the disease progresses. The mechanisms under which HIV-1 significantly
enhanced the inhibitory molecules expressed on primed T cells remained unknown. We
established that STAT3 was involved in the increase of inhibitory molecules on HIV-1
primed T cells. STAT3 was not activated through the well established STAT3 stimulants,
IL-6, IL-10, EGF, PDGF, VEGF, or G-CFS, rather through the intracellular P38MAPK
pathway. (See figure 11 for a schematic summary of inhibitory molecules).
41
Overall Findings/Conclusions
Figure 11: Summary of HIV-1 induced inhibitory molecules in DC T cell primed responses.
HIV-1 negatively modulates DC function by inducing increase expression of inhibitory molecules on primed T cells, which can act
both on other APCs and on T cells. HIV-1 binding and fusion to DCs is an important requirement to render the DCs capable of
inducing expression of inhibitory molecules on the primed T cells.
42
Recommendations and Future Challenges
RECOMMENDATIONS AND FUTURE CHALLENGES
The prime aim or motive of HIV research; be it epidemiological or scientific research is
to contribute to knowledge that can open the way forward towards the discovery of
vaccines or drugs that can cure this disease. Therefore, the major challenge today is the
quest towards an effective vaccine that will successfully eliminate HIV-1 and the
numerous emerging subtypes. This cannot be achieved without an in depth understanding
of HIV-1 structure and pathogenesis or the mechanisms of action.
In this study, we have focused on the type of specific T cell immune responses generated
against HIV-1 both in vitro and in vivo. This knowledge has revealed a possibility that
HIV-1 specific T cell immune responses can be generated in vitro and therefore, has
opened a window for future studies at improving the generation of in vitro specific
responses and how these responses could be adapted in vivo in patients to ameliorate or
cure their conditions.
We also identified a broad array of inhibitory molecules thought to be partially
responsible for the decline of immune functions in HIV-1 and the underlying mechanisms
responsible for the enhanced expression of these inhibitory molecules. Future studies are
needed to understand these attributes and possibly develop methods to prevent the
upsurge of inhibitory molecules or prevent cell responses as a result to these molecules.
These mechanisms are very important to understand rather than developing blockers for
these inhibitory molecules as these numerous molecules may be difficult to block
simultaneously in patients without causing severe systemic damage to the host.
The mechanisms under which these molecules elicit their inhibitory effects on other cells
are also of prime importance to study as this may shed more light on possible preventive
tactics implored to abrogate their activity.
43
Recommendations and Future Challenges
44
Lay language Summary
LAY LANGUAGE SUMMARY
Human Immunodeficiency Virus type 1 (HIV-1) is the virus that causes AIDS known in
full as Acquired immune deficiency syndrome. HIV-1 infects the body primarily through
unprotected sexual intercourse and till date there is no cure for this infection, only drug
regimens that keep the infection under control. White blood cells are one part of the host
defense against diseases (the immune system) and these cells recognize and fight
infections and have failed to successfully kill the virus and free the body from HIV-1.
However, some individuals are able to live with the HIV-1 infection for a long time
without any symptoms and they are called long term non progressors. These individuals’
white blood cells are able to control the disease by blocking the production of new
viruses. This protects them from obtaining other infections (opportunistic infections),
such as tuberculosis, diarrhea, fungi, parasites, and cancers, which normally are easy to
acquire due to the HIV-1 infection. This gives evidence that a strong immune response
mounted against HIV-1 infection is beneficial as it can control infection. Therefore,
finding ways of creating strong immune responses in infected individuals could greatly
control the disease.
When HIV-1 enters the body, it is picked up by specialized white blood cells called
dendritic cells. These cells are experts in activating the body’s defense against infections.
The dendritic cells will degrade and expose pieces of HIV-1 on their surface to another
type of white blood cell called T cells and instruct them what kind of responses they
should mount to fight the infection.
The aim of this thesis was to understand the type of T cell responses generated by
dendritic cells during the HIV-1 infection and to understand why the immune system
becomes dysfunctional and fails to eradicate HIV-1 from the body.
We showed that when we exposed dendritic cells to HIV-1, the dendritic cells activated T
cells that recognized HIV-1 and these T cells had the potential to kill HIV-1 infected
cells. These cells responded to HIV-1 by producing factors (IL-2, MIP-1, TNF-α, and
IFN-γ) with the ability to suppress this virus. The T cell responses in the HIV-1 infected
individuals gradually declined as the disease progressed.
We went on to show that one reason behind the impaired immune responses was the
increased expression of suppressive molecules (PD-1, CTLA4, TRAIL, LAG3, TIM3,
BLIMP-1, FOXP3, and DXT1) on the T cells. HIV-1 induction of these suppressor
molecules was dependent on signaling events (P38MAPK/STAT3) inside the T cells.
45
Lay language Summary
These findings show that it is possible for dendritic cells to generate strong immune
responses against HIV-1 outside the body and this knowledge can be applied in vaccine
design. Our results also suggested that the decline in the T cell functionality was partly
due to increased expression of immune suppressive molecules. The impairment could be
reversed by blocking the effects of these suppressive molecules or even better by
preventing the signaling events that lead to their production.
46
Acknowledgements
ACKNOWLEDGEMENTS
I cannot conclude without acknowledging the support of a few people who stood by me
to enable this dream come true.
To my supervisor Prof. Marie Larsson, I will forever remain indebted to you for the
wonderful opportunity you gave me. You saw in me what myself and others could not see
and you help tap out those qualities for which I am very thankful today. Marie, working
with you did not only improve my academic career but also upgraded a moral side in me.
You are not only a great scientist and supervisor, but also a wonder role model.
To my cosupervisors Prof. Jormah Hinkula, I will always remember your willingness to
accommodate me from the first day we met in the department. Since then, you have
supported me academically and morally throughout these years. And to you Prof. KarlEric Magnusson, thank you for being there and ready to attain to me and thanks for your
constant curiosity on the prospects of my work.
To my coworkers in Marie’s group, Veronica, Vegard, Shankar, Rada, Sundar and all
the masters’ students who have been in and out of the group, I appreciate every moment
we spent together. Veronica, you have been a source of comfort and encouragement. We
shared common difficulties and due to that, you could best understand my frustrations
and made it a duty to help when you could. To you Vegard, your kindness and simplicity
eased the working environment and enabled the freedom to share whatever I had in mind
with you. This quality was just what I needed to function properly from the first to the
last day at work. To you Shankar, coming to the lab was a blessing as you help in the
experiments and your special skills in editing the manuscripts. I will always be thankful
to have worked with you. Rada, I thank God for, making you join Marie Larsson’s
group. You have been an asset not only to the group but also to the entire department
with your exotic computer skills. You never said no to my requests and always did your
work with a lot of passion. Thank you for the wonderful pictures you made for this thesis.
And to Sundar, thanks for your support in the lab and editing of the manuscripts. Your
curiosity in understanding the subject matter enabled me to learn many more things as we
discussed science together.
To you Carolin Jonson and Lena Svensoon, you have not only been of help in the lab
but also outside the lab. You have consistently been there for the last 5 years and showed
no signs of resilience in your efforts to help me succeed. Your interest in the well being
of my family has been a corner stone for my admiration towards you. I owe my Swedish
skills to you Lena
47
Acknowledgements
To my fellow work mates whom I now call friends in Lennart svensson’s Lab, Johan I
am truly grateful to have had you in my life. Elin, thanks for the chats we had at work
and parties we attended. Your sharpness truly shaped me to be a better person. Beatice,
you are truly a nice person and I enjoyed each moment of our discussions. To Marlin,
your composure during your work and the writing of your thesis truly inspired me to be
strong as my turn approached. To Marie Hagbom, thanks for your friendship and
concern over my family. To Filemon, we had wonderful moments each time you visited
Sweden to complete your PhD and I am truly grateful for having you as a friend. To
Sumit alias Tum Casiho, thanks for your friendship and helping me with some Hindi.
To crown it all, special thanks Prof Lennart svensson. “Learning without playing makes
Jack a doll boy”. Under your guide, you made sure we had good working conditions and
the refreshments badly needed to rekindle the brains.
To Lotta and Pia, I thank you for your kind help with micro arrays and western blotting
and your persistence to make my Swedish better. I owe my Swedish skills to you.
To Hong, Sasan, Magnus, Daniel, Ryan, Anika, Lisa, Olivia, Gosia, Helen, Matthias
and Anna, I have enjoyed every moment I spent with you at work and the parties we
attended and am grateful for that.
To you Chi Celestine, Alain Asongwec and wife Matilda, Gilbert Nche, Muluh
Clifford, Ngeh Jonathan and Akenji Valentine, you occupy a special place in my heart
for what we share as friends and family. To my “other brother” Xavier Ekolle NdodeEkane, life in Sweden would have been miserable without you as we went through thick
and thin in the same boat but still kept our cool for each other. Your calmness and
confidence for the future has truly guided me during times of indecision. Good luck with
the last phase of your PhD too. To all the “Paysans”, with whom we have had special
moments here in Linköping, Fidelis, Elvis, Eric, Pride, Leon, Justice, Laurence,
Gerald, Sam, Atem, Pascal, Paul, Ebot, Alisine, Pauline, Challote and Millat, thanks
for making me feel Ndzemabeah was very close even though I was far away in Sweden..
To my Family, special thanks go to my Father (RIP) and mother Akumah Shadrack and
Magdalene respectively who gave me the life and education I have now. Daddy, this is
it!!! and you should smile down on us as we celebrate this day for it is for you. To my
step mothers Mami and Mangieh, I owe you my loyalty for the wonderful support I
have always received from you. My brothers Ernest for the incredible moments we have
had while growing up and thanks for being an example of hard work which has also
inspired the rest of our siblings. To Hilarious for being such a wonderful “big” brother
teaching me to be happy even when there was nothing to be happy about. Special thanks
to Watson and Sanna. Watson we share the world together and that has helped us to
48
Acknowledgements
grow stronger in spirit and I hope we keep the candle light glowing. To Addison,
Kenneth, Sebastian, Elvis, Mabi, Akumah, and mankah thanks for being such
wonderful and supportive siblings.
Million thanks go to my uncles and wives Mr. and Mrs. Akumah Gideon for assuming
the role of a father and mother and special thanks for the support during my college days.
I will always remain indebted to you. To Mr. and Mrs. Akumah Ruben, you have
always been there each time I needed you and no words can express how thankful I am
for that. Uncle, you told me not to return to Cameroon without a PhD and today, I am
proud to have respected that order.
To the Engstroms, Rosens, Nordins, Gogina Mores and Kakembo, you have been my
family here in Sweden. Words cannot express how thankful I am for your hospitality and
love. To Tina and Bjorn, I thank God for making us friends. Doing things with you
Tina Rosen and Tina wood has helped feel many holes in my life as I have learned so
much from you. Special thanks to all my choir members as singing with you helped
release tension and made my focus at work much better
To crown it all, special thanks to my wife Suzan Che. Wards cannot express how much
you mean to me as a wife, friend, and coach and it is my singular pleasure to have you
in my life. Excuse my stubbornness and irrationality sometimes for that has simply been
a source of strength for both of us. To my sons Mbumah Karl-Jeremy Che and FruMbonneh Desmond Che to whom I dedicate my thesis to, you are why I do the things I
do and you mean everything to me.
49
Acknowledgements
50
References
REFERENCES
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
Barre-Sinoussi F, Chermann JC, Rey F, et al. (1983) Isolation of a T-lymphotropic
retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS).
Science (New York, N.Y 220: 868-871.
Clavel F, Guetard D, Brun-Vezinet F, et al. (1986) Isolation of a new human retrovirus
from West African patients with AIDS. Science (New York, N.Y 233: 343-346.
Robertson DL, Anderson JP, Bradac JA, et al. (2000) HIV-1 nomenclature proposal.
Science (New York, N.Y 288: 55-56.
Plantier JC, Leoz M, Dickerson JE, et al. (2009) A new human immunodeficiency virus
derived from gorillas. Nature medicine 15: 871-872.
Lemey P, Pybus OG, Wang B, Saksena NK, Salemi M, Vandamme AM. (2003) Tracing the
origin and history of the HIV-2 epidemic. Proceedings of the National Academy of
Sciences of the United States of America 100: 6588-6592.
Chen Z, Luckay A, Sodora DL, et al. (1997) Human immunodeficiency virus type 2 (HIV-2)
seroprevalence and characterization of a distinct HIV-2 genetic subtype from the natural
range of simian immunodeficiency virus-infected sooty mangabeys. Journal of virology
71: 3953-3960.
Worobey M, Telfer P, Souquiere S, et al. Island biogeography reveals the deep history of
SIV. Science (New York, N.Y 329: 1487.
Hahn BH, Shaw GM, De Cock KM, Sharp PM. (2000) AIDS as a zoonosis: scientific and
public health implications. Science (New York, N.Y 287: 607-614.
Bailes E, Gao F, Bibollet-Ruche F, et al. (2003) Hybrid origin of SIV in chimpanzees.
Science (New York, N.Y 300: 1713.
Folks TM. (2000) Chimpanzees as original source for HIV. Jama 283: 310.
Zhu T, Korber BT, Nahmias AJ, Hooper E, Sharp PM, Ho DD. (1998) An African HIV-1
sequence from 1959 and implications for the origin of the epidemic. Nature 391: 594597.
Chiu IM, Yaniv A, Dahlberg JE, et al. (1985) Nucleotide sequence evidence for
relationship of AIDS retrovirus to lentiviruses. Nature 317: 366-368.
Fanales-Belasio E, Raimondo M, Suligoi B, Butto S. HIV virology and pathogenetic
mechanisms of infection: a brief overview. Annali dell'Istituto superiore di sanita 46: 514.
Redel L, Le Douce V, Cherrier T, et al. HIV-1 regulation of latency in the monocytemacrophage lineage and in CD4+ T lymphocytes. Journal of leukocyte biology 87: 575588.
Collman RG, Perno CF, Crowe SM, Stevenson M, Montaner LJ. (2003) HIV and cells of
macrophage/dendritic lineage and other non-T cell reservoirs: new answers yield new
questions. Journal of leukocyte biology 74: 631-634.
Aquaro S, Calio R, Balzarini J, Bellocchi MC, Garaci E, Perno CF. (2002) Macrophages and
HIV infection: therapeutical approaches toward this strategic virus reservoir. Antiviral
research 55: 209-225.
Carter CC, Onafuwa-Nuga A, McNamara LA, et al. HIV-1 infects multipotent progenitor
cells causing cell death and establishing latent cellular reservoirs. Nature medicine 16:
446-451.
Glass JD, Johnson RT. (1996) Human immunodeficiency virus and the brain. Annual
review of neuroscience 19: 1-26.
51
References
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
Nottet HS, van Dijk SJ, Fanoy EB, et al. (2009) HIV-1 can persist in aged memory CD4+ T
lymphocytes with minimal signs of evolution after 8.3 years of effective highly active
antiretroviral therapy. Journal of acquired immune deficiency syndromes (1999) 50: 345353.
Smith BA, Gartner S, Liu Y, et al. (2001) Persistence of infectious HIV on follicular
dendritic cells. J Immunol 166: 690-696.
Sonza S, Mutimer HP, Oelrichs R, et al. (2001) Monocytes harbour replicationcompetent, non-latent HIV-1 in patients on highly active antiretroviral therapy. AIDS
(London, England) 15: 17-22.
Gomez C, Hope TJ. (2005) The ins and outs of HIV replication. Cellular microbiology 7:
621-626.
Zaitseva M, Blauvelt A, Lee S, et al. (1997) Expression and function of CCR5 and CXCR4
on human Langerhans cells and macrophages: implications for HIV primary infection.
Nature medicine 3: 1369-1375.
Ruiz ME, Cicala C, Arthos J, et al. (1998) Peripheral blood-derived CD34+ progenitor
cells: CXC chemokine receptor 4 and CC chemokine receptor 5 expression and infection
by HIV. J Immunol 161: 4169-4176.
Rubbert A, Combadiere C, Ostrowski M, et al. (1998) Dendritic cells express multiple
chemokine receptors used as coreceptors for HIV entry. J Immunol 160: 3933-3941.
Suligoi B, Raimondo M, Fanales-Belasio E, Butto S. The epidemic of HIV infection and
AIDS, promotion of testing, and innovative strategies. Annali dell'Istituto superiore di
sanita 46: 15-23.
Bjorndal A, Deng H, Jansson M, et al. (1997) Coreceptor usage of primary human
immunodeficiency virus type 1 isolates varies according to biological phenotype. Journal
of virology 71: 7478-7487.
Scarlatti G, Tresoldi E, Bjorndal A, et al. (1997) In vivo evolution of HIV-1 co-receptor
usage and sensitivity to chemokine-mediated suppression. Nature medicine 3: 12591265.
Connor RI, Sheridan KE, Ceradini D, Choe S, Landau NR. (1997) Change in coreceptor use
correlates with disease progression in HIV-1--infected individuals. The Journal of
experimental medicine 185: 621-628.
Shankarappa R, Margolick JB, Gange SJ, et al. (1999) Consistent viral evolutionary
changes associated with the progression of human immunodeficiency virus type 1
infection. Journal of virology 73: 10489-10502.
Tersmette M, de Goede RE, Al BJ, et al. (1988) Differential syncytium-inducing capacity
of human immunodeficiency virus isolates: frequent detection of syncytium-inducing
isolates in patients with acquired immunodeficiency syndrome (AIDS) and AIDS-related
complex. Journal of virology 62: 2026-2032.
Berger EA. (1997) HIV entry and tropism: the chemokine receptor connection. AIDS
(London, England) 11 Suppl A: S3-16.
Grivel JC, Margolis LB. (1999) CCR5- and CXCR4-tropic HIV-1 are equally cytopathic for
their T-cell targets in human lymphoid tissue. Nature medicine 5: 344-346.
Borrow P, Lewicki H, Hahn BH, Shaw GM, Oldstone MB. (1994) Virus-specific CD8+
cytotoxic T-lymphocyte activity associated with control of viremia in primary human
immunodeficiency virus type 1 infection. Journal of virology 68: 6103-6110.
Bangham CR. (2009) CTL quality and the control of human retroviral infections.
European journal of immunology 39: 1700-1712.
52
References
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
Butto S, Raimondo M, Fanales-Belasio E, Suligoi B. Suggested strategies for the
laboratory diagnosis of HIV infection in Italy. Annali dell'Istituto superiore di sanita 46:
34-41.
Butto S, Suligoi B, Fanales-Belasio E, Raimondo M. Laboratory diagnostics for HIV
infection. Annali dell'Istituto superiore di sanita 46: 24-33.
Gupta KK. (1993) Acute immunosuppression with HIV seroconversion. The New England
journal of medicine 328: 288-289.
Kahn JO, Walker BD. (1998) Acute human immunodeficiency virus type 1 infection. The
New England journal of medicine 339: 33-39.
Ford ES, Puronen CE, Sereti I. (2009) Immunopathogenesis of asymptomatic chronic HIV
Infection: the calm before the storm. Current opinion in HIV and AIDS 4: 206-214.
Brenchley JM, Schacker TW, Ruff LE, et al. (2004) CD4+ T cell depletion during all stages
of HIV disease occurs predominantly in the gastrointestinal tract. The Journal of
experimental medicine 200: 749-759.
Mehandru S, Poles MA, Tenner-Racz K, et al. (2004) Primary HIV-1 infection is associated
with preferential depletion of CD4+ T lymphocytes from effector sites in the
gastrointestinal tract. The Journal of experimental medicine 200: 761-770.
Brooks JT, Kaplan JE, Holmes KK, Benson C, Pau A, Masur H. (2009) HIV-associated
opportunistic infections--going, going, but not gone: the continued need for prevention
and treatment guidelines. Clin Infect Dis 48: 609-611.
Siliciano JD, Kajdas J, Finzi D, et al. (2003) Long-term follow-up studies confirm the
stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nature medicine 9: 727728.
Keele BF, Tazi L, Gartner S, et al. (2008) Characterization of the follicular dendritic cell
reservoir of human immunodeficiency virus type 1. Journal of virology 82: 5548-5561.
Spiegel H, Herbst H, Niedobitek G, Foss HD, Stein H. (1992) Follicular dendritic cells are a
major reservoir for human immunodeficiency virus type 1 in lymphoid tissues facilitating
infection of CD4+ T-helper cells. The American journal of pathology 140: 15-22.
Lambotte O, Taoufik Y, de Goer MG, Wallon C, Goujard C, Delfraissy JF. (2000) Detection
of infectious HIV in circulating monocytes from patients on prolonged highly active
antiretroviral therapy. Journal of acquired immune deficiency syndromes (1999) 23: 114119.
McElrath MJ, Steinman RM, Cohn ZA. (1991) Latent HIV-1 infection in enriched
populations of blood monocytes and T cells from seropositive patients. The Journal of
clinical investigation 87: 27-30.
Mikovits JA, Lohrey NC, Schulof R, Courtless J, Ruscetti FW. (1992) Activation of
infectious virus from latent human immunodeficiency virus infection of monocytes in
vivo. The Journal of clinical investigation 90: 1486-1491.
Zhu T, Muthui D, Holte S, et al. (2002) Evidence for human immunodeficiency virus type
1 replication in vivo in CD14(+) monocytes and its potential role as a source of virus in
patients on highly active antiretroviral therapy. Journal of virology 76: 707-716.
Aquaro S, Bagnarelli P, Guenci T, et al. (2002) Long-term survival and virus production in
human primary macrophages infected by human immunodeficiency virus. Journal of
medical virology 68: 479-488.
Crowe SM, Mills J, Kirihara J, Boothman J, Marshall JA, McGrath MS. (1990) Full-length
recombinant CD4 and recombinant gp120 inhibit fusion between HIV infected
macrophages and uninfected CD4-expressing T-lymphoblastoid cells. AIDS research and
human retroviruses 6: 1031-1037.
53
References
53.
54.
55.
56.
57.
58.
59.
60.
61.
62.
63.
64.
65.
66.
67.
68.
69.
70.
Orenstein JM, Fox C, Wahl SM. (1997) Macrophages as a source of HIV during
opportunistic infections. Science (New York, N.Y 276: 1857-1861.
Wightman F, Solomon A, Khoury G, et al. Both CD31(+) and CD31 naive CD4(+) T cells are
persistent HIV type 1-infected reservoirs in individuals receiving antiretroviral therapy.
The Journal of infectious diseases 202: 1738-1748.
Quayle AJ, Xu C, Mayer KH, Anderson DJ. (1997) T lymphocytes and macrophages, but
not motile spermatozoa, are a significant source of human immunodeficiency virus in
semen. The Journal of infectious diseases 176: 960-968.
Zhang H, Dornadula G, Beumont M, et al. (1998) Human immunodeficiency virus type 1
in the semen of men receiving highly active antiretroviral therapy. The New England
journal of medicine 339: 1803-1809.
Liu R, Paxton WA, Choe S, et al. (1996) Homozygous defect in HIV-1 coreceptor accounts
for resistance of some multiply-exposed individuals to HIV-1 infection. Cell 86: 367-377.
Dean M, Carrington M, Winkler C, et al. (1996) Genetic restriction of HIV-1 infection and
progression to AIDS by a deletion allele of the CKR5 structural gene. Hemophilia Growth
and Development Study, Multicenter AIDS Cohort Study, Multicenter Hemophilia Cohort
Study, San Francisco City Cohort, ALIVE Study. Science (New York, N.Y 273: 1856-1862.
Kaul R, Trabattoni D, Bwayo JJ, et al. (1999) HIV-1-specific mucosal IgA in a cohort of
HIV-1-resistant Kenyan sex workers. AIDS (London, England) 13: 23-29.
Rowland-Jones SL, Dong T, Fowke KR, et al. (1998) Cytotoxic T cell responses to multiple
conserved HIV epitopes in HIV-resistant prostitutes in Nairobi. The Journal of clinical
investigation 102: 1758-1765.
Parham P, Ohta T. (1996) Population biology of antigen presentation by MHC class I
molecules. Science (New York, N.Y 272: 67-74.
Schellens IM, Kesmir C, Miedema F, van Baarle D, Borghans JA. (2008) An unanticipated
lack of consensus cytotoxic T lymphocyte epitopes in HIV-1 databases: the contribution
of prediction programs. AIDS (London, England) 22: 33-37.
Kiepiela P, Leslie AJ, Honeyborne I, et al. (2004) Dominant influence of HLA-B in
mediating the potential co-evolution of HIV and HLA. Nature 432: 769-775.
Migueles SA, Sabbaghian MS, Shupert WL, et al. (2000) HLA B*5701 is highly associated
with restriction of virus replication in a subgroup of HIV-infected long term
nonprogressors. Proceedings of the National Academy of Sciences of the United States of
America 97: 2709-2714.
de Groot NG, Heijmans CM, Zoet YM, et al. AIDS-protective HLA-B*27/B*57 and
chimpanzee MHC class I molecules target analogous conserved areas of HIV-1/SIVcpz.
Proceedings of the National Academy of Sciences of the United States of America 107:
15175-15180.
Carrington M, Nelson GW, Martin MP, et al. (1999) HLA and HIV-1: heterozygote
advantage and B*35-Cw*04 disadvantage. Science (New York, N.Y 283: 1748-1752.
Martin MP, Carrington M. (2005) Immunogenetics of viral infections. Current opinion in
immunology 17: 510-516.
Carrington M, O'Brien SJ. (2003) The influence of HLA genotype on AIDS. Annual review
of medicine 54: 535-551.
Julg B, Moodley ES, Qi Y, et al. Possession of HLA class II DRB1*1303 associates with
reduced viral loads in chronic HIV-1 clade C and B infection. The Journal of infectious
diseases 203: 803-809.
Ndung'u T, Gaseitsiwe S, Sepako E, et al. (2005) Major histocompatibility complex class II
(HLA-DRB and -DQB) allele frequencies in Botswana: association with human
54
References
71.
72.
73.
74.
75.
76.
77.
78.
79.
80.
81.
82.
83.
84.
85.
immunodeficiency virus type 1 infection. Clinical and diagnostic laboratory immunology
12: 1020-1028.
Moss AR, Bacchetti P, Osmond D, et al. (1988) Seropositivity for HIV and the
development of AIDS or AIDS related condition: three year follow up of the San
Francisco General Hospital cohort. British medical journal (Clinical research ed 296: 745750.
Bacchetti P, Osmond D, Chaisson RE, et al. (1988) Survival patterns of the first 500
patients with AIDS in San Francisco. The Journal of infectious diseases 157: 1044-1047.
Royce RA, Winkelstein W, Jr. (1990) HIV infection, cigarette smoking and CD4+ Tlymphocyte counts: preliminary results from the San Francisco Men's Health Study. AIDS
(London, England) 4: 327-333.
Moseson M, Zeleniuch-Jacquotte A, Belsito DV, Shore RE, Marmor M, Pasternack B.
(1989) The potential role of nutritional factors in the induction of immunologic
abnormalities in HIV-positive homosexual men. Journal of acquired immune deficiency
syndromes (1999) 2: 235-247.
Burack JH, Barrett DC, Stall RD, Chesney MA, Ekstrand ML, Coates TJ. (1993) Depressive
symptoms and CD4 lymphocyte decline among HIV-infected men. Jama 270: 2568-2573.
Ho DD, Toyoshima T, Mo H, et al. (1994) Characterization of human immunodeficiency
virus type 1 variants with increased resistance to a C2-symmetric protease inhibitor.
Journal of virology 68: 2016-2020.
Borman AM, Paulous S, Clavel F. (1996) Resistance of human immunodeficiency virus
type 1 to protease inhibitors: selection of resistance mutations in the presence and
absence of the drug. The Journal of general virology 77 ( Pt 3): 419-426.
Kaleebu P, French N, Mahe C, et al. (2002) Effect of human immunodeficiency virus (HIV)
type 1 envelope subtypes A and D on disease progression in a large cohort of HIV-1positive persons in Uganda. The Journal of infectious diseases 185: 1244-1250.
Poropatich K, Sullivan DJ, Jr. Human immunodeficiency virus type 1 long-term nonprogressors: the viral, genetic and immunological basis for disease non-progression. The
Journal of general virology 92: 247-268.
Cao Y, Qin L, Zhang L, Safrit J, Ho DD. (1995) Virologic and immunologic characterization
of long-term survivors of human immunodeficiency virus type 1 infection. The New
England journal of medicine 332: 201-208.
Munoz A, Kirby AJ, He YD, et al. (1995) Long-term survivors with HIV-1 infection:
incubation period and longitudinal patterns of CD4+ lymphocytes. J Acquir Immune Defic
Syndr Hum Retrovirol 8: 496-505.
Pantaleo G, Menzo S, Vaccarezza M, et al. (1995) Studies in subjects with long-term
nonprogressive human immunodeficiency virus infection. The New England journal of
medicine 332: 209-216.
Madec Y, Boufassa F, Avettand-Fenoel V, et al. (2009) Early control of HIV-1 infection in
long-term nonprogressors followed since diagnosis in the ANRS SEROCO/HEMOCO
cohort. Journal of acquired immune deficiency syndromes (1999) 50: 19-26.
Rodes B, Toro C, Paxinos E, et al. (2004) Differences in disease progression in a cohort of
long-term non-progressors after more than 16 years of HIV-1 infection. AIDS (London,
England) 18: 1109-1116.
Lefrere JJ, Morand-Joubert L, Mariotti M, et al. (1997) Even individuals considered as
long-term nonprogressors show biological signs of progression after 10 years of human
immunodeficiency virus infection. Blood 90: 1133-1140.
55
References
86.
87.
88.
89.
90.
91.
92.
93.
94.
95.
96.
97.
98.
99.
100.
101.
102.
103.
Deeks SG, Walker BD. (2007) Human immunodeficiency virus controllers: mechanisms of
durable virus control in the absence of antiretroviral therapy. Immunity 27: 406-416.
Lambotte O, Boufassa F, Madec Y, et al. (2005) HIV controllers: a homogeneous group of
HIV-1-infected patients with spontaneous control of viral replication. Clin Infect Dis 41:
1053-1056.
Walker BD. (2007) Elite control of HIV Infection: implications for vaccines and treatment.
Top HIV Med 15: 134-136.
Emu B, Sinclair E, Hatano H, et al. (2008) HLA class I-restricted T-cell responses may
contribute to the control of human immunodeficiency virus infection, but such
responses are not always necessary for long-term virus control. Journal of virology 82:
5398-5407.
Okulicz JF, Marconi VC, Landrum ML, et al. (2009) Clinical outcomes of elite controllers,
viremic controllers, and long-term nonprogressors in the US Department of Defense HIV
natural history study. The Journal of infectious diseases 200: 1714-1723.
Blankson JN. Effector mechanisms in HIV-1 infected elite controllers: highly active
immune responses? Antiviral research 85: 295-302.
Kaufmann GR, Bloch M, Zaunders JJ, Smith D, Cooper DA. (2000) Long-term
immunological response in HIV-1-infected subjects receiving potent antiretroviral
therapy. AIDS (London, England) 14: 959-969.
Staszewski S, Miller V, Sabin C, et al. (1999) Determinants of sustainable CD4
lymphocyte count increases in response to antiretroviral therapy. AIDS (London,
England) 13: 951-956.
Smith CJ, Sabin CA, Lampe FC, et al. (2003) The potential for CD4 cell increases in HIVpositive individuals who control viraemia with highly active antiretroviral therapy. AIDS
(London, England) 17: 963-969.
Piacenti FJ. (2006) An update and review of antiretroviral therapy. Pharmacotherapy 26:
1111-1133.
Dybul M, Fauci AS, Bartlett JG, Kaplan JE, Pau AK. (2002) Guidelines for using
antiretroviral agents among HIV-infected adults and adolescents. Recommendations of
the Panel on Clinical Practices for Treatment of HIV. MMWR Recomm Rep 51: 1-55.
Hammer SM, Saag MS, Schechter M, et al. (2006) Treatment for adult HIV infection:
2006 recommendations of the International AIDS Society-USA panel. Jama 296: 827843.
Thompson MA, Aberg JA, Cahn P, et al. Antiretroviral treatment of adult HIV infection:
2010 recommendations of the International AIDS Society-USA panel. Jama 304: 321333.
Kuhmann SE, Hartley O. (2008) Targeting chemokine receptors in HIV: a status report.
Annual review of pharmacology and toxicology 48: 425-461.
Biswas P, Tambussi G, Lazzarin A. (2007) Access denied? The status of co-receptor
inhibition to counter HIV entry. Expert opinion on pharmacotherapy 8: 923-933.
Ryser HJ, Fluckiger R. (2005) Progress in targeting HIV-1 entry. Drug discovery today 10:
1085-1094.
Lieberman-Blum SS, Fung HB, Bandres JC. (2008) Maraviroc: a CCR5-receptor antagonist
for the treatment of HIV-1 infection. Clinical therapeutics 30: 1228-1250.
Savarino A. (2006) A historical sketch of the discovery and development of HIV-1
integrase inhibitors. Expert opinion on investigational drugs 15: 1507-1522.
56
References
104.
105.
106.
107.
108.
109.
110.
111.
112.
113.
114.
115.
116.
117.
118.
119.
120.
121.
122.
FDA.
(2009)
FDA
approval
of
Isentress
(raltegravir).
http://www.fda.gov/ForConsumers/ByAudience/ForPatientAdvocates/HIVandAIDSActivi
ties/ucm124040.htm.
Barr SD, Smiley JR, Bushman FD. (2008) The interferon response inhibits HIV particle
production by induction of TRIM22. PLoS pathogens 4: e1000007.
Stoddart CA, Joshi P, Sloan B, et al. (2007) Potent activity of the HIV-1 maturation
inhibitor bevirimat in SCID-hu Thy/Liv mice. PloS one 2: e1251.
Baba M. (2006) Recent advances of CCR5 antagonists. Current opinion in HIV and AIDS 1:
367-372.
Emmelkamp JM, Rockstroh JK. (2007) CCR5 antagonists: comparison of efficacy, side
effects, pharmacokinetics and interactions--review of the literature. European journal of
medical research 12: 409-417.
Bangsberg DR, Hecht FM, Charlebois ED, et al. (2000) Adherence to protease inhibitors,
HIV-1 viral load, and development of drug resistance in an indigent population. AIDS
(London, England) 14: 357-366.
Haubrich RH, Little SJ, Currier JS, et al. (1999) The value of patient-reported adherence
to antiretroviral therapy in predicting virologic and immunologic response. California
Collaborative Treatment Group. AIDS (London, England) 13: 1099-1107.
Hecht FM, Grant RM, Petropoulos CJ, et al. (1998) Sexual transmission of an HIV-1
variant resistant to multiple reverse-transcriptase and protease inhibitors. The New
England journal of medicine 339: 307-311.
McNabb J, Ross JW, Abriola K, Turley C, Nightingale CH, Nicolau DP. (2001) Adherence to
highly active antiretroviral therapy predicts virologic outcome at an inner-city human
immunodeficiency virus clinic. Clin Infect Dis 33: 700-705.
Steinman RM, Witmer MD. (1978) Lymphoid dendritic cells are potent stimulators of the
primary mixed leukocyte reaction in mice. Proceedings of the National Academy of
Sciences of the United States of America 75: 5132-5136.
Banchereau J, Briere F, Caux C, et al. (2000) Immunobiology of dendritic cells. Annual
review of immunology 18: 767-811.
Banchereau J, Steinman RM. (1998) Dendritic cells and the control of immunity. Nature
392: 245-252.
Garrett WS, Chen LM, Kroschewski R, et al. (2000) Developmental control of endocytosis
in dendritic cells by Cdc42. Cell 102: 325-334.
Hugues S, Boissonnas A, Amigorena S, Fetler L. (2006) The dynamics of dendritic cell-T
cell interactions in priming and tolerance. Current opinion in immunology 18: 491-495.
von Boehmer H, Aifantis I, Gounari F, et al. (2003) Thymic selection revisited: how
essential is it? Immunological reviews 191: 62-78.
Baba T, Nakamoto Y, Mukaida N. (2009) Crucial contribution of thymic Sirp alpha+
conventional dendritic cells to central tolerance against blood-borne antigens in a CCR2dependent manner. J Immunol 183: 3053-3063.
Mueller DL. Mechanisms maintaining peripheral tolerance. Nature immunology 11: 2127.
Ueno H, Klechevsky E, Schmitt N, et al. Targeting human dendritic cell subsets for
improved vaccines. Seminars in immunology 23: 21-27.
Maldonado-Lopez R, De Smedt T, Michel P, et al. (1999) CD8alpha+ and CD8alphasubclasses of dendritic cells direct the development of distinct T helper cells in vivo. The
Journal of experimental medicine 189: 587-592.
57
References
123.
124.
125.
126.
127.
128.
129.
130.
131.
132.
133.
134.
135.
136.
137.
138.
139.
140.
Pulendran B, Smith JL, Caspary G, et al. (1999) Distinct dendritic cell subsets
differentially regulate the class of immune response in vivo. Proceedings of the National
Academy of Sciences of the United States of America 96: 1036-1041.
Dudziak D, Kamphorst AO, Heidkamp GF, et al. (2007) Differential antigen processing by
dendritic cell subsets in vivo. Science (New York, N.Y 315: 107-111.
Tew JG, Wu J, Fakher M, Szakal AK, Qin D. (2001) Follicular dendritic cells: beyond the
necessity of T-cell help. Trends in immunology 22: 361-367.
Hart DN, McKenzie JL. (1990) Interstitial dendritic cells. International reviews of
immunology 6: 127-138.
Valladeau J, Saeland S. (2005) Cutaneous dendritic cells. Seminars in immunology 17:
273-283.
Valladeau J, Duvert-Frances V, Pin JJ, et al. (1999) The monoclonal antibody DCGM4
recognizes Langerin, a protein specific of Langerhans cells, and is rapidly internalized
from the cell surface. European journal of immunology 29: 2695-2704.
Wu L, KewalRamani VN. (2006) Dendritic-cell interactions with HIV: infection and viral
dissemination. Nature reviews 6: 859-868.
Klechevsky E, Liu M, Morita R, et al. (2009) Understanding human myeloid dendritic cell
subsets for the rational design of novel vaccines. Human immunology 70: 281-288.
Breitfeld D, Ohl L, Kremmer E, et al. (2000) Follicular B helper T cells express CXC
chemokine receptor 5, localize to B cell follicles, and support immunoglobulin
production. The Journal of experimental medicine 192: 1545-1552.
Schaerli P, Willimann K, Lang AB, Lipp M, Loetscher P, Moser B. (2000) CXC chemokine
receptor 5 expression defines follicular homing T cells with B cell helper function. The
Journal of experimental medicine 192: 1553-1562.
Klechevsky E, Morita R, Liu M, et al. (2008) Functional specializations of human
epidermal Langerhans cells and CD14+ dermal dendritic cells. Immunity 29: 497-510.
Alvarez D, Harder G, Fattouh R, et al. (2005) Cutaneous antigen priming via gene gun
leads to skin-selective Th2 immune-inflammatory responses. J Immunol 174: 1664-1674.
Furio L, Briotet I, Journeaux A, Billard H, Peguet-Navarro J. Human langerhans cells are
more efficient than CD14(-)CD1c(+) dermal dendritic cells at priming naive CD4(+) T
cells. The Journal of investigative dermatology 130: 1345-1354.
Klechevsky E, Flamar AL, Cao Y, et al. Cross-priming CD8+ T cells by targeting antigens to
human dendritic cells through DCIR. Blood 116: 1685-1697.
Ratzinger G, Baggers J, de Cos MA, et al. (2004) Mature human Langerhans cells derived
from CD34+ hematopoietic progenitors stimulate greater cytolytic T lymphocyte activity
in the absence of bioactive IL-12p70, by either single peptide presentation or crosspriming, than do dermal-interstitial or monocyte-derived dendritic cells. J Immunol 173:
2780-2791.
Kadowaki N, Ho S, Antonenko S, et al. (2001) Subsets of human dendritic cell precursors
express different toll-like receptors and respond to different microbial antigens. The
Journal of experimental medicine 194: 863-869.
Jarrossay D, Napolitani G, Colonna M, Sallusto F, Lanzavecchia A. (2001) Specialization
and complementarity in microbial molecule recognition by human myeloid and
plasmacytoid dendritic cells. European journal of immunology 31: 3388-3393.
Poulin LF, Salio M, Griessinger E, et al. Characterization of human DNGR-1+ BDCA3+
leukocytes as putative equivalents of mouse CD8alpha+ dendritic cells. The Journal of
experimental medicine 207: 1261-1271.
58
References
141.
142.
143.
144.
145.
146.
147.
148.
149.
150.
151.
152.
153.
154.
155.
156.
157.
158.
159.
160.
Bachem A, Guttler S, Hartung E, et al. Superior antigen cross-presentation and XCR1
expression define human CD11c+CD141+ cells as homologues of mouse CD8+ dendritic
cells. The Journal of experimental medicine 207: 1273-1281.
Cao W, Rosen DB, Ito T, et al. (2006) Plasmacytoid dendritic cell-specific receptor ILT7-Fc
epsilonRI gamma inhibits Toll-like receptor-induced interferon production. The Journal
of experimental medicine 203: 1399-1405.
Siegal FP, Kadowaki N, Shodell M, et al. (1999) The nature of the principal type 1
interferon-producing cells in human blood. Science (New York, N.Y 284: 1835-1837.
Matsui T, Connolly JE, Michnevitz M, et al. (2009) CD2 distinguishes two subsets of
human plasmacytoid dendritic cells with distinct phenotype and functions. J Immunol
182: 6815-6823.
Romani N, Gruner S, Brang D, et al. (1994) Proliferating dendritic cell progenitors in
human blood. The Journal of experimental medicine 180: 83-93.
Shortman K, Naik SH. (2007) Steady-state and inflammatory dendritic-cell development.
Nature reviews 7: 19-30.
Rossi M, Young JW. (2005) Human dendritic cells: potent antigen-presenting cells at the
crossroads of innate and adaptive immunity. J Immunol 175: 1373-1381.
Sebzda E, Mariathasan S, Ohteki T, Jones R, Bachmann MF, Ohashi PS. (1999) Selection
of the T cell repertoire. Annual review of immunology 17: 829-874.
Mempel TR, Henrickson SE, Von Andrian UH. (2004) T-cell priming by dendritic cells in
lymph nodes occurs in three distinct phases. Nature 427: 154-159.
Sallusto F, Geginat J, Lanzavecchia A. (2004) Central memory and effector memory T cell
subsets: function, generation, and maintenance. Annual review of immunology 22: 745763.
Lanzavecchia A, Sallusto F. (2000) Dynamics of T lymphocyte responses: intermediates,
effectors, and memory cells. Science (New York, N.Y 290: 92-97.
Peck A, Mellins ED. Plasticity of T-cell phenotype and function: the T helper type 17
example. Immunology 129: 147-153.
Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coffman RL. (1986) Two types of
murine helper T cell clone. I. Definition according to profiles of lymphokine activities and
secreted proteins. J Immunol 136: 2348-2357.
Chen Y, Kuchroo VK, Inobe J, Hafler DA, Weiner HL. (1994) Regulatory T cell clones
induced by oral tolerance: suppression of autoimmune encephalomyelitis. Science (New
York, N.Y 265: 1237-1240.
Groux H, O'Garra A, Bigler M, et al. (1997) A CD4+ T-cell subset inhibits antigen-specific
T-cell responses and prevents colitis. Nature 389: 737-742.
Kalinski P, Hilkens CM, Wierenga EA, Kapsenberg ML. (1999) T-cell priming by type-1 and
type-2 polarized dendritic cells: the concept of a third signal. Immunology today 20: 561567.
Kapsenberg ML. (2003) Dendritic-cell control of pathogen-driven T-cell polarization.
Nature reviews 3: 984-993.
Barry M, Bleackley RC. (2002) Cytotoxic T lymphocytes: all roads lead to death. Nature
reviews 2: 401-409.
Mosmann TR, Li L, Sad S. (1997) Functions of CD8 T-cell subsets secreting different
cytokine patterns. Seminars in immunology 9: 87-92.
Delfs MW, Furukawa Y, Mitchell RN, Lichtman AH. (2001) CD8+ T cell subsets TC1 and
TC2 cause different histopathologic forms of murine cardiac allograft rejection.
Transplantation 71: 606-610.
59
References
161.
162.
163.
164.
165.
166.
167.
168.
169.
170.
171.
172.
173.
174.
175.
176.
177.
Smith-Garvin JE, Koretzky GA, Jordan MS. (2009) T cell activation. Annual review of
immunology 27: 591-619.
Jenkins MK, Schwartz RH. (1987) Antigen presentation by chemically modified
splenocytes induces antigen-specific T cell unresponsiveness in vitro and in vivo. The
Journal of experimental medicine 165: 302-319.
Cunningham AJ, Lafferty KJ. (1977) A simple conservative explanation of the H-2
restriction of interactions between lymphocytes. Scandinavian journal of immunology 6:
1-6.
Bettelli E, Carrier Y, Gao W, et al. (2006) Reciprocal developmental pathways for the
generation of pathogenic effector TH17 and regulatory T cells. Nature 441: 235-238.
Kunkel EJ, Butcher EC. (2002) Chemokines and the tissue-specific migration of
lymphocytes. Immunity 16: 1-4.
Campbell JJ, Butcher EC. (2000) Chemokines in tissue-specific and microenvironmentspecific lymphocyte homing. Current opinion in immunology 12: 336-341.
Agace WW. (2006) Tissue-tropic effector T cells: generation and targeting opportunities.
Nature reviews 6: 682-692.
DeNucci CC, Pagan AJ, Mitchell JS, Shimizu Y. Control of alpha4beta7 integrin expression
and CD4 T cell homing by the beta1 integrin subunit. J Immunol 184: 2458-2467.
Picker LJ, Michie SA, Rott LS, Butcher EC. (1990) A unique phenotype of skin-associated
lymphocytes in humans. Preferential expression of the HECA-452 epitope by benign and
malignant T cells at cutaneous sites. The American journal of pathology 136: 1053-1068.
Bos JD, de Boer OJ, Tibosch E, Das PK, Pals ST. (1993) Skin-homing T lymphocytes:
detection of cutaneous lymphocyte-associated antigen (CLA) by HECA-452 in normal
human skin. Archives of dermatological research 285: 179-183.
Lubong Sabado R, Kavanagh DG, Kaufmann DE, et al. (2009) In vitro priming
recapitulates in vivo HIV-1 specific T cell responses, revealing rapid loss of virus reactive
CD4 T cells in acute HIV-1 infection. PloS one 4: e4256.
Chitnis V, Pahwa R, Pahwa S. (2003) Determinants of HIV-specific CD8 T-cell responses in
HIV-infected pediatric patients and enhancement of HIV-gag-specific responses with
exogenous IL-15. Clinical immunology (Orlando, Fla 107: 36-45.
Klein MR, van Baalen CA, Holwerda AM, et al. (1995) Kinetics of Gag-specific cytotoxic T
lymphocyte responses during the clinical course of HIV-1 infection: a longitudinal
analysis of rapid progressors and long-term asymptomatics. The Journal of experimental
medicine 181: 1365-1372.
Rosenberg ES, Billingsley JM, Caliendo AM, et al. (1997) Vigorous HIV-1-specific CD4+ T
cell responses associated with control of viremia. Science (New York, N.Y 278: 14471450.
Wahren B, Morfeldt-Mansson L, Biberfeld G, et al. (1987) Characteristics of the specific
cell-mediated immune response in human immunodeficiency virus infection. Journal of
virology 61: 2017-2023.
Martin N, Welsch S, Jolly C, Briggs JA, Vaux D, Sattentau QJ. Virological synapsemediated spread of human immunodeficiency virus type 1 between T cells is sensitive to
entry inhibition. Journal of virology 84: 3516-3527.
Chen P, Hubner W, Spinelli MA, Chen BK. (2007) Predominant mode of human
immunodeficiency virus transfer between T cells is mediated by sustained Envdependent neutralization-resistant virological synapses. Journal of virology 81: 1258212595.
60
References
178.
179.
180.
181.
182.
183.
184.
185.
186.
187.
188.
189.
190.
191.
192.
193.
194.
Felts RL, Narayan K, Estes JD, et al. 3D visualization of HIV transfer at the virological
synapse between dendritic cells and T cells. Proceedings of the National Academy of
Sciences of the United States of America 107: 13336-13341.
Zoeteweij JP, Blauvelt A. (1998) HIV-Dendritic cell interactions promote efficient viral
infection of T cells. Journal of biomedical science 5: 253-259.
Granelli-Piperno A, Golebiowska A, Trumpfheller C, Siegal FP, Steinman RM. (2004) HIV1-infected monocyte-derived dendritic cells do not undergo maturation but can elicit IL10 production and T cell regulation. Proceedings of the National Academy of Sciences of
the United States of America 101: 7669-7674.
Kawamura T, Gatanaga H, Borris DL, Connors M, Mitsuya H, Blauvelt A. (2003)
Decreased stimulation of CD4+ T cell proliferation and IL-2 production by highly
enriched populations of HIV-infected dendritic cells. J Immunol 170: 4260-4266.
Smed-Sorensen A, Lore K, Walther-Jallow L, Andersson J, Spetz AL. (2004) HIV-1-infected
dendritic cells up-regulate cell surface markers but fail to produce IL-12 p70 in response
to CD40 ligand stimulation. Blood 104: 2810-2817.
Thoulouze MI, Sol-Foulon N, Blanchet F, Dautry-Varsat A, Schwartz O, Alcover A. (2006)
Human immunodeficiency virus type-1 infection impairs the formation of the
immunological synapse. Immunity 24: 547-561.
Douek DC, Brenchley JM, Betts MR, et al. (2002) HIV preferentially infects HIV-specific
CD4+ T cells. Nature 417: 95-98.
Miedema F, Meyaard L, Koot M, et al. (1994) Changing virus-host interactions in the
course of HIV-1 infection. Immunological reviews 140: 35-72.
Palmer BE, Blyveis N, Fontenot AP, Wilson CC. (2005) Functional and phenotypic
characterization of CD57+CD4+ T cells and their association with HIV-1-induced T cell
dysfunction. J Immunol 175: 8415-8423.
Zaunders JJ, Ip S, Munier ML, et al. (2006) Infection of CD127+ (interleukin-7 receptor+)
CD4+ cells and overexpression of CTLA-4 are linked to loss of antigen-specific CD4 T cells
during primary human immunodeficiency virus type 1 infection. Journal of virology 80:
10162-10172.
Larsson M. (2005) HIV-1 and the hijacking of dendritic cells: a tug of war. Springer
seminars in immunopathology 26: 309-328.
Conti L, Fantuzzi L, Del Corno M, Belardelli F, Gessani S. (2004) Immunomodulatory
effects of the HIV-1 gp120 protein on antigen presenting cells: implications for AIDS
pathogenesis. Immunobiology 209: 99-115.
McCune JM. (2001) The dynamics of CD4+ T-cell depletion in HIV disease. Nature 410:
974-979.
Barron MA, Blyveis N, Palmer BE, MaWhinney S, Wilson CC. (2003) Influence of plasma
viremia on defects in number and immunophenotype of blood dendritic cell subsets in
human immunodeficiency virus 1-infected individuals. The Journal of infectious diseases
187: 26-37.
Pacanowski J, Kahi S, Baillet M, et al. (2001) Reduced blood CD123+ (lymphoid) and
CD11c+ (myeloid) dendritic cell numbers in primary HIV-1 infection. Blood 98: 30163021.
Williams MA, Trout R, Spector SA. (2002) HIV-1 gp120 modulates the immunological
function and expression of accessory and co-stimulatory molecules of monocyte-derived
dendritic cells. Journal of hematotherapy & stem cell research 11: 829-847.
Chaudhry A, Das SR, Hussain A, et al. (2005) The Nef protein of HIV-1 induces loss of cell
surface costimulatory molecules CD80 and CD86 in APCs. J Immunol 175: 4566-4574.
61
References
195.
196.
197.
198.
199.
200.
201.
202.
203.
204.
205.
206.
207.
208.
209.
210.
211.
212.
213.
214.
215.
Muthumani K, Hwang DS, Choo AY, et al. (2005) HIV-1 Vpr inhibits the maturation and
activation of macrophages and dendritic cells in vitro. International immunology 17:
103-116.
Zhang R, Fichtenbaum CJ, Hildeman DA, Lifson JD, Chougnet C. (2004) CD40 ligand
dysregulation in HIV infection: HIV glycoprotein 120 inhibits signaling cascades upstream
of CD40 ligand transcription. J Immunol 172: 2678-2686.
Chen L. (2004) Co-inhibitory molecules of the B7-CD28 family in the control of T-cell
immunity. Nature reviews 4: 336-347.
Croft M. (2003) Co-stimulatory members of the TNFR family: keys to effective T-cell
immunity? Nature reviews 3: 609-620.
Croft M. (2003) Costimulation of T cells by OX40, 4-1BB, and CD27. Cytokine & growth
factor reviews 14: 265-273.
Salek-Ardakani S, Song J, Halteman BS, et al. (2003) OX40 (CD134) controls memory T
helper 2 cells that drive lung inflammation. The Journal of experimental medicine 198:
315-324.
Schoenberger SP, Toes RE, van der Voort EI, Offringa R, Melief CJ. (1998) T-cell help for
cytotoxic T lymphocytes is mediated by CD40-CD40L interactions. Nature 393: 480-483.
Ridge JP, Di Rosa F, Matzinger P. (1998) A conditioned dendritic cell can be a temporal
bridge between a CD4+ T-helper and a T-killer cell. Nature 393: 474-478.
Baecher-Allan C, Anderson DE. (2006) Regulatory cells and human cancer. Seminars in
cancer biology 16: 98-105.
Chatila TA. (2005) Role of regulatory T cells in human diseases. The Journal of allergy
and clinical immunology 116: 949-959; quiz 960.
Mills KH. (2004) Regulatory T cells: friend or foe in immunity to infection? Nature
reviews 4: 841-855.
Battaglia M, Roncarolo MG. (2006) Induction of transplantation tolerance via regulatory
T cells. Inflammation & allergy drug targets 5: 157-165.
Zenclussen AC. (2006) Regulatory T cells in pregnancy. Springer seminars in
immunopathology 28: 31-39.
Roncarolo MG, Bacchetta R, Bordignon C, Narula S, Levings MK. (2001) Type 1 T
regulatory cells. Immunological reviews 182: 68-79.
Weiner HL. (2001) Induction and mechanism of action of transforming growth factorbeta-secreting Th3 regulatory cells. Immunological reviews 182: 207-214.
Sakaguchi S. (2000) Regulatory T cells: key controllers of immunologic self-tolerance.
Cell 101: 455-458.
Chatenoud L, Salomon B, Bluestone JA. (2001) Suppressor T cells--they're back and
critical for regulation of autoimmunity! Immunological reviews 182: 149-163.
Shevach EM. (2002) CD4+ CD25+ suppressor T cells: more questions than answers.
Nature reviews 2: 389-400.
Takahashi T, Kuniyasu Y, Toda M, et al. (1998) Immunologic self-tolerance maintained by
CD25+CD4+ naturally anergic and suppressive T cells: induction of autoimmune disease
by breaking their anergic/suppressive state. International immunology 10: 1969-1980.
Sakaguchi S, Yamaguchi T, Nomura T, Ono M. (2008) Regulatory T cells and immune
tolerance. Cell 133: 775-787.
Zhu C, Anderson AC, Schubart A, et al. (2005) The Tim-3 ligand galectin-9 negatively
regulates T helper type 1 immunity. Nature immunology 6: 1245-1252.
62
References
216.
217.
218.
219.
220.
221.
222.
223.
224.
225.
226.
227.
228.
229.
230.
231.
232.
Workman CJ, Cauley LS, Kim IJ, Blackman MA, Woodland DL, Vignali DA. (2004)
Lymphocyte activation gene-3 (CD223) regulates the size of the expanding T cell
population following antigen activation in vivo. J Immunol 172: 5450-5455.
Shin H, Blackburn SD, Intlekofer AM, et al. (2009) A role for the transcriptional repressor
Blimp-1 in CD8(+) T cell exhaustion during chronic viral infection. Immunity 31: 309-320.
Hsiao HW, Liu WH, Wang CJ, et al. (2009) Deltex1 is a target of the transcription factor
NFAT that promotes T cell anergy. Immunity 31: 72-83.
Che KF, Sabado RL, Shankar EM, et al. HIV-1 impairs in vitro priming of naive T cells and
gives rise to contact-dependent suppressor T cells. European journal of immunology 40:
2248-2258.
Shankar EM, Che KF, Messmer D, Lifson JD, Larsson M. Expression of a Broad Array of
Negative Costimulatory Molecules and Blimp-1 in T-cells Following Priming by HIV-1
Pulsed Dendritic Cells. Molecular medicine (Cambridge, Mass.
Kaufmann DE, Walker BD. (2009) PD-1 and CTLA-4 inhibitory cosignaling pathways in
HIV infection and the potential for therapeutic intervention. J Immunol 182: 5891-5897.
Walunas TL, Bakker CY, Bluestone JA. (1996) CTLA-4 ligation blocks CD28-dependent T
cell activation. The Journal of experimental medicine 183: 2541-2550.
Keir ME, Francisco LM, Sharpe AH. (2007) PD-1 and its ligands in T-cell immunity.
Current opinion in immunology 19: 309-314.
Kinter A, McNally J, Riggin L, Jackson R, Roby G, Fauci AS. (2007) Suppression of HIVspecific T cell activity by lymph node CD25+ regulatory T cells from HIV-infected
individuals. Proceedings of the National Academy of Sciences of the United States of
America 104: 3390-3395.
Weiss L, Donkova-Petrini V, Caccavelli L, Balbo M, Carbonneil C, Levy Y. (2004) Human
immunodeficiency virus-driven expansion of CD4+CD25+ regulatory T cells, which
suppress HIV-specific CD4 T-cell responses in HIV-infected patients. Blood 104: 32493256.
Day CL, Kaufmann DE, Kiepiela P, et al. (2006) PD-1 expression on HIV-specific T cells is
associated with T-cell exhaustion and disease progression. Nature 443: 350-354.
Kaufmann DE, Kavanagh DG, Pereyra F, et al. (2007) Upregulation of CTLA-4 by HIVspecific CD4+ T cells correlates with disease progression and defines a reversible
immune dysfunction. Nature immunology 8: 1246-1254.
Sehrawat S, Suryawanshi A, Hirashima M, Rouse BT. (2009) Role of Tim-3/galectin-9
inhibitory interaction in viral-induced immunopathology: shifting the balance toward
regulators. J Immunol 182: 3191-3201.
Sanchez-Fueyo A, Tian J, Picarella D, et al. (2003) Tim-3 inhibits T helper type 1mediated auto- and alloimmune responses and promotes immunological tolerance.
Nature immunology 4: 1093-1101.
Jones RB, Ndhlovu LC, Barbour JD, et al. (2008) Tim-3 expression defines a novel
population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1
infection. The Journal of experimental medicine 205: 2763-2779.
Cai G, Anumanthan A, Brown JA, Greenfield EA, Zhu B, Freeman GJ. (2008) CD160
inhibits activation of human CD4+ T cells through interaction with herpesvirus entry
mediator. Nature immunology 9: 176-185.
Yamamoto T, Price DA, Casazza JP, et al. Surface expression patterns of negative
regulatory molecules identify determinants of virus-specific CD8+ T cell exhaustion in
HIV infection. Blood.
63
References
233.
234.
235.
236.
237.
238.
239.
240.
241.
242.
243.
244.
245.
246.
247.
248.
249.
250.
Li Q, Smith AJ, Schacker TW, et al. (2009) Microarray analysis of lymphatic tissue reveals
stage-specific, gene expression signatures in HIV-1 infection. J Immunol 183: 1975-1982.
Li N, Wang Y, Forbes K, et al. (2007) Metalloproteases regulate T-cell proliferation and
effector function via LAG-3. The EMBO journal 26: 494-504.
Price P, Keane N, Gray L, Lee S, Gorry PR, French MA. (2006) CXCR4 or CCR5 tropism of
human immunodeficiency virus type 1 isolates does not determine the immunological
milieu in patients responding to antiretroviral therapy. Viral immunology 19: 734-740.
Huang CT, Workman CJ, Flies D, et al. (2004) Role of LAG-3 in regulatory T cells.
Immunity 21: 503-513.
Blackburn SD, Shin H, Haining WN, et al. (2009) Coregulation of CD8+ T cell exhaustion
by multiple inhibitory receptors during chronic viral infection. Nature immunology 10:
29-37.
Lunemann JD, Waiczies S, Ehrlich S, et al. (2002) Death ligand TRAIL induces no
apoptosis but inhibits activation of human (auto)antigen-specific T cells. J Immunol 168:
4881-4888.
Wang SH, Chen GH, Fan Y, Van Antwerp M, Baker JR, Jr. (2009) Tumor necrosis factorrelated apoptosis-inducing ligand inhibits experimental autoimmune thyroiditis by the
expansion of CD4+CD25+ regulatory T cells. Endocrinology 150: 2000-2007.
Oestreich KJ, Yoon H, Ahmed R, Boss JM. (2008) NFATc1 regulates PD-1 expression upon
T cell activation. J Immunol 181: 4832-4839.
Gibson HM, Hedgcock CJ, Aufiero BM, et al. (2007) Induction of the CTLA-4 gene in
human lymphocytes is dependent on NFAT binding the proximal promoter. J Immunol
179: 3831-3840.
Martins GA, Cimmino L, Shapiro-Shelef M, et al. (2006) Transcriptional repressor Blimp-1
regulates T cell homeostasis and function. Nature immunology 7: 457-465.
Martins GA, Cimmino L, Liao J, Magnusdottir E, Calame K. (2008) Blimp-1 directly
represses Il2 and the Il2 activator Fos, attenuating T cell proliferation and survival. The
Journal of experimental medicine 205: 1959-1965.
Ott DE, Nigida SM, Jr., Henderson LE, Arthur LO. (1995) The majority of cells are
superinfected in a cloned cell line that produces high levels of human immunodeficiency
virus type 1 strain MN. Journal of virology 69: 2443-2450.
Rossio JL, Esser MT, Suryanarayana K, et al. (1998) Inactivation of human
immunodeficiency virus type 1 infectivity with preservation of conformational and
functional integrity of virion surface proteins. Journal of virology 72: 7992-8001.
Sedgwick JD, Holt PG. (1983) A solid-phase immunoenzymatic technique for the
enumeration of specific antibody-secreting cells. Journal of immunological methods 57:
301-309.
Lyons AB, Parish CR. (1994) Determination of lymphocyte division by flow cytometry.
Journal of immunological methods 171: 131-137.
Lu W, Arraes LC, Ferreira WT, Andrieu JM. (2004) Therapeutic dendritic-cell vaccine for
chronic HIV-1 infection. Nature medicine 10: 1359-1365.
Huang XL, Fan Z, Zheng L, et al. (2003) Priming of human immunodeficiency virus type 1
(HIV-1)-specific CD8+ T cell responses by dendritic cells loaded with HIV-1 proteins. The
Journal of infectious diseases 187: 315-319.
Zarling AL, Johnson JG, Hoffman RW, Lee DR. (1999) Induction of primary human CD8+ T
lymphocyte responses in vitro using dendritic cells. J Immunol 162: 5197-5204.
64
References
251.
252.
253.
254.
255.
256.
257.
258.
259.
260.
261.
262.
263.
264.
265.
266.
Gruber A, Kan-Mitchell J, Kuhen KL, Mukai T, Wong-Staal F. (2000) Dendritic cells
transduced by multiply deleted HIV-1 vectors exhibit normal phenotypes and functions
and elicit an HIV-specific cytotoxic T-lymphocyte response in vitro. Blood 96: 1327-1333.
Van Gulck ER, Vanham G, Heyndrickx L, et al. (2008) Efficient in vitro expansion of
human immunodeficiency virus (HIV)-specific T-cell responses by gag mRNAelectroporated dendritic cells from treated and untreated HIV type 1-infected
individuals. Journal of virology 82: 3561-3573.
Huang XL, Fan Z, Borowski L, Rinaldo CR. (2008) Maturation of dendritic cells for
enhanced activation of anti-HIV-1 CD8(+) T cell immunity. Journal of leukocyte biology
83: 1530-1540.
Lapenta C, Santini SM, Logozzi M, et al. (2003) Potent immune response against HIV-1
and protection from virus challenge in hu-PBL-SCID mice immunized with inactivated
virus-pulsed dendritic cells generated in the presence of IFN-alpha. The Journal of
experimental medicine 198: 361-367.
Yoshida A, Tanaka R, Murakami T, et al. (2003) Induction of protective immune
responses against R5 human immunodeficiency virus type 1 (HIV-1) infection in hu-PBLSCID mice by intrasplenic immunization with HIV-1-pulsed dendritic cells: possible
involvement of a novel factor of human CD4(+) T-cell origin. Journal of virology 77:
8719-8728.
Wilson CC, Palmer B, Southwood S, et al. (2001) Identification and antigenicity of
broadly cross-reactive and conserved human immunodeficiency virus type 1-derived
helper T-lymphocyte epitopes. Journal of virology 75: 4195-4207.
Frahm N, Korber BT, Adams CM, et al. (2004) Consistent cytotoxic-T-lymphocyte
targeting of immunodominant regions in human immunodeficiency virus across multiple
ethnicities. Journal of virology 78: 2187-2200.
Kaufmann DE, Bailey PM, Sidney J, et al. (2004) Comprehensive analysis of human
immunodeficiency virus type 1-specific CD4 responses reveals marked
immunodominance of gag and nef and the presence of broadly recognized peptides.
Journal of virology 78: 4463-4477.
Addo MM, Altfeld M, Rosenberg ES, et al. (2001) The HIV-1 regulatory proteins Tat and
Rev are frequently targeted by cytotoxic T lymphocytes derived from HIV-1-infected
individuals. Proceedings of the National Academy of Sciences of the United States of
America 98: 1781-1786.
McDonald D, Wu L, Bohks SM, KewalRamani VN, Unutmaz D, Hope TJ. (2003)
Recruitment of HIV and its receptors to dendritic cell-T cell junctions. Science (New York,
N.Y 300: 1295-1297.
Zaunders JJ, Munier ML, Kaufmann DE, et al. (2005) Early proliferation of CCR5(+)
CD38(+++) antigen-specific CD4(+) Th1 effector cells during primary HIV-1 infection.
Blood 106: 1660-1667.
Brenchley JM, Price DA, Schacker TW, et al. (2006) Microbial translocation is a cause of
systemic immune activation in chronic HIV infection. Nature medicine 12: 1365-1371.
Letvin NL. (2006) Progress and obstacles in the development of an AIDS vaccine. Nature
reviews 6: 930-939.
Robinson HL. (2007) HIV/AIDS vaccines: 2007. Clinical pharmacology and therapeutics
82: 686-693.
McMichael AJ. (2006) HIV vaccines. Annual review of immunology 24: 227-255.
Thorner AR, Barouch DH. (2007) HIV-1 vaccine development: progress and prospects.
Current infectious disease reports 9: 71-75.
65
References
267.
268.
269.
270.
271.
272.
273.
274.
275.
276.
277.
278.
279.
280.
281.
282.
Robinson HL, Amara RR. (2005) T cell vaccines for microbial infections. Nature medicine
11: S25-32.
Quaranta MG, Mattioli B, Giordani L, Viora M. (2004) HIV-1 Nef equips dendritic cells to
reduce survival and function of CD8+ T cells: a mechanism of immune evasion. Faseb J
18: 1459-1461.
Herbeuval JP, Boasso A, Grivel JC, et al. (2005) TNF-related apoptosis-inducing ligand
(TRAIL) in HIV-1-infected patients and its in vitro production by antigen-presenting cells.
Blood 105: 2458-2464.
Gondek DC, Lu LF, Quezada SA, Sakaguchi S, Noelle RJ. (2005) Cutting edge: contactmediated suppression by CD4+CD25+ regulatory cells involves a granzyme B-dependent,
perforin-independent mechanism. J Immunol 174: 1783-1786.
Grossman WJ, Verbsky JW, Barchet W, Colonna M, Atkinson JP, Ley TJ. (2004) Human T
regulatory cells can use the perforin pathway to cause autologous target cell death.
Immunity 21: 589-601.
Salomon B, Lenschow DJ, Rhee L, et al. (2000) B7/CD28 costimulation is essential for the
homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune
diabetes. Immunity 12: 431-440.
McMahan RH, Golden-Mason L, Nishimura MI, et al. Tim-3 expression on PD-1+ HCVspecific human CTLs is associated with viral persistence, and its blockade restores
hepatocyte-directed in vitro cytotoxicity. The Journal of clinical investigation 120: 45464557.
del Rio ML, Lucas CL, Buhler L, Rayat G, Rodriguez-Barbosa JI. HVEM/LIGHT/BTLA/CD160
cosignaling pathways as targets for immune regulation. Journal of leukocyte biology 87:
223-235.
Golden-Mason L, Palmer BE, Kassam N, et al. (2009) Negative immune regulator Tim-3 is
overexpressed on T cells in hepatitis C virus infection and its blockade rescues
dysfunctional CD4+ and CD8+ T cells. Journal of virology 83: 9122-9130.
Agnellini P, Wolint P, Rehr M, Cahenzli J, Karrer U, Oxenius A. (2007) Impaired NFAT
nuclear translocation results in split exhaustion of virus-specific CD8+ T cell functions
during chronic viral infection. Proceedings of the National Academy of Sciences of the
United States of America 104: 4565-4570.
Muthumani K, Choo AY, Shedlock DJ, et al. (2008) Human immunodeficiency virus type 1
Nef induces programmed death 1 expression through a p38 mitogen-activated protein
kinase-dependent mechanism. Journal of virology 82: 11536-11544.
Kortylewski M, Yu H. (2008) Role of Stat3 in suppressing anti-tumor immunity. Current
opinion in immunology 20: 228-233.
Wang T, Niu G, Kortylewski M, et al. (2004) Regulation of the innate and adaptive
immune responses by Stat-3 signaling in tumor cells. Nature medicine 10: 48-54.
Kortylewski M, Kujawski M, Wang T, et al. (2005) Inhibiting Stat3 signaling in the
hematopoietic system elicits multicomponent antitumor immunity. Nature medicine 11:
1314-1321.
Pallandre JR, Brillard E, Crehange G, et al. (2007) Role of STAT3 in CD4+CD25+FOXP3+
regulatory lymphocyte generation: implications in graft-versus-host disease and
antitumor immunity. J Immunol 179: 7593-7604.
Bromberg J, Wang TC. (2009) Inflammation and cancer: IL-6 and STAT3 complete the
link. Cancer cell 15: 79-80.
66
References
283.
284.
285.
286.
287.
288.
289.
290.
291.
292.
293.
294.
295.
Tanaka N, Hoshino Y, Gold J, et al. (2005) Interleukin-10 induces inhibitory C/EBPbeta
through STAT-3 and represses HIV-1 transcription in macrophages. American journal of
respiratory cell and molecular biology 33: 406-411.
Cheng F, Wang HW, Cuenca A, et al. (2003) A critical role for Stat3 signaling in immune
tolerance. Immunity 19: 425-436.
Nishiki S, Hato F, Kamata N, et al. (2004) Selective activation of STAT3 in human
monocytes stimulated by G-CSF: implication in inhibition of LPS-induced TNF-alpha
production. American journal of physiology 286: C1302-1311.
Aggarwal BB, Sethi G, Ahn KS, et al. (2006) Targeting signal-transducer-and-activator-oftranscription-3 for prevention and therapy of cancer: modern target but ancient
solution. Annals of the New York Academy of Sciences 1091: 151-169.
Lim CP, Cao X. (2001) Regulation of Stat3 activation by MEK kinase 1. The Journal of
biological chemistry 276: 21004-21011.
Turkson J, Bowman T, Adnane J, et al. (1999) Requirement for Ras/Rac1-mediated p38
and c-Jun N-terminal kinase signaling in Stat3 transcriptional activity induced by the Src
oncoprotein. Molecular and cellular biology 19: 7519-7528.
Kassu A, Marcus RA, D'Souza MB, et al. Regulation of virus-specific CD4+ T cell function
by multiple costimulatory receptors during chronic HIV infection. J Immunol 185: 30073018.
Hori S, Nomura T, Sakaguchi S. (2003) Control of regulatory T cell development by the
transcription factor Foxp3. Science (New York, N.Y 299: 1057-1061.
Bromberg J, Chen X. (2001) STAT proteins: signal tranducers and activators of
transcription. Methods in enzymology 333: 138-151.
Obata T, Brown GE, Yaffe MB. (2000) MAP kinase pathways activated by stress: the p38
MAPK pathway. Critical care medicine 28: N67-77.
Zarubin T, Han J. (2005) Activation and signaling of the p38 MAP kinase pathway. Cell
research 15: 11-18.
Callahan JE, Herman A, Kappler JW, Marrack P. (1990) Stimulation of B10.BR T cells with
superantigenic staphylococcal toxins. J Immunol 144: 2473-2479.
Li H, Llera A, Malchiodi EL, Mariuzza RA. (1999) The structural basis of T cell activation by
superantigens. Annual review of immunology 17: 435-466.
67