Volume 13, number 4
 Views: (Visited 480 times, 1 visits today)    PDF Downloads: 1420

Alieh Farshbaf A, Esmaeilzadeh A. CCR5 as a Novel Cell and Gene Therapy Strategies Based on Induction of Resistance to HIV. Biosci Biotech Res Asia 2016;13(4).
Manuscript received on : 11 February 2016
Manuscript accepted on : 17 April 2016
Published online on:  --

Plagiarism Check: Yes

How to Cite    |   Publication History    |   PlumX Article Matrix

CCR5 as a Novel Cell and Gene Therapy Strategies Based on Induction of Resistance to HIV

Alieh Farshbafand Abdolreza Esmaeilzadeh2,3

1Department of Genetic and Molecular Medicine, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.

2Department of Immunology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.

3Cancer Gene Therapy Research Center, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.

Corresponding Author E-mail: a46reza@zums.ac.ir 

DOI : http://dx.doi.org/10.13005/bbra/2377

ABSTRACT: Cc-chemokine receptor-5 (CCR5) is known as a main co-receptor in human immunodeficiency virus-1 (HIV-1) infection. So, it could be a target for inhibition of HIV-1 entry into CD 4+ immune cells. Many studies showed homozygote individual with 32bp deletion in CCR5 gene had nature resistance to HIV-1. In this manner, recent treatments are focused on inducing this resistance to HIV-1 infected patients withCCR5. Berlin and Boston patients transplanted with allogeneic hematopoietic stem cell (HSC) and demonstrated effective cure for HIV-1 infection. In addition, zinc finger nuclease (ZFN) eliminated some problems of Berlin and Boston patients by site-specific CCR5 gene modification. These recent strategies declined highly-active anti-retroviral therapy (HAART) restrictions such as toxicity, low safety, the side effects following long-term consuming and virus reloading immediately after cut the drugs off. In this review, in addition of introductorybiologic and immune-genetic roles of CCR5, we consider novel treatment strategies for HIV-1 infected patient by CCR5gene targeted therapy.

KEYWORDS: CCR5, HIV-1; hematopoietic stem cell therapy; gene therapy; genemodification; nuclease

Download this article as: 
Copy the following to cite this article:

Alieh Farshbaf A, Esmaeilzadeh A. CCR5 as a Novel Cell and Gene Therapy Strategies Based on Induction of Resistance to HIV. Biosci Biotech Res Asia 2016;13(4).

Copy the following to cite this URL:

Alieh Farshbaf A, Esmaeilzadeh A. CCR5 as a Novel Cell and Gene Therapy Strategies Based on Induction of Resistance to HIV. Biosci Biotech Res Asia 2016;13(4). Available from: https://www.biotech-asia.org/?p=16562

Introduction

It is demonstratedthata cc-chemokine receptor (CCR)-based targeted therapy canplaycritical role in more effective cure of patients infected by human immune-deficiency virus-1 (HIV-1) [1-4]. CCRs are a group ofG-protein coupled family of receptors(GPCR) andapproximately 800 genes have beenintroduced that encoded functional GPCR and made up about %1 of human genome[5, 6].It is demonstrated that about half of clinical drugs designCCR5 as a Novel Cell and Gene Therapy for GPCRs, with blocking their ligands or increasing ligands accessibility. Sotheir rolesin our bodiesare very important[7].

Human GPCRs consist of six families whose major groups are A,B and C andtheir largest group is A family. One of the most popular subfamilies of class A is CCR5 which for many years hasattracted scientists. The vast majority of investigations have focused to provide the novel approaches to the treatment of HIV based on targeted therapy. We have previously suggested induction of resistance to HIV by CCR5 gene therapy and stem cell transplantation [8, 9]. Here we present most recently discovered approaches to the treatment of HIV, based on CCR5, which may define the better treatment, especially target therapy.

CCR5 genetic basis, structure and molecular signaling

CCR5 is encoded by CMKBR5 gene located on p21.31 region of human chromosome 3[10].Protein productof this gene includes 3 sections[11, 12]: 1) A seven helical trans-membrane domain which provide 3 extra-cellular and 3 intra-cellular hydrophilic loops. 2) C-terminal residue that regulates receptor by serine-threonine phosphorylation. 3) N-terminal residue that is bound to ligand. MIP 1α,β (also knownas CCL3, CCL4), RANTES (also known as CCL5) and the other β-chemokines areligands that are bound to NT site in CCR5 [12, 13]. After ligand binding to NT or 3 extracellular loops, a molecular mechanism activate that cause receptor conformation change and the dissociation of G-protein subunits. Gα-GTP and Gβγ subunits are freed and regulate enzymesactivity such as adenylate cyclase, phospholipase C isoforms and ion channels. These enzymes regulate protein kinase function, finally increase intracellular ca2+ ions and stimulate chemotaxis[14].Pharmacologist have named GPCR as “spare receptor” because full biologic response happen after 5%occupancy [15]. Thus GPCRs associatewithcell metabolism, growth, migration, differentiation and death of multiple cell type (apoptosis)[16, 17].

Resistance to HIV-1 infection byCCR5

Any change in the sequence of gene variants causes alternation in HIV infection or AIDS process, named as AIDS Restricted Gene (ARG). Some of these variants are detected in special group population which causes natural selection and/or genetic drift different ARG polymorphism/mutation. To this date, more than 35 ARGs have been identified. First of them is CCR5Δ32 and [18-22].This32bp deletion(CCR5Δ32) in the single-coding exon provides resistance mechanism to HIV/AIDS infection[12, 23].CCR5Δ32 is caused by replicative slippage by RNA pol due to the presence of a direct repeat flanking the deleted region which leads to elimination of second extracellular loop on receptor. Therefore, HIV-1 enters to bloodstream but could not infect CD4+ T cell and macrophage[12]. A number of studieshave shown thatindividual with homozygote CCR5Δ32allele exhibits a natural resistance to HIV and Acquired immunodeficiency syndrome(AIDS)[12, 23, 24].It is also worth mentioning that heterozygote individual after exposing to human immunodeficiency virus display slower progression to AIDSthan homozygote individualfor the wild type allele [25, 26].In Northern Europe, Caucasian population indicate the highestCCR5Δ32, 20% of whom are heterozygote and 1% homozygote for this mutation, respectively [27]. Because of these remarkable detections, CCR5 isknown as a major co-receptor, even when immune-cells presentCD4 on their surface.If CCR5 isnot presentedon cell surface (following knock-out it or when using its inhibitors) HIV-1couldnot contaminate cells [28, 29].

CCR5Δ32is highly resistant to HIV-1 infection, but not completely. SinceCXCR4is another co-receptor for HIV-1 entries. So CCR5 is not the only co-receptor for HIV-1 infection and one of the reasons that HIV/AIDS have epidemiological heterogeneity is genetic variants in host receptor and co-receptors (CCR5 or CXCR4) that HIV can switch itself to per variant[30].

resistance to HIV-1 infection or delay to AIDSprocess byAnother polymorphisms

In addition to CCR5Δ32,Some other mutations/polymorphisms have been discoveredwhichprovidethe resistance mechanism, suchasCCR5/m303 (table 1)[31], IDH1C[32]. But although some ofthe otherslikeCCR5Δ32 heterozygote are susceptible to HIV infection,theydelay the AIDS progression,such as CCR2 64I[33], SDF1 3A[25], HLA B*57[18], HLA B27[34], KIR 3DS1[35, 36], PROX1 Hap-CGT[37], ACSM4 A[38].

Thus, the variant of host genotype may change the function or alternate gene expression after HIV-1 contamination, known as HIV-dependency factors (HDFs). HDFs are needed for HIV-1 infection process, transmission, viral loading and challenge with immune-system[39]. However, host genetic background is a part of factors that affect HIV-1infection. The others are HIV acquisition, immune-system condition and HAART (highly-active-anti-retroviral therapy)results.

Current therapeutic methods: benefits and limitations

To date, several drugs have been introduced for inhibition of AIDS progression, like enfuviritude (T20) and maraviroc. Maravirocisa co-receptor antagonistblocks interaction between CCR5 and envelope (env) proteincoating HIV-1 surface[40].Enfuviritudesact asfusion inhibitordisrupting conformation change in glycoprotein-41 (gp41)[40]. In addition to,highly active anti-retroviral therapy (HAART)is emerging which targets virus enzymes (Table 2). It isshowed that ARThave significantpositive effect on the reduction of latent reservoir virus in immune system atan early stage or immediately after informing of HIV-1 infection. For example,when an infant was burn from a mother that infected by HIV-1,after 30 hours of birth undergoneART and continued until 18 months. In the months30, no pro-viral DNA or plasma RNA of HIV was detected in peripheral mononuclear cells[41].

One of the problems in HIV-1 treatment by ART is “drugs resistance”. This phenomenon mainly caused by new mutation patternsin HIV-1 genes that virus need for its essential proteins like protease[42]. Hence,HAART could not eradicate virus, although reduce its replication significance. In addition, theyshouldbe used long timeand maybe discontinueof themin any time causeRNA and DNA virusrebounding from latent reservoirs. Moreover, HAART cannot act in different individuals by the same efficiency. High cost and some side effects following long-term therapy are another restrictionthe use of HAART[43, 44].

Recent treatment strategies are focused on gene therapy especially against HIV and other refractory disease, we present some study in this field previously[8, 9, 45-49]. Here, we present most recently discovered approaches to the treatment of HIV, based on CCR5, which may define the better treatment, especially target therapy.

Novel therapeutics approaches:stem cell transplantation and gene-modification

Novel treatment strategies with stem cell transplantation (SCT) overcome to some problems that observed in ART[8, 9].Successful allogeneic SCT haveperformed for acute myeloid leukemia (AML) in a patient co-infected by HIV and HCV andhad been undergone HAART in 2002[50]. Following this, Hutteret al. in 2009 transplanted allogeneic (CCR5 Δ32/Δ32) stem cell as a treatment for “Berlin patient”whowas suffering from AML and HIV infection[1]. The patient was undergoing HAART for 10 years and discontinued them after SCT[1].Rebounding of HIV may beobserved if HAART discontinued because of another CXCR4 co-receptorexistence[51], but in this patient after 20 months not observed signs of virus activating or replicating [1].Hutteret al. sequenced CXCR4 patient and checked its variants, but not detected any trail of CXCR4 and concluded HIV-1 in this patient was not binding with CXCR4 as a co-receptor. Number of CD 4+ T-cells after HSCT for CCR5 Δ32/Δ32 increased such as normal range in health people and HIV DNA or RNA was undetectable gradually [1]. Result of this treatment showedno existence of RNA or DNA virusafter 3.5 years, even when HAART had been discontinued[2, 52].Likewise, it was reported Berlin patient body remained free of HIV-1 after 5.5 years[53].

In 2012,Henrichet al. performed HSCT for two patients, Boston patients,who have heterozygote genotype forCCR5 (CCR5 WT/CCR5Δ32) and transplanted with WT CCR5 cells. Result of this study suggested that replication of DNA or RNA virus had been suppressed and HIV-1 reservoir reducedafter transplantation [3].

Although results of Hutteret al. and Henrichet al. made a revolution in HIV-1 treatment, but there were problems such as low frequency of homozygote CCR5Δ32 in population and founding a suitable HLA match donor with target patient. These problems encouraged Duarteet al. in 2015 to transplanted hematopoietic stem cell of umbilical cord blood (CB) to a patient infected by HIV-1 from CCR5 -/- donor that have AML [54-56]. HCT from CB not needed to stringent HLA matching like as HSCT with bone marrow. This study showed peripheral mononuclear cells were resistance to HIV-1 infection.

As describe above, artificial disruption methodswith “CCR5gene modification”were exerted widely and eliminated some problems of allogeneic HSCT[57]. Gene modification is permanent, inheritable and transmitted HIV-1 resistant cells to next generation.

For example for site-specific nuclease,zinc finger protein (ZFP) surveyed extensive. ZFP is a transcription factor that binding to Fok I restriction enzyme domains with their zinc finger motifs and provide zinc finger nuclease (ZFN)[58, 59].Theoretically, following DSB in DNA by Fok I, repair DNA mechanism applied error prone non-homologous end joining (NHEJ) [60] or homology directly repair by homologous recombination (HR) [61]. Then, small nucleotide deletion or addition observed and result in disruption of reading frame and gene expression. It could engineer with nucleases artificially. Predominant repair with ZFN is error-prone NHEJ.

First in 2005, alternation ofCCR5 with ZFN in in vitro conditionwas showed [62]. ThenCCR5 disruption by ZFN in mouse model was applied and exhibited specify and sufficient in vivofunctions for induction of resistance to HIV[63].In this manner, genetic modification with ZFNs utilized in various living organism and cell lines;Including primary T cell, HSC and humanize mice[64-67].Afterwards, CCR5 was modified by ZFN with adenovirus vector on ex vivo condition and performance wasefficiently in healthy and HIV infected CD4+ T cellin 2013[68].Knock out ofCCR5 by ZFP modification artificiallyand infusion of autologous CD4+ T cell transplantation showed safety and immune reconstitution with increasing in CD4+T cells,In 2014[4]. In this studyDNA virus level decreased in most patients and RNA virus level were undetectable in one patient andmodified T cells for CCR5were stable[4].

Another kind of site-specific nuclease is TALEN (Transcription Activator-like Effectors Nuclease). In comparison toZFN, TALEN has lower cytotoxicity and reduce off-target activityin CCR5 locus. But both of them could disrupt genes about 45%[69].In comparison toZFN more delivery problems observed, due to largeTALEN protein size. But had been showed that TALEN expressed by adenoviral vector [70].

In addition to ZFN andTALEN, CRISPR/Cas9 is another targeted gene disruption in HIV therapy.Clustered regularly interspaced palindromic repeats (CRISPRs) are short direct repeat (21-47 nt) with vary intervening spacer sequence that surround by CRISPR associated gene (Cas9) in bacteria. When CRISPR istranscribed, pre-crRNA converts to crRNA byRNase III and associate with trans-acting RNA (tracRNA) that diagnose target DNA.Then, this RNA duplex bind to Cas9 and the crRNA guide complex to target DNA that is complementary to spacer sequence. This ribonucleotide complex cleave target DNA with Cas9 and cause DSB[71].Some studies suggested CRISPR/Cas9 could efficientlyablate the viral genome from latently HIV-1 infected cells [72, 73]. CCR5 in primary human CD4+ T cells and CD34+ hematopoietic is targeted progenitor and stem cells and demonstrated ablate viral genes with minimal off-target mutagenesis by CRISPR/Cas9,In 2014[74].

T cell recovery and suppression of HIV-1 are achieved by gene modification and/or cell therapy. Recent approaches provide effective cure of HIV-1 but there are some challenges in CCR5 candidate for HIV treatment and need to more investigations.

Table 1: CCR5 polymorphisms that affected HIV-1infection

Type of polymorphism Heterozygote

Or Homozygote

Effect on

HIV/AIDS

CCR5*Δ32/Δ32 Homozygote Create resistance to HIV-1 infection.
CCR5Δ32/WT Heterozygote Infected by HIV-1 but delay to AIDS for 2-4 years.
CCR5Δ32/**m303 Heterozygote Create resistance to HIV-1 infection.

 

*CCR5Δ32: 32bp deletion in exon 1 CCR5 gene that cause second extracellular loop be defected and truncate protein created.

**CCR5 m303 (C101X): Transversion of T to A in 303 nucleotide open reading frame that created nonsense mutation in 101 amino-acid at first extracellular loop CCR5.

 

Table 2: Some of the current drugs for HIV treatment

Drug(s) Function Reference
Maraviroc HIV entry inhibitor

(CCR5 antagonist)

[40]
Enfuviritude HIV entry inhibitor

(fusion inhibitor)

[40]
Abacavir,  Didanosine,  Emtricitabine,  Lamivudine,  Stavudine,  Tenofovirdisoproxil fumarate (DF),  Zalcitabine,  Zidovudine Nucleotide reverse transcriptase (RT) inhibitors [75]
Rilpivirine, Nevirapine,  Etravirine,  Efavirenz,  Delavirdine Non-nucleotide reverse transcriptase (RT) inhibitors [75]
Raltegravir and Elvitegravir Integrase inhibitor [75]
Vorinostat Histone deacetylase inhibitor [76]
Atazanavir, Darunavir, Fosamprenavir, Indinavir, Lopinavir/ritonavir, Nelfinavir, Ritonavir, Saquinavir, Tipranavir Protease inhibitor [75]
Elvitegravir/Cobicistat/Emtricitabine/Tenofovir combinations of reverse transcriptase and integrase inhibitors [75]
Abacavir/Lamivudine

Abacavir/Lamivudine/Zidovudine

Emtricitabine/Tenofovir DF

Lamivudine/Zidovudine

Reverse transcriptase inhibitors (fixed-dose combinations of nucleotide analogues) [75]
Efavirenz/Emtricitabine/Tenofovir DF

Emtricitabine/Rilpivirine/Tenofovir DF

Reverse transcriptase inhibitors (fixed-dose combinations of both types of inhibitors) [75]

 

Conclusion

Results of many studies have shown that CCR5 can be a therapeutic target for treatment of HIV-1.A new horizon of stem cell therapy (such as cord blood stem cell) is shifted to obtain more effective and easier methods that can apply for many people with no HLA-matching problems and donor finding restrictions. We suggest identification of another unknown polymorphism to apply more effective treatment of HIVby find out more genetic factors that promote or restrict HIV replication. We think also it would be interesting to study the therapeutic effect of autologous embryonic stem cell transplantation that modify by human artificial chromosome (HAC) and ZFN gene. It can cleavages CCR5 gene specifically and induces the resistance to HIV-1 infection.

References

  1. Hutter G, Nowak D, Mossner M, Ganepola S, Mussig A, et al. (2009) Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N Engl J Med. 360: 692-8.
    CrossRef
  2. Hutter G, Thiel E (2011) Allogeneic transplantation of CCR5-deficient progenitor cells in a patient with HIV infection: an update after 3 years and the search for patient no. 2. AIDS. 25: 273-4.
    CrossRef
  3. Henrich TJ, Hu Z, Li JZ, Sciaranghella G, Busch MP, et al. (2013) Long-term reduction in peripheral blood HIV type 1 reservoirs following reduced-intensity conditioning allogeneic stem cell transplantation. J Infect Dis. 207: 1694-702.
    CrossRef
  4. Tebas P, Stein D, Tang WW, Frank I, Wang SQ, et al. (2014) Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N Engl J Med. 370: 901-10.
    CrossRef
  5. Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, et al. (2001) Initial sequencing and analysis of the human genome. Nature. 409: 860-921.
    CrossRef
  6. Venter JC, Adams MD, Myers EW, Li PW, Mural RJ, et al. (2001) The sequence of the human genome. Science. 291: 1304-51.
    CrossRef
  7. Flower DR (1999) Modelling G-protein-coupled receptors for drug design. Biochim Biophys Acta. 1422: 207-34.
    CrossRef
  8. Esmaeilzadeh A, Farshbaf A, Erfanmanesh M (2015) Autologous Hematopoietic Stem Cells transplantation and genetic modification of CCR5 m303/m303 mutant patient for HIV/AIDS. Medical Hypotheses. 84: 216-18.
    CrossRef
  9. Esmaeilzadeh A, Farshbaf A (2015) Novel Approaches Based on Autologous Stem Cell Engineering and Gene-Modification; Evidence for the Cure of HIV/AIDS. Genetic Syndromes & Gene Therapy. 6:1.
    CrossRef
  10. Samson M, Soularue P, Vassart G, Parmentier M (1996) The genes encoding the human CC-chemokine receptors CC-CKR1 to CC-CKR5 (CMKBR1-CMKBR5) are clustered in the p21.3-p24 region of chromosome 3. Genomics. 36: 522-6.
    CrossRef
  11. Samson M, Labbe O, Mollereau C, Vassart G, Parmentier M (1996) Molecular cloning and functional expression of a new human CC-chemokine receptor gene. Biochemistry. 35: 3362-7.
    CrossRef
  12. Samson M, Libert F, Doranz BJ, Rucker J, Liesnard C, et al. (1996) Resistance to HIV-1 infection in caucasian individuals bearing mutant alleles of the CCR-5 chemokine receptor gene. Nature. 382: 722-5.
    CrossRef
  13. Cocchi F, DeVico AL, Garzino-Demo A, Arya SK, Gallo RC, et al. (1995) Identification of RANTES, MIP-1 alpha, and MIP-1 beta as the major HIV-suppressive factors produced by CD8+ T cells. Science. 270: 1811-5.
    CrossRef
  14. Abbas W, Herbein G (2014) Plasma membrane signaling in HIV-1 infection. Biochim Biophys Acta. 1838: 1132-42.
    CrossRef
  15. Luttrell LM (2008) Reviews in molecular biology and biotechnology: transmembrane signaling by G protein-coupled receptors. Mol Biotechnol. 39: 239-64.
    CrossRef
  16. Murdoch C, Finn A (2000) Chemokine receptors and their role in inflammation and infectious diseases. Blood. 95: 3032-43.
  17. Vlahakis SR, Villasis-Keever A, Gomez T, Vanegas M, Vlahakis N, et al. (2002) G protein-coupled chemokine receptors induce both survival and apoptotic signaling pathways. J Immunol. 169: 5546-54.
    CrossRef
  18. Carrington M, O’Brien SJ (2003) The influence of HLA genotype on AIDS. Annu Rev Med. 54: 535-51.
    CrossRef
  19. An P, Winkler CA (2010) Host genes associated with HIV/AIDS: advances in gene discovery. Trends Genet. 26: 119-31.
    CrossRef
  20. Hutcheson HB, Lautenberger JA, Nelson GW, Pontius JU, Kessing BD, et al. (2008) Detecting AIDS restriction genes: from candidate genes to genome-wide association discovery. Vaccine. 26: 2951-65.
    CrossRef
  21. O’Brien SJ, Nelson GW (2004) Human genes that limit AIDS. Nat Genet. 36: 565-74.
    CrossRef
  22. O’Brien SJ, Hendrickson SL (2013) Host genomic influences on HIV/AIDS. Genome Biol. 14: 201.
    CrossRef
  23. Liu R, Paxton WA, Choe S, Ceradini D, Martin SR, et al. (1996) Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiply-exposed individuals to HIV-1 infection. Cell. 86: 367-77.
    CrossRef
  24. Zagury D, Lachgar A, Chams V, Fall LS, Bernard J, et al. (1998) C-C chemokines, pivotal in protection against HIV type 1 infection. Proc Natl Acad Sci U S A. 95: 3857-61.
    CrossRef
  25. Dean M, Carrington M, Winkler C, Huttley GA, Smith MW, et al. (1996) Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene. Hemophilia Growth and Development Study, Multicenter AIDS Cohort Study, Multicenter Hemophilia Cohort Study, San Francisco City Cohort, ALIVE Study. Science. 273: 1856-62.
    CrossRef
  26. Huang Y, Paxton WA, Wolinsky SM, Neumann AU, Zhang L, et al. (1996) The role of a mutant CCR5 allele in HIV-1 transmission and disease progression. Nat Med. 2: 1240-3.
    CrossRef
  27. Naif HM (2013) Pathogenesis of HIV Infection. Infect Dis Rep. 5: e6.
    CrossRef
  28. Deng H, Liu R, Ellmeier W, Choe S, Unutmaz D, et al. (1996) Identification of a major co-receptor for primary isolates of HIV-1. Nature. 381: 661-6.
    CrossRef
  29. Dragic T, Litwin V, Allaway GP, Martin SR, Huang Y, et al. (1996) HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature. 381: 667-73.
    CrossRef
  30. Arenzana-Seisdedos F, Parmentier M (2006) Genetics of resistance to HIV infection: Role of co-receptors and co-receptor ligands. Semin Immunol. 18: 387-403.
    CrossRef
  31. Quillent C, Oberlin E, Braun J, Rousset D, Gonzalez-Canali G, et al. (1998) HIV-1-resistance phenotype conferred by combination of two separate inherited mutations of CCR5 gene. Lancet. 351: 14-8.
    CrossRef
  32. Chinn LW, Tang M, Kessing BD, Lautenberger JA, Troyer JL, et al. (2010) Genetic associations of variants in genes encoding HIV-dependency factors required for HIV-1 infection. J Infect Dis. 202: 1836-45.
    CrossRef
  33. Smith MW, Dean M, Carrington M, Winkler C, Huttley GA, et al. (1997) Contrasting genetic influence of CCR2 and CCR5 variants on HIV-1 infection and disease progression. Hemophilia Growth and Development Study (HGDS), Multicenter AIDS Cohort Study (MACS), Multicenter Hemophilia Cohort Study (MHCS), San Francisco City Cohort (SFCC), ALIVE Study. Science. 277: 959-65.
    CrossRef
  34. Gao X, Bashirova A, Iversen AK, Phair J, Goedert JJ, et al. (2005) AIDS restriction HLA allotypes target distinct intervals of HIV-1 pathogenesis. Nat Med. 11: 1290-2.
    CrossRef
  35. Martin MP, Gao X, Lee JH, Nelson GW, Detels R, et al. (2002) Epistatic interaction between KIR3DS1 and HLA-B delays the progression to AIDS. Nat Genet. 31: 429-34.
    CrossRef
  36. Martin MP, Qi Y, Gao X, Yamada E, Martin JN, et al. (2007) Innate partnership of HLA-B and KIR3DL1 subtypes against HIV-1. Nat Genet. 39: 733-40.
    CrossRef
  37. Herbeck JT, Gottlieb GS, Winkler CA, Nelson GW, An P, et al. (2010) Multistage genomewide association study identifies a locus at 1q41 associated with rate of HIV-1 disease progression to clinical AIDS. J Infect Dis. 201: 618-26.
    CrossRef
  38. Hendrickson SL, Lautenberger JA, Chinn LW, Malasky M, Sezgin E, et al. (2010) Genetic variants in nuclear-encoded mitochondrial genes influence AIDS progression. PLoS One. 5: e12862.
    CrossRef
  39. Hutter G, Bluthgen C, Neumann M, Reinwald M, Nowak D, et al. (2013) Coregulation of HIV-1 dependency factors in individuals heterozygous to the CCR5-delta32 deletion. AIDS Res Ther. 10: 26.
    CrossRef
  40. Haqqani AA, Tilton JC (2013) Entry inhibitors and their use in the treatment of HIV-1 infection. Antiviral Res. 98: 158-70.
    CrossRef
  41. Persaud D, Gay H, Ziemniak C, Chen YH, Piatak M, Jr., et al. (2013) Absence of detectable HIV-1 viremia after treatment cessation in an infant. N Engl J Med. 369: 1828-35.
    CrossRef
  42. Kozyryev I, Zhang J (2015) Bayesian Analysis of Complex Interacting Mutations in HIV Drug Resistance and Cross-Resistance. Adv Exp Med Biol. 827: 367-83.
    CrossRef
  43. Zaccarelli M, Tozzi V, Lorenzini P, Trotta MP, Forbici F, et al. (2005) Multiple drug class-wide resistance associated with poorer survival after treatment failure in a cohort of HIV-infected patients. AIDS. 19: 1081-9.
    CrossRef
  44. Carr A (2003) Toxicity of antiretroviral therapy and implications for drug development. Nat Rev Drug Discov. 2: 624-34.
    CrossRef
  45. Erfan Manesh M, Esmaeilzadeh A, Hajikhan Mirzaei M (2015) IL-24: A Novel Gene Therapy Candidate For Immune System Up-Regulation in Hodgkin’s Lymphoma. Journal of Medical Hypotheses and Ideas. 9: 61-6.
    CrossRef
  46. Hajikhan Mirzaei M, Esmaeilzadeh A (2014) Overexpression of MDA-7/IL-24 as an anticancer cytokine in gene therapy of thyroid carcinoma. Journal of Medical Hypotheses and Ideas. 8: 7-13.
    CrossRef
  47. Piri Z, Esmaeilzadeh A, Hajikhanmirzaei M (2012) Interleukin-25 as a candidate gene in immunogene therapy of pancreatic cancer. Journal of Medical Hypotheses and Ideas. 6: 75-9.
    CrossRef
  48. Mazaheri T, Esmaeilzadeh A, Mirzaei MH (2012) Introducing the immunomodulatory effects of mesenchymal stem cells in an experimental model of Behçet’s disease. Journal of Medical Hypotheses and Ideas. 6: 23-7.
    CrossRef
  49. Mirzamohammadi F, Esmaeilzadeh A (2007) A Novel Method in Gene Therapy of HIV. iranian journal of public health. 36:
  50. Sora F, Antinori A, Piccirillo N, De Luca A, Chiusolo P, et al. (2002) Highly active antiretroviral therapy and allogeneic CD34(+) peripheral blood progenitor cells transplantation in an HIV/HCV coinfected patient with acute myeloid leukemia. Exp Hematol. 30: 279-84.
    CrossRef
  51. Jubault V, Burgard M, Le Corfec E, Costagliola D, Rouzioux C, et al. (1998) High rebound of plasma and cellular HIV load after discontinuation of triple combination therapy. AIDS. 12: 2358-9.
  52. Allers K, Hutter G, Hofmann J, Loddenkemper C, Rieger K, et al. (2011) Evidence for the cure of HIV infection by CCR5Delta32/Delta32 stem cell transplantation. Blood. 117: 2791-9.
    CrossRef
  53. Hutter G, Ganepola S (2011) Eradication of HIV by transplantation of CCR5-deficient hematopoietic stem cells. ScientificWorldJournal. 11: 1068-76.
    CrossRef
  54. Duarte RF, Salgado M, Sanchez-Ortega I, Arnan M, Canals C, et al. (2015) CCR5 Delta32 homozygous cord blood allogeneic transplantation in a patient with HIV: a case report. Lancet HIV. 2: e236-42
    CrossRef
  55. Petz LD, Redei I, Bryson Y, Regan D, Kurtzberg J, et al. (2013) Hematopoietic cell transplantation with cord blood for cure of HIV infections. Biol Blood Marrow Transplant. 19: 393-7.
    CrossRef
  56. Petz L (2013) Cord blood transplantation for cure of HIV infections. Stem Cells Transl Med. 2: 635-7.
    CrossRef
  57. Manjunath N, Yi G, Dang Y, Shankar P (2013) Newer gene editing technologies toward HIV gene therapy. Viruses. 5: 2748-66.
    CrossRef
  58. Pavletich NP, Pabo CO (1991) Zinc finger-DNA recognition: crystal structure of a Zif268-DNA complex at 2.1 A. Science. 252: 809-17.
    CrossRef
  59. Bitinaite J, Wah DA, Aggarwal AK, Schildkraut I (1998) FokI dimerization is required for DNA cleavage. Proc Natl Acad Sci U S A. 95: 10570-5.
    CrossRef
  60. Lieber MR (2010) The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. Annu Rev Biochem. 79: 181-211.
    CrossRef
  61. Moynahan ME, Jasin M (2010) Mitotic homologous recombination maintains genomic stability and suppresses tumorigenesis. Nat Rev Mol Cell Biol. 11: 196-207.
    CrossRef
  62. Mani M, Kandavelou K, Dy FJ, Durai S, Chandrasegaran S (2005) Design, engineering, and characterization of zinc finger nucleases. Biochem Biophys Res Commun. 335: 447-57.
    CrossRef
  63. Perez EE, Wang J, Miller JC, Jouvenot Y, Kim KA, et al. (2008) Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat Biotechnol. 26: 808-16.
    CrossRef
  64. Wilen CB, Wang J, Tilton JC, Miller JC, Kim KA, et al. (2011) Engineering HIV-resistant human CD4+ T cells with CXCR4-specific zinc-finger nucleases. PLoS Pathog. 7: e1002020.
    CrossRef
  65. Yuan J, Wang J, Crain K, Fearns C, Kim KA, et al. (2012) Zinc-finger nuclease editing of human cxcr4 promotes HIV-1 CD4(+) T cell resistance and enrichment. Mol Ther. 20: 849-59.
    CrossRef
  66. Doyon Y, Vo TD, Mendel MC, Greenberg SG, Wang J, et al. (2011) Enhancing zinc-finger-nuclease activity with improved obligate heterodimeric architectures. Nat Methods. 8: 74-9.
    CrossRef
  67. Voit RA, McMahon MA, Sawyer SL, Porteus MH (2013) Generation of an HIV resistant T-cell line by targeted “stacking” of restriction factors. Mol Ther. 21: 786-95.
    CrossRef
  68. Maier DA, Brennan AL, Jiang S, Binder-Scholl GK, Lee G, et al. (2013) Efficient clinical scale gene modification via zinc finger nuclease-targeted disruption of the HIV co-receptor CCR5. Hum Gene Ther. 24: 245-58.
    CrossRef
  69. Mussolino C, Morbitzer R, Lutge F, Dannemann N, Lahaye T, et al. (2011) A novel TALE nuclease scaffold enables high genome editing activity in combination with low toxicity. Nucleic Acids Res. 39: 9283-93.
    CrossRef
  70. Holkers M, Maggio I, Liu J, Janssen JM, Miselli F, et al. (2013) Differential integrity of TALE nuclease genes following adenoviral and lentiviral vector gene transfer into human cells. Nucleic Acids Res. 41: e63.
    CrossRef
  71. Wiedenheft B, Lander GC, Zhou K, Jore MM, Brouns SJ, et al. (2011) Structures of the RNA-guided surveillance complex from a bacterial immune system. Nature. 477: 486-9.
    CrossRef
  72. Ebina H, Misawa N, Kanemura Y, Koyanagi Y (2013) Harnessing the CRISPR/Cas9 system to disrupt latent HIV-1 provirus. Scientific Reports. 3: 2510.
    CrossRef
  73. Hu W, Kaminski R, Yang F, Zhang Y, Cosentino L, et al. (2014) RNA-directed gene editing specifically eradicates latent and prevents new HIV-1 infection. Proc Natl Acad Sci U S A. 111: 11461-6.
    CrossRef
  74. Mandal PK, Ferreira LM, Collins R, Meissner TB, Boutwell CL, et al. (2014) Efficient ablation of genes in human hematopoietic stem and effector cells using CRISPR/Cas9. Cell Stem Cell. 15: 643-52.
    CrossRef
  75. Menendez-Arias L (2013) Molecular basis of human immunodeficiency virus type 1 drug resistance: overview and recent developments. Antiviral Res. 98: 93-120.
    CrossRef
  76. Archin NM, Liberty AL, Kashuba AD, Choudhary SK, Kuruc JD, et al. (2012) Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature. 487: 482-5.
    CrossRef
(Visited 480 times, 1 visits today)

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.