Download HIV - Wikipedia, the free encyclopedia

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

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

Document related concepts

Pandemic wikipedia , lookup

HIV trial in Libya wikipedia , lookup

Zinc finger nuclease wikipedia , lookup

Infection wikipedia , lookup

Viral phylodynamics wikipedia , lookup

Diseases of poverty wikipedia , lookup

Syndemic wikipedia , lookup

Epidemiology of HIV/AIDS wikipedia , lookup

HIV and pregnancy wikipedia , lookup

Index of HIV/AIDS-related articles wikipedia , lookup

Transcript
HIV - Wikipedia, the free encyclopedia
HIV
From Wikipedia, the free encyclopedia
Human immunodeficiency virus (HIV) is a lentivirus (a member of the
retrovirus family) that causes acquired immunodeficiency syndrome (AIDS), a
condition in humans in which the immune system begins to fail, leading to lifethreatening opportunistic infections. Infection with HIV occurs by the transfer
of blood, semen, vaginal fluid, pre-ejaculate, or breast milk. Within these bodily
fluids, HIV is present as both free virus particles and virus within infected
immune cells. The four major routes of transmission are unsafe sex,
contaminated needles, breast milk, and transmission from an infected mother
to her baby at birth (Vertical transmission). Screening of blood products for
HIV has largely eliminated transmission through blood transfusions or infected
blood products in the developed world.
HIV infection in humans is considered pandemic by the World Health
Organization (WHO). From its discovery in 1981 to 2006, AIDS killed more than
25 million people.[1] HIV infects about 0.6% of the world's population.[1] In
2005 alone, AIDS claimed an estimated 2.4–3.3 million lives, of which more
than 570,000 were children. A third of these deaths are occurring in subSaharan Africa, retarding economic growth and increasing poverty.[2]
According to current estimates, HIV is set to infect 90 million people in Africa,
resulting in a minimum estimate of 18 million orphans.[3] Antiretroviral
treatment reduces both the mortality and the morbidity of HIV infection, but
routine access to antiretroviral medication is not available in all countries. [4]
Human immunodeficiency
virus
Scanning electron micrograph
of HIV-1 (in green) budding
from cultured lymphocyte.
Multiple round bumps on cell
surface represent sites of
assembly and budding of
virions.
Virus classification
Group: Group VI (ssRNART)
Family: Retroviridae
Genus: Lentivirus
Species
Human
immunodeficiency
virus 1
Human
immunodeficiency
virus 2
HIV infects primarily vital cells in the human immune system such as helper T
cells (to be specific, CD4 + T cells), macrophages, and dendritic cells. HIV
infection leads to low levels of CD4 + T cells through three main mechanisms:
First, direct viral killing of infected cells; second, increased rates of apoptosis in
infected cells; and third, killing of infected CD4 + T cells by CD8 cytotoxic
lymphocytes that recognize infected cells. When CD4 + T cell numbers decline
below a critical level, cell-mediated immunity is lost, and
International Statistical Classification
the body becomes progressively more susceptible to
of Diseases and Related Health
opportunistic infections.
Problems Codes
Classification and external resources
Most people infected with HIV eventually develop AIDS.
These individuals mostly die from opportunistic
ICD-10 B20-B24
infections or malignancies associated with the
ICD-9 042
progressive failure of the immune system. [5] HIV
(http://www.icd9data.com/getICD9Code.ashx?
progresses to AIDS at a variable rate affected by viral,
icd9=042) -044
host, and environmental factors; HIV-specific treatment
(http://www.icd9data.com/getICD9Code.ashx?
delays this process. Most will progress to AIDS within 10
years of HIV infection: some will have progressed much
icd9=044)
[6][7]
sooner, and some will take much longer.
Treatment
with anti-retrovirals increases the life expectancy of people infected with HIV. Even after HIV has progressed
to diagnosable AIDS, the average survival time with antiretroviral therapy was estimated to be more than
[8]
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
[9]
HIV - Wikipedia, the free encyclopedia
5 years as of 2005.
Without antiretroviral therapy, someone who has AIDS typically dies within a year.
Contents
1 Classification
2 Signs and symptoms
2.1 Acute HIV infection
2.2 Latency stage
2.3 AIDS
3 Pathophysiology
3.1 Transmission
3.1.1 Sexual
3.1.2 Blood or blood product
3.1.3 Mother-to-child
3.1.4 Multiple infection
3.2 Structure and genome
3.3 Tropism
3.4 Replication cycle
3.4.1 Entry to the cell
3.4.2 Replication and transcription
3.4.3 Assembly and release
3.5 Genetic variability
4 Diagnosis
5 Treatment
5.1 Treatments in development
5.2 HIV latent reservoir
6 Prognosis
7 Epidemiology
8 History
8.1 Origins
8.2 Discovery
9 AIDS denialism
10 See also
11 References
12 External links
Classification
HIV is a member of the genus Lentivirus,[10] part of the family of Retroviridae.[11] Lentiviruses have many
common morphologies and biological properties. Many species are infected by lentiviruses, which are
characteristically responsible for long-duration illnesses with a long incubation period.[12] Lentiviruses are
transmitted as single-stranded, positive-sense, enveloped RNA viruses. Upon entry of the target cell, the viral
RNA genome is converted to double-stranded DNA by a virally encoded reverse transcriptase that is present in
the virus particle. This viral DNA is then integrated into the cellular DNA by a virally encoded integrase, along
with host cellular co-factors,[13] so that the genome can be transcribed. After the virus has infected the cell,
two pathways are possible: either the virus becomes latent and the infected cell continues to function or the
virus becomes active and replicates, and a large number of virus particles that can then infect other cells are
liberated.
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
There are two species of HIV known to exist: HIV-1 and HIV-2. HIV-1 is the virus that was initially discovered
and termed LAV. It is more virulent, more infective,[14] and is the cause of the majority of HIV infections
globally. The lower infectivity of HIV-2 compared to HIV-1 implies that fewer of those exposed to HIV-2 will
be infected per exposure. Because of its relatively poor capacity for transmission, HIV-2 is largely confined to
West Africa.[15]
Comparison of HIV species
Species Virulence Infectivity Prevalence
Inferred origin
HIV-1 High
High
Global
Common Chimpanzee
HIV-2 Lower
Low
West Africa Sooty Mangabey
Signs and symptoms
Infection with HIV-1 is associated with a progressive decrease
of the CD4 + T cell count and an increase in viral load. The
stage of infection can be determined by measuring the patient's
CD4 + T cell count, and the level of HIV in the blood.
HIV infection has basically four stages: incubation period, acute
infection, latency stage and AIDS. The initial incubation period
upon infection is asymptomatic and usually lasts between two
and four weeks. The second stage, acute infection, which lasts
an average of 28 days and can include symptoms such as
fever, lymphadenopathy (swollen lymph nodes),
pharyngitis (sore throat), rash, myalgia (muscle pain), malaise,
and mouth and esophageal sores.
The latency stage, which occurs third, shows few or no
symptoms and can last anywhere from two weeks to twenty
years and beyond. AIDS, the fourth and final stage of HIV
infection shows as symptoms of various opportunistic infections.
A generalized graph of the relationship
between HIV copies (viral load) and CD4
counts over the average course of untreated
HIV infection; any particular individual's
disease course may vary considerably.
CD4+ T cell count (cells per µL)
HIV RNA copies per mL of plasma
A study of French hospital patients found that approximately 0.5% of HIV-1 infected individuals retain high
levels of CD4 T-Cells and a low or clinically undetectable viral load without anti-retroviral treatment. These
individuals are classified as HIV controllers or Long-term nonprogressors.[16]
Acute HIV infection
Main article: Acute HIV infection
The initial infection with HIV generally occurs after transfer of
body fluids from an infected person to an uninfected one. The
first stage of infection, the primary, or acute infection, is a
period of rapid viral replication that immediately follows the
individual's exposure to HIV leading to an abundance of virus
in the peripheral blood with levels of HIV commonly
approaching several million viruses per mL. [17]
This response is accompanied by a marked drop in the
numbers of circulating CD4 + T cells. This acute viremia is
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
associated in virtually all patients with the activation of CD8 + T
cells, which kill HIV-infected cells, and subsequently with
antibody production, or seroconversion. The CD8 + T cell
response is thought to be important in controlling virus levels,
which peak and then decline, as the CD4 + T cell counts
Main symptoms of acute HIV infection.
rebound to around 800 cells per µL (the normal blood value is
1200 cells per µL ). A good CD8 + T cell response has been linked to slower disease progression and a better
prognosis, though it does not eliminate the virus. [18]
During this period (usually 2–4 weeks post-exposure) most individuals (80 to 90%) develop an influenza or
mononucleosis-like illness called acute HIV infection, the most common symptoms of which may include fever,
lymphadenopathy, pharyngitis, rash, myalgia, malaise, mouth and esophagal sores, and may also include, but
less commonly, headache, nausea and vomiting, enlarged liver/spleen, weight loss, thrush, and neurological
symptoms. Infected individuals may experience all, some, or none of these symptoms. The duration of
symptoms varies, averaging 28 days and usually lasting at least a week. [19]
Because of the nonspecific nature of these symptoms, they are often not recognized as signs of HIV infection.
Even if patients go to their doctors or a hospital, they will often be misdiagnosed as having one of the more
common infectious diseases with the same symptoms. As a consequence, these primary symptoms are not
used to diagnose HIV infection, as they do not develop in all cases and because many are caused by other
more common diseases. However, recognizing the syndrome can be important because the patient is much
more infectious during this period. [20]
Latency stage
A strong immune defense reduces the number of viral particles in the blood stream, marking the start of the
infection's clinical latency stage. Clinical latency can vary between two weeks and 20 years. During this early
phase of infection, HIV is active within lymphoid organs, where large amounts of virus become trapped in the
follicular dendritic cells (FDC) network.[21]
The surrounding tissues that are rich in CD4 + T cells may also become infected, and viral particles accumulate
both in infected cells and as free virus. Individuals who are in this phase are still infectious. During this time,
CD4 + CD45RO + T cells carry most of the proviral load. [22]
AIDS
Main article: AIDS
For more details on this topic, see AIDS Diagnosis, AIDS Symptoms and WHO Disease Staging System
for HIV Infection and Disease
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
When CD4 + T cell numbers decline below a critical level of 200 cells per µL, cell-mediated immunity is lost,
and infections with a variety of opportunistic microbes appear. The first symptoms often include moderate
and unexplained weight loss, recurring respiratory tract infections (such as sinusitis, bronchitis, otitis media,
pharyngitis), prostatitis, skin rashes, and oral ulcerations.
Common opportunistic infections and tumors, most of which are normally controlled by robust CD4 + T cellmediated immunity then start to affect the patient. Typically, resistance is lost early on to oral Candida
species and to Mycobacterium tuberculosis, which leads to an increased susceptibility to oral
candidiasis(thrush) and tuberculosis. Later, reactivation of latent herpes viruses may cause worsening
recurrences of herpes simplex eruptions, shingles, Epstein-Barr virus-induced B-cell lymphomas, or Kaposi's
sarcoma.
Pneumonia caused by the fungus Pneumocystis jirovecii is common and often fatal. In the final stages of
AIDS, infection with cytomegalovirus (another herpes virus) or Mycobacterium avium complex is more
prominent. Not all patients with AIDS get all these infections or tumors, and there are other tumors and
infections that are less prominent but still significant.
Pathophysiology
Transmission
Three main transmission routes for HIV have
been identified. HIV-2 is transmitted much less
frequently by the mother-to-child and sexual
route than HIV-1.
Sexual
The majority of HIV infections are acquired
through unprotected sexual relations. Sexual
transmission can occur when infected sexual
secretions of one partner come into contact with
the genital, oral, or rectal mucous membranes of
another. In high-income countries, the risk of
female-to-male transmission is 0.04% per act
and male-to-female transmission is 0.08% per
act. For various reasons, these rates are 4 to 10
times higher in low-income countries. [31]
The correct and consistent use of latex condoms
reduces the risk of sexual transmission of HIV by
about 85%.[32] However, spermicide may
actually increase the transmission
rate.[33][34][35]
Estimated per-act risk for acquisition
of HIV by exposure route[23]
Estimated infections
Exposure Route
per 10,000 exposures
to an infected source
Blood Transfusion
9,000[24]
Childbirth
2,500[25]
Needle-sharing injection drug
67 [26]
use
Percutaneous needle stick
30 [27]
Receptive anal intercourse*
50 [28][29]
Insertive anal intercourse*
Receptive penile-vaginal
intercourse*
Insertive penile-vaginal
intercourse*
6.5 [28][29]
Receptive oral intercourse*§
1†[29]
Insertive oral intercourse*§
0.5 †[29]
10 [28][29][30]
5[28][29]
*
§
assuming no condom use
source refers to oral intercourse
performed on a man
†
"best-guess estimate"
A meta-analysis of 27 observational studies
conducted prior to 1999 in sub-Saharan Africa indicated that male circumcision reduces the risk of HIV
infection.[36] However, a subsequent review indicated that the correlation between circumcision and HIV in
these observational studies may have been due to confounding factors.[37] In addition, concerns were raised
about the potential for spread of HIV by unsterilized blades during ritual circumcision.[38] Later trials, in which
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
uncircumcised men were randomly assigned to be medically circumcised in sterile conditions and given
counseling and other men were not circumcised, have been conducted in South Africa,[39] Kenya,[40] and
Uganda[41] showing reductions in female-to-male sexual HIV transmission of 60%, 53%, and 51%
respectively. As a result, a panel of experts convened by WHO and the UNAIDS Secretariat has
"recommended that male circumcision now be recognized as an additional important intervention to reduce
the risk of heterosexually acquired HIV infection in men." [42]
Blood or blood product
In general if infected blood comes into contact with any open wound, HIV may be transmitted. This
transmission route can account for infections in intravenous drug users,hemophiliacs, and recipients of blood
transfusions (though most transfusions are checked for HIV in the developed world) and blood products. It is
also of concern for persons receiving medical care in regions where there is prevalent substandard hygiene in
the use of injection equipment, such as the reuse of needles in Third World countries. Health care workers
such as nurses, laboratory workers, and doctors have also been infected, although this occurs more rarely.
Since transmission of HIV by blood became known medical personnel are required to protect themselves from
contact with blood by the use of Universal precautions. People who give and receive tattoos,piercings, and
scarification procedures can also be at risk of infection.
HIV has been found at low concentrations in the saliva, tears and urine of infected individuals, but there are
no recorded cases of infection by these secretions and the potential risk of transmission is negligible.[43] It is
not possible for mosquitoes to transmit HIV. [44]
Mother-to-child
The transmission of the virus from the mother to the child can occur in utero (during pregnancy), intrapartum
(at childbirth), or via breast feeding. In the absence of treatment, the transmission rate up to birth between
the mother and child is around 25%.[25] However, where combination antiretroviral drug treatment and
Cesarian section are available, this risk can be reduced to as low as one percent.[25] Postnatal mother-tochild transmission may be largely prevented by complete avoidance of breast feeding; however, this has
significant associated morbidity. Exclusive breast feeding and the provision of extended antiretroviral
prophylaxis to the infant are also efficacious in avoiding transmission. [45]
Multiple infection
Main article: HIV superinfection
Unlike some other viruses, infection with HIV does not provide immunity against additional infections,
particularly in the case of more genetically distant viruses. Both inter- and intra-clade multiple infections have
been reported,[46] and even associated with more rapid disease progression.[47] Multiple infections are
divided into two categories depending on the timing of the acquisition of the second strain. Coinfection refers
to two strains that appear to have been acquired at the same time (or too close to distinguish). Reinfection
(or superinfection ) is infection with a second strain at a measurable time after the first. Both forms of dual
infection have been reported for HIV in both acute and chronic infection around the world.[48][49][50][51]
Structure and genome
Main article: Structure and genome of HIV
HIV is different in structure from other retroviruses. It is
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
roughly spherical [52] with a diameter of about 120 nm, around
60 times smaller than a red blood cell, yet large for a virus. [53]
It is composed of two copies of positive single-stranded RNA
that codes for the virus's nine genes enclosed by a conical
capsid composed of 2,000 copies of the viral protein p24.[54]
The single-stranded RNA is tightly bound to nucleocapsid
proteins, p7 and enzymes needed for the development of the
virion such as reverse transcriptase, proteases, ribonuclease
and integrase. A matrix composed of the viral protein p17
surrounds the capsid ensuring the integrity of the virion
Diagram of HIV
particle. [54] This is, in turn, surrounded by the viral envelope
that is composed of two layers of fatty molecules called
phospholipids taken from the membrane of a human cell when a newly formed virus particle buds from the
cell. Embedded in the viral envelope are proteins from the host cell and about 70 copies of a complex HIV
protein that protrudes through the surface of the virus particle. [54] This protein, known as Env, consists of a
cap made of three molecules called glycoprotein (gp) 120, and a stem consisting of three gp41 molecules that
anchor the structure into the viral envelope. [55] This glycoprotein complex enables the virus to attach to and
fuse with target cells to initiate the infectious cycle.[55] Both these surface proteins, especially gp120, have
been considered as targets of future treatments or vaccines against HIV. [56]
The RNA genome consists of at least seven structural landmarks (LTR,TAR, RRE, PE, SLIP, CRS, and INS) and
nine genes ( gag , pol , and env , tat , rev , nef , vif, vpr, vpu, and tev) encoding 19 proteins. Three of these
genes, gag , pol , and env , contain information needed to make the structural proteins for new virus
particles. [54] For example, env codes for a protein called gp160 that is broken down by a viral enzyme to form
gp120 and gp41. The six remaining genes, tat , rev , nef , vif, vpr, and vpu (or vpx in the case of HIV-2), are
regulatory genes for proteins that control the ability of HIV to infect cells, produce new copies of virus
(replicate), or cause disease.[54] The two Tat proteins (p16 and p14) are transcriptional transactivators for the
LTR promoter acting by binding the TAR RNA element. The TAR may also be processed into microRNAs that
regulate the apoptosis genes ERCC1 and IER3.[57][58] The Rev protein (p19) is involved in shuttling RNAs
from the nucleus and the cytoplasm by binding to the RRE RNA element. The Vif protein (p23) prevents the
action of APOBEC3G (a cell protein that deaminates DNA:RNA hybrids and/or interferes with the Pol protein).
The Vpr protein (p14) arrests cell division at G2/M. The Nef protein (p27) down-regulates CD4 (the major viral
receptor), as well as the MHC class I and class II molecules.[59][60][61] Nef also interacts with SH3 domains.
The Vpu protein (p16) influences the release of new virus particles from infected cells. [54] The ends of each
strand of HIV RNA contain an RNA sequence called the long terminal repeat (LTR). Regions in the LTR act as
switches to control production of new viruses and can be triggered by proteins from either HIV or the host
cell. The Psi element is involved in viral genome packaging and recognized by Gag and Rev proteins. The SLIP
element (TTTTTT) is involved in the frameshift in the Gag-Pol reading frame required to make functional
Pol. [54]
Tropism
Main article: HIV tropism
The term viral tropism refers to which cell types HIV infects. HIV can infect a variety of immune cells such as
CD4 + T cells, macrophages, and microglial cells. HIV-1 entry to macrophages and CD4 + T cells is mediated
through interaction of the virion envelope glycoproteins (gp120) with the CD4 molecule on the target cells
and also with chemokine coreceptors.[55]
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
Macrophage (M-tropic) strains of HIV-1, or non-syncitia-inducing strains (NSI) use the β-chemokine receptor
CCR5 for entry and are thus able to replicate in macrophages and CD4 + T cells. [62] This CCR5 coreceptor is
used by almost all primary HIV-1 isolates regardless of viral genetic subtype. Indeed, macrophages play a key
role in several critical aspects of HIV infection. They appear to be the first cells infected by HIV and perhaps
the source of HIV production when CD4 + cells become depleted in the patient. Macrophages and microglial
cells are the cells infected by HIV in the central nervous system. In tonsils and adenoids of HIV-infected
patients, macrophages fuse into multinucleated giant cells that produce huge amounts of virus.
T-tropic isolates, or syncitia-inducing (SI) strains replicate in primary CD4 + T cells as well as in macrophages
and use the α-chemokine receptor, CXCR4, for entry. [62][63][64] Dual-tropic HIV-1 strains are thought to be
transitional strains of the HIV-1 virus and thus are able to use both CCR5 and CXCR4 as co-receptors for viral
entry.
The α-chemokine SDF-1, a ligand for CXCR4, suppresses replication of T-tropic HIV-1 isolates. It does this by
down-regulating the expression of CXCR4 on the surface of these cells. HIV that use only the CCR5 receptor
are termed R5; those that only use CXCR4 are termed X4, and those that use both, X4R5. However, the use
of coreceptor alone does not explain viral tropism, as not all R5 viruses are able to use CCR5 on macrophages
for a productive infection [62] and HIV can also infect a subtype of myeloid dendritic cells,[65] which probably
constitute a reservoir that maintains infection when CD4 + T cell numbers have declined to extremely low
levels.
Some people are resistant to certain strains of HIV. [66] For example people with the CCR5-Δ32 mutation are
resistant to infection with R5 virus as the mutation stops HIV from binding to this coreceptor, reducing its
ability to infect target cells.
Sexual intercourse is the major mode of HIV transmission. Both X4 and R5 HIV are present in the seminal
fluid, which is passed from a male to his sexual partner. The virions can then infect numerous cellular targets
and disseminate into the whole organism. However, a selection process leads to a predominant transmission
of the R5 virus through this pathway.[67][68][69] How this selective process works is still under investigation,
but one model is that spermatozoa may selectively carry R5 HIV as they possess both CCR3 and CCR5 but
not CXCR4 on their surface [70] and that genital epithelial cells preferentially sequester X4 virus. [71] In patients
infected with subtype B HIV-1, there is often a co-receptor switch in late-stage disease and T-tropic variants
appear that can infect a variety of T cells through CXCR4. [72] These variants then replicate more aggressively
with heightened virulence that causes rapid T cell depletion, immune system collapse, and opportunistic
infections that mark the advent of AIDS. [73] Thus, during the course of infection, viral adaptation to the use
of CXCR4 instead of CCR5 may be a key step in the progression to AIDS. A number of studies with subtype Binfected individuals have determined that between 40 and 50% of AIDS patients can harbour viruses of the
SI, and presumably the X4, phenotype. [74][75]
HIV-2 is much less pathogenic than HIV-1 and is restricted in its worldwide distribution. The adoption of
"accessory genes" by HIV-2 and its more promiscuous pattern of coreceptor usage (including CD4independence) may assist the virus in its adaptation to avoid innate restriction factors present in host cells.
Adaptation to use normal cellular machinery to enable transmission and productive infection has also aided
the establishment of HIV-2 replication in humans. A survival strategy for any infectious agent is not to kill its
host but ultimately become a commensal organism. Having achieved a low pathogenicity, over time, variants
more successful at transmission will be selected. [76]
Replication cycle
Entry to the cell
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
HIV enters macrophages and CD4 + T cells by the adsorption of
glycoproteins on its surface to receptors on the target cell
followed by fusion of the viral envelope with the cell membrane
and the release of the HIV capsid into the cell.[77][78]
Entry to the cell begins through interaction of the trimeric
envelope complex (gp160 spike) and both CD4 and a
chemokine receptor (generally either CCR5 or CXCR4, but
others are known to interact) on the cell surface.[77][78] gp120
binds to integrin α4 β7 activating LFA-1 the central integrin
involved in the establishment of virological synapses, which
facilitate efficient cell-to-cell spreading of HIV-1.[79] The gp160
spike contains binding domains for both CD4 and chemokine
receptors.[77][78] The first step in fusion involves the highaffinity attachment of the CD4 binding domains of gp120 to
CD4. Once gp120 is bound with the CD4 protein, the envelope
complex undergoes a structural change, exposing the
chemokine binding domains of gp120 and allowing them to
interact with the target chemokine receptor. [77][78] This allows
for a more stable two-pronged attachment, which allows the Nterminal fusion peptide gp41 to penetrate the cell
membrane. [77][78] Repeat sequences in gp41, HR1 and HR2
then interact, causing the collapse of the extracellular portion of
gp41 into a hairpin. This loop structure brings the virus and cell
membranes close together, allowing fusion of the membranes
and subsequent entry of the viral capsid. [77][78]
After HIV has bound to the target cell, the HIV RNA and
various enzymes, including reverse transcriptase, integrase,
ribonuclease, and protease, are injected into the cell.[77] During
the microtubule based transport to the nucleus, the viral single
strand RNA genome is transcribed into double strand DNA,
which is then integrated into a host chromosome.
The HIV replication cycle
HIV can infect dendritic cells (DCs) by this CD4-CCR5 route, but another route using mannose-specific C-type
lectin receptors such as DC-SIGN can also be used. [80] DCs are one of the first cells encountered by the virus
during sexual transmission. They are currently thought to play an important role by transmitting HIV to T-cells
when the virus is captured in the mucosa by DCs. [80] The presence of FEZ-1, which occurs naturally in
neurons, is believed to prevent the infection of cells by HIV. [81]
Replication and transcription
Shortly after the viral capsid enters the cell, an enzyme called reverse transcriptase liberates the singlestranded (+)RNA genome from the attached viral proteins and copies it into a complementary DNA
molecule. [82] The process of reverse transcription is extremely error-prone, and the resulting mutations may
cause drug resistance or allow the virus to evade the body's immune system. The reverse transcriptase also
has ribonuclease activity that degrades the viral RNA during the synthesis of cDNA, as well as DNA-dependent
DNA polymerase activity that copies the sense cDNA strand into an antisense DNA.[83] Together, the cDNA
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
and its complement form a double-stranded viral DNA that is then transported into the cell nucleus. The
integration of the viral DNA into the host cell's genome is carried out by another viral enzyme called
integrase.[82]
This integrated viral DNA may then lie dormant, in the latent
stage of HIV infection.[82] To actively produce the virus, certain
cellular transcription factors need to be present, the most
important of which is NF-κB (NF kappa B), which is upregulated
when T-cells become activated.[84] This means that those cells
most likely to be killed by HIV are those currently fighting
infection.
During viral replication, the integrated DNA provirus is
transcribed into mRNA, which is then spliced into smaller
pieces. These small pieces are exported from the nucleus into
the cytoplasm, where they are translated into the regulatory
proteins Tat (which encourages new virus production) and Rev.
As the newly produced Rev protein accumulates in the nucleus,
it binds to viral mRNAs and allows unspliced RNAs to leave the
Reverse transcription of the HIV genome into
nucleus, where they are otherwise retained until spliced.[85] At
double strand DNA
this stage, the structural proteins Gag and Env are produced
from the full-length mRNA. The full-length RNA is actually the virus genome; it binds to the Gag protein and
is packaged into new virus particles.
HIV-1 and HIV-2 appear to package their RNA differently; HIV-1 will bind to any appropriate RNA, whereas
HIV-2 will preferentially bind to the mRNA that was used to create the Gag protein itself. This may mean that
HIV-1 is better able to mutate (HIV-1 infection progresses to AIDS faster than HIV-2 infection and is
responsible for the majority of global infections).
Assembly and release
The final step of the viral cycle, assembly of new HIV-1 virons, begins at the plasma membrane of the host
cell. The Env polyprotein (gp160) goes through the endoplasmic reticulum and is transported to the Golgi
complex where it is cleaved by protease and processed into the two HIV envelope glycoproteins gp41 and
gp120. These are transported to the plasma membrane of the host cell where gp41 anchors the gp120 to the
membrane of the infected cell. The Gag (p55) and Gag-Pol (p160) polyproteins also associate with the inner
surface of the plasma membrane along with the HIV genomic RNA as the forming virion begins to bud from
the host cell. Maturation either occurs in the forming bud or in the immature virion after it buds from the host
cell. During maturation, HIV proteases cleave the polyproteins into individual functional HIV proteins and
enzymes. The various structural components then assemble to produce a mature HIV virion. [86] This cleavage
step can be inhibited by protease inhibitors. The mature virus is then able to infect another cell.
Genetic variability
Further information: Subtypes of HIV
HIV differs from many viruses in that it has very high genetic
variability. This diversity is a result of its fast replication cycle,
with the generation of 10 9 to 10 10 virions every day, coupled
with a high mutation rate of approximately 3 x 10 −5 per
nucleotide base per cycle of replication and recombinogenic
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
properties of reverse transcriptase. [87][88]
This complex scenario leads to the generation of many variants
of HIV in a single infected patient in the course of one day. [87]
This variability is compounded when a single cell is
simultaneously infected by two or more different strains of HIV.
When simultaneous infection occurs, the genome of progeny
virions may be composed of RNA strands from two different
strains. This hybrid virion then infects a new cell where it
The phylogenetic tree of the SIV and HIV.
undergoes replication. As this happens, the reverse
transcriptase, by jumping back and forth between the two
different RNA templates, will generate a newly synthesized retroviral DNA sequence that is a recombinant
between the two parental genomes.[87] This recombination is most obvious when it occurs between
subtypes. [87]
The closely related simian immunodeficiency virus (SIV) has evolved into many strains, classified by the
natural host species. SIV strains of the African green monkey (SIVagm) and sooty mangabey (SIVsmm) are
thought to have a long evolutionary history with their hosts. These hosts have adapted to the presence of the
virus, [89] which is present at high levels in the host's blood but evokes only a mild immune response, [90] does
not cause the development of simian AIDS, [91] and does not undergo the extensive mutation and
recombination typical of HIV infection in humans.[92] In contrast, when these strains infect species that have
not adapted to SIV ("heterologous" hosts such as rhesus or cynomologus macaques), the animals develop
AIDS and the virus generates genetic diversity similar to what is seen in human HIV infection.[93] Chimpanzee
SIV (SIVcpz), the closest genetic relative of HIV-1, is associated with increased mortality and AIDS-like
symptoms in its natural host.[94] Both SIVcpz and HIV-1 appear to have been transmitted relatively recently
to chimpanzee and human populations, so their hosts have not yet adapted to the virus. [89] Both viruses have
also lost a function of the Nef gene that is present in most SIVs; without this function, T cell depletion is more
likely, leading to immunodeficiency.[94]
Three groups of HIV-1 have been identified on the basis of differences in the envelope ( env ) region: M, N,
and O. [95] Group M is the most prevalent and is subdivided into eight subtypes (or clades), based on the
whole genome, which are geographically distinct. [96] The most prevalent are subtypes B (found mainly in
North America and Europe), A and D (found mainly in Africa), and C (found mainly in Africa and Asia); these
subtypes form branches in the phylogenetic tree representing the lineage of the M group of HIV-1. Coinfection
with distinct subtypes gives rise to circulating recombinant forms (CRFs). In 2000, the last year in which an
analysis of global subtype prevalence was made, 47.2% of infections worldwide were of subtype C, 26.7%
were of subtype A/CRF02_AG, 12.3% were of subtype B, 5.3% were of subtype D, 3.2% were of CRF_AE,
and the remaining 5.3% were composed of other subtypes and CRFs.[97] Most HIV-1 research is focused on
subtype B; few laboratories focus on the other subtypes. [98] The existence of a fourth group, "P", has been
hypothesised based on a virus isolated in 2009.[99][100][101] The strain is apparently derived from gorilla SIV
(SIVgor), first isolated from western lowland gorillas in 2006.[99]
The genetic sequence of HIV-2 is only partially homologous to HIV-1 and more closely resembles that of
SIVsmm.
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
Diagnosis
Main article: HIV test
Many HIV-positive people are unaware that they are infected with the virus. [102] For example, less than 1%
of the sexually active urban population in Africa have been tested and this proportion is even lower in rural
populations.[102] Furthermore, only 0.5% of pregnant women attending urban health facilities are counselled,
tested or receive their test results. [102] Again, this proportion is even lower in rural health facilities. [102] Since
donors may therefore be unaware of their infection, donor blood and blood products used in medicine and
medical research are routinely screened for HIV. [103]
HIV-1 testing consists of initial screening with an enzyme-linked immunosorbent assay (ELISA) to detect
antibodies to HIV-1. Specimens with a nonreactive result from the initial ELISA are considered HIV-negative
unless new exposure to an infected partner or partner of unknown HIV status has occurred. Specimens with
a reactive ELISA result are retested in duplicate. [104] If the result of either duplicate test is reactive, the
specimen is reported as repeatedly reactive and undergoes confirmatory testing with a more specific
supplemental test (e.g., Western blot or, less commonly, an immunofluorescence assay (IFA)). Only
specimens that are repeatedly reactive by ELISA and positive by IFA or reactive by Western blot are
considered HIV-positive and indicative of HIV infection. Specimens that are repeatedly ELISA-reactive
occasionally provide an indeterminate Western blot result, which may be either an incomplete antibody
response to HIV in an infected person, or nonspecific reactions in an uninfected person.[105] Although IFA can
be used to confirm infection in these ambiguous cases, this assay is not widely used. Generally, a second
specimen should be collected more than a month later and retested for persons with indeterminate Western
blot results. Although much less commonly available, nucleic acid testing (e.g., viral RNA or proviral DNA
amplification method) can also help diagnosis in certain situations. [104] In addition, a few tested specimens
might provide inconclusive results because of a low quantity specimen. In these situations, a second
specimen is collected and tested for HIV infection.
Modern HIV testing is extremely accurate. The chance of a false-positive result in the two-step testing
protocol is estimated to be 0.0004% to 0.0007% in the general U.S. population.[106][107][108][109]
Treatment
See also Antiretroviral drug
There is currently no publicly available vaccine or cure for HIV or AIDS. [110][111]
However, a vaccine that is a combination of two previously unsuccessful vaccine
candidates was reported in September 2009 to have resulted in a 30%
reduction in infections in a trial conducted in Thailand.[112] Additionally, a course
of antiretroviral treatment administered immediately after exposure, referred to
as post-exposure prophylaxis, is believed to reduce the risk of infection if begun
as quickly as possible. [113] However, due to the incomplete protection provided
by the vaccine and/or post-exposure prophylaxis, the avoidance of exposure to
the virus is expected to remain the only reliable way to escape infection for
some time yet. Current treatment for HIV infection consists of highly active
antiretroviral therapy, or HAART. [114] This has been highly beneficial to many
HIV-infected individuals since its introduction in 1996, when the protease
inhibitor-based HAART initially became available. [115] Current HAART options are
combinations (or "cocktails") consisting of at least three drugs belonging to at
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
Abacavir - a nucleoside
analog reverse transcriptase
inhibitors (NARTIs or
NRTIs)
HIV - Wikipedia, the free encyclopedia
least two types, or "classes," of antiretroviral agents. Typically, these classes are
two nucleoside analogue reverse transcriptase inhibitors (NARTIs or NRTIs) plus either a protease inhibitor or
a non-nucleoside reverse transcriptase inhibitor (NNRTI). New classes of drugs such as entry inhibitors
provide treatment options for patients who are infected with viruses already resistant to common therapies,
although they are not widely available and not typically accessible in resource-limited settings. Because AIDS
progression in children is more rapid and less predictable than in adults, particularly in young infants, more
aggressive treatment is recommended for children than adults. [116] In developed countries where HAART is
available, doctors assess their patients thoroughly: measuring the viral load, how fast CD4 declines, and
patient readiness. They then decide when to recommend starting treatment. [117]
HAART neither cures the patient nor does it uniformly remove all symptoms; high levels of HIV-1, often
HAART resistant, return if treatment is stopped.[118][119] Moreover, it would take more than a lifetime for HIV
infection to be cleared using HAART. [120] Despite this, many HIV-infected individuals have experienced
remarkable improvements in their general health and quality of life, which has led to a large reduction in HIVassociated morbidity and mortality in the developed world.[115][121][122] One study suggests the average life
expectancy of an HIV infected individual is 32 years from the time of infection if treatment is started when the
CD4 count is 350/µL. [123] In the absence of HAART, progression from HIV infection to AIDS has been
observed to occur at a median of between nine to ten years and the median survival time after developing
AIDS is only 9.2 months. [9] However, HAART sometimes achieves far less than optimal results, in some
circumstances being effective in less than fifty percent of patients. This is due to a variety of reasons such as
medication intolerance/side effects, prior ineffective antiretroviral therapy and infection with a drug-resistant
strain of HIV. However, non-adherence and non-persistence with antiretroviral therapy is the major reason
most individuals fail to benefit from HAART. [124] The reasons for non-adherence and non-persistence with
HAART are varied and overlapping. Major psychosocial issues, such as poor access to medical care,
inadequate social supports, psychiatric disease and drug abuse contribute to non-adherence. The complexity
of these HAART regimens, whether due to pill number, dosing frequency, meal restrictions or other issues
along with side effects that create intentional non-adherence also contribute to this problem. [125][126][127]
The side effects include lipodystrophy, dyslipidemia, insulin resistance, an increase in cardiovascular risks, and
birth defects.[128][129]
The timing for starting HIV treatment is still debated. There is no question that treatment should be started
before the patient's CD4 count falls below 200, and most national guidelines say to start treatment once the
CD4 count falls below 350; but there is some evidence from cohort studies that treatment should be started
before the CD4 count falls below 350. [121][130] In those countries where CD4 counts are not available,
patients with WHO stage III or IV disease [131] should be offered treatment.
Anti-retroviral drugs are expensive, and the majority of the world's infected individuals do not have access to
medications and treatments for HIV and AIDS. [132] Research to improve current treatments includes
decreasing side effects of current drugs, further simplifying drug regimens to improve adherence, and
determining the best sequence of regimens to manage drug resistance. Unfortunately, only a vaccine is
thought to be able to halt the pandemic. This is because a vaccine would cost less, thus being affordable for
developing countries, and would not require daily treatment. [132] However, after over 20 years of research,
HIV-1 remains a difficult target for a vaccine.[132]
Treatments in development
Media reports in 2008 and a publication in the New England Journal of Medicine in 2009 described the
anecdotal case of an HIV-positive patient of a Berlin doctor, Gero Hütter. The patient, who had both acute
myelogenous leukemia (AML) and HIV infection, was said by some to be "functionally cured" of his HIV
following a bone marrow transplant for AML. The bone marrow donor had been selected as homozygous for a
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
CCR5-Δ32 mutation (which confers resistance to "almost all strains of HIV").[133][134] After 600 days without
antiretroviral drug treatment, HIV levels in the patient's blood, bone marrow and bowel were below the limit
of detection, although the authors note that the virus is likely present in other tissues. Researchers cautioned
that it would be premature to consider this treatment a possible cure because of its anecdotal nature, the
mortality risk associated with bone marrow transplants and other concerns. [135][136]
HIV latent reservoir
Despite the success of highly active antiretroviral therapy (HAART) in controlling HIV infection and reducing
HIV-associated mortality, current drug regimens are unable to completely eradicate HIV infection. Many
people on HAART achieve suppression of HIV to levels below the limit of detection of standard clinical assays
for many years. However, upon withdrawal of HAART, HIV viral loads rebound quickly with a concomitant
decline in CD4+ T-Cells, which, in most cases, absent a resumption of treatment, leads to AIDS.
To successfully reproduce itself, HIV must convert its RNA genome to DNA, which is then imported into the
host cell's nucleus and inserted into the host genome through the action of HIV integrase. Because HIV's
primary cellular target, CD4+ T-Cells, function as the memory cells of the immune system, integrated HIV can
remain dormant for the duration of these cell's lifetime. Memory T-Cells may survive for many years and
possibly for decades. The latent HIV reservoir can be measured by co-culturing CD4+ T-Cells from infected
patients with CD4+ T-Cells from uninfected donors and measuring HIV protein or RNA. [137]
The failure of vaccine candidates to protect against HIV infection and progression to AIDS has led to a
renewed focus on the biological mechanisms responsible for HIV latency. A limited period of therapy
combining anti-retrovirals with drugs targeting the latent reservoir may one day allow for total eradication of
HIV infection.[138]
Prognosis
Without treatment, the net median survival time after infection with HIV is estimated to be 9 to 11 years,
depending on the HIV subtype, [139] and the median survival rate after diagnosis of AIDS in resource-limited
settings where treatment is not available ranges between 6 and 19 months, depending on the study. [140] In
areas where it is widely available, the development of HAART as effective therapy for HIV infection and AIDS
reduced the death rate from this disease by 80%, and raised the life expectancy for a newly diagnosed HIVinfected person to 20–50 years. [141][142]
As new treatments continue to be developed and because HIV continues to evolve resistance to treatments,
estimates of survival time are likely to continue to change. Without antiretroviral therapy, death normally
occurs within a year after the individual progresses to AIDS. [9] Most patients die from opportunistic infections
or malignancies associated with the progressive failure of the immune system. [5] The rate of clinical disease
progression varies widely between individuals and has been shown to be affected by many factors such as
host susceptibility and immune function[66][143][144] health care and co-infections, [5][9] as well as which
particular strain of the virus is involved.[145][146][147]
Epidemiology
Main article: AIDS pandemic
UNAIDS and the WHO estimate that AIDS has killed more than 25 million people
since it was first recognized in 1981, making it one of the most destructive
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
pandemics in recorded history. Despite recent improved access to antiretroviral
treatment and care in many regions of the world, the AIDS pandemic claimed an
estimated 2.8 million (between 2.4 and 3.3 million) lives in 2005 of which more
than half a million (570,000) were children. [1]
In 2007, between 30.6 and 36.1 million people were believed to live with HIV,
and it killed an estimated 2.1 million people that year, including 330,000
children; there were 2.5 million new infections. [139]
Sub-Saharan Africa remains by far the worst-affected region, with an estimated
21.6 to 27.4 million people currently living with HIV. Two million [1.5–
3.0 million] of them are children younger than 15 years of age. More than 64%
of all people living with HIV are in sub-Saharan Africa, as are more than three
quarters of all women living with HIV. In 2005, there were 12.0 million [10.6–
13.6 million] AIDS orphans living in sub-Saharan Africa 2005.[1]
South & South East Asia are second-worst affected with 15% of the total. AIDS
accounts for the deaths of 500,000 children in this region. South Africa has the
largest number of HIV patients in the world followed by Nigeria.[148] India has
an estimated 2.5 million infections (0.23% of population), making India the
country with the third largest population of HIV patients. In the 35 African
nations with the highest prevalence, average life expectancy is 48.3 years—6.5
years less than it would be without the disease.[149]
Estimated prevalence of
HIV among young adults
(15-49) per country at the
end of 2005.
Disability-adjusted life year
for HIV and AIDS per
100,000 inhabitants.
no data
≤ 10
10-25
25-50
50-100
100-500
500-1000
1000-2500
2500-5000
5000-7500
7500-10000
10000-50000
≥ 50000
The latest evaluation report of the World Bank's Operations Evaluation
Department assesses the development effectiveness of the World Bank's
country-level HIV/AIDS assistance defined as policy dialogue, analytic work, and lending with the explicit
objective of reducing the scope or impact of the AIDS epidemic. [150] This is the first comprehensive
evaluation of the World Bank's HIV/AIDS support to countries, from the beginning of the epidemic through
mid-2004. Because the Bank aims to assist in implementation of national government programmes, their
experience provides important insights on how national AIDS programmes can be made more effective.
The development of HAART as effective therapy for HIV infection has substantially reduced the death rate
from this disease in those areas where these drugs are widely available. [115] As the life expectancy of persons
with HIV has increased in countries where HAART is widely used, the continuing spread of the disease has
caused the number of persons living with HIV to increase substantially.
In Africa, the number of MTCT and the prevalence of AIDS is beginning to reverse decades of steady progress
in child survival. Countries such as Uganda are attempting to curb the MTCT epidemic by offering VCT
(voluntary counselling and testing), PMTCT (prevention of mother-to-child transmission) and ANC (ante-natal
care) services, which include the distribution of antiretroviral therapy.
History
Origins
Main article: Origin of AIDS
See History of known cases and spread for early cases of HIV / AIDS
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
HIV is thought to have originated in non-human primates in sub-Saharan Africa and was transferred to
humans late in the 19th or early in the 20th century.[151] The first paper recognizing a pattern of
opportunistic infections characteristic of AIDS was published in 1981.[152]
Both HIV-1 and HIV-2 are believed to have originated in West-Central Africa and to have jumped species (a
process known as zoonosis) from non-human primates to humans. HIV-1 appears to have originated in
southern Cameroon through the evolution of SIV(cpz), a simian immunodeficiency virus (SIV) that infects wild
chimpanzees ( Pan troglodytes troglodytes).[153][154] The closest relative of HIV-2 is SIV(agm), a virus of the
sooty mangabey ( Cercocebus atys), an Old World monkey of Guinea-Bissau, Gabon, and Cameroon.[15] New
World monkeys such as the owl monkey are resistant to HIV-1 infection, possibly because of a genomic fusion
of two viral resistance genes.[155]
Discovery
AIDS was first clinically observed between late 1980 and early 1981. A group of five men showed symptoms
of Pneumocystis carinii pneumonia (PCP), a rare oppourtinistic infection that was known to present itself in
people with very compromised immune systems. Soon thereafter, another set of men developed a rare skin
cancer called Kaposi’s sarcoma (KP). Many more cases of PCP and KP quickly emerged, alerting U.S. Centers
for Disease Control and Prevention (CDC). A CDC task force was formed to monitored the outbreak. After
recognizing a pattern of anomalous symptoms presenting themselves in patients, the task force named the
condition acquired immune deficiency syndrome (AIDS). [156]
In 1983, two separate research groups lead by Robert Gallo and Luc Montagnier independently declared that
a novel retrovirus may have been infecting AIDS patients, and published their findings in the same issue of
the journal Science .[157][158] Gallo claimed that a virus his group had isolated from an AIDS patient was
strikingly similar in shape to other human T-lymphotropic viruses (HLTVs) his group had been the first to
isolate. Gallo's group called their newly isolated virus HLTV-III. At the same time, Montagnier's group isolated
a virus from a patient presenting lymphadenopathy (swelling of the lymph nodes) of the neck and physical
weakness, two classic symptoms of AIDS. Contradicting the report from Gallo's group, Montagnier and his
colleagues showed that core proteins of this virus were immunologically different from those of HTLV-I.
Montagnier's group named their isolated virus lymphadenopathy-associated virus (LAV). [156]
Whether Gallo or Montagnier deserve more credit for the discovery of the virus that causes AIDS has been a
matter of considerable controversy. Together with his colleague Françoise Barré-Sinoussi, Montagnier was
awarded one half of the 2008 Nobel Prize in Physiology or Medicine for his "discovery of human
immunodeficiency virus".[159] Harald zur Hausen also shared the Prize for his discovery that human papilloma
virus leads to cervical cancer, but Gallo was left out.[160] Gallo said that it was "a disappointment" that he
was not named a co-recipient. [161] Montagnier said he was "surprised" Gallo was not recognized by the Nobel
Committee: "It was important to prove that HIV was the cause of AIDS, and Gallo had a very important role
in that. I'm very sorry for Robert Gallo." [160]
AIDS denialism
Main article: AIDS denialism
Some individuals, including some scientists who are not recognized experts on HIV, [162] question the
connection between HIV and AIDS, [163] the existence of HIV itself, or the validity of HIV testing and
treatment methods.[162][164] These claims, known as AIDS denialism, have been examined and rejected by
the worldwide scientific community,[165] although they have had a political impact, particularly in South Africa,
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
where the government's official promotion of AIDS denialism was responsible for its ineffective response to
that country's AIDS epidemic. [166][167][168]
See also
Higher Ground (support group) for HIV-AIDS infected persons
HIV blood screening
HIV disease-related drug reaction
HIV Prevention Act of 1997 (in the US)
HIV-associated pruritus
Photosensitivity with HIV infection
Discovery and development of CCR5 receptor antagonists
References
1. ^ a b c d Joint United Nations Programme on HIV/AIDS (2006). "Overview of the global AIDS epidemic"
(http://data.unaids.org/pub/GlobalReport/2006/2006_GR_CH02_en.pdf) (PDF). 2006 Report on the global AIDS
epidemic. Retrieved 2006-06-08.
2. ^ Greener, R. (2002). "AIDS and macroeconomic impact" (http://db.jhuccp.org/ics-wpd/exec/icswppro.dll?
BU=http://db.jhuccp.org/icswpd/exec/icswppro.dll&QF0=DocNo&QI0=285428&TN=Popline&AC=QBE_QUERY&MR=30%25DL=1&&RL=1&&RF=Lon
gRecordDisplay&DF=LongRecordDisplay) . in S, Forsyth (ed.). State of The Art: AIDS and Economics. IAEN. pp. 49–55.
3. ^ Joint United Nations Programme on HIV/AIDS. "AIDS epidemic update, 2005"
(http://www.unaids.org/epi/2005/doc/EPIupdate2005_pdf_en/epi-update2005_en.pdf) (PDF). Retrieved 2006-02-28.
4. ^ Palella, F. J. Jr, Delaney, K. M., Moorman, A. C., Loveless, M. O., Fuhrer, J., Satten, G. A., Aschman and D. J.,
Holmberg, S. D. (1998). "Declining morbidity and mortality among patients with advanced human immunodeficiency
virus infection. HIV Outpatient Study Investigators". N. Engl. J. Med 338 (13): 853–860.
doi:10.1056/NEJM199803263381301 (http://dx.doi.org/10.1056%2FNEJM199803263381301) . PMID 9516219
(http://www.ncbi.nlm.nih.gov/pubmed/9516219) .
5. ^ a b c Lawn SD (2004). "AIDS in Africa: the impact of coinfections on the pathogenesis of HIV-1 infection". J. Infect.
Dis. 48 (1): 1–12. doi:10.1016/j.jinf.2003.09.001 (http://dx.doi.org/10.1016%2Fj.jinf.2003.09.001) . PMID 14667787
(http://www.ncbi.nlm.nih.gov/pubmed/14667787) .
6. ^ Buchbinder SP, Katz MH, Hessol NA, O'Malley PM, Holmberg SD. (1994). "Long-term HIV-1 infection without
immunologic progression". AIDS 8 (8): 1123–8. doi:10.1097/00002030-199408000-00014
(http://dx.doi.org/10.1097%2F00002030-199408000-00014) . PMID 7986410
(http://www.ncbi.nlm.nih.gov/pubmed/7986410) .
7. ^ "Time from HIV-1 seroconversion to AIDS and death before widespread use of highly-active antiretroviral therapy: a
collaborative re-analysis. Collaborative Group on AIDS Incubation and HIV Survival including the CASCADE EU Concerted
Action. Concerted Action on SeroConversion to AIDS and Death in Europe". Lancet 355 (9210): 1131–7. April 2000.
doi:10.1016/S0140-6736(00)02061-4 (http://dx.doi.org/10.1016%2FS0140-6736%2800%2902061-4) . PMID 10791375
(http://www.ncbi.nlm.nih.gov/pubmed/10791375) .
8. ^ Schneider MF, Gange SJ, Williams CM, Anastos K, Greenblatt RM, Kingsley L, Detels R, Munoz A (2005). "Patterns of
the hazard of death after AIDS through the evolution of antiretroviral therapy: 1984–2004". AIDS 19 (17): 2009–18.
PMID 16260908 (http://www.ncbi.nlm.nih.gov/pubmed/16260908) .
9. ^ a b c d Morgan D, Mahe C, Mayanja B, Okongo JM, Lubega R, Whitworth JA (2002). "HIV-1 infection in rural Africa: is
there a difference in median time to AIDS and survival compared with that in industrialized countries?". AIDS 16 (4):
597–632. PMID 11873003 (http://www.ncbi.nlm.nih.gov/pubmed/11873003) .
10. ^ International Committee on Taxonomy of Viruses. "61.0.6. Lentivirus"
(http://www.ncbi.nlm.nih.gov/ICTVdb/ICTVdB/61060000.htm) . National Institutes of Health. Retrieved 2006-02-28.
11. ^ International Committee on Taxonomy of Viruses. "61. Retroviridae"
(http://www.ncbi.nlm.nih.gov/ICTVdb/ICTVdB/61000000.htm) . National Institutes of Health. Retrieved 2006-02-28.
12. ^ Lévy, J. A. (1993). "HIV pathogenesis and long-term survival". AIDS 7 (11): 1401–10. doi:10.1097/00002030199311000-00001 (http://dx.doi.org/10.1097%2F00002030-199311000-00001) . PMID 8280406
(http://www.ncbi.nlm.nih.gov/pubmed/8280406) .
13. ^ Smith, Johanna A.; Daniel, René (Division of Infectious Diseases, Center for Human Virology, Thomas Jefferson
University, Philadelphia) (2006). "Following the path of the virus: the exploitation of host DNA repair mechanisms by
retroviruses". ACS Chem Biol 1 (4): 217–26. doi:10.1021/cb600131q (http://dx.doi.org/10.1021%2Fcb600131q) . PMID
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
17163676 (http://www.ncbi.nlm.nih.gov/pubmed/17163676) .
14. ^ Gilbert, PB et al (28 February 2003). "Comparison of HIV-1 and HIV-2 infectivity from a prospective cohort study in
Senegal". Statistics in Medicine 22 (4): 573–593. PMID 12590415 (http://www.ncbi.nlm.nih.gov/pubmed/12590415) .
15. ^ a b Reeves, J. D. and Doms, R. W (2002). "Human Immunodeficiency Virus Type 2". J. Gen. Virol. 83 (Pt 6): 1253–
65. PMID 12029140 (http://www.ncbi.nlm.nih.gov/pubmed/12029140) .
16. ^ Grabar, S., Selinger-Leneman, H., Abgrall, S., Pialoux, G., Weiss, L., Costagliola, D. (2009). "Prevalence and
comparative characteristics of long-term nonprogressors and HIV controller patients in the French Hospital Database on
HIV". AIDS 23 (9): 1163–1169. doi:10.1097/QAD.0b013e32832b44c8
(http://dx.doi.org/10.1097%2FQAD.0b013e32832b44c8) . PMID 19444075
(http://www.ncbi.nlm.nih.gov/pubmed/19444075) .
17. ^ Piatak, M., Jr, Saag, M. S., Yang, L. C., Clark, S. J., Kappes, J. C., Luk, K. C., Hahn, B. H., Shaw, G. M. and Lifson,
J.D. (1993). "High levels of HIV-1 in plasma during all stages of infection determined by competitive PCR". Science 259
(5102): 1749–1754. doi:10.1126/science.8096089 (http://dx.doi.org/10.1126%2Fscience.8096089) . PMID 8096089
(http://www.ncbi.nlm.nih.gov/pubmed/8096089) .
18. ^ Pantaleo G, Demarest JF, Schacker T, Vaccarezza M, Cohen OJ, Daucher M, Graziosi C, Schnittman SS, Quinn TC,
Shaw GM, Perrin L, Tambussi G, Lazzarin A, Sekaly RP, Soudeyns H, Corey L, Fauci AS. (1997). "The qualitative nature
of the primary immune response to HIV infection is a prognosticator of disease progression independent of the initial
level of plasma viremia". Proc Natl Acad Sci U S A. 94 (1): 254–258. doi:10.1073/pnas.94.1.254
(http://dx.doi.org/10.1073%2Fpnas.94.1.254) . PMID 8990195 (http://www.ncbi.nlm.nih.gov/pubmed/8990195) .
19. ^ Kahn, J. O. and Walker, B. D. (1998). "Acute Human Immunodeficiency Virus type 1 infection". N. Engl. J. Med. 331
(1): 33–39. doi:10.1056/NEJM199807023390107 (http://dx.doi.org/10.1056%2FNEJM199807023390107) . PMID
9647878 (http://www.ncbi.nlm.nih.gov/pubmed/9647878) .
20. ^ Daar ES, Little S, Pitt J, et al. (2001). "Diagnosis of primary HIV-1 infection. Los Angeles County Primary HIV
Infection Recruitment Network". Ann. Intern. Med. 134 (1): 25–9. PMID 11187417
(http://www.ncbi.nlm.nih.gov/pubmed/11187417) .
21. ^ Burton GF, Keele BF, Estes JD, Thacker TC, Gartner S. (2002). "Follicular dendritic cell contributions to HIV
pathogenesis". Semin Immunol. 14 (4): 275–284. doi:10.1016/S1044-5323(02)00060-X
(http://dx.doi.org/10.1016%2FS1044-5323%2802%2900060-X) . PMID 12163303
(http://www.ncbi.nlm.nih.gov/pubmed/12163303) .
22. ^ Clapham PR, McKnight A. (2001). "HIV-1 receptors and cell tropism". Br Med Bull. 58 (4): 43–59.
doi:10.1093/bmb/58.1.43 (http://dx.doi.org/10.1093%2Fbmb%2F58.1.43) . PMID 11714623
(http://www.ncbi.nlm.nih.gov/pubmed/11714623) .
23. ^ Smith DK, Grohskopf LA, Black RJ, et al. (January 2005). "Antiretroviral postexposure prophylaxis after sexual,
injection-drug use, or other nonoccupational exposure to HIV in the United States: recommendations from the U.S.
Department of Health and Human Services" (http://www.cdc.gov/mmwr/preview/mmwrhtml/rr5402a1.htm#tab1) .
MMWR Recomm Rep 54 (RR-2): 1–20. PMID 15660015 (http://www.ncbi.nlm.nih.gov/pubmed/15660015) . Retrieved
2009-03-31.
24. ^ Donegan E, Stuart M, Niland JC, et al. (1990). "Infection with human immunodeficiency virus type 1 (HIV-1) among
recipients of antibody-positive blood donations". Ann. Intern. Med. 113 (10): 733–739. PMID 2240875
(http://www.ncbi.nlm.nih.gov/pubmed/2240875) .
25. ^ a b c Coovadia H (2004). "Antiretroviral agents—how best to protect infants from HIV and save their mothers from
AIDS". N. Engl. J. Med. 351 (3): 289–292. doi:10.1056/NEJMe048128 (http://dx.doi.org/10.1056%2FNEJMe048128) .
PMID 15247337 (http://www.ncbi.nlm.nih.gov/pubmed/15247337) .
26. ^ Kaplan EH, Heimer R (1995). "HIV incidence among New Haven needle exchange participants: updated estimates
from syringe tracking and testing data". J. Acquir. Immune Defic. Syndr. Hum. Retrovirol. 10 (2): 175–176. PMID
7552482 (http://www.ncbi.nlm.nih.gov/pubmed/7552482) .
27. ^ Bell DM (1997). "Occupational risk of human immunodeficiency virus infection in healthcare workers: an overview.".
Am. J. Med. 102 (5B): 9–15. PMID 9845490 (http://www.ncbi.nlm.nih.gov/pubmed/9845490) .
28. ^ a b c d European Study Group on Heterosexual Transmission of HIV (1992). "Comparison of female to male and male
to female transmission of HIV in 563 stable couples" (http://www.pubmedcentral.nih.gov/articlerender.fcgi?
tool=pmcentrez&artid=1881672) . BMJ. 304 (6830): 809–813. PMID 1392708
(http://www.ncbi.nlm.nih.gov/pubmed/1392708) .
29. ^ a b c d e f Varghese B, Maher JE, Peterman TA, Branson BM,Steketee RW (2002). "Reducing the risk of sexual HIV
transmission: quantifying the per-act risk for HIV on the basis of choice of partner, sex act, and condom use". Sex.
Transm. Dis. 29 (1): 38–43. PMID 11773877 (http://www.ncbi.nlm.nih.gov/pubmed/11773877) .
30. ^ Leynaert B, Downs AM, de Vincenzi I (1998). "Heterosexual transmission of human immunodeficiency virus:
variability of infectivity throughout the course of infection. European Study Group on Heterosexual Transmission of HIV".
Am. J. Epidemiol. 148 (1): 88–96. PMID 9663408 (http://www.ncbi.nlm.nih.gov/pubmed/9663408) .
31. ^ Boily M-C, Baggaley RF, Wang L, et al. (2009). "Heterosexual risk of HIV-1 infection per sexual act: systematic
review and meta-analysis of observational studies". Lancet Infect Dis 9 (2): 118–129. doi:10.1016/S1473file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
3099(09)70021-0 (http://dx.doi.org/10.1016%2FS1473-3099%2809%2970021-0) . PMID 19179227
(http://www.ncbi.nlm.nih.gov/pubmed/19179227) .
^ National Institute of Allergy and Infectious Diseases; National Institutes of Health, Department of Health and Human
Services (2001-07-20). "Workshop Summary: Scientific Evidence on Condom Effectiveness for Sexually Transmitted
Disease (STD) Prevention" (http://www3.niaid.nih.gov/about/organization/dmid/PDF/condomReport.pdf) (PDF). Hyatt
Dulles Airport, Herndon, Virginia. pp. 13–15. Retrieved 2009-01-08.
^ "Should spermicides be used with condoms?"
(http://www.fda.gov/ForConsumers/byAudience/ForPatientAdvocates/HIVandAIDSActivities/ucm126372.htm) . Condoms
and Sexually Transmitted Diseases, Brochure . United States Food and Drug Administration. 2009-04-30. Retrieved
2009-07-23.
^ Global Campaign for Microbicides : Rectal Use of N-9 (http://www.global-campaign.org/rectalN9.htm#rectal) checked
2009-07-22
^ Nonoxynol-9 Spermicide on HIV Risk List (http://www.global-campaign.org/clientfiles/GFN.pdf) checked 2009-07-22
^ Weiss, H.A.; M.A. Quigley, R.J. Hayes (2000). "Male circumcision and risk of HIV infection in sub-Saharan Africa: A
systematic review and meta-analysis". AIDS 14 (15): 2361–70. doi:10.1097/00002030-200010200-00018
(http://dx.doi.org/10.1097%2F00002030-200010200-00018) . PMID 11089625
(http://www.ncbi.nlm.nih.gov/pubmed/11089625) .
^ Siegfried, N., Muller, M., Deeks, J., Volmink, J., Egger, M., Low, N., Walker, S. and Williamson, P. (2005). "HIV and
male circumcision--a systematic review with assessment of the quality of studies". Lancet Infect. Dis. 5 (3): 165–73.
PMID 15766651 (http://www.ncbi.nlm.nih.gov/pubmed/15766651) .
^ Various (2005). "Repeated Use of Unsterilized Blades in Ritual Circumcision Might Contribute to HIV Spread in S.
Africa, Doctors Say" (http://www.kaisernetwork.org/daily_reports/rep_index.cfm?DR_ID=31199) . Kaisernetwork.org.
Retrieved 2006-03-28.
^ Williams BG, Lloyd-Smith JO, Gouws E, Hankins C, Getz WM, Hargrove J, de Zoysa I, Dye C, Auvert B. (2006). "The
Potential Impact of Male Circumcision on HIV in Sub-Saharan Africa.". PLoS Med 3 (7): e262.
doi:10.1371/journal.pmed.0030262 (http://dx.doi.org/10.1371%2Fjournal.pmed.0030262) . PMID 16822094
(http://www.ncbi.nlm.nih.gov/pubmed/16822094) .
^ Bailey RC, Moses S, Parker CB, et al. (2007). "Male circumcision for HIV prevention in young men in Kisumu, Kenya:
a randomised controlled trial". Lancet 369 (9562): 643–56. doi:10.1016/S0140-6736(07)60312-2
(http://dx.doi.org/10.1016%2FS0140-6736%2807%2960312-2) . PMID 17321310
(http://www.ncbi.nlm.nih.gov/pubmed/17321310) .
^ Gray RH et al. (February 24, 2007). "Male circumcision for HIV prevention in men in Rakai, Uganda: a randomised
trial". Lancet 369 (9562): 657–66. doi:10.1016/S0140-6736(07)60313-4 (http://dx.doi.org/10.1016%2FS01406736%2807%2960313-4) . PMID 17321311 (http://www.ncbi.nlm.nih.gov/pubmed/17321311) .
^ WHO (2007). "WHO and UNAIDS announce recommendations from expert consultation on male circumcision for HIV
prevention" (http://www.who.int/hiv/mediacentre/news68/en/index.html) . WHO.int. Retrieved 2007-07-13.
^ Lifson AR (1988). "Do alternate modes for transmission of human immunodeficiency virus exist? A review". JAMA
259 (9): 1353–6. doi:10.1001/jama.259.9.1353 (inactive 2009-08-10). PMID 2963151
(http://www.ncbi.nlm.nih.gov/pubmed/2963151) .
^ http://www.rci.rutgers.edu/%7Einsects/aids.htm
^ Cochrane Systematic Review on interventions for prevention of late postnatal mother-to-child transmission of HIV
http://www.cochrane.org/reviews/en/ab006734.html
^ Smith D, Richman D, Little S (2005). "HIV Superinfection". Journal of Infectious Diseases 192: 438–44.
doi:10.1086/431682 (http://dx.doi.org/10.1086%2F431682) . PMID 15995957
(http://www.ncbi.nlm.nih.gov/pubmed/15995957) .
^ Gottlieb, et al. (2004). "Dual HIV-1 infection associated with rapid disease progression". Lancet 363 (9049): 619–22.
doi:10.1016/S0140-6736(04)15596-7 (http://dx.doi.org/10.1016%2FS0140-6736%2804%2915596-7) . PMID 14987889
(http://www.ncbi.nlm.nih.gov/pubmed/14987889) .
^ Smith et al. (2004). "Incidence of HIV superinfection following primary infection". JAMA 292 (10): 1177–8.
doi:10.1001/jama.292.10.1177 (http://dx.doi.org/10.1001%2Fjama.292.10.1177) . PMID 15353529
(http://www.ncbi.nlm.nih.gov/pubmed/15353529) .
^ Chohan B, Lavreys L, Rainwater SM, Overbaugh J (August 2005). "Evidence for frequent reinfection with human
immunodeficiency virus type 1 of a different subtype" (http://www.pubmedcentral.nih.gov/articlerender.fcgi?
tool=pmcentrez&artid=1182664) . J. Virol. 79 (16): 10701–8. doi:10.1128/JVI.79.16.10701-10708.2005
(http://dx.doi.org/10.1128%2FJVI.79.16.10701-10708.2005) . PMID 16051862
(http://www.ncbi.nlm.nih.gov/pubmed/16051862) .
^ Piantadosi A, Chohan B, Chohan V, McClelland RS, Overbaugh J (November 2007). "Chronic HIV-1 infection frequently
fails to protect against superinfection" (http://www.pubmedcentral.nih.gov/articlerender.fcgi?
tool=pmcentrez&artid=2077901) . PLoS Pathog. 3 (11): e177. doi:10.1371/journal.ppat.0030177
(http://dx.doi.org/10.1371%2Fjournal.ppat.0030177) . PMID 18020705
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
(http://www.ncbi.nlm.nih.gov/pubmed/18020705) .
51. ^ Hu DJ, Subbarao S, Vanichseni S, et al. (February 2005). "Frequency of HIV-1 dual subtype infections, including
intersubtype superinfections, among injection drug users in Bangkok, Thailand" (http://meta.wkhealth.com/pt/ptcore/template-journal/lwwgateway/media/landingpage.htm?an=00002030-200502180-00009) . AIDS 19 (3): 303–8.
PMID 15718841 (http://www.ncbi.nlm.nih.gov/pubmed/15718841) . Retrieved 2009-03-31.
52. ^ McGovern SL, Caselli E, Grigorieff N, Shoichet BK (2002). "A common mechanism underlying promiscuous inhibitors
from virtual and high-throughput screening". J Med Chem 45 (8): 1712–22. doi:10.1021/jm010533y
(http://dx.doi.org/10.1021%2Fjm010533y) . PMID 11931626 (http://www.ncbi.nlm.nih.gov/pubmed/11931626) .
53. ^ Compared with overview in: Fisher, Bruce; Harvey, Richard P.; Champe, Pamela C. (2007). Lippincott's Illustrated
Reviews: Microbiology (Lippincott's Illustrated Reviews Series) . Hagerstown, MD: Lippincott Williams & Wilkins. ISBN 07817-8215-5. Page 3
54. ^ a b c d e f g Various (2008) (PDF). HIV Sequence Compendium 2008 Introduction
(http://www.hiv.lanl.gov/content/sequence/HIV/COMPENDIUM/2008/frontmatter.pdf) . Retrieved 2009-03-31.
55. ^ a b c Chan, DC., Fass, D., Berger, JM., Kim, PS. (1997). "Core Structure of gp41 from the HIV Envelope Glycoprotein"
(http://www.its.caltech.edu/%7Echanlab/PDFs/Chan_Cell_1997.pdf) (PDF). Cell 89: 263–73. PMID 9108481
(http://www.ncbi.nlm.nih.gov/pubmed/9108481) . Retrieved 2009-03-31.
56. ^ National Institute of Health (June 17, 1998). "Crystal Structure of Key HIV Protein Reveals New Prevention,
Treatment Targets" (http://www3.niaid.nih.gov/news/newsreleases/1998/hivprotein.htm) . Retrieved 2006-09-14.
57. ^ Ouellet DL, Plante I, Landry P, et al. (April 2008). "Identification of functional microRNAs released through
asymmetrical processing of HIV-1 TAR element" (http://nar.oxfordjournals.org/cgi/content/full/36/7/2353) . Nucleic
Acids Res. 36 (7): 2353–65. doi:10.1093/nar/gkn076 (http://dx.doi.org/10.1093%2Fnar%2Fgkn076) . PMID 18299284
(http://www.ncbi.nlm.nih.gov/pubmed/18299284) . PMC 2367715
(http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2367715) .
58. ^ Klase Z, Winograd R, Davis J, et al. (2009). "HIV-1 TAR miRNA protects against apoptosis by altering cellular gene
expression" (http://www.retrovirology.com/content/6/1/18) . Retrovirology 6: 18. doi:10.1186/1742-4690-6-18
(http://dx.doi.org/10.1186%2F1742-4690-6-18) . PMID 19220914 (http://www.ncbi.nlm.nih.gov/pubmed/19220914) .
PMC 2654423 (http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2654423) .
59. ^ Garcia JV, Miller AD (April 1991). "Serine phosphorylation-independent downregulation of cell-surface CD4 by nef".
Nature 350 (6318): 508–11. doi:10.1038/350508a0 (http://dx.doi.org/10.1038%2F350508a0) . PMID 2014052
(http://www.ncbi.nlm.nih.gov/pubmed/2014052) .
60. ^ Schwartz O, Maréchal V, Le Gall S, Lemonnier F, Heard JM (March 1996). "Endocytosis of major histocompatibility
complex class I molecules is induced by the HIV-1 Nef protein". Nat. Med. 2 (3): 338–42. doi:10.1038/nm0396-338
(http://dx.doi.org/10.1038%2Fnm0396-338) . PMID 8612235 (http://www.ncbi.nlm.nih.gov/pubmed/8612235) .
61. ^ Stumptner-Cuvelette P, Morchoisne S, Dugast M, et al. (October 2001). "HIV-1 Nef impairs MHC class II antigen
presentation and surface expression" (http://www.pubmedcentral.nih.gov/articlerender.fcgi?
tool=pmcentrez&artid=59782) . Proc. Natl. Acad. Sci. U.S.A. 98 (21): 12144–9. doi:10.1073/pnas.221256498
(http://dx.doi.org/10.1073%2Fpnas.221256498) . PMID 11593029 (http://www.ncbi.nlm.nih.gov/pubmed/11593029) .
62. ^ a b c Coakley, E., Petropoulos, C. J. and Whitcomb, J. M. (2005). "Assessing ch vbgemokine co-receptor usage in
HIV". Curr. Opin. Infect. Dis. 18 (1): 9–15. PMID 15647694 (http://www.ncbi.nlm.nih.gov/pubmed/15647694) .
63. ^ Deng H, Liu R, Ellmeier W, Choe S, Unutmaz D, Burkhart M, Di Marzio P, Marmon S, Sutton RE, Hill CM, Davis CB,
Peiper SC, Schall TJ, Littman DR, Landau NR. (1996). "Identification of a major co-receptor for primary isolates of HIV1". Nature 381 (6584): 661–6. doi:10.1038/381661a0 (http://dx.doi.org/10.1038%2F381661a0) . PMID 8649511
(http://www.ncbi.nlm.nih.gov/pubmed/8649511) .
64. ^ Feng Y, Broder CC, Kennedy PE, Berger EA. (1996). "HIV-1 entry cofactor: functional cDNA cloning of a seventransmembrane, G protein-coupled receptor". Science 272 (5263): 872–7. doi:10.1126/science.272.5263.872
(http://dx.doi.org/10.1126%2Fscience.272.5263.872) . PMID 8629022 (http://www.ncbi.nlm.nih.gov/pubmed/8629022)
.
65. ^ Knight, S. C., Macatonia, S. E. and Patterson, S. (1990). "HIV I infection of dendritic cells". Int. Rev. Immunol. 6 (23): 163–75. doi:10.3109/08830189009056627 (http://dx.doi.org/10.3109%2F08830189009056627) . PMID 2152500
(http://www.ncbi.nlm.nih.gov/pubmed/2152500) .
66. ^ a b Tang, J. and Kaslow, R. A. (2003). "The impact of host genetics on HIV infection and disease progression in the
era of highly active antiretroviral therapy". AIDS 17 (Suppl 4): S51–S60. PMID 15080180
(http://www.ncbi.nlm.nih.gov/pubmed/15080180) .
67. ^ Zhu T, Mo H, Wang N, Nam DS, Cao Y, Koup RA, Ho DD. (1993). "Genotypic and phenotypic characterization of HIV1 patients with primary infection". Science 261 (5125): 1179–81. doi:10.1126/science.8356453
(http://dx.doi.org/10.1126%2Fscience.8356453) . PMID 8356453 (http://www.ncbi.nlm.nih.gov/pubmed/8356453) .
68. ^ van’t Wout AB, Kootstra NA, Mulder-Kampinga GA, Albrecht-van Lent N, Scherpbier HJ, Veenstra J, Boer K, Coutinho
RA, Miedema F, Schuitemaker H. (1994). "Macrophage-tropic variants initiate human immunodeficiency virus type 1
infection after sexual, parenteral, and vertical transmission". J Clin Invest 94 (5): 2060–7. doi:10.1172/JCI117560
(http://dx.doi.org/10.1172%2FJCI117560) . PMID 7962552 (http://www.ncbi.nlm.nih.gov/pubmed/7962552) .
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
69. ^ Zhu T, Wang N, Carr A, Nam DS, Moor-Jankowski R, Cooper DA, Ho DD. (1996). "Genetic characterization of human
immunodeficiency virus type 1 in blood and genital secretions: evidence for viral compartmentalization and selection
during sexual transmission" (http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=190172) . J
Virol 70 (5): 3098–107. PMID 8627789 (http://www.ncbi.nlm.nih.gov/pubmed/8627789) .
70. ^ Muciaccia B, Padula F, Vicini E, Gandini L, Lenzi A, Stefanini M. (2005). "Beta-chemokine receptors 5 and 3 are
expressed on the head region of human spermatozoon". FASEB J 19 (14): 2048–50. doi:10.1096/fj.05-3962fje
(http://dx.doi.org/10.1096%2Ffj.05-3962fje) . PMID 16174786 (http://www.ncbi.nlm.nih.gov/pubmed/16174786) .
71. ^ Berlier W, Bourlet T, Lawrence P, Hamzeh H, Lambert C, Genin C, Verrier B, Dieu-Nosjean MC, Pozzetto B, Delezay O.
(2005). "Selective sequestration of X4 isolates by human genital epithelial cells: Implication for virus tropism selection
process during sexual transmission of HIV". J Med Virol. 77 (4): 465–74. doi:10.1002/jmv.20478
(http://dx.doi.org/10.1002%2Fjmv.20478) . PMID 16254974 (http://www.ncbi.nlm.nih.gov/pubmed/16254974) .
72. ^ Clevestig P, Maljkovic I, Casper C, Carlenor E, Lindgren S, Naver L, Bohlin AB, Fenyo EM, Leitner T, Ehrnst A. (2005).
"The X4 phenotype of HIV type 1 evolves from R5 in two children of mothers, carrying X4, and is not linked to
transmission". AIDS Res Hum Retroviruses 5 (21): 371–8. doi:10.1089/aid.2005.21.371
(http://dx.doi.org/10.1089%2Faid.2005.21.371) . PMID 15929699 (http://www.ncbi.nlm.nih.gov/pubmed/15929699) .
73. ^ Moore JP. (1997). "Coreceptors: implications for HIV pathogenesis and therapy". Science 276 (5309): 51–2.
doi:10.1126/science.276.5309.51 (http://dx.doi.org/10.1126%2Fscience.276.5309.51) . PMID 9122710
(http://www.ncbi.nlm.nih.gov/pubmed/9122710) .
74. ^ Karlsson A, Parsmyr K, Aperia K, Sandstrom E, Fenyo EM, Albert J. (1994). "MT-2 cell tropism of human
immunodeficiency virus type 1 isolates as a marker for response to treatment and development of drug resistance". J
Infect Dis. 170 (6): 1367–75. PMID 7995974 (http://www.ncbi.nlm.nih.gov/pubmed/7995974) .
75. ^ Koot M, van 't Wout AB, Kootstra NA, de Goede RE, Tersmette M, Schuitemaker H. (1996). "Relation between
changes in cellular load, evolution of viral phenotype, and the clonal composition of virus populations in the course of
human immunodeficiency virus type 1 infection". J Infect Dis. 173 (2): 349–54. PMID 8568295
(http://www.ncbi.nlm.nih.gov/pubmed/8568295) .
76. ^ Cheney, K and McKnight, A (2010). "HIV-2 Tropism and Disease". Lentiviruses and Macrophages: Molecular and
Cellular Interactions . Caister Academic Press. ISBN 978-1-904455-60-8.
77. ^ a b c d e f g Chan D, Kim P (1998). "HIV entry and its inhibition". Cell 93 (5): 681–4. doi:10.1016/S00928674(00)81430-0 (http://dx.doi.org/10.1016%2FS0092-8674%2800%2981430-0) . PMID 9630213
(http://www.ncbi.nlm.nih.gov/pubmed/9630213) .
78. ^ a b c d e f Wyatt R, Sodroski J (1998). "The HIV-1 envelope glycoproteins: fusogens, antigens, and immunogens".
Science 280 (5371): 1884–8. doi:10.1126/science.280.5371.1884
(http://dx.doi.org/10.1126%2Fscience.280.5371.1884) . PMID 9632381
(http://www.ncbi.nlm.nih.gov/pubmed/9632381) .
79. ^ Arthos J, Cicala C, Martinelli E, Macleod K, Van Ryk D, Wei D, Xiao Z, Veenstra TD, Conrad TP, Lempicki RA,
McLaughlin S, Pascuccio M, Gopaul R, McNally J, Cruz CC, Censoplano N, Chung E, Reitano KN, Kottilil S, Goode DJ,
Fauci AS. (2008). "HIV-1 envelope protein binds to and signals through integrin alpha(4)beta(7), the gut mucosal
homing receptor for peripheral T cells". Nature Immunol. In Press: 301. doi:10.1038/ni1566
(http://dx.doi.org/10.1038%2Fni1566) . PMID 18264102 (http://www.ncbi.nlm.nih.gov/pubmed/18264102) .
80. ^ a b Pope M, Haase A (2003). "Transmission, acute HIV-1 infection and the quest for strategies to prevent infection".
Nat Med 9 (7): 847–52. doi:10.1038/nm0703-847 (http://dx.doi.org/10.1038%2Fnm0703-847) . PMID 12835704
(http://www.ncbi.nlm.nih.gov/pubmed/12835704) .
81. ^ Haedicke, J.; Brown, C.; Naghavi, H. (Aug 2009). "The brain-specific factor FEZ1 is a determinant of neuronal
susceptibility to HIV-1 infection" (http://www.pubmedcentral.nih.gov/articlerender.fcgi?
tool=pmcentrez&artid=2729016) . Proceedings of the National Academy of Sciences 106 (33): 14040.
doi:10.1073/pnas.0900502106 (http://dx.doi.org/10.1073%2Fpnas.0900502106) . ISSN 0027-8424
(http://worldcat.org/issn/0027-8424) . PMID 19667186 (http://www.ncbi.nlm.nih.gov/pubmed/19667186) .
82. ^ a b c Zheng, Y. H., Lovsin, N. and Peterlin, B. M. (2005). "Newly identified host factors modulate HIV replication".
Immunol. Lett. 97 (2): 225–34. doi:10.1016/j.imlet.2004.11.026 (http://dx.doi.org/10.1016%2Fj.imlet.2004.11.026) .
PMID 15752562 (http://www.ncbi.nlm.nih.gov/pubmed/15752562) .
83. ^ Doc Kaiser's Microbiology Home Page > IV. VIRUSES > F. ANIMAL VIRUS LIFE CYCLES > 3. The Life Cycle of HIV
(http://student.ccbcmd.edu/courses/bio141/lecguide/unit3/viruses/hivlc.html) Community College of Baltimore County.
Updated: Jan., 2008
84. ^ Hiscott J, Kwon H, Genin P. (2001). "Hostile takeovers: viral appropriation of the NF-kappaB pathway". J Clin Invest.
107 (2): 143–151. doi:10.1172/JCI11918 (http://dx.doi.org/10.1172%2FJCI11918) . PMID 11160127
(http://www.ncbi.nlm.nih.gov/pubmed/11160127) .
85. ^ Pollard, V. W. and Malim, M. H. (1998). "The HIV-1 Rev protein". Annu. Rev. Microbiol. 52: 491–532.
doi:10.1146/annurev.micro.52.1.491 (http://dx.doi.org/10.1146%2Fannurev.micro.52.1.491) . PMID 9891806
(http://www.ncbi.nlm.nih.gov/pubmed/9891806) .
86. ^ Gelderblom, H. R (1997). "Fine structure of HIV and SIV"
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
87.
88.
89.
90.
91.
92.
93.
94.
95.
96.
97.
98.
99.
100.
101.
102.
103.
104.
105.
(http://www.hiv.lanl.gov/content/sequence/HIV/COMPENDIUM/1997/partIII/Gelderblom.pdf) . in Los Alamos National
Laboratory (ed.) (PDF). HIV Sequence Compendium . Los Alamos, New Mexico: Los Alamos National Laboratory. pp. 31–
44.
^ a b c d Robertson DL, Hahn BH, Sharp PM. (1995). "Recombination in AIDS viruses". J Mol Evol. 40 (3): 249–59.
doi:10.1007/BF00163230 (http://dx.doi.org/10.1007%2FBF00163230) . PMID 7723052
(http://www.ncbi.nlm.nih.gov/pubmed/7723052) .
^ Rambaut, A et al (January 2004). "The causes and consequences of HIV evolution"
(http://tree.bio.ed.ac.uk/downloadPaper.php?id=242) . Nature Reviews Genetics 5. doi:10.1038/nrg1246
(http://dx.doi.org/10.1038%2Fnrg1246) . PMID 14708016 (http://www.ncbi.nlm.nih.gov/pubmed/14708016) .
^ a b Sodora, D.; Allan, J.; Apetrei, C.; Brenchley, J.; Douek, D.; Else, J.; Estes, J.; Hahn, B. et al . (2009). "Toward an
AIDS vaccine: lessons from natural simian immunodeficiency virus infections of African nonhuman primate hosts"
(http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2782707) . Nature medicine 15 (8): 861–
865. doi:10.1038/nm.2013 (http://dx.doi.org/10.1038%2Fnm.2013) . PMID 19661993
(http://www.ncbi.nlm.nih.gov/pubmed/19661993) .
^ Holzammer S, Holznagel E, Kaul A, Kurth R, Norley S (2001). "High virus loads in naturally and experimentally
SIVagm-infected African green monkeys". Virology 283 (2): 324–31. doi:10.1006/viro.2001.0870
(http://dx.doi.org/10.1006%2Fviro.2001.0870) . PMID 11336557 (http://www.ncbi.nlm.nih.gov/pubmed/11336557) .
^ Kurth, R. and Norley, S. (1996) Why don't the natural hosts of SIV develop simian AIDS?, J. NIH Res. 8, 33-37.
^ Baier M, Dittmar MT, Cichutek K, Kurth R (1991). "Development of vivo of genetic variability of simian
immunodeficiency virus". Proc. Natl. Acad. Sci. U.S.A. 88 (18): 8126–30. doi:10.1073/pnas.88.18.8126
(http://dx.doi.org/10.1073%2Fpnas.88.18.8126) . PMID 1896460 (http://www.ncbi.nlm.nih.gov/pubmed/1896460) .
^ Daniel MD, King NW, Letvin NL, Hunt RD, Sehgal PK, Desrosiers RC (1984). "A new type D retrovirus isolated from
macaques with an immunodeficiency syndrome". Science 223 (4636): 602–5. doi:10.1126/science.6695172
(http://dx.doi.org/10.1126%2Fscience.6695172) . PMID 6695172 (http://www.ncbi.nlm.nih.gov/pubmed/6695172) .
^ a b Keele, B.; Jones, J.; Terio, K.; Estes, J.; Rudicell, R.; Wilson, M.; Li, Y.; Learn, G. et al . (2009). "Increased
mortality and AIDS-like immunopathology in wild chimpanzees infected with SIVcpz". Nature 460 (7254): 515–519.
doi:10.1038/nature08200 (http://dx.doi.org/10.1038%2Fnature08200) . PMID 19626114
(http://www.ncbi.nlm.nih.gov/pubmed/19626114) .
^ Thomson, M. M., Perez-Alvarez, L. and Najera, R. (2002). "Molecular epidemiology of HIV-1 genetic forms and its
significance for vaccine development and therapy". Lancet Infect. Dis. 2 (8): 461–471. doi:10.1016/S14733099(02)00343-2 (http://dx.doi.org/10.1016%2FS1473-3099%2802%2900343-2) . PMID 12150845
(http://www.ncbi.nlm.nih.gov/pubmed/12150845) .
^ Carr, J. K.; Foley, B. T., Leitner, T., Salminen, M., Korber, B. and McCutchan, F. (1998). "Reference Sequences
Representing the Principal Genetic Diversity of HIV-1 in the Pandemic"
(http://www.hiv.lanl.gov/content/sequence/HIV/COMPENDIUM/1998/III/Carr.pdf) . in Los Alamos National Laboratory
(ed.) (PDF). HIV Sequence Compendium . Los Alamos, New Mexico: Los Alamos National Laboratory. pp. 10–19.
^ Osmanov S, Pattou C, Walker N, Schwardlander B, Esparza J; WHO-UNAIDS Network for HIV Isolation and
Characterization. (2002). "Estimated global distribution and regional spread of HIV-1 genetic subtypes in the year 2000".
Acquir. Immune. Defic. Syndr. 29 (2): 184–190. PMID 11832690 (http://www.ncbi.nlm.nih.gov/pubmed/11832690) .
^ Perrin L, Kaiser L, Yerly S. (2003). "Travel and the spread of HIV-1 genetic variants". Lancet Infect Dis. 3 (1): 22–27.
doi:10.1016/S1473-3099(03)00484-5 (http://dx.doi.org/10.1016%2FS1473-3099%2803%2900484-5) . PMID 12505029
(http://www.ncbi.nlm.nih.gov/pubmed/12505029) .
^ a b Plantier JC, Leoz M, Dickerson JE, et al. (August 2009). "A new human immunodeficiency virus derived from
gorillas". Nat. Med. 15 (8): 871–2. doi:10.1038/nm.2016 (http://dx.doi.org/10.1038%2Fnm.2016) . PMID 19648927
(http://www.ncbi.nlm.nih.gov/pubmed/19648927) .
^ Smith, L (2009-08-03). "Woman found carrying new strain of HIV from gorillas" (http://www.independent.co.uk/lifestyle/health-and-families/health-news/woman-found-carrying-new-strain-of-hiv-from-gorillas-1766627.html) . The
Independent. Retrieved 2009-08-04.
^ "Gorillas may be a source of AIDS, researchers find" (http://in.reuters.com/article/worldNews/idINIndia41490620090803) . Reuters India. 2009-08-03. Retrieved 2009-08-04.
^ a b c d Kumaranayake, L. and Watts, C. (2001). "Resource allocation and priority setting of HIV/AIDS interventions:
addressing the generalized epidemic in sub-Saharan Africa". J. Int. Dev. 13 (4): 451–466. doi:10.1002/jid.797
(http://dx.doi.org/10.1002%2Fjid.797) .
^ Kleinman, S. (September 2004). "Patient information: Blood donation and transfusion"
(http://www.uptodate.com/patients/content/topic.do?topicKey=blod_dis/2419) . Uptodate. Retrieved 2008-04-03.
^ a b Centers for Disease Control and Prevention. (2001). "Revised guidelines for HIV counseling, testing, and referral".
MMWR Recomm Rep. 50 (RR-19): 1–57. PMID 11718472 (http://www.ncbi.nlm.nih.gov/pubmed/11718472) .
^ Celum CL, Coombs RW, Lafferty W, Inui TS, Louie PH, Gates CA, McCreedy BJ, Egan R, Grove T, Alexander S, et al.
(1991). "Indeterminate human immunodeficiency virus type 1 western blots: seroconversion risk, specificity of
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
106.
107.
108.
109.
110.
111.
112.
113.
114.
115.
116.
117.
118.
119.
120.
121.
122.
123.
supplemental tests, and an algorithm for evaluation". J Infect Dis. 164 (4): 656–664. PMID 1894929
(http://www.ncbi.nlm.nih.gov/pubmed/1894929) .
^ Chou R, Huffman LH, Fu R, Smits AK, Korthuis PT (July 2005). "Screening for HIV: a review of the evidence for the
U.S. Preventive Services Task Force" (http://www.annals.org/cgi/content/full/143/1/55) . Ann. Intern. Med. 143 (1):
55–73. PMID 15998755 (http://www.ncbi.nlm.nih.gov/pubmed/15998755) .
^ Kleinman S, Busch MP, Hall L, et al. for the Retrovirus Epidemiology Donor Study (1998). "False-positive HIV-1 test
results in a low-risk screening setting of voluntary blood donation". JAMA 280: 1080–1085.
doi:10.1001/jama.280.12.1080 (http://dx.doi.org/10.1001%2Fjama.280.12.1080) . PMID 9757856
(http://www.ncbi.nlm.nih.gov/pubmed/9757856) .
^ Wood RW, Dunphy C, Okita K, Swenson P (2003). "Two "HIV-Infected" Persons Not Really Infected". Arch Intern Med
163: 1857–1859. doi:10.1001/archinte.163.15.1857 (http://dx.doi.org/10.1001%2Farchinte.163.15.1857) . PMID
12912724 (http://www.ncbi.nlm.nih.gov/pubmed/12912724) .
^ Padeh YC, Rubinstein A, Shliozberg J (2005). "Common variable immunodeficiency and testing for HIV-1". N Engl J
Med 353: 1074–1075. doi:10.1056/NEJMc051339 (http://dx.doi.org/10.1056%2FNEJMc051339) . PMID 16148302
(http://www.ncbi.nlm.nih.gov/pubmed/16148302) .
^ http://www.lanl.gov/discover/curing_aids
^ Robb ML (2008). "Failure of the Merck HIV vaccine: an uncertain step forward". Lancet 372 (9653): 1857–1858.
doi:10.1016/S0140-6736(08)61593-7 (http://dx.doi.org/10.1016%2FS0140-6736%2808%2961593-7) .
^ http://news.bbc.co.uk/2/hi/health/8272113.stm
^ Fan, H., Conner, R. F. and Villarreal, L. P. eds, ed (2005). AIDS : science and society (4th ed.). Boston, MA: Jones and
Bartlett Publishers. ISBN 0-7637-0086-X.
^ Department of Health and Human Services (January 2005). "A Pocket Guide to Adult HIV/AIDS Treatment January
2005 edition" (http://www.hab.hrsa.gov/tools/HIVpocketguide05/PktGARTtables.htm) . Retrieved 2006-01-17.
^ a b c Palella, F. J., Delaney, K. M., Moorman, A. C., Loveless, M. O., Fuhrer, J., Satten, G. A., Aschman, D. J. and
Holmberg, S. D. (1998). "Declining morbidity and mortality among patients with advanced human immunodeficiency
virus infection". N. Engl. J. Med. 338 (13): 853–860. doi:10.1056/NEJM199803263381301
(http://dx.doi.org/10.1056%2FNEJM199803263381301) . PMID 9516219
(http://www.ncbi.nlm.nih.gov/pubmed/9516219) .
^ Department of Health and Human Services Working Group on Antiretroviral Therapy and Medical Management of
HIV-Infected Children (November 3, 2005). "Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection"
(http://www.aidsinfo.nih.gov/ContentFiles/PediatricGuidelines_PDA.pdf) (PDF). Retrieved 2006-01-17.
^ Department of Health and Human Services Panel on Clinical Practices for Treatment of HIV Infection (October 6,
2005). "Guidelines for the Use of Antiretroviral Agents in HIV-1-Infected Adults and Adolescents"
(http://aidsinfo.nih.gov/ContentFiles/AdultandAdolescentGL.pdf) (PDF). Retrieved 2006-01-17.
^ Martinez-Picado, J., DePasquale, M. P., Kartsonis, N., Hanna, G. J., Wong, J., Finzi, D., Rosenberg, E., Gunthard,
H.F., Sutton, L., Savara, A., Petropoulos, C. J., Hellmann, N., Walker, B. D., Richman, D. D., Siliciano, R. and D'Aquila, R.
T. (2000). "Antiretroviral resistance during successful therapy of human immunodeficiency virus type 1 infection". Proc.
Natl. Acad. Sci. U. S. A. 97 (20): 10948–10953. doi:10.1073/pnas.97.20.10948
(http://dx.doi.org/10.1073%2Fpnas.97.20.10948) . PMID 11005867 (http://www.ncbi.nlm.nih.gov/pubmed/11005867) .
^ Dybul, M., Fauci, A. S., Bartlett, J. G., Kaplan, J. E., Pau, A. K.; Panel on Clinical Practices for Treatment of HIV.
(2002). "Guidelines for using antiretroviral agents among HIV-infected adults and adolescents". Ann. Intern. Med. 137
(5 Pt 2): 381–433. PMID 12617573 (http://www.ncbi.nlm.nih.gov/pubmed/12617573) .
^ Blankson, J. N., Persaud, D., Siliciano, R. F. (2002). "The challenge of viral reservoirs in HIV-1 infection". Annu. Rev.
Med. 53: 557–593. doi:10.1146/annurev.med.53.082901.104024
(http://dx.doi.org/10.1146%2Fannurev.med.53.082901.104024) . PMID 11818490
(http://www.ncbi.nlm.nih.gov/pubmed/11818490) .
^ a b Wood, E., Hogg, R. S., Yip, B., Harrigan, P. R., O'Shaughnessy, M. V. and Montaner, J. S. (2003). "Is there a
baseline CD4 cell count that precludes a survival response to modern antiretroviral therapy?". AIDS 17 (5): 711–720.
doi:10.1097/01.aids.0000050854.71999.d8 (http://dx.doi.org/10.1097%2F01.aids.0000050854.71999.d8) . PMID
12646794 (http://www.ncbi.nlm.nih.gov/pubmed/12646794) .
^ Chene, G., Sterne, J. A., May, M., Costagliola, D., Ledergerber, B., Phillips, A. N., Dabis, F., Lundgren, J., D'Arminio
Monforte, A., de Wolf, F., Hogg, R., Reiss, P., Justice, A., Leport, C., Staszewski, S., Gill, J., Fatkenheuer, G., Egger, M.
E. and the Antiretroviral Therapy Cohort Collaboration. (2003). "Prognostic importance of initial response in HIV-1
infected patients starting potent antiretroviral therapy: analysis of prospective studies". Lancet 362 (9385): 679–686.
PMID 12957089 (http://www.ncbi.nlm.nih.gov/pubmed/12957089) .
^ A computer based study in 2006, following the 2004 United States treatment guidelines: Schackman BR, Gebo KA,
Walensky RP, Losina E, Muccio T, Sax PE, Weinstein MC, Seage GR 3rd, Moore RD, Freedberg KA. (2006). "The lifetime
cost of current HIV care in the United States". Med Care 44 (11): 990–997. doi:10.1097/01.mlr.0000228021.89490.2a
(http://dx.doi.org/10.1097%2F01.mlr.0000228021.89490.2a) . PMID 17063130
(http://www.ncbi.nlm.nih.gov/pubmed/17063130) .
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
124. ^ Becker SL, Dezii CM, Burtcel B, Kawabata H, Hodder S. (2002). "Young HIV-infected adults are at greater risk for
medication nonadherence". MedGenMed. 4 (3): 21. PMID 12466764 (http://www.ncbi.nlm.nih.gov/pubmed/12466764) .
125. ^ Nieuwkerk, P., Sprangers, M., Burger, D., Hoetelmans, R. M., Hugen, P. W., Danner, S. A., van Der Ende, M. E.,
Schneider, M. M., Schrey, G., Meenhorst, P. L., Sprenger, H. G., Kauffmann, R. H., Jambroes, M., Chesney, M. A., de
Wolf, F., Lange, J. M. and the ATHENA Project. (2001). "Limited Patient Adherence to Highly Active Antiretroviral
Therapy for HIV-1 Infection in an Observational Cohort Study". Arch. Intern. Med. 161 (16): 1962–1968. PMID
11525698 (http://www.ncbi.nlm.nih.gov/pubmed/11525698) .
126. ^ Kleeberger, C., Phair, J., Strathdee, S., Detels, R., Kingsley, L. and Jacobson, L. P. (2001). "Determinants of
Heterogeneous Adherence to HIV-Antiretroviral Therapies in the Multicenter AIDS Cohort Study". J. Acquir. Immune
Defic. Syndr. 26 (1): 82–92. PMID 11176272 (http://www.ncbi.nlm.nih.gov/pubmed/11176272) .
127. ^ Heath, K. V., Singer, J., O'Shaughnessy, M. V., Montaner, J. S. and Hogg, R. S. (2002). "Intentional Nonadherence
Due to Adverse Symptoms Associated With Antiretroviral Therapy". J. Acquir. Immune Defic. Syndr. 31 (2): 211–217.
PMID 12394800 (http://www.ncbi.nlm.nih.gov/pubmed/12394800) .
128. ^ Montessori, V., Press, N., Harris, M., Akagi, L., Montaner, J. S. (2004). "Adverse effects of antiretroviral therapy for
HIV infection." (http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=315530) . CMAJ 170 (2):
229–238. PMID 14734438 (http://www.ncbi.nlm.nih.gov/pubmed/14734438) .
129. ^ Saitoh, A., Hull, A. D., Franklin, P. and Spector, S. A. (2005). "Myelomeningocele in an infant with intrauterine
exposure to efavirenz". J. Perinatol. 25 (8): 555–556. doi:10.1038/sj.jp.7211343
(http://dx.doi.org/10.1038%2Fsj.jp.7211343) . PMID 16047034 (http://www.ncbi.nlm.nih.gov/pubmed/16047034) .
130. ^ Wang C, Vlahov D, Galai N, et al. (2004). "Mortality in HIV-seropositive versus seronegative persons in the era of
highly active antiretroviral therapy.". J. Infect. Dis. 190: 1046–54. doi:10.1086/422848
(http://dx.doi.org/10.1086%2F422848) . PMID 15319852 (http://www.ncbi.nlm.nih.gov/pubmed/15319852) .
131. ^ World Health Organisation (2006). "WHO case definitions of HIV for surveillance and revised clinical staging and
immunological classification" (http://www.who.int/hiv/pub/guidelines/WHO%20HIV%20Staging.pdf) (PDF). Retrieved
2006-12-27.
132. ^ a b c Ferrantelli F, Cafaro A, Ensoli B (December 2004). "Nonstructural HIV proteins as targets for prophylactic or
therapeutic vaccines". Curr. Opin. Biotechnol. 15 (6): 543–56. doi:10.1016/j.copbio.2004.10.008
(http://dx.doi.org/10.1016%2Fj.copbio.2004.10.008) . PMID 15560981
(http://www.ncbi.nlm.nih.gov/pubmed/15560981) .
133. ^ Mark Schoofs (2008). "A Doctor, a Mutation and a Potential Cure for AIDS"
(http://online.wsj.com/article/SB122602394113507555.html) . The Wall Street Journal. Retrieved 2008-11-09.
134. ^ Hütter G, Nowak D, Mossner M, Ganepola S, Ganepola A, Allers K, Schneider T, Hofmann J, Kücherer C, Blau O, Blau
IW, Hofmann WK, Thiel E (2009). "Long-Term Control of HIV by CCR5 Delta32/Delta32 Stem-Cell Transplantation"
(http://content.nejm.org/cgi/content/abstract/360/7/692) . N Engl J Med 360 (7): 692–698.
doi:10.1056/NEJMoa0802905 (http://dx.doi.org/10.1056%2FNEJMoa0802905) . PMID 19213682
(http://www.ncbi.nlm.nih.gov/pubmed/19213682) . Retrieved 2009-03-31.
135. ^ Levy JA (2009). "Not an HIV Cure, but Encouraging New Directions"
(http://content.nejm.org/cgi/content/full/360/7/724) . N Engl J Med 360 (7): 724–725. doi:10.1056/NEJMe0810248
(http://dx.doi.org/10.1056%2FNEJMe0810248) . PMID 19213687 (http://www.ncbi.nlm.nih.gov/pubmed/19213687) .
Retrieved 2009-03-31.
136. ^ Glass WG, McDermott DH, Lim JK, et al. (January 2006). "CCR5 deficiency increases risk of symptomatic West Nile
virus infection" (http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2118086) . J. Exp. Med.
203 (1): 35–40. doi:10.1084/jem.20051970 (http://dx.doi.org/10.1084%2Fjem.20051970) . PMID 16418398
(http://www.ncbi.nlm.nih.gov/pubmed/16418398) .
137. ^ Archin NM, Eron JJ, Palmer S, Hartmann-Duff A, Martinson JA, Wiegand A, Bandarenko N, Schmitz JL, Bosch RJ,
Landay AL, Coffin JM, Margolis DM. (2008). "Valproic acid without intensified antiviral therapy has limited impact on
persistent HIV infection of resting CD4+ T cells.". AIDS 22 (10): 1131–1135. doi:10.1097/QAD.0b013e3282fd6df4
(http://dx.doi.org/10.1097%2FQAD.0b013e3282fd6df4) . PMID 18525258
(http://www.ncbi.nlm.nih.gov/pubmed/18525258) .
138. ^ Bowman MC, Archin NM, Margolis DM. (2009). "Pharmaceutical approaches to eradication of persistent HIV
infection.". Expert Reviews in Molecular Medicine 11 (e6).
139. ^ a b UNAIDS, WHO (December 2007). "2007 AIDS epidemic update"
(http://data.unaids.org/pub/EPISlides/2007/2007_epiupdate_en.pdf) (PDF). Retrieved 2008-03-12.
140. ^ Zwahlen M, Egger M (2006) (PDF). Progression and mortality of untreated HIV-positive individuals living in resourcelimited settings: update of literature review and evidence synthesis
(http://data.unaids.org/pub/Periodical/2006/zwahlen_unaids_hq_05_422204_2007_en.pdf) . UNAIDS Obligation
HQ/05/422204. Retrieved 2008-03-19.
141. ^ Knoll B, Lassmann B, Temesgen Z (2007). "Current status of HIV infection: a review for non-HIV-treating
physicians". Int J Dermatol 46 (12): 1219–28. doi:10.1111/j.1365-4632.2007.03520.x (inactive 2009-08-10). PMID
18173512 (http://www.ncbi.nlm.nih.gov/pubmed/18173512) .
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
142. ^ Antiretroviral Therapy Cohort Collaboration (2008). "Life expectancy of individuals on combination antiretroviral
therapy in high-income countries: a collaborative analysis of 14 cohort studies". Lancet 372 (9635): 293–9.
doi:10.1016/S0140-6736(08)61113-7 (http://dx.doi.org/10.1016%2FS0140-6736%2808%2961113-7) . PMID 18657708
(http://www.ncbi.nlm.nih.gov/pubmed/18657708) .
143. ^ Clerici M, Balotta C, Meroni L, et al. (1996). "Type 1 cytokine production and low prevalence of viral isolation correlate
with long-term non progression in HIV infection". AIDS Res. Hum. Retroviruses. 12 (11): 1053–1061. PMID 8827221
(http://www.ncbi.nlm.nih.gov/pubmed/8827221) .
144. ^ Morgan D, Mahe C, Mayanja B, Whitworth JA (2002). "Progression to symptomatic disease in people infected with
HIV-1 in rural Uganda: prospective cohort study". BMJ 324 (7331): 193–196. doi:10.1136/bmj.324.7331.193
(http://dx.doi.org/10.1136%2Fbmj.324.7331.193) . PMID 11809639 (http://www.ncbi.nlm.nih.gov/pubmed/11809639) .
145. ^ Campbell GR, Pasquier E, Watkins J, et al. (2004). "The glutamine-rich region of the HIV-1 Tat protein is involved in
T-cell apoptosis". J. Biol. Chem. 279 (46): 48197–48204. doi:10.1074/jbc.M406195200
(http://dx.doi.org/10.1074%2Fjbc.M406195200) . PMID 15331610 (http://www.ncbi.nlm.nih.gov/pubmed/15331610) .
146. ^ Campbell GR, Watkins JD, Esquieu D, Pasquier E, Loret EP, Spector SA (2005). "The C terminus of HIV-1 Tat
modulates the extent of CD178-mediated apoptosis of T cells". J. Biol. Chem. 280 (46): 38376–39382. PMID 16155003
(http://www.ncbi.nlm.nih.gov/pubmed/16155003) .
147. ^ Senkaali D, Muwonge R, Morgan D, Yirrell D, Whitworth J, Kaleebu P (2005). "The relationship between HIV type 1
disease progression and V3 serotype in a rural Ugandan cohort". AIDS Res. Hum. Retroviruses. 20 (9): 932–937. PMID
15585080 (http://www.ncbi.nlm.nih.gov/pubmed/15585080) .
148. ^ McNeil, Jr., Donald (November 20, 2007). "U.N. Agency to Say It Overstated Extent of H.I.V. Cases by Millions"
(http://query.nytimes.com/gst/fullpage.html?res=9C01EEDF103BF933A15752C1A9619C8B63&n) . The New York Times.
Retrieved 2008-01-16.
149. ^ UNAIDS (2001). "Special Session of the General Assembly on HIV/AIDS Round table 3 Socio-economic impact of the
epidemic and the strengthening of national capacities to combat HIV/AIDS"
(http://data.unaids.org/Publications/External-Documents/GAS26-rt3_en.pdf) (PDF). Retrieved 2006-06-15.
150. ^ World Bank (2005). "Evaluating the World Bank's Assistance for Fighting the HIV/AIDS Epidemic"
(http://www.worldbank.org/oed/aids/main_report.html) . Retrieved 2006-01-17.
151. ^ Worobey M, Gemmel M, Teuwen DE, et al. (October 2008). "Direct evidence of extensive diversity of HIV-1 in
Kinshasa by 1960". Nature 455 (7213): 661–4. doi:10.1038/nature07390 (http://dx.doi.org/10.1038%2Fnature07390) .
PMID 18833279 (http://www.ncbi.nlm.nih.gov/pubmed/18833279) .
152. ^ "Pneumocystis Pneumonia -- Los Angeles" (http://www.cdc.gov/MMWR/preview/mmwrhtml/00043494.htm) .
Retrieved 2008-05-05.
153. ^ Gao F, Bailes E, Robertson DL, et al. (February 1999). "Origin of HIV-1 in the chimpanzee Pan troglodytes
troglodytes". Nature 397 (6718): 436–41. doi:10.1038/17130 (http://dx.doi.org/10.1038%2F17130) . PMID 9989410
(http://www.ncbi.nlm.nih.gov/pubmed/9989410) .
154. ^ Keele, B. F., van Heuverswyn, F., Li, Y. Y., Bailes, E., Takehisa, J., Santiago, M. L., Bibollet-Ruche, F., Chen, Y., Wain,
L. V., Liegois, F., Loul, S., Mpoudi Ngole, E., Bienvenue, Y., Delaporte, E., Brookfield, J. F. Y., Sharp, P. M., Shaw, G. M.,
Peeters, M., and Hahn, B. H. (28 July 2006). "Chimpanzee Reservoirs of Pandemic and Nonpandemic HIV-1"
(http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=PMC2442710) . Science 313 (5786
pages=523–526). doi:10.1126/science.1126531 (http://dx.doi.org/10.1126%2Fscience.1126531) . PMID 16728595
(http://www.ncbi.nlm.nih.gov/pubmed/16728595) .
155. ^ Goodier, J., and Kazazian, H. (2008). "Retrotransposons Revisited: The Restraint and Rehabilitation of Parasites". Cell
135 (1): 23–35. doi:10.1016/j.cell.2008.09.022 (http://dx.doi.org/10.1016%2Fj.cell.2008.09.022) . PMID 18854152
(http://www.ncbi.nlm.nih.gov/pubmed/18854152) .
156. ^ a b Basavapathruni, A; Anderson, KS (December 2007). "Reverse transcription of the HIV-1 pandemic". The FASEB
Journal 21 (14): 3795–3808. doi:10.1096/fj.07-8697rev (http://dx.doi.org/10.1096%2Ffj.07-8697rev) . PMID 17639073
(http://www.ncbi.nlm.nih.gov/pubmed/17639073) .
157. ^ RC Gallo, PS Sarin, EP Gelmann, M Robert-Guroff, E Richardson, VS Kalyanaraman, D Mann, GD Sidhu, RE Stahl, S
Zolla-Pazner, J Leibowitch, and M Popovic (1983). "Isolation of human T-cell leukemia virus in acquired immune
deficiency syndrome (AIDS)". Science 220: 865–867. doi:10.1126/science.6189183
(http://dx.doi.org/10.1126%2Fscience.6189183) .
158. ^ F Barre-Sinoussi, JC Chermann, F Rey, MT Nugeyre, S Chamaret, J Gruest, C Dauguet, C Axler-Blin, F Vezinet-Brun,
C Rouzioux, W Rozenbaum, and L Montagnier (1983). "Isolation of a T-lymphotropic retrovirus from a patient at risk for
acquired immune deficiency syndrome (AIDS)". Science 220: 868–870. doi:10.1126/science.6601823
(http://dx.doi.org/10.1126%2Fscience.6601823) .
159. ^ "The Nobel Prize in Physiology or Medicine 2008"
(http://nobelprize.org/nobel_prizes/medicine/laureates/2008/index.html) . Nobel Foundation. Retrieved October 28,
2009.
160. ^ a b Cohen, J; Enserink, Martin (10 October 2008). "Nobel Prize in Physiology or Medicine: HIV, HPV researchers
honored, but one scientist is left out". Science 322 (5899): 149–175. doi:10.1126/science.322.5899.174
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
161.
162.
163.
164.
165.
(http://dx.doi.org/10.1126%2Fscience.322.5899.174) . PMID 18845715
(http://www.ncbi.nlm.nih.gov/pubmed/18845715) .
^ Altman, Lawrence (2008-10-06). "Three Europeans Win the 2008 Nobel for Medicine"
(http://www.nytimes.com/2008/10/07/health/07nobel.html) . New York Times. Retrieved 2008-10-06.
^ a b Kalichman, Seth (2009). Denying AIDS: Conspiracy Theories, Pseudoscience, and Human Tragedy
(http://books.google.ca/books?
id=_mtDBCDwxugC&printsec=frontcover&source=gbs_navlinks_s#v=onepage&q=&f=false) . New York: Copernicus
Books (Springer Science+Business Media). ISBN 978-0-387-79475-4.
^ Duesberg, P. H. (1988). "HIV is not the cause of AIDS". Science 241 (4865): 514, 517. doi:10.1126/science.3399880
(http://dx.doi.org/10.1126%2Fscience.3399880) . PMID 3399880 (http://www.ncbi.nlm.nih.gov/pubmed/3399880) .
^ Smith TC, Novella SP (August 2007). "HIV denial in the Internet era" (http://medicine.plosjournals.org/perlserv/?
request=get-document&doi=10.1371/journal.pmed.0040256&ct=1&SESSID=3d4baa1a64e57d8ff33e9d41eb2335a1) .
PLoS Med. 4 (8): e256. doi:10.1371/journal.pmed.0040256 (http://dx.doi.org/10.1371%2Fjournal.pmed.0040256) .
PMID 17713982 (http://www.ncbi.nlm.nih.gov/pubmed/17713982) . Retrieved 2009-11-07.
^ For evidence that HIV is the cause of AIDS, see (for example):
, (2000). "The Durban Declaration". Nature 406 (6791): 15–6. doi:10.1038/35017662
(http://dx.doi.org/10.1038%2F35017662) . PMID 10894520 (http://www.ncbi.nlm.nih.gov/pubmed/10894520) ..
Cohen, J. (1994). "The Controversy over HIV and AIDS" (http://www.sciencemag.org/feature/data/cohen/2665191-1642a.pdf) (PDF). Science 266 (5191): 1642–1649. doi:10.1126/science.7992043
(http://dx.doi.org/10.1126%2Fscience.7992043) . PMID 7992043
(http://www.ncbi.nlm.nih.gov/pubmed/7992043) . Retrieved 2009-03-31.
Various. "Resources and Links, HIV-AIDS Connection"
(http://www3.niaid.nih.gov/topics/HIVAIDS/Understanding/How+HIV+Causes+AIDS/connection.htm) . National
Institute of Allergy and Infectious Diseases. Retrieved 2009-02-22.
O'Brien SJ, Goedert JJ (1996). "HIV causes AIDS: Koch's postulates fulfilled". Curr. Opin. Immunol. 8 (5): 613–8.
doi:10.1016/S0952-7915(96)80075-6 (http://dx.doi.org/10.1016%2FS0952-7915%2896%2980075-6) . PMID
8902385 (http://www.ncbi.nlm.nih.gov/pubmed/8902385) .
Galéa P, Chermann JC (1998). "HIV as the cause of AIDS and associated diseases". Genetica 104 (2): 133–42.
doi:10.1023/A:1003432603348 (http://dx.doi.org/10.1023%2FA%3A1003432603348) . PMID 10220906
(http://www.ncbi.nlm.nih.gov/pubmed/10220906) .
166. ^ Watson J (2006). "Scientists, activists sue South Africa's AIDS 'denialists'". Nat. Med. 12 (1): 6. doi:10.1038/nm01066a (http://dx.doi.org/10.1038%2Fnm0106-6a) . PMID 16397537 (http://www.ncbi.nlm.nih.gov/pubmed/16397537) .
167. ^ Baleta A (2003). "S Africa's AIDS activists accuse government of murder". Lancet 361 (9363): 1105.
doi:10.1016/S0140-6736(03)12909-1 (http://dx.doi.org/10.1016%2FS0140-6736%2803%2912909-1) . PMID 12672319
(http://www.ncbi.nlm.nih.gov/pubmed/12672319) .
168. ^ Cohen J (2000). "South Africa's new enemy". Science 288 (5474): 2168–70. doi:10.1126/science.288.5474.2168
(http://dx.doi.org/10.1126%2Fscience.288.5474.2168) . PMID 10896606
(http://www.ncbi.nlm.nih.gov/pubmed/10896606) .
External links
HIV/AIDS
(http://www.dmoz.org/Health/Conditions_and_Diseases/Immune_Disorders/Immune_Deficiency/AIDS//)
at the Open Directory Project
"AIDSinfo - HIV/AIDS Treatment Information" (http://aidsinfo.nih.gov/) . US Department of Health and
Human Services. Retrieved 2008-03-21.
"UNAIDS: The Joint United Nations Programme on HIV/AIDS" (http://www.unaids.org/en/) . UNAIDS.
Retrieved 2008-03-21.
"AIDS.gov: Portal to all Federal HIV/AIDS information" (http://www.aids.gov/) . AIDS.gov. Retrieved
2008-05-20.
"HIV News and Resources" (http://www.thebody.com/) . The Body. Retrieved 2008-07-18.
Retrieved from "http://en.wikipedia.org/wiki/HIV"
Categories: HIV/AIDS | Retroviruses | Sexually transmitted diseases and infections | Immunodeficiency |
Discovery and invention controversies | Initialisms | Causes of death
This page was last modified on 5 February 2010 at 19:58.
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]
HIV - Wikipedia, the free encyclopedia
Text is available under the Creative Commons Attribution-ShareAlike License; additional terms may
apply. See Terms of Use for details.
Wikipedia® is a registered trademark of the Wikimedia Foundation, Inc., a non-profit organization.
file:///C|/Documents%20and%20Settings/joe/Desktop/HIV%20Information%20Sheet.htm[2/8/2010 4:43:27 PM]