Skip to main content

25 years of HIV-1 research – progress and perspectives

Abstract

Twenty-five years after the discovery and isolation of the human immunodeficiency virus by French and American scientists, much progress has been made in basic research, clinical treatment, and public health prevention measures for acquired immunodeficiency syndrome. Here, we summarize, in brief, advances that have been achieved and provide some perspectives on future challenges.

Peer Review reports

Background

The first cases of acquired immunodeficiency syndrome (AIDS) were described in homosexual men in the US in 1981 [1]. Several years later in 1983 and 1984, respectively, French and American scientists confirmed that the causative agent for AIDS was a retrovirus, the human immunodeficiency virus (HIV) [24]. Today, 25 years after the isolation of HIV-1, approximately 25 million individuals have died from AIDS, a number exceeding by 60 times the total number of American casualties in World War II; and over 33 million people globally are infected and living with HIV-1. In 2007, 2.7 million individuals became newly infected with HIV-1, and 2 million AIDS deaths occurred [5]. Regrettably, half of all people who are infected with HIV acquire the infection before the age of 25 years, and are killed by AIDS before they turn 35. More than 95% of new HIV-1 infections arise in low and middle income nations, populations least likely to have access to antiretroviral therapy. In the face of these daunting statistics and an unabated pandemic, we look back on progress achieved in HIV-1 research, treatment, and policies, and look forward to the challenges that confront AIDS scientists, clinicians and decision-makers for the next 25 years.

Achievements from basic research

The basic research of HIV has made great strides over the past quarter century. Key discoveries, reviewed here in brief, have clarified the intricate steps used by the virus from entry into and exit from the cell (Figure 1). HIV-1 is an enveloped virus with glycoproteins on its surface [6, 7], responsible for viral entry into cells. The cell's primary receptor for the virus is the CD4 molecule, and its co-receptors are members of a family of chemokine receptors [8, 9]. Using the different co-receptors, HIV-1 can differentially infect T-lymphocytes and/or macrophages [10, 11].

Figure 1
figure 1

A schematic illustration of the life-cycle of HIV-1 during the infection of a human cell. Entry, reverse transcription, nuclear entry, integration, transcription, translation, virus assembly and egress are shown.

Entry of the virus into the cytoplasm initiates the disassembly of the HIV core. At this juncture, the virus can be susceptible to intrinsic cellular antiviral mechanisms which include tripartite motif protein 5a (TRIM5α), a protein with a tripartite motif containing a RING domain, a B-box type 1, and a B-box type 2, followed by a coiled-coil region [12, 13], apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3G (APOBEC3G) [14, 15], mammalian RNAi (RNA interference) [16, 17] and methylation by a cellular methylase enzyme, protein arginine methyltransferase 6 (PRMT6) of the arginine residues [18, 19] of several viral proteins. TRIM5α can target the viral capsid protein [13]. APOBEC3G can edit HIV RNA through its cytidine deaminase [20] activity, and in turn HIV encodes an accessory protein called Vif, which counteracts the effect of APOBEC3G [15, 21]. Similarly, methylation of viral proteins by cellular methyltransferases like PRMT6 renders them less active and decreases viral replication [22]. Finally, small cellular RNAs appear to contribute to a mammalian cellular defense against retroviral infection [23].

The uncoated HIV-1 reverse transcribes its genomic RNA in the cytoplasm into a DNA copy (a provirus), which is then transported into the nucleus for integration into host chromosomes. Several viral and cellular proteins have been characterized to participate in these processes [24, 25]. Once integrated, the provirus is transcribed by the cell's RNA polymerase II machinery with contributions from cellular transcription factors such as Sp1 and NF-κB [26]. Later, viral transcription enters a second stage after the synthesis of the HIV-1 trans-activator of transcription (Tat), an RNA-binding protein. Tat recognizes a viral trans-activation responsive RNA leader [27, 28], and together with the cellular positive transcription elongation factor b complex, made up of cyclin-dependant kinase 9 and cyclin T1[29, 30], enhances the initiation and processivity of HIV-1 transcription by several hundred fold. Transcribed viral RNAs are next subject to post-transcriptional regulation by HIV-1 Rev and cellular factors Crm1 and RNA helicases [31, 32] to export unspliced and partially spliced RNAs from the nucleus into the cytoplasm for translation.

The viral unspliced RNA (around nine kilobases in size) is also the genomic RNA, which is encapsidated into the newly assembled viral particle as a dimer [33, 34]. Encapsidation of the dimeric HIV-1 RNA depends on structured RNA motifs [35, 36]. The RNA dimer is captured by the viral Gag polyprotein. Thereafter, the assembly and budding of the virus transit via the cell's late endosomal pathway through multi-vesicular bodies [37, 38] employing cellular ESCRT (endosomal sorting complex required for transport, proteins that are in multivesicular bodies inside cells) trafficking proteins [39].

Diagnostics

There has been important progress in regard to HIV diagnostics. From the initial availability of an antibody test, the field has now progressed to be able to monitor directly viral load in the blood using sensitive polymerase chain reaction (PCR) assays. The advent of PCR testing has also represented an important safeguard for the blood supply, since it is now possible to screen all donated blood samples for HIV using this sensitive technique [40].

Epidemiology

Much has been accomplished also in regard to better understanding the epidemiology of HIV transmission. On a worldwide basis, heterosexual transmission represents the most important means of HIV acquisition [5]. In contrast, however, gay men remain at extremely high risk in regard to new infections in developed country settings. Many observers have questioned whether this may be due to complacency among individuals whose attitude may be that the drugs will work to prolong their lives on an effective basis, regardless of whether or not they eventually do become infected by HIV-1 [5]. It is to be hoped that better awareness and education will provide the basis for diminution of the numbers of new infections, as the realization sets in that drug resistance will likely become a reality over the long term and that the drugs may not continue to work forever. A second consideration is the epidemiologic observation that large numbers of HIV-infected individuals may become prone to a wide array of cancers (for example, lymphomas and anal carcinomas). This is an additional reason for individuals to remain HIV-negative rather than abandoning precautions that would otherwise protect them against HIV-1. The occurrence of high numbers of cancers in HIV-positive individuals is relevant to both developing as well as developed countries.

Progress in treatment

Without doubt, the most significant headway in the battle against HIV/AIDS has been the development of effective antiretroviral drugs that provide important treatment options for patients infected by HIV. These drugs now exist in a variety of categories based on the enzymatic targets and/or cellular targets that are the targets of the drugs themselves (Table 1). The first drugs were targeted against viral reverse transcriptase but, in short order, we have seen the development of anti-HIV drugs that are targeted against protease, and most recently, integrase inhibitors. The development of combination therapies has been key in assuring prolongation of life, to an extent that HIV disease in almost all developed countries today can be considered to be a chronic manageable condition, unlike the death sentence that existed during the initial years of the epidemic through to the mid 1990s when triple therapies first became available [41, 42].

Table 1 Antiretroviral drugs approved by the US Food and Drug Administration

Of course, it is also fair to state that treatments have improved over the years, not only because of the effectiveness of the drugs themselves but also because newer compounds developed to treat HIV disease are far less toxic than those initially used during the 1980s and 1990s. In addition, simplicity of dosing has become commonplace, such that patients can, in some cases, take only one pill per day, based on a co-formulation that involves two nucleoside/nucleotide reverse transcriptase inhibitors coupled with a non-nucleoside reverse transcriptase inhibitor. The ability to simplify dosing, coupled with current treatment protocols, make current therapies far less toxic than their predecessors, and this has also had important implications in another area, that is, today's treatment regimens are far less prone to the development of drug resistance than were the initially available combination therapies [43].

It will be recalled that the first use of PIs was based on compounds such as saquinavir and indinavir, which at that time were used in their native form. The subsequent development of yet another HIV protease, ritonavir, has been important for the field, since it has been shown that ritonavir can significantly boost plasma levels of other PIs [44]. This has had the consequence of imparting to the newer generation of PIs a much higher genetic barrier for resistance, such that an accumulation of multiple mutations is almost always now required in order for a drug to cease being active against its intended target. This progress has only been possible as a result of continuous research over the past decades that have improved our understanding of drug mechanism of action, and pharmacokinetics and other factors responsible for the development of drug resistance. It is important as well, to recognize the contributions of social science research in this context and the fact that proper adherence to antiretroviral drug regimens has turned out to be a key factor in both the long-term efficacy of the drugs employed in therapy as well as in minimizing the likelihood that drug resistance will develop [45].

Drug resistance

Drug resistance is probably the single most critical factor responsible for failure of antiretroviral therapy and has ensured that drug companies and scientists are strongly motivated to continue to move forward with new anti-HIV drug discovery programs [46]. In recent years, the search for anti-HIV drugs has been extended to compounds that might act against cellular rather than viral targets. Indeed, one such drug now approved for therapy works by blocking the CCR5 co-receptor that is required for HIV to gain entry into susceptible CD4 lymphocytes [8]. The advantage of a drug that targets cellular proteins rather than viral enzymes is that this should theoretically minimize the likelihood that viral mutagenesis might give rise to drug resistance. Indeed, we now know that other cellular factors might also become important targets for anti-HIV chemotherapy [47]. Good examples of strong leads include the APOBEC3G series of proteins that are impacted by viral proteins such as Vif, and cellular LEDGF proteins that are essential for the proper function of the viral integrase enzyme [15, 47].

Perspectives and challenges

Looking forward, multiple challenges confront HIV-1 researchers, clinicians and policy makers. Below, we highlight a few of these issues.

Sustainability of antiretroviral treatment

The United Nations estimate that only 7% of people in developing countries have access to antiretroviral drugs, and that only 20% of people worldwide are reached by HIV-1 prevention programs [5]. At recent international conferences on AIDS, the point has repeatedly been made that more people become newly infected by HIV each year than the number that are newly able to gain access to antiretroviral drugs. The drug access that is now being provided by the Global Fund to Fight AIDS, TB, and Malaria and the US-sponsored President's Emergency Plan for AIDS Relief are having major impact in this regard [48]. Nevertheless, it must be recognized that HIV-1 drug treatment is chronic, unabated life-long therapy. Thoughtful policies that save today's lives and countenance the sustained resources needed to continue future long-term treatment have to be developed.

Prevention strategies, microbicides, and vaccines

Although HIV treatment has improved steadily since the introduction of the first antiretroviral drugs, for example, AZT and ddI, during the late 1980s, most of this progress has been limited to developed countries. In contrast, access to anti-HIV drugs continues to be problematic in most of the developing world. Sadly, for millions of HIV-infected individuals, access to anti-HIV therapy is not yet a reality [5]. Anti-HIV drugs may contribute much to the prevention of infection. As an example, it is well-known that the most important correlate of HIV transmission is the level of plasma viremia within an infected individual. Since anti-HIV drugs act to limit viral replication, they also lower plasma viremia, and it is hoped to levels that are non-detectable by very sensitive PCR assays [49]. As a consequence, patients whose plasma viremia is well suppressed can potentially be regarded as non-infectious for sexual contacts. This statement, although somewhat controversial in terms of public health recommendations, establishes rationale to treat patients on as widespread a basis as possible, in order to lower viral loads and hopefully diminish overall levels of HIV transmission. This is important, since practical progress in regard to prevention strategies involving HIV-1 has been almost negligible [50].

In regard to preventive HIV vaccines, two phase 3 clinical trials, one involving the viral protein gp160, and a second, involving a recombinant adenovirus vector containing viral proteins, have failed [51]. Indeed, the latter trial may have resulted in a greater number of individuals in the experimental rather than in the placebo arm becoming infected. There is widespread consensus that it will be at least another decade before phase 3 trials are ready to begin once again with vaccines that are designed to prevent new HIV transmissions.

Similarly, there has been frustration in regard to the use of anti-HIV vaginal microbicides to prevent infection. A large number of phase 3 clinical trials, involving non-specific polyanionic substances, have failed [50]. These have included trials of nonoyxynol-9, sulfated polysaccharides, and other substances; in which again, it appears that the women who received the anti-HIV microbicides may actually have suffered greater numbers of infections than women in the control arm of the studies, perhaps because the substances used were toxic to vaginal mucosal tissue, thus promoting infection in the experimental arm.

It is now unlikely that further studies using non-specific approaches of the sort mentioned above will be undertaken. Rather, there is strong consensus that future microbicide trials should be conducted with antiretroviral drugs that could perhaps be formulated as vaginal gels and that might protect against HIV transmission from an infected male partner. A type of compound that probably has the strongest rationale in this context is a CCR5 antagonist; several such compounds have now been shown in animal models to be protective against transmission of simian immunodeficiency virus. Whether or not CCR5 antagonists formulated as gels will also protect against HIV transmission is not yet known. In addition, there is also rationale to develop certain nucleoside(tide) and/or non-nucleoside reverse transcriptase inhibitors as anti-HIV vaginal microbicides. Research in this area is continuing apace to evaluate these various prevention strategies [52].

An approach similar to the above, also now undergoing phase 3 clinical testing, is that of pre-exposure prophylaxis (PREP) [53]. People who are susceptible to HIV infection are asked to take antiretroviral drugs on a prophylactic rather than therapeutic basis. Clinical studies now underway are evaluating the co-formulation of tenofovir together with FTC (Truvada). The reason for choosing these two drugs in combination is that both of them have relatively long half-lives, meaning that dosing is only required once per day. The hope is that these drugs will be able to interfere with early reverse transcription events that may transpire immediately after infection, thereby preventing the virus from being able to take root within the body. Results of these studies should be available within 1 to 2 years.

One issue that must be considered, however, is drug resistance. Of course, it may be difficult to assess whether or not antiretroviral drugs when used as preventives will work, if the viruses being transmitted by infected partners are themselves drug resistant. Also, there is a danger that an individual may choose to avail themselves of this PREP approach while being in the window period for seroconversion, at a time that high viral loads are present, thus representing a scenario whereby selection of drug resistance might occur. This notwithstanding, many observers now consider the PREP approach to be the most promising of all strategies for prevention of HIV transmission [52].

Pediatric infections

Almost all HIV-infected pregnant women in developed countries now receive triple therapy during the course of pregnancy to prevent mother-to-child transmission of the virus. This has resulted in the virtual elimination of the pandemic of native-born pediatric AIDS in all developed country settings. Sadly, such progress has not been realized in developing countries [5].

Circumcision

Another success story in relation to prevention has been the understanding that male circumcision may help to reduce female-to-male transmission of HIV by as much as 60% [54]. This is presumably because circumcision eliminates the foreskin that contains penile tissue rich in Langerhans cells that are thought to be important portals of entry for HIV into the body. Hence, elimination of this tissue may diminish the likelihood of transmission.

Conclusion

The theme of World AIDS Day in 2007 was 'leadership'. As AIDS moves from the 20th into the 21st century, the world today with its increased globalization is much different than it was in 1983; and over the next 25 years, it will change again. For example, several well-regarded economic sources have projected that China will have overtaken the US by 2040 to become the world's largest economy, with India capturing third place. Although China's official AIDS statistics in 2005 reveal a relatively few, 650,000, HIV-cases, against a total population of 1.3 billion people, China's future HIV numbers will undoubtedly increase significantly [55]. It seems inescapable that a long-term solution for HIV/AIDS must include a commitment of leadership and resources from China, on a scale no weaker than the recent support of the Olympic Games.

Our achievements over the past 25 years, as outlined above, have come, in part, because of outstanding, capable leaders in all facets of HIV-1 research. A concern discussed at the International AIDS Society's XVII International AIDS Conference in Mexico City was how to develop, nurture, and avoid a shortfall in new AIDS leaders. Realistically, major initiatives such as the development of successful prophylactic HIV-vaccines and effective topical antiviral microbicides require perseverance that spans a decade or more with little reason to expect short-term success. Thus, looking ahead, we must attain sustainability of economic resources in order to assure delivery of antiretroviral treatment in developing countries, while not neglecting the need to develop the new human leadership that will be essential to sustain and accelerate the global campaign against HIV/AIDS.

Abbreviations

AIDS:

acquired immunodeficiency syndrome

APOBEC3G:

apolipoprotein B mRNA-editing enzyme: catalytic polypeptide-like 3G

ESCRT:

endosomal sorting complex required for transport

HIV:

human immunodeficiency virus

PCR:

polymerase chain reaction

PI:

protease inhibitor

PREP:

pre-exposure prophylaxis

PRMT6:

protein arginine methyltransferase 6

Tat:

trans-activator of transcription

TRIM5α:

tripartite motif protein 5a.

References

  1. Gottlieb MS, Schroff R, Schanker HM, Weisman JD, Fan PT, Wolf RA, Saxon A: Pneumocystis carinii pneumonia and mucosal candidiasis in previously healthy homosexual men: evidence of a new acquired cellular immunodeficiency. N Engl J Med. 1981, 305: 1425-1431.

    Article  CAS  PubMed  Google Scholar 

  2. Barre-Sinoussi F, Chermann JC, Rey F, Nugeyre MT, Chamaret S, Gruest J, Dauguet C, Axler-Blin C, Vezinet-Brun F, Rouzioux C, Rozenbaum W, Montagnier L: Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS). Science. 1983, 220: 868-871. 10.1126/science.6189183.

    Article  CAS  PubMed  Google Scholar 

  3. Popovic M, Sarngadharan MG, Read E, Gallo RC: Detection, isolation, and continuous production of cytopathic retroviruses (HTLV-III) from patients with AIDS and pre-AIDS. Science. 1984, 224: 497-500. 10.1126/science.6200935.

    Article  CAS  PubMed  Google Scholar 

  4. Gallo RC: A reflection on HIV/AIDS research after 25 years. Retrovirology. 2006, 3: 72-10.1186/1742-4690-3-72.

    Article  PubMed  PubMed Central  Google Scholar 

  5. UNAIDS. [http://www.unaids.org/en/knowledgecentre/hivdata/epidemiology/epislides.asp]

  6. Kwong PD, Wyatt R, Robinson J, Sweet RW, Sodroski J, Hendrickson WA: Structure of an HIV gp120 envelope glycoprotein in complex with the CD4 receptor and a neutralizing human antibody. Nature. 1998, 393: 648-659. 10.1038/31405.

    Article  CAS  PubMed  Google Scholar 

  7. Stricher F, Huang CC, Descours A, Duquesnoy S, Combes O, Decker JM, Kwon YD, Lusso P, Shaw GM, Vita C, Kwong PD, Martin L: Combinatorial optimization of a CD4-mimetic miniprotein and cocrystal structures with HIV-1 gp120 envelope glycoprotein. J Mol Biol. 2008, 382: 510-524. 10.1016/j.jmb.2008.06.069.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lusso P: HIV and the chemokine system: 10 years later. EMBO J. 2006, 25: 447-456. 10.1038/sj.emboj.7600947.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Clapham PR, McKnight A: Cell surface receptors, virus entry and tropism of primate lentiviruses. J Gen Virol. 2002, 83: 1809-1829.

    Article  CAS  PubMed  Google Scholar 

  10. Alkhatib G, Combadiere C, Broder CC, Feng Y, Kennedy PE, Murphy PM, Berger EA: CC CKR5: a RANTES, MIP-1alpha, MIP-1beta receptor as a fusion cofactor for macrophage-tropic HIV-1. Science. 1996, 272: 1955-1958. 10.1126/science.272.5270.1955.

    Article  CAS  PubMed  Google Scholar 

  11. Feng Y, Broder CC, Kennedy PE, Berger EA: HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor. Science. 1996, 272: 872-877. 10.1126/science.272.5263.872.

    Article  CAS  PubMed  Google Scholar 

  12. Towers GJ: The control of viral infection by tripartite motif proteins and cyclophilin A. Retrovirology. 2007, 4: 40-10.1186/1742-4690-4-40.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Stremlau M, Owens CM, Perron MJ, Kiessling M, Autissier P, Sodroski J: The cytoplasmic body component TRIM5alpha restricts HIV-1 infection in Old World monkeys. Nature. 2004, 427: 848-853. 10.1038/nature02343.

    Article  CAS  PubMed  Google Scholar 

  14. Sheehy AM, Gaddis NC, Choi JD, Malim MH: Isolation of a human gene that inhibits HIV-1 infection and is suppressed by the viral Vif protein. Nature. 2002, 418: 646-650. 10.1038/nature00939.

    Article  CAS  PubMed  Google Scholar 

  15. Goila-Gaur R, Strebel K: HIV-1 Vif, APOBEC, and intrinsic immunity. Retrovirology. 2008, 5: 51-10.1186/1742-4690-5-51.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Yeung ML, Benkirane M, Jeang KT: Small non-coding RNAs, mammalian cells, and viruses: regulatory interactions?. Retrovirology. 2007, 4: 74-10.1186/1742-4690-4-74.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Scaria V, Hariharan M, Maiti S, Pillai B, Brahmachari SK: Host-virus interaction: a new role for microRNAs. Retrovirology. 2006, 3: 68-10.1186/1742-4690-3-68.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Xie B, Invernizzi CF, Richard S, Wainberg MA: Arginine methylation of the human immunodeficiency virus type 1 Tat protein by PRMT6 negatively affects Tat interactions with both cyclin T1 and the Tat transactivation region. J Virol. 2007, 81: 4226-4234. 10.1128/JVI.01888-06.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Invernizzi CF, Xie B, Richard S, Wainberg MA: PRMT6 diminishes HIV-1 Rev binding to and export of viral RNA. Retrovirology. 2006, 3: 93-10.1186/1742-4690-3-93.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Newman EN, Holmes RK, Craig HM, Klein KC, Lingappa JR, Malim MH, Sheehy AM: Antiviral function of APOBEC3G can be dissociated from cytidine deaminase activity. Curr Biol. 2005, 15: 166-170. 10.1016/j.cub.2004.12.068.

    Article  CAS  PubMed  Google Scholar 

  21. Marin M, Rose KM, Kozak SL, Kabat D: HIV-1 Vif protein binds the editing enzyme APOBEC3G and induces its degradation. Nat Med. 2003, 9: 1398-1403. 10.1038/nm946.

    Article  CAS  PubMed  Google Scholar 

  22. Yedavalli VR, Jeang KT: Methylation: a regulator of HIV-1 replication?. Retrovirology. 2007, 4: 9-10.1186/1742-4690-4-9.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Grassmann R, Jeang KT: The roles of microRNAs in mammalian virus infection. Biochim Biophys Acta. 2008, 1779: 706-11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Sherman MP, Greene WC: Slipping through the door: HIV entry into the nucleus. Microbes Infect. 2002, 4: 67-73. 10.1016/S1286-4579(01)01511-8.

    Article  CAS  PubMed  Google Scholar 

  25. Iordanskiy S, Berro R, Altieri M, Kashanchi F, Bukrinsky M: Intracytoplasmic maturation of the human immunodeficiency virus type 1 reverse transcription complexes determines their capacity to integrate into chromatin. Retrovirology. 2006, 3: 4-10.1186/1742-4690-3-4.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Berkhout B, Jeang KT: Functional roles for the TATA promoter and enhancers in basal and Tat-induced expression of the human immunodeficiency virus type 1 long terminal repeat. J Virol. 1992, 66: 139-149.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Berkhout B, Silverman RH, Jeang KT: Tat trans-activates the human immunodeficiency virus through a nascent RNA target. Cell. 1989, 59: 273-282. 10.1016/0092-8674(89)90289-4.

    Article  CAS  PubMed  Google Scholar 

  28. Dingwall C, Ernberg I, Gait MJ, Green SM, Heaphy S, Karn J, Lowe AD, Singh M, Skinner MA, Valerio R: Human immunodeficiency virus 1 tat protein binds trans-activation-responsive region (TAR) RNA in vitro. Proc Natl Acad Sci USA. 1989, 86: 6925-6929. 10.1073/pnas.86.18.6925.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Garber ME, Wei P, KewalRamani VN, Mayall TP, Herrmann CH, Rice AP, Littman DR, Jones KA: The interaction between HIV-1 Tat and human cyclin T1 requires zinc and a critical cysteine residue that is not conserved in the murine CycT1 protein. Genes Dev. 1998, 12: 3512-3527. 10.1101/gad.12.22.3512.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Marshall NF, Price DH: Control of formation of two distinct classes of RNA polymerase II elongation complexes. Mol Cell Biol. 1992, 12: 2078-2090.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lever AM, Jeang KT: Replication of human immunodeficiency virus type 1 from entry to exit. Int J Hematol. 2006, 84: 23-30. 10.1532/IJH97.06112.

    Article  CAS  PubMed  Google Scholar 

  32. Yedavalli VS, Neuveut C, Chi YH, Kleiman L, Jeang KT: Requirement of DDX3 DEAD box RNA helicase for HIV-1 Rev-RRE export function. Cell. 2004, 119: 381-392. 10.1016/j.cell.2004.09.029.

    Article  CAS  PubMed  Google Scholar 

  33. Butsch M, Boris-Lawrie K: Destiny of unspliced retroviral RNA: ribosome and/or virion?. J Virol. 2002, 76: 3089-3094. 10.1128/JVI.76.7.3089-3094.2002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Dorman N, Lever A: Comparison of viral genomic RNA sorting mechanisms in human immunodeficiency virus type 1 (HIV-1), HIV-2, and Moloney murine leukemia virus. J Virol. 2000, 74: 11413-11417. 10.1128/JVI.74.23.11413-11417.2000.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Zeffman A, Hassard S, Varani G, Lever A: The major HIV-1 packaging signal is an extended bulged stem loop whose structure is altered on interaction with the Gag polyprotein. J Mol Biol. 2000, 297: 877-893. 10.1006/jmbi.2000.3611.

    Article  CAS  PubMed  Google Scholar 

  36. De Guzman RN, Wu ZR, Stalling CC, Pappalardo L, Borer PN, Summers MF: Structure of the HIV-1 nucleocapsid protein bound to the SL3 psi-RNA recognition element. Science. 1998, 279: 384-388. 10.1126/science.279.5349.384.

    Article  CAS  PubMed  Google Scholar 

  37. Pelchen-Matthews A, Kramer B, Marsh M: Infectious HIV-1 assembles in late endosomes in primary macrophages. J Cell Biol. 2003, 162: 443-455. 10.1083/jcb.200304008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ono A, Freed EO: Cell-type-dependent targeting of human immunodeficiency virus type 1 assembly to the plasma membrane and the multivesicular body. J Virol. 2004, 78: 1552-1563. 10.1128/JVI.78.3.1552-1563.2004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Martin-Serrano J, Zang T, Bieniasz PD: Role of ESCRT-I in retroviral budding. J Virol. 2003, 77: 4794-4804. 10.1128/JVI.77.8.4794-4804.2003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Mellors JW, Munoz A, Giorgi JV, Margolick JB, Tassoni CJ, Gupta P, Kingsley LA, Todd JA, Saah AJ, Detels R, Phair JP, Rinaldo CR: Plasma viral load and CD4+ lymphocytes as prognostic markers of HIV-1 infection. Ann Intern Med. 1997, 126: 946-954.

    Article  CAS  PubMed  Google Scholar 

  41. Brenner BG, Roger M, Routy JP, Moisi D, Ntemgwa M, Matte C, Baril JG, Thomas R, Rouleau D, Bruneau J, Leblanc R, Legault M, Tremblay C, Charest H, Wainberg MA: High rates of forward transmission events after acute/early HIV-1 infection. J Infect Dis. 2007, 195: 951-959. 10.1086/512088.

    Article  CAS  PubMed  Google Scholar 

  42. Murphy EL, Collier AC, Kalish LA, Assmann SF, Para MF, Flanigan TP, Kumar PN, Mintz L, Wallach FR, Nemo GJ, for the Viral Activation Transfusion Study Investigation: Highly active antiretroviral therapy decreases mortality and morbidity in patients with advanced HIV disease. Ann Intern Med. 2001, 135: 17-26.

    Article  CAS  PubMed  Google Scholar 

  43. Palella FJ, Delaney KM, Moorman AC, Loveless MO, Fuhrer J, Satten GA, Aschman DJ, Holmberg SD: Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med. 1998, 338: 853-860. 10.1056/NEJM199803263381301.

    Article  PubMed  Google Scholar 

  44. Martinez-Cajas JL, Wainberg MA: Antiretroviral therapy: optimal sequencing of therapy to avoid resistance. Drugs. 2008, 68: 43-72.

    Article  CAS  PubMed  Google Scholar 

  45. Walmsley S, Bernstein B, King M, Arribas J, Beall G, Ruane P, Johnson M, Johnson D, Lalonde R, Japour A, Brun S, Sun E: Lopinavir-ritonavir versus nelfinavir for the initial treatment of HIV infection. N Engl J Med. 2002, 346: 2039-2046. 10.1056/NEJMoa012354.

    Article  CAS  PubMed  Google Scholar 

  46. Larder BA, Kemp SD: Multiple mutations in HIV-1 reverse transcriptase confer high-level resistance to zidovudine (AZT). Science. 1989, 246: 1155-1158. 10.1126/science.2479983.

    Article  CAS  PubMed  Google Scholar 

  47. Lama J, Planelles V: Host factors influencing susceptibility to HIV infection and AIDS progression. Retrovirology. 2007, 4: 52-10.1186/1742-4690-4-52.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Institute of Medicine: PEPFAR Implementation: Progress and Promise. Committee for the Evaluation of the President's Emergency Plan for AIDS Relief (PEPFAR) Implementation. Washington. 2007

    Google Scholar 

  49. Quinn TC, Wawer MJ, Sewankambo N, Serwadda D, Li C, Wabwire-Mangen F, Meehan MO, Lutalo T, Gray RH: Viral load and heterosexual transmission of human immunodeficiency virus type 1. Rakai Project Study Group. N Engl J Med. 2000, 342: 921-929. 10.1056/NEJM200003303421303.

    Article  CAS  PubMed  Google Scholar 

  50. Grant RM, Hamer D, Hope T, Johnston R, Lange J, Lederman MM, Lieberman J, Miller CJ, Moore JP, Mosier DE, Richman DD, Schooley RT, Springer MS, Veazey RS, Wainberg MA: Whither or wither microbicides?. Science. 2008, 321: 532-534. 10.1126/science.1160355.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Fauci AS, Johnston MI, Dieffenbach CW, Burton DR, Hammer SM, Hoxie JA, Martin M, Overbaugh J, Watkins DI, Mahmoud A, Greene WC: HIV vaccine research: the way forward. Science. 2008, 321: 530-532. 10.1126/science.1161000.

    Article  CAS  PubMed  Google Scholar 

  52. Cohen M: Prevention of the sexual transmission of HIV-1: preparing for success. J Inform Security.

  53. Youle M, Wainberg MA: Could chemoprophylaxis be used as an HIV prevention strategy while we wait for an effective vaccine?. AIDS. 2003, 17: 937-938. 10.1097/00002030-200304110-00027.

    Article  PubMed  Google Scholar 

  54. Auvert B, Marseille E, Korenromp EL, Lloyd-Smith J, Sitta R, Taljaard D, Pretorius C, Williams B, Kahn JG: Estimating the resources needed and savings anticipated from roll-out of adult male circumcision in Sub-Saharan Africa. PLoS ONE. 2008, 3: e2679-10.1371/journal.pone.0002679.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Shao Y: AIDS epidemic at age 25 and control efforts in China. Retrovirology. 2006, 3: 87-10.1186/1742-4690-3-87.

    Article  PubMed  PubMed Central  Google Scholar 

Pre-publication history

Download references

Acknowledgements

Research in the laboratory of K-TJ is supported by intramural funds from the National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), and the Intramural AIDS Targeted Antiretroviral Program from the Office of the Director, NIH; research in the laboratory of MAW is supported by grants from the Canadian Institutes for Health Research, the NIH and the International Partnership on Microbicides

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Kuan-Teh Jeang.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

Both MAW and KTJ contributed equally to the writing of this commentary.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Wainberg, M.A., Jeang, KT. 25 years of HIV-1 research – progress and perspectives. BMC Med 6, 31 (2008). https://doi.org/10.1186/1741-7015-6-31

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1741-7015-6-31

Keywords