1
|
Djataou P, Djuidje Ngounoue M, Nkenfou-Tchinda CN, Ngoufack MN, Elong E, Tiga A, Muluh C, Kadji Kameni J, Djaouda M, Ndjolo A, Nkenfou CN. Low prevalence of HIV in the northern Cameroon: contribution of some AIDS restriction genes and potential implications for gene therapy. Front Genet 2024; 15:1447971. [PMID: 39346778 PMCID: PMC11427317 DOI: 10.3389/fgene.2024.1447971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/22/2024] [Indexed: 10/01/2024] Open
Abstract
Background HIV infection and its progression to AIDS depend on several factors including host genetic factors. The immunological mechanisms of host resistance to HIV infection greatly influence the prevalence of HIV in a given region. Worldwide, Cameroon not exempted, the frequency of AIDS-associated genes varies and may influence this prevalence. The North and Far North Regions of Cameroon have had the lowest HIV prevalence in the country for many years despite risky behaviors associated with their customs and habits. In this work, we seek to explore the contribution of host genes to the HIV low prevalence in these regions. Methodology Five genes variants previously described as HIV AIDS related were studied. These genes are: CCR5Δ32, CCR5promoter59029G, CCR2-64I, SDF1-3'A and Trim5α(R136Q). A total of 384 consented participants were included in this study. The HIV serological status was confirmed using national algorithm. Genomic DNA was extracted from the buffy coats and used for genotyping. The results obtained were compiled in Excel 2016, Epi Info 7.1 and snpStats software and Chi two tests allowed us to compare the frequencies of the AIDS related alleles in the North with those in other Regions of Cameroon and to measure the impact of these ARGs on protection against HIV. Results The frequency of protective alleles CCR5Δ32, CCR5promoter59029G, CCR2-64I, SDF1-3'A and Trim5α(R136Q) was the allelic frequencies should be expressed as percentages i.e. 0.52%; 37.56%; 36.46%; 25.19% and 69.33%. These allelic frequencies exhibited a significant difference when compared to those obtained in other regions of Cameroon (p < 0.01). Protective alleles were predominant in the Northern region compared to others and were associated with resistance to HIV [(p < 0.0001); OR = 2.02 CI, 95%]. Conclusion The higher frequency of HIV-protective alleles in the northern regions may be a contributing factor to the lower prevalence of HIV. Nevertheless, this should be reinforced by other preventive and surveillance methods to guarantee the sustained low prevalence. HIV can develop resistance through the process of mutation, but the host targets themselves are genetically stable. The study of these host genetic restriction factors is of great value in the design of a practical cure for HIV infection or an effective vaccine.
Collapse
Affiliation(s)
- Patrice Djataou
- Chantal BIYA International Reference Center (CBIRC), Yaounde, Cameroon
- Department of Biochemistry, Faculty of Science, University of Yaounde I, Yaounde, Cameroon
| | | | - Carine Nguefeu Nkenfou-Tchinda
- Department of Biochemistry, Faculty of Medicine and Pharmaceutical Sciences of Sangmelima, Ebolowa University, Sangmelima, Cameroon
| | | | - Elise Elong
- Chantal BIYA International Reference Center (CBIRC), Yaounde, Cameroon
| | - Aline Tiga
- Chantal BIYA International Reference Center (CBIRC), Yaounde, Cameroon
| | | | | | - Moussa Djaouda
- Department of Life and Earth Sciences, University of Maroua, Maroua, Cameroon
| | - Alexis Ndjolo
- Chantal BIYA International Reference Center (CBIRC), Yaounde, Cameroon
| | - Celine Nguefeu Nkenfou
- Chantal BIYA International Reference Center (CBIRC), Yaounde, Cameroon
- Department of Biological Sciences, Higher Teachers Training College, Yaounde, Cameroon
| |
Collapse
|
2
|
Faivre N, Verollet C, Dumas F. The chemokine receptor CCR5: multi-faceted hook for HIV-1. Retrovirology 2024; 21:2. [PMID: 38263120 PMCID: PMC10807162 DOI: 10.1186/s12977-024-00634-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/13/2024] [Indexed: 01/25/2024] Open
Abstract
Chemokines are cytokines whose primary role is cellular activation and stimulation of leukocyte migration. They perform their various functions by interacting with G protein-coupled cell surface receptors (GPCRs) and are involved in the regulation of many biological processes such as apoptosis, proliferation, angiogenesis, hematopoiesis or organogenesis. They contribute to the maintenance of the homeostasis of lymphocytes and coordinate the function of the immune system. However, chemokines and their receptors are sometimes hijacked by some pathogens to infect the host organism. For a given chemokine receptor, there is a wide structural, organizational and conformational diversity. In this review, we describe the evidence for structural variety reported for the chemokine receptor CCR5, how this variability can be exploited by HIV-1 to infect its target cells and what therapeutic solutions are currently being developed to overcome this problem.
Collapse
Affiliation(s)
- Natacha Faivre
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Buenos Aires, Argentina
| | - Christel Verollet
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Buenos Aires, Argentina
| | - Fabrice Dumas
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France.
| |
Collapse
|
3
|
Alkhatib B, Jabari M, Bilasy S, Abdul-Rahman H, Sandhu K, Lai S, Alkhatib G. Resistance to Human Immunodeficiency Virus 1 Infection Conferred by a Compound CCR5Δ32 and CCR5 C20S Heterozygote. J Infect Dis 2023; 228:116-121. [PMID: 36912158 PMCID: PMC11009460 DOI: 10.1093/infdis/jiad062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/23/2023] [Accepted: 03/10/2023] [Indexed: 03/14/2023] Open
Abstract
We analyzed findings in a same-gender couple discordant in their human immunodeficiency virus (HIV) status. The HIV+ partner was homozygous for CCR5 while his receptive HIV- partner was a CCR5Δ32 heterozygote with a C20S missense mutation in his CCR5 allele. The cells from the HIV- partner showed significant resistance to R5 fusion/infection and had no chemotactic response to CCL4 (macrophage inflammatory protein 1β). We demonstrated abundant CCR5-specific RNA in the HIV- partner's cells but no detectable CCR5 protein. CCR5 promoter region cloned from each partner's DNA indicated no significant impact on RNA transcription. The compound effect of CCR5Δ32 and C20S mutation impaired CCR5 coreceptor function and conferred resistance to HIV-1.
Collapse
Affiliation(s)
- Bashar Alkhatib
- Department of Basic Sciences, California Northstate University College of Medicine, Elk Grove, California, USA
| | - Mary Jabari
- Department of Basic Sciences, California Northstate University College of Medicine, Elk Grove, California, USA
| | - Shymaa Bilasy
- Department of Basic Sciences, California Northstate University College of Medicine, Elk Grove, California, USA
| | - Husni Abdul-Rahman
- Department of Basic Sciences, California Northstate University College of Medicine, Elk Grove, California, USA
| | - Kamal Sandhu
- Department of Basic Sciences, California Northstate University College of Medicine, Elk Grove, California, USA
| | - Stephen Lai
- Department of Basic Sciences, California Northstate University College of Medicine, Elk Grove, California, USA
| | - Ghalib Alkhatib
- Department of Basic Sciences, California Northstate University College of Medicine, Elk Grove, California, USA
| |
Collapse
|
4
|
Hsu J, Besien KV, Glesby MJ, Pahwa S, Coletti A, Warshaw MG, Petz L, Moore TB, Chen YH, Pallikkuth S, Dhummakupt A, Cortado R, Golner A, Bone F, Baldo M, Riches M, Mellors JW, Tobin NH, Browning R, Persaud D, Bryson Y, International Maternal Pediatric Adolescent AIDS Clinical Trials Network (IMPAACT) P1107 Team. HIV-1 remission and possible cure in a woman after haplo-cord blood transplant. Cell 2023; 186:1115-1126.e8. [PMID: 36931242 PMCID: PMC10616809 DOI: 10.1016/j.cell.2023.02.030] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/01/2022] [Accepted: 02/17/2023] [Indexed: 03/18/2023]
Abstract
Previously, two men were cured of HIV-1 through CCR5Δ32 homozygous (CCR5Δ32/Δ32) allogeneic adult stem cell transplant. We report the first remission and possible HIV-1 cure in a mixed-race woman who received a CCR5Δ32/Δ32 haplo-cord transplant (cord blood cells combined with haploidentical stem cells from an adult) to treat acute myeloid leukemia (AML). Peripheral blood chimerism was 100% CCR5Δ32/Δ32 cord blood by week 14 post-transplant and persisted through 4.8 years of follow-up. Immune reconstitution was associated with (1) loss of detectable replication-competent HIV-1 reservoirs, (2) loss of HIV-1-specific immune responses, (3) in vitro resistance to X4 and R5 laboratory variants, including pre-transplant autologous latent reservoir isolates, and (4) 18 months of HIV-1 control with aviremia, off antiretroviral therapy, starting at 37 months post-transplant. CCR5Δ32/Δ32 haplo-cord transplant achieved remission and a possible HIV-1 cure for a person of diverse ancestry, living with HIV-1, who required a stem cell transplant for acute leukemia.
Collapse
Affiliation(s)
- Jingmei Hsu
- Department of Medicine, Division of Hematology& Oncology, Weill Cornell Medicine / New York Presbyterian Hospital, New York, NY, 10021, USA
| | - Koen Van Besien
- Department of Medicine, Division of Hematology& Oncology, Weill Cornell Medicine / New York Presbyterian Hospital, New York, NY, 10021, USA
| | - Marshall J. Glesby
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine / New York Presbyterian Hospital, New York, NY, 10021, USA
| | - Savita Pahwa
- Department of Microbiology and Immunology, Pediatrics and Medicine, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Anne Coletti
- Family Health International 360, Durham, NC, 27761, USA
| | - Meredith G Warshaw
- Center for Biostatistics in AIDS Research, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
| | - Larry Petz
- StemCyte International Cord Blood Center, Baldwin Park, California, 91706, USA
| | - Theodore B. Moore
- Department of Pediatrics, Division of Hematology& Oncology, Ronald Reagan UCLA Medical Center, Los Angeles, CA, 90095, USA
| | - Ya Hui Chen
- Department of Pediatrics, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, 21025, USA
| | - Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Adit Dhummakupt
- Department of Pediatrics, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, 21025, USA
| | - Ruth Cortado
- Department of Pediatrics, Division of Infectious Diseases, Mattel Children’s, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Amanda Golner
- Frontier Science & Technology Research Foundation, Inc, Amherst, NY, 14226, USA
| | - Frederic Bone
- Frontier Science & Technology Research Foundation, Inc, Amherst, NY, 14226, USA
| | - Maria Baldo
- Department of Medicine, Division of Hematology& Oncology, Weill Cornell Medicine / New York Presbyterian Hospital, New York, NY, 10021, USA
| | - Marcie Riches
- Center for International Blood and Marrow Transplant Research (CIBMTR), Medical College of Wisconsin Clinical Cancer Center, Milwaukee, WI, 53226, USA
| | - John W. Mellors
- Department of Medicine, Division of Infectious Diseases, University of Pittsburg School of Medicine, Pittsburgh, PA, 15261, USA
| | - Nicole H. Tobin
- Department of Pediatrics, Division of Infectious Diseases, Mattel Children’s, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Renee Browning
- National Institutes of Health, National Institute of Allergy and Infectious Diseases, Bethesda, MD, 20892, USA
| | - Deborah Persaud
- Department of Pediatrics, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, 21025, USA
| | - Yvonne Bryson
- Department of Pediatrics, Division of Infectious Diseases, Mattel Children’s, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | | |
Collapse
|
5
|
Jai J, Shirleen D, Hanbali C, Wijaya P, Anginan TB, Husada W, Pratama MY. Multiplexed shRNA-miRs as a candidate for anti HIV-1 therapy: strategies, challenges, and future potential. J Genet Eng Biotechnol 2022; 20:172. [PMID: 36576612 PMCID: PMC9797628 DOI: 10.1186/s43141-022-00451-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 12/04/2022] [Indexed: 12/29/2022]
Abstract
The spread of HIV is on the rise and has become a global issue, especially for underdeveloped and developing countries. This is due to the fact that HIV majorly occurs asymptomatically and is implausible for early diagnosis. Recent advances in research and science have enabled the investigation of a new potential treatment involving gene-based therapy, known as RNA interference (RNAi) that will direct gene silencing and further compensate for natural variants and viral mutants. Several types of small regulatory RNA are discussed in this present study, including microRNA (miRNA), small interfering RNA (siRNA), and short hairpin RNA (shRNA).This paper examines the mechanism of RNAi as a viable HIV therapy, using a minimum of four shRNAs to target both dispensable host components (CCR5) and viral genes (Gag, Env, Tat, Pol I, Pol II and Vif). Moreover, a multiplexed mechanism of shRNAs and miRNA is known to be effective in preventing viral escape due to mutation as the miRNA develops a general polycistronic platform for the expression of a large amount of shRNA-miRs. Several administration methods as well as the advantages of this RNAi treatment are also discussed in this study. The administration methods include (1) ex vivo delivery with the help of viral vectors, nanoparticles, and electroporation, (2) nonspecific in vivo delivery using non-viral carriers including liposomes, dendrimers and aptamers, as well as (3) targeted delivery that uses antibodies, modified nanoparticles, nucleic acid aptamers, and tissue-specific serotypes of AAV. Moreover, the advantages of this treatment are related to the effectiveness in silencing the HIV gene, which is more compatible compared to other gene therapy treatments, such as ZFN, TALEN, and CRISPR/Cas9.
Collapse
Affiliation(s)
- Jyotsna Jai
- grid.504251.70000 0004 7706 8927Department of Biotechnology, Indonesia International Institute for Life-Sciences (i3L), Jakarta, Indonesia
| | - Deborah Shirleen
- grid.504251.70000 0004 7706 8927Department of Biotechnology, Indonesia International Institute for Life-Sciences (i3L), Jakarta, Indonesia
| | - Christian Hanbali
- grid.504251.70000 0004 7706 8927Department of Biomedicine, Indonesia International Institute for Life-Sciences (i3L), Jakarta, Indonesia
| | - Pamela Wijaya
- grid.504251.70000 0004 7706 8927Department of Biomedicine, Indonesia International Institute for Life-Sciences (i3L), Jakarta, Indonesia
| | - Theresia Brigita Anginan
- grid.504251.70000 0004 7706 8927Department of Biomedicine, Indonesia International Institute for Life-Sciences (i3L), Jakarta, Indonesia
| | - William Husada
- grid.504251.70000 0004 7706 8927Department of Biotechnology, Indonesia International Institute for Life-Sciences (i3L), Jakarta, Indonesia
| | - Muhammad Yogi Pratama
- grid.504251.70000 0004 7706 8927Department of Biomedicine, Indonesia International Institute for Life-Sciences (i3L), Jakarta, Indonesia ,grid.240324.30000 0001 2109 4251Division of Vascular Surgery, Department of Surgery, New York University Medical Center, New York, USA
| |
Collapse
|
6
|
Ferrero MR, Tavares LP, Garcia CC. The Dual Role of CCR5 in the Course of Influenza Infection: Exploring Treatment Opportunities. Front Immunol 2022; 12:826621. [PMID: 35126379 PMCID: PMC8810482 DOI: 10.3389/fimmu.2021.826621] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/31/2021] [Indexed: 12/12/2022] Open
Abstract
Influenza is one of the most relevant respiratory viruses to human health causing annual epidemics, and recurrent pandemics. Influenza disease is principally associated with inappropriate activation of the immune response. Chemokine receptor 5 (CCR5) and its cognate chemokines CCL3, CCL4 and CCL5 are rapidly induced upon influenza infection, contributing to leukocyte recruitment into the airways and a consequent effective antiviral response. Here we discuss the existing evidence for CCR5 role in the host immune responses to influenza virus. Complete absence of CCR5 in mice revealed the receptor’s role in coping with influenza via the recruitment of early memory CD8+ T cells, B cell activation and later recruitment of activated CD4+ T cells. Moreover, CCR5 contributes to inflammatory resolution by enhancing alveolar macrophages survival and reprogramming macrophages to pro-resolving phenotypes. In contrast, CCR5 activation is associated with excessive recruitment of neutrophils, inflammatory monocytes, and NK cells in models of severe influenza pneumonia. The available data suggests that, while CCL5 can play a protective role in influenza infection, CCL3 may contribute to an overwhelming inflammatory process that can harm the lung tissue. In humans, the gene encoding CCR5 might contain a 32-base pair deletion, resulting in a truncated protein. While discordant data in literature regarding this CCR5 mutation and influenza severity, the association of CCR5delta32 and HIV resistance fostered the development of different CCR5 inhibitors, now being tested in lung inflammation therapy. The potential use of CCR5 inhibitors to modulate the inflammatory response in severe human influenza infections is to be addressed.
Collapse
Affiliation(s)
- Maximiliano Ruben Ferrero
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
- *Correspondence: Maximiliano Ruben Ferrero,
| | - Luciana Pádua Tavares
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Cristiana Couto Garcia
- Laboratory of Respiratory Virus and Measles, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| |
Collapse
|
7
|
Bauss J, Morris M, Shankar R, Olivero R, Buck LN, Stenger CL, Hinds D, Mills J, Eby A, Zagorski JW, Smith C, Cline S, Hartog NL, Chen B, Huss J, Carcillo JA, Rajasekaran S, Bupp CP, Prokop JW. CCR5 and Biological Complexity: The Need for Data Integration and Educational Materials to Address Genetic/Biological Reductionism at the Interface of Ethical, Legal, and Social Implications. Front Immunol 2021; 12:790041. [PMID: 34925370 PMCID: PMC8674737 DOI: 10.3389/fimmu.2021.790041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/18/2021] [Indexed: 01/02/2023] Open
Abstract
In the age of genomics, public understanding of complex scientific knowledge is critical. To combat reductionistic views, it is necessary to generate and organize educational material and data that keep pace with advances in genomics. The view that CCR5 is solely the receptor for HIV gave rise to demand to remove the gene in patients to create host HIV resistance, underestimating the broader roles and complex genetic inheritance of CCR5. A program aimed at providing research projects to undergraduates, known as CODE, has been expanded to build educational material for genes such as CCR5 in a rapid approach, exposing students and trainees to large bioinformatics databases and previous experiments for broader data to challenge commitment to biological reductionism. Our students organize expression databases, query environmental responses, assess genetic factors, generate protein models/dynamics, and profile evolutionary insights into a protein such as CCR5. The knowledgebase generated in the initiative opens the door for public educational information and tools (molecular videos, 3D printed models, and handouts), classroom materials, and strategy for future genetic ideas that can be distributed in formal, semiformal, and informal educational environments. This work highlights that many factors are missing from the reductionist view of CCR5, including the role of missense variants or expression of CCR5 with neurological phenotypes and the role of CCR5 and the delta32 variant in complex critical care patients with sepsis. When connected to genomic stories in the news, these tools offer critically needed Ethical, Legal, and Social Implication (ELSI) education to combat biological reductionism.
Collapse
Affiliation(s)
- Jacob Bauss
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Michele Morris
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, United States
| | - Rama Shankar
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Rosemary Olivero
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Infectious Disease, Helen DeVos Children's Hospital, Grand Rapids, MI, United States
| | - Leah N Buck
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Department of Mathematics, University of North Alabama, Florence, AL, United States
| | - Cynthia L Stenger
- Department of Mathematics, University of North Alabama, Florence, AL, United States
| | - David Hinds
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,HudsonAlpha Institute for Biotechnology, Huntsville, AL, United States
| | - Joshua Mills
- Department of Biology, Grand Valley State University, Allendale, MI, United States
| | - Alexandra Eby
- Department of Science, Davenport University, Grand Rapids, MI, United States
| | - Joseph W Zagorski
- Office of Research, Spectrum Health, Grand Rapids, MI, United States
| | - Caitlin Smith
- Department of Biology, Athens State University, Athens, AL, United States
| | - Sara Cline
- Department of Biology, Athens State University, Athens, AL, United States
| | - Nicholas L Hartog
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Allergy & Immunology, Spectrum Health, Grand Rapids, MI, United States
| | - Bin Chen
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - John Huss
- Department of Philosophy, The University of Akron, Akron, OH, United States
| | - Joseph A Carcillo
- Department of Critical Care Medicine and Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Surender Rajasekaran
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Office of Research, Spectrum Health, Grand Rapids, MI, United States.,Pediatric Intensive Care Unit, Helen DeVos Children's Hospital, Grand Rapids, MI, United States
| | - Caleb P Bupp
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Medical Genetics, Spectrum Health, Grand Rapids, MI, United States
| | - Jeremy W Prokop
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
8
|
Kulmann-Leal B, Ellwanger JH, Chies JAB. CCR5Δ32 in Brazil: Impacts of a European Genetic Variant on a Highly Admixed Population. Front Immunol 2021; 12:758358. [PMID: 34956188 PMCID: PMC8703165 DOI: 10.3389/fimmu.2021.758358] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/23/2021] [Indexed: 01/10/2023] Open
Abstract
The genetic background of Brazilians encompasses Amerindian, African, and European components as a result of the colonization of an already Amerindian inhabited region by Europeans, associated to a massive influx of Africans. Other migratory flows introduced into the Brazilian population genetic components from Asia and the Middle East. Currently, Brazil has a highly admixed population and, therefore, the study of genetic factors in the context of health or disease in Brazil is a challenging and remarkably interesting subject. This phenomenon is exemplified by the genetic variant CCR5Δ32, a 32 base-pair deletion in the CCR5 gene. CCR5Δ32 originated in Europe, but the time of origin as well as the selective pressures that allowed the maintenance of this variant and the establishment of its current frequencies in the different human populations is still a field of debates. Due to its origin, the CCR5Δ32 allele frequency is high in European-derived populations (~10%) and low in Asian and African native human populations. In Brazil, the CCR5Δ32 allele frequency is intermediate (4-6%) and varies on the Brazilian States, depending on the migratory history of each region. CCR5 is a protein that regulates the activity of several immune cells, also acting as the main HIV-1 co-receptor. The CCR5 expression is influenced by CCR5Δ32 genotypes. No CCR5 expression is observed in CCR5Δ32 homozygous individuals. Thus, the CCR5Δ32 has particular effects on different diseases. At the population level, the effect that CCR5Δ32 has on European populations may be different than that observed in highly admixed populations. Besides less evident due to its low frequency in admixed groups, the effect of the CCR5Δ32 variant may be affected by other genetic traits. Understanding the effects of CCR5Δ32 on Brazilians is essential to predict the potential use of pharmacological CCR5 modulators in Brazil. Therefore, this study reviews the impacts of the CCR5Δ32 on the Brazilian population, considering infectious diseases, inflammatory conditions, and cancer. Finally, this article provides a general discussion concerning the impacts of a European-derived variant, the CCR5Δ32, on a highly admixed population.
Collapse
Affiliation(s)
| | | | - José Artur Bogo Chies
- Laboratório de Imunobiologia e Imunogenética, Programa de Pós-Graduação em Genética e Biologia Molecular (PPGBM), Departamento de Genética, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| |
Collapse
|
9
|
Abstract
The CD8+ T cell noncytotoxic antiviral response (CNAR) was discovered during studies of asymptomatic HIV-infected subjects more than 30 years ago. In contrast to CD8+ T cell cytotoxic lymphocyte (CTL) activity, CNAR suppresses HIV replication without target cell killing. This activity has characteristics of innate immunity: it acts on all retroviruses and thus is neither epitope specific nor HLA restricted. The HIV-associated CNAR does not affect other virus families. It is mediated, at least in part, by a CD8+ T cell antiviral factor (CAF) that blocks HIV transcription. A variety of assays used to measure CNAR/CAF and the effects on other retrovirus infections are described. Notably, CD8+ T cell noncytotoxic antiviral responses have now been observed with other virus families but are mediated by different cytokines. Characterizing the protein structure of CAF has been challenging despite many biologic, immunologic, and molecular studies. It represents a low-abundance protein that may be identified by future next-generation sequencing approaches. Since CNAR/CAF is a natural noncytotoxic activity, it could provide promising strategies for HIV/AIDS therapy, cure, and prevention.
Collapse
Affiliation(s)
- Maelig G Morvan
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Fernando C Teque
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | | | - Jay A Levy
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
10
|
Morvan MG, Teque F, Ye L, Moreno ME, Wang J, VandenBerg S, Stoddart CA, Kan YW, Levy JA. Genetically edited CD34 + cells derived from human iPS cells in vivo but not in vitro engraft and differentiate into HIV-resistant cells. Proc Natl Acad Sci U S A 2021; 118:e2102404118. [PMID: 33975958 PMCID: PMC8158014 DOI: 10.1073/pnas.2102404118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Genetic editing of induced pluripotent stem (iPS) cells represents a promising avenue for an HIV cure. However, certain challenges remain before bringing this approach to the clinic. Among them, in vivo engraftment of cells genetically edited in vitro needs to be achieved. In this study, CD34+ cells derived in vitro from iPS cells genetically modified to carry the CCR5Δ32 mutant alleles did not engraft in humanized immunodeficient mice. However, the CD34+ cells isolated from teratomas generated in vivo from these genetically edited iPS cells engrafted in all experiments. These CD34+ cells also gave rise to peripheral blood mononuclear cells in the mice that, when inoculated with HIV in cell culture, were resistant to HIV R5-tropic isolates. This study indicates that teratomas can provide an environment that can help evaluate the engraftment potential of CD34+ cells derived from the genetically modified iPS cells in vitro. The results further confirm the possibility of using genetically engineered iPS cells to derive engraftable hematopoietic stem cells resistant to HIV as an approach toward an HIV cure.
Collapse
Affiliation(s)
- Maelig G Morvan
- Department of Medicine, Division of Hematology and Oncology, University of California, San Francisco, CA 94143-1270
| | - Fernando Teque
- Department of Medicine, Division of Hematology and Oncology, University of California, San Francisco, CA 94143-1270
| | - Lin Ye
- Department of Medicine, Institute of Human Genetics, University of California, San Francisco, CA 94143
| | - Mary E Moreno
- Department of Medicine, Division of Experimental Medicine, San Francisco General Hospital, University of California, San Francisco, CA 94110
| | - Jiaming Wang
- Department of Medicine, Institute of Human Genetics, University of California, San Francisco, CA 94143
| | - Scott VandenBerg
- Helen Diller Family Comprehensive Cancer Center, Biorepository and Tissue Biomarker Technology Core, University of California, San Francisco, CA 94143-0875
| | - Cheryl A Stoddart
- Department of Medicine, Division of Experimental Medicine, San Francisco General Hospital, University of California, San Francisco, CA 94110
| | - Yuet Wai Kan
- Department of Medicine, Institute of Human Genetics, University of California, San Francisco, CA 94143;
| | - Jay A Levy
- Department of Medicine, Division of Hematology and Oncology, University of California, San Francisco, CA 94143-1270;
| |
Collapse
|
11
|
|
12
|
Zamai L. Unveiling Human Non-Random Genome Editing Mechanisms Activated in Response to Chronic Environmental Changes: I. Where Might These Mechanisms Come from and What Might They Have Led To? Cells 2020; 9:E2362. [PMID: 33121045 PMCID: PMC7693803 DOI: 10.3390/cells9112362] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
This article challenges the notion of the randomness of mutations in eukaryotic cells by unveiling stress-induced human non-random genome editing mechanisms. To account for the existence of such mechanisms, I have developed molecular concepts of the cell environment and cell environmental stressors and, making use of a large quantity of published data, hypothesised the origin of some crucial biological leaps along the evolutionary path of life on Earth under the pressure of natural selection, in particular, (1) virus-cell mating as a primordial form of sexual recombination and symbiosis; (2) Lamarckian CRISPR-Cas systems; (3) eukaryotic gene development; (4) antiviral activity of retrotransposon-guided mutagenic enzymes; and finally, (5) the exaptation of antiviral mutagenic mechanisms to stress-induced genome editing mechanisms directed at "hyper-transcribed" endogenous genes. Genes transcribed at their maximum rate (hyper-transcribed), yet still unable to meet new chronic environmental demands generated by "pollution", are inadequate and generate more and more intronic retrotransposon transcripts. In this scenario, RNA-guided mutagenic enzymes (e.g., Apolipoprotein B mRNA editing catalytic polypeptide-like enzymes, APOBECs), which have been shown to bind to retrotransposon RNA-repetitive sequences, would be surgically targeted by intronic retrotransposons on opened chromatin regions of the same "hyper-transcribed" genes. RNA-guided mutagenic enzymes may therefore "Lamarkianly" generate single nucleotide polymorphisms (SNP) and gene copy number variations (CNV), as well as transposon transposition and chromosomal translocations in the restricted areas of hyper-functional and inadequate genes, leaving intact the rest of the genome. CNV and SNP of hyper-transcribed genes may allow cells to surgically explore a new fitness scenario, which increases their adaptability to stressful environmental conditions. Like the mechanisms of immunoglobulin somatic hypermutation, non-random genome editing mechanisms may generate several cell mutants, and those codifying for the most environmentally adequate proteins would have a survival advantage and would therefore be Darwinianly selected. Non-random genome editing mechanisms represent tools of evolvability leading to organismal adaptation including transgenerational non-Mendelian gene transmission or to death of environmentally inadequate genomes. They are a link between environmental changes and biological novelty and plasticity, finally providing a molecular basis to reconcile gene-centred and "ecological" views of evolution.
Collapse
Affiliation(s)
- Loris Zamai
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; ; Tel./Fax: +39-0722-304-319
- National Institute for Nuclear Physics (INFN)-Gran Sasso National Laboratory (LNGS), 67100 Assergi, L’Aquila, Italy
| |
Collapse
|
13
|
Almeida MJ, Matos A. Designer Nucleases: Gene-Editing Therapies using CCR5 as an Emerging Target in HIV. Curr HIV Res 2020; 17:306-323. [PMID: 31652113 DOI: 10.2174/1570162x17666191025112918] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/16/2019] [Accepted: 10/18/2019] [Indexed: 02/08/2023]
Abstract
Acquired Immunodeficiency Syndrome (AIDS), caused by the Human Immunodeficiency Virus (HIV), is a life-threatening disorder that persists worldwide as a severe health problem. Since it was linked with the HIV attachment process, the Chemokine receptor, CCR5, has been at the development leading edge of several gene-based therapies. Given the shortcomings of the current antiretroviral treatment procedure and the non-availability of a licensed vaccine, the aptitude to modify complex genomes with Designer Nucleases has had a noteworthy impact on biotechnology. Over the last years, ZFN, TALEN and CRISPR/Cas9 gene-editing technology have appeared as a promising solution that mimics the naturally occurring CCR5/Δ32 mutation and permanently guarantees the absence of CCR5-expression on the surface of HIV target-cells, leading to a continuous resistance to the virus entry and, ultimately, proving that cellular immunization from infection could be, in fact, a conceivable therapeutic approach to finally achieve the long-awaited functional cure of HIV.
Collapse
Affiliation(s)
- Maria João Almeida
- Laboratory of Microbiology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Ana Matos
- Laboratory of Microbiology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,Research Centre on Chemical Processes Engineering and Forest Products (CIEPQF), Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
14
|
Genetically-edited induced pluripotent stem cells derived from HIV-1-infected patients on therapy can give rise to immune cells resistant to HIV-1 infection. AIDS 2020; 34:1141-1149. [PMID: 32287059 DOI: 10.1097/qad.0000000000002539] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To assess the in-vitro CCR5---tropic and CXCR4---tropic HIV---1 infectivity of immune cells, particularly macrophages, derived from CCR5 gene---edited induced pluripotent stem cells (iPSCs) obtained from the peripheral blood mononuclear cells (PBMC) of HIV---infected patients on antiretroviral therapy (ART). DESIGN PBMC were obtained from six patients who had been HIV---infected for over 20 years and were on ART for 1---12 years prior to this study. METHODS The PBMC were derived into iPSCs and genetically edited with TALENs or CRISPR---cas9 endonucleases combined with PiggyBac technology to introduce the naturally occurring 32---bp deletion to the CCR5 gene. These iPSCs were differentiated into macrophages, and subsequently challenged with CCR5---tropic or CCR5/CXCR4 dual--- tropic HIV---1 strains. iPSC derivation, gene editing and immune cell differentiation were done in feeder---free, xeno---free in-vitro conditions. RESULTS Multiple unedited (wild---type) and CCR5 gene---edited (mutant) iPSCs were derived from patients' PBMC. When differentiated into immune cells and HIV---1 challenged, mutant iPSC lines were resistant to CCR5---tropic and to some extent to CCR5/CXCR4 dual---tropic HIV---1 infection when compared to wild---type iPSC lines. CONCLUSION Our study demonstrates that iPSC---derived, gene---edited immune cells are resistant to distinct HIV---1 strains. These findings have important implications for both in-vitro stem cell development and therapeutic approaches to cure HIV infection.
Collapse
|
15
|
Tajbakhsh A, Fazeli M, Rezaee M, Ghasemi F, Heravi MM, Gholoobi A, Meshkat Z. Prevalence of CCR5delta32 in Northeastern Iran. BMC MEDICAL GENETICS 2019; 20:184. [PMID: 31730458 PMCID: PMC6858674 DOI: 10.1186/s12881-019-0913-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 10/25/2019] [Indexed: 12/16/2022]
Abstract
Background A 32-base pair deletion (∆32) in the open reading frame (ORF) of C-C motif chemokine receptor 5 (CCR5) seems to be a protective variant against immune system diseases, especially human immunodeficiency virus type 1 (HIV-1). We aimed to assess the frequency of CCR5∆32 in the healthy Iranian population. Methods In this study, 400 normal samples from Khorasan, northeastern Iran, were randomly selected. The frequency of CCR5∆32 carriers was investigated using PCR analysis. Allele prevalence and the fit to the Hardy-Weinberg equilibrium were analyzed. Results The prevalence of CCR5∆32 in the northeastern population of Iran was 0.016. Four hundred samples were studied, among which one with CCR5∆32/∆32 and 11 with CCR5Wild/∆32 genotype were detected. Conclusion This study was the first investigation for an assessment of the prevalence of CCR5∆32 in northeastern Iran. The low prevalence of CCR5∆32 allele in the Iranian population may result in the increased susceptibility to HIV-1. In addition, this prevalence is the same as that of reported in East Asia, while is lower than that in the Europeans.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Department of Modern Sciences & Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mostafa Fazeli
- Department of Modern Sciences & Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Rezaee
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mastoureh Momen Heravi
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, P.O Box: 9196773117, Mashhad, IR, Iran
| | - Aida Gholoobi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Meshkat
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, P.O Box: 9196773117, Mashhad, IR, Iran.
| |
Collapse
|
16
|
Abstract
A recent report from Dr He Jiankui concerning the birth of twin girls harbouring mutations engineered by CRISPR/Cas nucleases has been met with international condemnation. Beside the serious ethical concerns, there are known technical risks associated with CRISPR/Cas gene editing which further raise questions about how these events could have been allowed to occur. Numerous studies have reported unexpected genomic mutation and mosaicism following the use of CRISPR/Cas nucleases, and it is currently unclear how prevalent these disadvantageous events are and how robust and sensitive the strategies to detect these unwanted events may be. Although Dr Jiankui's study appears to have involved certain checks to ascertain these risks, the decision to implant the manipulated embryos, given these unknowns, must nonetheless be considered reckless. Here I review the technical concerns surrounding genome editing and consider the available data from Dr Jiankui in this context. Although the data remains unpublished, preventing a thorough assessment of what was performed, it seems clear that the rationale behind the undertaking was seriously flawed; the procedures involved substantial technical risks which, when added to the serious ethical concerns, fully justify the widespread criticism that the events have received.
Collapse
Affiliation(s)
- Benjamin Davies
- Welcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| |
Collapse
|
17
|
Arendt V, Amand M, Iserentant G, Lemaire M, Masquelier C, Ndayisaba GF, Verhofstede C, Karita E, Allen S, Chevigné A, Schmit J, Bercoff DP, Seguin‐Devaux C. Predominance of the heterozygous CCR5 delta-24 deletion in African individuals resistant to HIV infection might be related to a defect in CCR5 addressing at the cell surface. J Int AIDS Soc 2019; 22:e25384. [PMID: 31486251 PMCID: PMC6727025 DOI: 10.1002/jia2.25384] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 07/31/2019] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION The chemokine receptor CCR5 is the main co-receptor for R5-tropic HIV-1 variants. We have previously described a novel 24-base pair deletion in the coding region of CCR5 among individuals from Rwanda. Here, we investigated the prevalence of hCCR5Δ24 in different cohorts and its impact on CCR5 expression and HIV-1 infection in vitro. METHODS We screened hCCR5Δ24 in a total of 3232 individuals which were either HIV-1 uninfected, high-risk HIV-1 seronegative and seropositive partners from serodiscordant couples, Long-Term Survivors, or HIV-1 infected volunteers from Africa (Rwanda, Kenya, Guinea-Conakry) and Luxembourg, using a real-time PCR assay. The role of the 24-base pair deletion on CCR5 expression and HIV infection was assessed in cell lines and PBMC using mRNA quantification, confocal analysis, flow and imaging cytometry. RESULTS AND DISCUSSION Among the 1661 patients from Rwanda, 12 individuals were heterozygous for hCCR5Δ24 but none were homozygous. Although heterozygosity for this allele may not confer complete resistance to HIV-1 infection, the prevalence of the mutation was 2.41% (95%CI: 0.43; 8.37) in 83 Long-Term Survivors (LTS) and 0.99% (95%CI: 0.45; 2.14) in 613 HIV-1 exposed seronegative members as compared with 0.35% (95% Cl: 0.06; 1.25) in 579 HIV-1 seropositive members. The prevalence of hCCR5Δ24 was 0.55% (95%CI: 0.15; 1.69) in 547 infants from Kenya but the mutation was not detected in 224 infants from Guinea-Conakry nor in 800 Caucasian individuals from Luxembourg. Expression of hCCR5Δ24 in cell lines and PBMC showed that the hCCR5Δ24 protein is stably expressed but is not transported to the plasma membrane due to a conformational change. Instead, the mutant receptor was retained intracellularly, colocalized with an endoplasmic reticulum marker and did not mediate HIV-1 infection. Co-transfection of hCCR5Δ24 and wtCCR5 did not indicate a transdominant negative effect of CCR5Δ24 on wtCCR5. CONCLUSIONS Our findings indicate that hCCR5Δ24 is not expressed at the cell surface. This could explain the higher prevalence of the heterozygous hCCR5Δ24 in LTS and HIV-1 exposed seronegative members from serodiscordant couples. Our data suggest an East-African localization of this deletion, which needs to be confirmed in larger cohorts from African and non-African countries.
Collapse
Affiliation(s)
- Vic Arendt
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
- Centre Hospitalier de LuxembourgNational Service of Infectious DiseasesLuxembourgLuxembourg
| | - Mathieu Amand
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
| | - Gilles Iserentant
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
| | - Morgane Lemaire
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
| | - Cécile Masquelier
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
| | | | - Chris Verhofstede
- Department of Clinical Chemistry, Microbiology and ImmunologyAIDS Reference LaboratoryGhent UniversityGhentBelgium
| | - Etienne Karita
- Department of Pathology and Laboratory MedicineEmory University School of MedicineAtlantaGAUSA
| | - Susan Allen
- Department of Pathology and Laboratory MedicineEmory University School of MedicineAtlantaGAUSA
| | - Andy Chevigné
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
| | - Jean‐Claude Schmit
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
| | - Danielle Perez Bercoff
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
| | - Carole Seguin‐Devaux
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
| |
Collapse
|
18
|
Ronsard L, Sood V, Yousif AS, Ramesh J, Shankar V, Das J, Sumi N, Rai T, Mohankumar K, Sridharan S, Dorschel A, Ramachandran VG, Banerjea AC. Genetic Polymorphisms in the Open Reading Frame of the CCR5 gene From HIV-1 Seronegative and Seropositive Individuals From National Capital Regions of India. Sci Rep 2019; 9:7594. [PMID: 31110236 PMCID: PMC6527560 DOI: 10.1038/s41598-019-44136-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 05/10/2019] [Indexed: 11/19/2022] Open
Abstract
C-C chemokine receptor type 5 (CCR5) serves as a co-receptor for Human immunodeficiency virus (HIV), enabling the virus to enter human CD4 T cells and macrophages. In the absence of CCR5, HIV strains that require CCR5 (R5 or M-tropic HIV) fail to successfully initiate infection. Various natural mutations of the CCR5 gene have been reported to interfere with the HIV-CCR5 interaction, which influences the rate of AIDS progression. Genetic characterization of the CCR5 gene in individuals from the National Capital Regions (NCRs) of India revealed several natural point mutations in HIV seropositive/negative individuals. Furthermore, we identified novel frame-shifts mutations in the CCR5 gene in HIV seronegative individuals, as well as the well reported CCR5Δ32 mutation. Additionally, we observed a number of mutations present only in HIV seropositive individuals. This is the first report to describe the genetic variations of CCR5 in individuals from the NCRs of India and demonstrates the utility of investigating understudied populations to identify novel CCR5 polymorphisms.
Collapse
Affiliation(s)
- Larance Ronsard
- Laboratory of Virology, National Institute of Immunology, New Delhi, India. .,Department of Microbiology, University College of Medical Sciences and Guru Teg Bahadur Hospital, Delhi, India. .,Ragon Institute of MGH, MIT and Harvard University, 400 Technology Square, Cambridge, MA, USA.
| | - Vikas Sood
- Laboratory of Virology, National Institute of Immunology, New Delhi, India.,Department of Microbiology, University College of Medical Sciences and Guru Teg Bahadur Hospital, Delhi, India
| | - Ashraf S Yousif
- Ragon Institute of MGH, MIT and Harvard University, 400 Technology Square, Cambridge, MA, USA
| | - Janani Ramesh
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vijay Shankar
- Laboratory of Virology, National Institute of Immunology, New Delhi, India
| | - Jishnu Das
- Ragon Institute of MGH, MIT and Harvard University, 400 Technology Square, Cambridge, MA, USA
| | - N Sumi
- Endocrinology & Toxicology Lab, Department of Zoology, University of Calicut, Kerala, India
| | - Tripti Rai
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, Delhi, India
| | | | - Subhashree Sridharan
- Department of Biochemistry and Molecular Biology, Pondicherry University, Pondicherry, India
| | | | | | - Akhil C Banerjea
- Laboratory of Virology, National Institute of Immunology, New Delhi, India.
| |
Collapse
|
19
|
Ledger S, Howe A, Turville S, Aggarwal A, Savkovic B, Ong A, Wolstein O, Boyd M, Millington M, Gorry PR, Murray JM, Symonds G. Analysis and dissociation of anti-HIV effects of shRNA to CCR5 and the fusion inhibitor C46. J Gene Med 2019; 20:e3006. [PMID: 29552747 DOI: 10.1002/jgm.3006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 12/31/2017] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The gene therapeutic Cal-1 comprises the anti-HIV agents: (i) sh5, a short hairpin RNA to CCR5 that down-regulates CCR5 expression and (ii) maC46 (C46), a peptide that inhibits viral fusion with the cell membrane. These constructs were assessed for inhibition of viral replication and selective cell expansion in a number of settings. METHODS HIV replication, selective outgrowth and cell surface viral binding were analysed with a single cycle infection assay of six pseudotyped HIV strains and a static and longitudinal passaging of MOLT4/CCR5 cells with HIV. Pronase digestion of surface virus and fluorescence microscopy assessed interactions between HIV virions and transduced cells. RESULTS Cal-1 reduced CCR5 expression in peripheral blood mononuclear cells to CCR5Δ32 heterozygote levels. Even low level transduction resulted in significant preferential expansion in MOLT4/CCR5 gene-containing cells over a 3-week HIV challenge regardless of viral suppression [12.5% to 47.0% (C46), 46.7% (sh5), 62.2% (Dual), respectively]. The sh5 and Dual constructs at > 95% transduction also significantly suppressed virus to day 12 in the passage assay and all constructs, at varying percentage transduction inhibited virus in static culture. No escape mutations were present through 9 weeks of challenge. The Dual construct significantly suppressed infection by a panel of CCR5-using viruses, with its efficacy being independently determined from the single constructs. Dual and sh5 inhibited virion internalisation, as determined via pronase digestion of surface bound virus, by 70% compared to 13% for C46. CONCLUSIONS The use of two anti-HIV genes allows optimal preferential survival and inhibition of HIV replication, with the impact on viral load being dependent on the percentage of gene marked cells.
Collapse
Affiliation(s)
- Scott Ledger
- Faculty of Medicine, UNSW Australia, Sydney, NSW, Australia.,St Vincent's Centre for Applied Medical Research, Darlinghurst, NSW, Australia
| | - Annett Howe
- School of Mathematics and Statistics, UNSW Australia, Sydney, NSW, Australia
| | | | | | - Borislav Savkovic
- School of Mathematics and Statistics, UNSW Australia, Sydney, NSW, Australia
| | - Andrew Ong
- School of Mathematics and Statistics, UNSW Australia, Sydney, NSW, Australia
| | | | - Maureen Boyd
- St Vincent's Centre for Applied Medical Research, Darlinghurst, NSW, Australia.,Calimmune Australia, Sydney, NSW, Australia
| | - Michelle Millington
- St Vincent's Centre for Applied Medical Research, Darlinghurst, NSW, Australia.,Calimmune Australia, Sydney, NSW, Australia
| | - Paul R Gorry
- School of Applied Sciences and Program in Metabolism, Exercise and Disease, Health Initiatives Research Institute, RMIT University, Melbourne, VIC, Australia.,Department of Infectious Diseases, Monash University, Melbourne, VIC, Australia.,Department of Microbiology and Immunology, University of Melbourne, Parkville, VIC, Australia
| | - John M Murray
- School of Mathematics and Statistics, UNSW Australia, Sydney, NSW, Australia
| | - Geoff Symonds
- Faculty of Medicine, UNSW Australia, Sydney, NSW, Australia.,St Vincent's Centre for Applied Medical Research, Darlinghurst, NSW, Australia.,Calimmune Australia, Sydney, NSW, Australia
| |
Collapse
|
20
|
The potential risks of C-C chemokine receptor 5-edited babies in bone development. Bone Res 2019; 7:4. [PMID: 30701110 PMCID: PMC6351561 DOI: 10.1038/s41413-019-0044-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 12/13/2018] [Indexed: 12/27/2022] Open
|
21
|
Tough RH, McLaren PJ. Interaction of the Host and Viral Genome and Their Influence on HIV Disease. Front Genet 2019; 9:720. [PMID: 30728828 PMCID: PMC6351501 DOI: 10.3389/fgene.2018.00720] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 12/21/2018] [Indexed: 01/23/2023] Open
Abstract
The course of Human Immunodeficiency Virus type 1 (HIV) infection is a dynamic interplay in which both host and viral genetic variation, among other factors, influence disease susceptibility and rate of progression. HIV set-point viral load (spVL), a key indicator of HIV disease progression, has an estimated 30% of variance attributable to common heritable effects and roughly 70% attributable to environmental factors and/or additional non-genetic factors. Genome-wide genotyping and sequencing studies have allowed for large-scale association testing studying host and viral genetic variants associated with infection and disease progression. Host genomics of HIV infection has been studied predominantly in Caucasian populations consistently identifying human leukocyte antigen (HLA) genes and C-C motif chemokine receptor 5 as key factors of HIV susceptibility and progression. However, these studies don’t fully assess all classes of genetic variation (e.g., very rare polymorphisms, copy number variants etc.) and do not inform on non-European ancestry groups. Additionally, viral sequence variability has been demonstrated to influence disease progression independently of host genetic variation. Viral sequence variation can be attributed to the rapid evolution of the virus within the host due to the selective pressure of the host immune response. As the host immune system responds to the virus, e.g., through recognition of HIV antigens, the virus is able to mitigate this response by evolving HLA-specific escape mutations. Diversity of viral genotypes has also been correlated with moderate to strong effects on CD4+ T cell decline and some studies showing weak to no correlation with spVL. There is evidence to support these viral genetic factors being heritable between individuals and the evolution of these factors having important consequences in the genetic epidemiology of HIV infection on a population level. This review will discuss the host-pathogen interaction of HIV infection, explore the importance of host and viral genetics for a better understanding of pathogenesis and identify opportunities for additional genetic studies.
Collapse
Affiliation(s)
- Riley H Tough
- JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Paul J McLaren
- JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
22
|
CCR5 Revisited: How Mechanisms of HIV Entry Govern AIDS Pathogenesis. J Mol Biol 2018; 430:2557-2589. [PMID: 29932942 DOI: 10.1016/j.jmb.2018.06.027] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/10/2018] [Accepted: 06/13/2018] [Indexed: 01/01/2023]
Abstract
The chemokine receptor CCR5 has been the focus of intensive studies since its role as a coreceptor for HIV entry was discovered in 1996. These studies lead to the development of small molecular drugs targeting CCR5, with maraviroc becoming in 2007 the first clinically approved chemokine receptor inhibitor. More recently, the apparent HIV cure in a patient transplanted with hematopoietic stem cells devoid of functional CCR5 rekindled the interest for inactivating CCR5 through gene therapy and pharmacological approaches. Fundamental research on CCR5 has also been boosted by key advances in the field of G-protein coupled receptor research, with the realization that CCR5 adopts a variety of conformations, and that only a subset of these conformations may be targeted by chemokine ligands. In addition, recent genetic and pathogenesis studies have emphasized the central role of CCR5 expression levels in determining the risk of HIV and SIV acquisition and disease progression. In this article, we propose to review the key properties of CCR5 that account for its central role in HIV pathogenesis, with a focus on mechanisms that regulate CCR5 expression, conformation, and interaction with HIV envelope glycoproteins.
Collapse
|
23
|
Hughes CE, Nibbs RJB. A guide to chemokines and their receptors. FEBS J 2018; 285:2944-2971. [PMID: 29637711 PMCID: PMC6120486 DOI: 10.1111/febs.14466] [Citation(s) in RCA: 876] [Impact Index Per Article: 125.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/25/2018] [Accepted: 04/03/2018] [Indexed: 12/12/2022]
Abstract
The chemokines (or chemotactic cytokines) are a large family of small, secreted proteins that signal through cell surface G protein-coupled heptahelical chemokine receptors. They are best known for their ability to stimulate the migration of cells, most notably white blood cells (leukocytes). Consequently, chemokines play a central role in the development and homeostasis of the immune system, and are involved in all protective or destructive immune and inflammatory responses. Classically viewed as inducers of directed chemotactic migration, it is now clear that chemokines can stimulate a variety of other types of directed and undirected migratory behavior, such as haptotaxis, chemokinesis, and haptokinesis, in addition to inducing cell arrest or adhesion. However, chemokine receptors on leukocytes can do more than just direct migration, and these molecules can also be expressed on, and regulate the biology of, many nonleukocytic cell types. Chemokines are profoundly affected by post-translational modification, by interaction with the extracellular matrix (ECM), and by binding to heptahelical 'atypical' chemokine receptors that regulate chemokine localization and abundance. This guide gives a broad overview of the chemokine and chemokine receptor families; summarizes the complex physical interactions that occur in the chemokine network; and, using specific examples, discusses general principles of chemokine function, focusing particularly on their ability to direct leukocyte migration.
Collapse
Affiliation(s)
- Catherine E Hughes
- Institute of Infection, Inflammation & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Robert J B Nibbs
- Institute of Infection, Inflammation & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| |
Collapse
|
24
|
CCR5 Δ32 homozygous cord blood allogeneic transplantation in a patient with HIV: a case report. Lancet HIV 2015; 2:e236-42. [PMID: 26423196 DOI: 10.1016/s2352-3018(15)00083-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 05/01/2015] [Accepted: 05/01/2015] [Indexed: 01/15/2023]
Abstract
BACKGROUND Allogeneic donor CCR5 Δ32 homozygous haemopoietic cell transplantation (HCT) provides the only evidence to date of long-term control of HIV infection. However, availability of conventional CCR5 Δ32 homozygous donors is insufficient to develop this as a therapeutic strategy further. METHODS We present a 37-year-old patient with HIV-1 infection and aggressive lymphoma who had disease progression after five lines of radiochemotherapy including an autologous HCT, and in the absence of matched sibling donors, received an allogeneic HCT with four of six HLA-matched CCR5 Δ32 homozygous cord blood cells (StemCyte, Covina, CA), supported with purified CD34+ cells from a haploidentical sibling. Blood or tissue samples were obtained before and weekly after HCT to monitor transplant and HIV infection, including chimerism analysis, CCR5 genotyping and viral tropism, viral isolation and sequence, viral reservoir analysis, immune activation and proliferation, and ex-vivo cell infectivity assays. Combined antiretroviral therapy continued during the procedure. FINDINGS The patient's HIV was CCR5-tropic by genotypic and phenotypic analyses. Baseline latent reservoir tests showed HIV DNA copies in bulk and resting CD4 T cells and in gut-associated lymphoid tissue, CD4 T-cell-associated HIV RNA, replication competent viral size of 2·1 copies per 10(7) CD4 T cells, and single copy assay of 303 copies per mL. After HCT, plasma HIV DNA load was undetectable by ultrasensitive analyses. Upon cord blood full chimerism, the patient's CCR5 Δ32 homozygous CD4 T cells responded to proliferation and activation stimuli and became resistant to infection by the patient's viral isolate and by laboratory-adapted HIV-1 strains. Death related to lymphoma progression regretfully prevented long-term monitoring of the patient's viral reservoir. INTERPRETATION CCR5 Δ32 homozygous cord blood reconstitution can successfully eliminate HIV-1 and render the allogeneic graft recipient's T lymphocytes resistant to HIV infection. Thus, they build on the evidence available to strongly support the use of cord blood as a strategic platform for a broader application of non-functional CCR5 transplantation to other infected individuals. FUNDING Spanish Secretariat of Research, the American Foundation for AIDS Research (amfAR).
Collapse
|
25
|
Shih YS, Fu E, Fu MM, Lin FG, Chiu HC, Shen EC, Chiang CY. Association of CCL5 and CCR5 gene polymorphisms with periodontitis in Taiwanese. J Periodontol 2014; 85:1596-602. [PMID: 25119558 DOI: 10.1902/jop.2014.130651] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND It has been suggested that genetic factors may predispose individuals to periodontal diseases. The present case-control study aims to test whether the -403 single nucleotide polymorphism of chemokine ligand 5 (CCL5-403) and the 32-bp deletion of CCR5 (CCR5Δ32) polymorphisms are associated with susceptibility to chronic and aggressive periodontitis. METHODS Taiwanese participants (N = 213) were grouped into control group (CG), generalized aggressive periodontitis (GAgP), or chronic periodontitis (CP) groups. DNA samples were obtained from peripheral blood. CCL5-403, evaluated by polymerase chain reaction-restriction fragment length polymorphism, and CCR5Δ32, evaluated by polymerase chain reaction, were compared among the three groups. RESULTS There was a significant association between type of periodontitis and having allele A or G in the CCL5-403 polymorphism. GAgP patients were 3.7 times more likely than CP patients and 2.0 times more likely than CG patients to have allele A, instead of allele G, in CCL5-403. GAgP patients were 3.1 times more likely than CG patients to have AG versus GG genotype. GAgP patients were also 5.0 and 19.8 times more likely than CP patients to have AG and AA genotypes, respectively, compared to GG. For the CCR5Δ32 polymorphism, no association was found between the type of periodontitis and having different genotype or allele distributions among GAgP, CP, or CG patients. CONCLUSION The single nucleotide polymorphism of CCL5-403 G substitution by A may play a role in AgP; however, the CCR5Δ32 polymorphism may not.
Collapse
Affiliation(s)
- Yo-Seng Shih
- Department of Periodontology, School of Dentistry, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|
26
|
Savkovic B, Nichols J, Birkett D, Applegate T, Ledger S, Symonds G, Murray JM. A quantitative comparison of anti-HIV gene therapy delivered to hematopoietic stem cells versus CD4+ T cells. PLoS Comput Biol 2014; 10:e1003681. [PMID: 24945407 PMCID: PMC4063676 DOI: 10.1371/journal.pcbi.1003681] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 05/05/2014] [Indexed: 02/07/2023] Open
Abstract
Gene therapy represents an alternative and promising anti-HIV modality to highly active antiretroviral therapy. It involves the introduction of a protective gene into a cell, thereby conferring protection against HIV. While clinical trials to date have delivered gene therapy to CD4+T cells or to CD34+ hematopoietic stem cells (HSC), the relative benefits of each of these two cellular targets have not been conclusively determined. In the present analysis, we investigated the relative merits of delivering a dual construct (CCR5 entry inhibitor + C46 fusion inhibitor) to either CD4+T cells or to CD34+ HSC. Using mathematical modelling, we determined the impact of each scenario in terms of total CD4+T cell counts over a 10 year period, and also in terms of inhibition of CCR5 and CXCR4 tropic virus. Our modelling determined that therapy delivery to CD34+ HSC generally resulted in better outcomes than delivery to CD4+T cells. An early one-off therapy delivery to CD34+ HSC, assuming that 20% of CD34+ HSC in the bone marrow were gene-modified (G+), resulted in total CD4+T cell counts ≥ 180 cells/ µL in peripheral blood after 10 years. If the uninfected G+ CD4+T cells (in addition to exhibiting lower likelihood of becoming productively infected) also exhibited reduced levels of bystander apoptosis (92.5% reduction) over non gene-modified (G-) CD4+T cells, then total CD4+T cell counts of ≥ 350 cells/ µL were observed after 10 years, even if initially only 10% of CD34+ HSC in the bone marrow received the protective gene. Taken together our results indicate that: 1.) therapy delivery to CD34+ HSC will result in better outcomes than delivery to CD4+T cells, and 2.) a greater impact of gene therapy will be observed if G+ CD4+T cells exhibit reduced levels of bystander apoptosis over G- CD4+T cells.
Collapse
Affiliation(s)
- Borislav Savkovic
- School of Mathematics and Statistics, University of New South Wales, Sydney, Australia
| | - James Nichols
- School of Mathematics and Statistics, University of New South Wales, Sydney, Australia
| | - Donald Birkett
- Department of Clinical Pharmacology, Flinders University, Adelaide, Australia
| | - Tanya Applegate
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Scott Ledger
- Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Geoff Symonds
- St Vincent's Centre for Applied Medical Research, Darlinghurst, New South Wales, Sydney, Australia
- Calimmune Pty Ltd, Darlinghurst, New South Wales, Australia
| | - John M. Murray
- School of Mathematics and Statistics, University of New South Wales, Sydney, Australia
- * E-mail:
| |
Collapse
|
27
|
Impact of chemotherapy for HIV-1 related lymphoma on residual viremia and cellular HIV-1 DNA in patients on suppressive antiretroviral therapy. PLoS One 2014; 9:e92118. [PMID: 24638072 PMCID: PMC3956871 DOI: 10.1371/journal.pone.0092118] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/17/2014] [Indexed: 11/19/2022] Open
Abstract
The first cure of HIV-1 infection was achieved through complex, multimodal therapy including myeloablative chemotherapy, total body irradiation, anti-thymocyte globulin, and allogeneic stem cell transplantation with a CCR5 delta32 homozygous donor. The contributions of each component of this therapy to HIV-1 eradication are unclear. To assess the impact of cytotoxic chemotherapy alone on HIV-1 persistence, we longitudinally evaluated low-level plasma viremia and HIV-1 DNA in PBMC from patients in the ACTG A5001/ALLRT cohort on suppressive antiretroviral therapy (ART) who underwent chemotherapy for HIV-1 related lymphoma without interrupting ART. Plasma HIV-1 RNA, total HIV-1 DNA and 2-LTR circles (2-LTRs) in PBMC were measured using sensitive qPCR assays. In the 9 patients who received moderately intensive chemotherapy for HIV-1 related lymphoma with uninterrupted ART, low-level plasma HIV-1 RNA did not change significantly with chemotherapy: median HIV-1 RNA was 1 copy/mL (interquartile range: 1.0 to 20) pre-chemotherapy versus 4 copies/mL (interquartile range: 1.0 to 7.0) post-chemotherapy. HIV-1 DNA levels also did not change significantly, with median pre-chemotherapy HIV-1 DNA of 355 copies/106 CD4+ cells versus 228 copies/106 CD4+ cells post-chemotherapy. 2-LTRs were detectable in 2 of 9 patients pre-chemotherapy and in 3 of 9 patients post-chemotherapy. In summary, moderately intensive chemotherapy for HIV-1 related lymphoma in the context of continuous ART did not have a prolonged impact on HIV-1 persistence. Clinical trials registration unique identifier: NCT00001137.
Collapse
|
28
|
Differential expression of the alternatively spliced OPRM1 isoform μ-opioid receptor-1K in HIV-infected individuals. AIDS 2014; 28:19-30. [PMID: 24413261 DOI: 10.1097/qad.0000000000000113] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE We previously examined the expression of specific C-terminal μ-opioid receptor (MOR) splice variants in human central nervous system cell types and HIV-infected brain tissue from individuals with neurocognitive impairment ± HIV encephalitis (HIVE). In the present study, we examined the N-terminal splice variant MOR-1K, which mediates excitatory cellular signaling. METHODS AND RESULTS We found segregation of expression ranging from undetectable to seemingly exclusive across nervous system cell types compared to the pool of C-terminal MOR splice variants using the real-time polymerase chain reaction (RT-PCR). Expression of MOR-1K mRNA was also increased in HIV-infected individuals with combined neurocognitive impairment and HIVE compared with the other groups. MOR-1K expression correlated with the level of patient neurocognitive impairment, whereas the pool of C-terminal MOR splice variants did not. HIVE was also associated with increased expression of the inflammatory mediators MCP-1, MCP-2, and RANTES, but not the host HIV coreceptors CXCR4 and CCR5 or the CD4 receptor using qRT-PCR. Network analysis of microarray data from these same patients revealed filamin A (FLNA) as a possible interaction partner with MOR-1K, and FLNA gene expression was also found to be upregulated in HIVE using qRT-PCR. Overexpression of FLNA in HEK293 cells redistributed MOR-1K from intracellular compartments to the cell surface. CONCLUSION These results suggest that HIVE, and neurocognitive impairment depending on its severity, are associated with enhanced MOR-1K signaling through both increased expression and trafficking to the cell surface, which may alter the contribution of MOR receptor isoforms and exacerbate the effects of MOR activation in neuroAIDS.
Collapse
|
29
|
Aiamkitsumrit B, Dampier W, Antell G, Rivera N, Martin-Garcia J, Pirrone V, Nonnemacher MR, Wigdahl B. Bioinformatic analysis of HIV-1 entry and pathogenesis. Curr HIV Res 2014; 12:132-61. [PMID: 24862329 PMCID: PMC4382797 DOI: 10.2174/1570162x12666140526121746] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 03/18/2014] [Accepted: 05/06/2014] [Indexed: 02/07/2023]
Abstract
The evolution of human immunodeficiency virus type 1 (HIV-1) with respect to co-receptor utilization has been shown to be relevant to HIV-1 pathogenesis and disease. The CCR5-utilizing (R5) virus has been shown to be important in the very early stages of transmission and highly prevalent during asymptomatic infection and chronic disease. In addition, the R5 virus has been proposed to be involved in neuroinvasion and central nervous system (CNS) disease. In contrast, the CXCR4-utilizing (X4) virus is more prevalent during the course of disease progression and concurrent with the loss of CD4(+) T cells. The dual-tropic virus is able to utilize both co-receptors (CXCR4 and CCR5) and has been thought to represent an intermediate transitional virus that possesses properties of both X4 and R5 viruses that can be encountered at many stages of disease. The use of computational tools and bioinformatic approaches in the prediction of HIV-1 co-receptor usage has been growing in importance with respect to understanding HIV-1 pathogenesis and disease, developing diagnostic tools, and improving the efficacy of therapeutic strategies focused on blocking viral entry. Current strategies have enhanced the sensitivity, specificity, and reproducibility relative to the prediction of co-receptor use; however, these technologies need to be improved with respect to their efficient and accurate use across the HIV-1 subtypes. The most effective approach may center on the combined use of different algorithms involving sequences within and outside of the env-V3 loop. This review focuses on the HIV-1 entry process and on co-receptor utilization, including bioinformatic tools utilized in the prediction of co-receptor usage. It also provides novel preliminary analyses for enabling identification of linkages between amino acids in V3 with other components of the HIV-1 genome and demonstrates that these linkages are different between X4 and R5 viruses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Brian Wigdahl
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA 19102.
| |
Collapse
|
30
|
Barmania F, Pepper MS. C-C chemokine receptor type five (CCR5): An emerging target for the control of HIV infection. Appl Transl Genom 2013; 2:3-16. [PMID: 27942440 PMCID: PMC5133339 DOI: 10.1016/j.atg.2013.05.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 05/19/2013] [Accepted: 05/21/2013] [Indexed: 12/25/2022]
Abstract
When HIV was initially discovered as the causative agent of AIDS, many expected to find a vaccine within a few years. This has however proven to be elusive; it has been approximately 30 years since HIV was first discovered, and a suitable vaccine is still not in effect. In 2009, a paper published by Hutter et al. reported on a bone marrow transplant performed on an HIV positive individual using stem cells that were derived from a donor who was homozygous for a mutation in the CCR5 gene known as CCR5 delta-32 (Δ32) (Hütter et al., 2009). The HIV positive individual became HIV negative and remained free of viral detection after transplantation despite having halted anti-retroviral (ARV) treatment. This review will focus on CCR5 as a key component in HIV immunity and will discuss the role of CCR5 in the control of HIV infection.
Collapse
Affiliation(s)
| | - Michael S. Pepper
- Corresponding author at: Dept. of Immunology, Faculty of Health Sciences, University of Pretoria, P.O. Box 2034, Pretoria 0001, South Africa. Tel.: + 27 12 319 2190; fax: + 27 12 319 2946.
| |
Collapse
|
31
|
Zwolińska K, Knysz B, Rybka K, Pazgan-Simon M, Gąsiorowski J, Sobczyński M, Gładysz A, Piasecki E. Protective effect of CCR5-Δ32 against HIV infection by the heterosexual mode of transmission in a Polish population. AIDS Res Hum Retroviruses 2013; 29:54-60. [PMID: 22957692 DOI: 10.1089/aid.2011.0362] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Effects of chemokine receptor alleles (CCR5-Δ32 and CCR2-64I) on susceptibility to human immunodeficiency virus (HIV) infection were studied in a Polish population. The CCR5 and CCR2 genotypes were determined for 311 healthy, HIV-negative individuals (control group), 121 exposed to HIV infection but uninfected (EU group), and 470 HIV-positive patients. The frequency of the alleles in the control group was calculated as 0.12 for both CCR5-Δ32 and CCR2-64I. The logistic regression method was used to analyze the effects of the described factors. A protective effect was observed for the CCR5-Δ32 allele but only in the case of heterosexual exposure. Prevalence of the CCR5-Δ32/+ genotype in HIV(+) patients infected via the heterosexual route (n=61; 8.2%) was much lower than in the control group (n=311; 21.5%); in the heterosexually exposed uninfected group it was slightly higher (n=28; 25%). This suggested that in this mode of infection, the native CCR5 expression level was crucial for establishment of infection. Individuals with the CCR5-Δ32 allele have more than three times less chance of infection in the case of HIV heterosexual exposure (odds ratio, 3.37; 95% confidence interval, 1.055-10.76). However, a protective effect of the CCR5-Δ32/+ genotype was not observed in the case of intravenous drug users (IDUs). The rates of the genotype were similar in HIV-infected IDU individuals (n=356; 17.7%) and in exposed uninfected patients (n=84; 15.5%), not significantly different from control group. No effect of the CCR2 genotype was observed. The analysis revealed that the important factor increasing infection risk was, in particular, hepatitis C virus (HCV) infection (odds ratio, 12.9). Moreover, the effect of HCV infection was found to be age dependent. Susceptibility to HIV infection resulting from HCV positivity became weaker (6% per year) with increasing age.
Collapse
Affiliation(s)
- Katarzyna Zwolińska
- Laboratory of Virology, Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Brygida Knysz
- Department and Clinic of Infectious Diseases, Hepatology, and Acquired Immune Deficiencies, Wrocław Medical University, Wrocław, Poland
| | - Katarzyna Rybka
- Laboratory of Virology, Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Monika Pazgan-Simon
- Department and Clinic of Infectious Diseases, Hepatology, and Acquired Immune Deficiencies, Wrocław Medical University, Wrocław, Poland
| | - Jacek Gąsiorowski
- Department and Clinic of Infectious Diseases, Hepatology, and Acquired Immune Deficiencies, Wrocław Medical University, Wrocław, Poland
| | - Maciej Sobczyński
- Department of Genomics, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - Andrzej Gładysz
- Department and Clinic of Infectious Diseases, Hepatology, and Acquired Immune Deficiencies, Wrocław Medical University, Wrocław, Poland
| | - Egbert Piasecki
- Laboratory of Virology, Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
- Institute of Chemistry, Environmental Protection, and Biotechnology, Jan Długosz University, Częstochowa, Poland
| |
Collapse
|
32
|
Messiaen P, De Spiegelaere W, Alcami J, Vervisch K, Van Acker P, Verhasselt B, Meuwissen P, Calonge E, Gonzalez N, Gutierrez-Rodero F, Rodriguez-Martín C, Sermijn E, Poppe B, Vogelaers D, Verhofstede C, Vandekerckhove L. Characterization of LEDGF/p75 genetic variants and association with HIV-1 disease progression. PLoS One 2012; 7:e50204. [PMID: 23226247 PMCID: PMC3511443 DOI: 10.1371/journal.pone.0050204] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 10/16/2012] [Indexed: 12/24/2022] Open
Abstract
Background As Lens epithelium-derived growth factor (LEDGF/p75) is an important co-factor involved in HIV-1 integration, the LEDGF/p75-IN interaction is a promising target for the new class of allosteric HIV integrase inhibitors (LEDGINs). Few data are available on the genetic variability of LEDGF/p75 and the influence on HIV disease in vivo. This study evaluated the relation between LEDGF/p75 genetic variation, mRNA expression and HIV-1 disease progression in order to guide future clinical use of LEDGINs. Methods Samples were derived from a therapy-naïve cohort at Ghent University Hospital and a Spanish long-term-non-progressor cohort. High-resolution melting curve analysis and Sanger sequencing were used to identify all single nucleotide polymorphisms (SNPs) in the coding region, flanking intronic regions and full 3′UTR of LEDGF/p75. In addition, two intronic tagSNPs were screened based on previous indication of influencing HIV disease. LEDGF/p75 mRNA was quantified in patient peripheral blood mononuclear cells (PBMC) using RT-qPCR. Results 325 samples were investigated from patients of Caucasian (n = 291) and African (n = 34) origin, including Elite (n = 49) and Viremic controllers (n = 62). 21 SNPs were identified, comprising five in the coding region and 16 in the non-coding regions and 3′UTR. The variants in the coding region were infrequent and had no major impact on protein structure according to SIFT and PolyPhen score. One intronic SNP (rs2737828) was significantly under-represented in Caucasian patients (P<0.0001) compared to healthy controls (HapMap). Two SNPs showed a non-significant trend towards association with slower disease progression but not with LEDGF/p75 expression. The observed variation in LEDGF/p75 expression was not correlated with disease progression. Conclusions LEDGF/p75 is a highly conserved protein. Two non-coding polymorphisms were identified indicating a correlation with disease outcome, but further research is needed to clarify phenotypic impact. The conserved coding region and the observed variation in LEDGF/p75 expression are important characteristics for clinical use of LEDGINs.
Collapse
Affiliation(s)
- Peter Messiaen
- HIV Translational Research Unit, Department of General Internal Medicine and Infectious Diseases, Ghent University Hospital, Ghent, Belgium
| | - Ward De Spiegelaere
- HIV Translational Research Unit, Department of General Internal Medicine and Infectious Diseases, Ghent University Hospital, Ghent, Belgium
| | - Jose Alcami
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Karen Vervisch
- HIV Translational Research Unit, Department of General Internal Medicine and Infectious Diseases, Ghent University Hospital, Ghent, Belgium
| | - Petra Van Acker
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Bruno Verhasselt
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Pieter Meuwissen
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Esther Calonge
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Nuria Gonzalez
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - Erica Sermijn
- HIV Translational Research Unit, Department of General Internal Medicine and Infectious Diseases, Ghent University Hospital, Ghent, Belgium
| | - Bruce Poppe
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Dirk Vogelaers
- HIV Translational Research Unit, Department of General Internal Medicine and Infectious Diseases, Ghent University Hospital, Ghent, Belgium
| | - Chris Verhofstede
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Linos Vandekerckhove
- HIV Translational Research Unit, Department of General Internal Medicine and Infectious Diseases, Ghent University Hospital, Ghent, Belgium
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
- * E-mail:
| |
Collapse
|
33
|
O'Hayre M, Salanga CL, Handel TM, Hamel DJ. Emerging concepts and approaches for chemokine-receptor drug discovery. Expert Opin Drug Discov 2012; 5:1109-22. [PMID: 21132095 DOI: 10.1517/17460441.2010.525633] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
IMPORTANCE OF THE FIELD Chemokine receptors are most noted for their role in cell migration. However, inappropriate utilization or regulation of these receptors is implicated in many inflammatory diseases, cancer and HIV, making them important drug targets. AREAS COVERED IN THIS REVIEW Allostery, oligomerization and ligand bias are presented as they pertain to chemokine receptors and their associated pathologies.Specific examples of each are described from the recent literature and their implications are discussed in terms of drug discovery efforts targeting chemokine receptors. WHAT THE READER WILL GAIN Insight into the expanding view of the multitude of pharmacological variables that need to be considered or that may be exploited in chemokine receptor drug discovery. TAKE HOME MESSAGE Since 2007, two drugs targeting chemokine receptors have been approved by the FDA, Maraviroc for preventing HIV infection and Mozobil™ for hematopoietic stem cell mobilization. While these successes permit optimism for chemokine receptors as drug targets, only recently has the complexity of this system begun to be appreciated. The concepts of allosteric inhibitors, biased ligands and functional selectivity raise the possibility that drugs with precisely-defined properties can be developed. Other complexities such as receptor oligomerization and tissue-specific functional states of receptors also offer opportunities for increased target and response specificity, although it will be more challenging to translate these ideas into approved therapeutics compared to traditional approaches.
Collapse
Affiliation(s)
- Morgan O'Hayre
- University of California, San Diego, Skaggs School of Pharmacy andPharmaceutical Sciences, La Jolla, CA 92093, USA
| | | | | | | |
Collapse
|
34
|
Savkovic B, Symonds G, Murray JM. Stochastic model of in-vivo X4 emergence during HIV infection: implications for the CCR5 inhibitor maraviroc. PLoS One 2012; 7:e38755. [PMID: 22866173 PMCID: PMC3398969 DOI: 10.1371/journal.pone.0038755] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 05/11/2012] [Indexed: 12/30/2022] Open
Abstract
The emergence of X4 tropic viral strains throughout the course of HIV infection is associated with poorer prognostic outcomes and faster progressions to AIDS than for patients in whom R5 viral strains predominate. Here we investigate a stochastic model to account for the emergence of X4 virus via mutational intermediates of lower fitness that exhibit dual/mixed (D/M) tropism, and employ the model to investigate whether the administration of CCR5 blockers in-vivo is likely to promote a shift towards X4 tropism. We show that the proposed stochastic model can account for X4 emergence with a median time of approximately 4 years post-infection as a result of: 1.) random stochastic mutations in the V3 region of env during the reverse transcription step of infection; 2.) increasing numbers of CXCR4-expressing activated naive CD4+ T cells with declining total CD4+ T cell counts, thereby providing increased numbers of activated target cells for productive infection by X4 virus. Our model indicates that administration of the CCR5 blocker maraviroc does not promote a shift towards X4 tropism, assuming sufficient efficacy of background therapy (BT). However our modelling also indicates that administration of maraviroc as a monotherapy or with BT of suboptimal efficacy can promote emergence of X4 tropic virus, resulting in accelerated progression to AIDS. Taken together, our results demonstrate that maraviroc is safe and effective if co-administered with sufficiently potent BT, but that suboptimal BT may promote X4 emergence and accelerated progression to AIDS. These results underscore the clinical importance for careful selection of BT when CCR5 blockers are administered in-vivo.
Collapse
Affiliation(s)
- Borislav Savkovic
- School of Mathematics and Statistics, University of New South Wales, Sydney, Australia.
| | | | | |
Collapse
|
35
|
Kuang YQ, Charette N, Frazer J, Holland PJ, Attwood KM, Dellaire G, Dupré DJ. Dopamine receptor-interacting protein 78 acts as a molecular chaperone for CCR5 chemokine receptor signaling complex organization. PLoS One 2012; 7:e40522. [PMID: 22815758 PMCID: PMC3398031 DOI: 10.1371/journal.pone.0040522] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 06/11/2012] [Indexed: 11/18/2022] Open
Abstract
Chemokine receptors are members of the G protein-coupled receptor (GPCR) family. CCR5 and CXCR4 act as co-receptors for human immunodeficiency virus (HIV) and several efforts have been made to develop ligands to inhibit HIV infection by blocking those receptors. Removal of chemokine receptors from the cell surface using polymorphisms or other means confers some levels of immunity against HIV infection. Up to now, very limited success has been obtained using ligand therapies so we explored potential avenues to regulate chemokine receptor expression at the plasma membrane. We identified a molecular chaperone, DRiP78, that interacts with both CXCR4 and CCR5, but not the heterodimer formed by these receptors. We further characterized the effects of DRiP78 on CCR5 function. We show that the molecular chaperone inhibits CCR5 localization to the plasma membrane. We identified the interaction region on the receptor, the F(x)6LL motif, and show that upon mutation of this motif the chaperone cannot interact with the receptor. We also show that DRiP78 is involved in the assembly of CCR5 chemokine signaling complex as a homodimer, as well as with the Gαi protein. Finally, modulation of DRiP78 levels will affect receptor functions, such as cell migration in cells that endogenously express CCR5. Our results demonstrate that modulation of the functions of a chaperone can affect signal transduction at the cell surface.
Collapse
Affiliation(s)
- Yi-Qun Kuang
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Nicholle Charette
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jennifer Frazer
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Patrick J. Holland
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kathleen M. Attwood
- Department of Pathology, Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Graham Dellaire
- Department of Pathology, Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Denis J. Dupré
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- * E-mail:
| |
Collapse
|
36
|
Borinskaya S, Kozhekbaeva Z, Zalesov A, Olseeva E, Maksimov A, Kutsev S, Garaev M, Rubanovich A, Yankovsky N. Risk of HIV Infection and Lethality Are Decreased in CCR5del32 Heterozygotes: Focus Nosocomial Infection Study and Meta-analysis. Acta Naturae 2012; 4:42-52. [PMID: 22708062 PMCID: PMC3372986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
CCR5del32 Homozygous deletion in the chemokine receptor R5 gene provides almost complete protection to individuals against HIV infection. However, data relating to the protective effect forCCR5del32 heterozygous individuals have been contradictory. The frequency of theCCR5del32allele in population control cohorts was compared with that of a group of children (27 Kalmyks and 50 Russians) infected by G-subtype HIV-1 in a nosocomial outbreak. The frequency of theCCR5del32allele was shown to be lower among the infected children in comparison with that of the control group; however, the difference was small and statistically insignificant. Similar results were obtained in a number of earlier studies. The insignificance of the small differences could be a result of one of two reasons. (i) The fact that there is no protective effect of the heterozygous state, and that the phenomenon depends only on the fluctuation of allele frequencies. In this case, there would be no differences even if the infected cohort is enlarged. (ii)The protective effect of the heterozygous state is real; however, the size of the studied cohort is insufficient to demonstrate it. In order to discern between these two reasons, a meta-analysis of data from 25 published articles (a total of 5,963 HIV-infected individuals and 5,048 individuals in the control group, including the authors' own data) was undertaken. A conclusion was drawn from the meta-analysis that theCCR5del32 allele protects individuals against the HIV infection even in a heterozygous state (OR=1.22, 95%CI=1.10-1.36). The risk of HIV infection forCCR5 wt/del32 heterozygotes was lower by at least 13% as compared to that for wild typeCCR5 wt/wthomozygotes. Prior to this study, no data of the type or any conclusions had been published for Caucasians. The mortality rate in the 15 years following the infection was found to be approximately 40% lower forCCR5del32 heterozygotes in comparison with that for the wild type homozygotes in the studied group. The size of the studied group was insufficient to claim difference validity (OR=2.0;p= 0.705), even though the effect quantitatively matched the published data. The features of the meta-analysis influencing the threshold level and the statistical validity of the effects are being discussed. The level of theCCR5del32 protective effect on the chances to be infected with HIV and on the outcome of the HIV infection was assessed for various ethnic groups.
Collapse
Affiliation(s)
- S.A. Borinskaya
- Vavilov Institute of General Genetics, Russian Academy of Sciences
| | | | - A.V. Zalesov
- Vavilov Institute of General Genetics, Russian Academy of Sciences
- Moscow Institute of Physics and Technology
| | - E.V. Olseeva
- Ministry of Health and Social Development of the Republic of
Kalmykia
| | - A.R. Maksimov
- Vavilov Institute of General Genetics, Russian Academy of Sciences
| | | | - M.M. Garaev
- Ivanovsky Research Institute of Virology, Russian Academy of Medical
Sciences
| | - A.V. Rubanovich
- Vavilov Institute of General Genetics, Russian Academy of Sciences
| | - N.K. Yankovsky
- Vavilov Institute of General Genetics, Russian Academy of Sciences
- Moscow Institute of Physics and Technology
- Faculty of Biology, Lomonosov Moscow State University
| |
Collapse
|
37
|
Charette N, Holland P, Frazer J, Allen H, Dupré DJ. Dependence on different Rab GTPases for the trafficking of CXCR4 and CCR5 homo or heterodimers between the endoplasmic reticulum and plasma membrane in Jurkat cells. Cell Signal 2011; 23:1738-49. [DOI: 10.1016/j.cellsig.2011.06.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 06/07/2011] [Accepted: 06/15/2011] [Indexed: 10/18/2022]
|
38
|
Madlala P, Gijsbers R, Christ F, Hombrouck A, Werner L, Mlisana K, An P, Abdool Karim SS, Winkler CA, Debyser Z, Ndung'u T. Association of polymorphisms in the LEDGF/p75 gene (PSIP1) with susceptibility to HIV-1 infection and disease progression. AIDS 2011; 25:1711-9. [PMID: 21681054 PMCID: PMC3233670 DOI: 10.1097/qad.0b013e328349c693] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE LEDGF/p75, encoded by the PSIP1 gene, interacts with HIV-1 integrase and targets HIV-1 integration into active genes. We investigated the influence of polymorphisms in PSIP1 on HIV-1 acquisition and disease progression in black South Africans. METHODS Integrase binding domain of LEDGF/p75 was sequenced in 126 participants. Four haplotype tagging SNPs rs2277191, rs1033056, rs12339417 and rs10283923 referred to as SNP1, SNP2, SNP3 and SNP4, respectively, and one exonic SNP rs61744944 (SNP5, Q472L) were genotyped in 195 HIV-1 seronegative, 52 primary and 403 chronically infected individuals using TaqMan assays. LEDGF/p75 expression was quantified by real-time RT-PCR. The impact of Q472L mutation on the interaction with HIV_1 IN was measured by AlphaScreen. RESULTS rs2277191 (SNP1) A was more frequent among seropositives (P = 0.06, Fisher's exact test). Among individuals followed longitudinally SNP1A trended towards association with higher likelihood of HIV-1 acquisition [relative hazard (RH) = 2.21, P = 0.08; Cox model] and it was also associated with rapid disease progression (RH = 5.98, P = 0.04; Cox model) in the recently infected (primary infection) cohort. rs12339417 (SNP3)C was associated with slower decline of CD4(+) T cells (P = 0.02) and lower messenger RNA (mRNA) levels of LEDGF/p75 (P < 0.01). Seroconverters had higher preinfection mRNA levels of LEDGF/p75 (P < 0.01) and these levels decreased after HIV-1 infection (P = 0.02). CONCLUSIONS Genetic variants of PSIP1 may affect HIV-1 outcomes. Further studies are needed to confirm the effect of genetic variation of PSIP1 on HIV-1 pathogenesis in different cohorts.
Collapse
Affiliation(s)
- Paradise Madlala
- HIV Pathogenesis Programme, University of KwaZulu-Natal, Durban, South Africa
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
CD4+ T cells from elite suppressors are more susceptible to HIV-1 but produce fewer virions than cells from chronic progressors. Proc Natl Acad Sci U S A 2011; 108:E689-98. [PMID: 21873218 DOI: 10.1073/pnas.1108866108] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Elite suppressors/controllers (ES) are HIV-1-infected individuals who maintain stable CD4(+) T-cell counts and viral loads of <50 copies/mL without antiretroviral therapy. Research has predominantly focused on immune factors contributing to the control of viral replication in these patients. A more fundamental question, however, is whether there are differences in the nature of CD4(+) T-cell infection in ES compared with viremic patients. Here, we compare chronic progressor (CP), ES, and uninfected donors in terms of three aspects of CD4(+) T-cell infection: cellular susceptibility to infection, death of infected cells, and production of virus from infected cells. Using multiple methods of infection and both single-cycle and replication-competent virus, we show that unmanipulated CD4(+) T-cell populations from ES are actually more susceptible to HIV-1 infection than those populations from CP. Depletion of highly susceptible cells in CP may contribute to this difference. Using 7AAD and AnnexinV staining, we show that infected cells die more rapidly than uninfected cells, but the increased death of infected cells from CP and ES is proportional. Finally, using an assay for measuring virus production, we show that virus production by cells from CP is high compared with virus production by cells from ES or uninfected donors. This higher virus production is linked to cellular activation levels. These data identify fundamental differences in chronic infection of ES and CP that likely contribute to differential HIV-1 disease progression.
Collapse
|
40
|
Jin Q, Altenburg JD, Hossain MM, Alkhatib G. Role for the conserved N-terminal cysteines in the anti-chemokine activities by the chemokine-like protein MC148R1 encoded by Molluscum contagiosum virus. Virology 2011; 417:449-56. [PMID: 21802105 DOI: 10.1016/j.virol.2011.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 04/21/2011] [Accepted: 07/05/2011] [Indexed: 11/29/2022]
Abstract
Molluscum contagiosum poxvirus (MCV) type 1 and type 2 encode two chemokine-like proteins MC148R1 and MC148R2. It is believed that MC148R proteins function by blocking the inflammatory response. However, the mechanism of the proposed biological activities of MC148R proteins and the role of the additional C-terminal cysteines that do not exist in other chemokines are not understood. Here, we demonstrated in two different assay systems that His-tagged MC148R1 displaces the interaction between CXCL12α and CXCR4. The N-terminal cysteines but not the additional C-terminal cysteines modulate this displacement. His-tagged MC148R1 blocked both CXCL12α-mediated and MIP-1α-mediated chemotaxis. In contrast, MC148R2 blocked MIP-1α-mediated but not CXCL12α-mediated chemotaxis. Immunoprecipitation by antibodies to MC148R1 or CXCL12α followed by immunoblotting and detection by antibodies to the other protein demonstrated physical interaction of His-tagged CXCL12α and His-tagged MC148R1. Interaction with chemokines might mask the receptor interaction site resulting in decreased binding and impairment of the biological activities.
Collapse
Affiliation(s)
- Qingwen Jin
- Department of Biomedical Sciences, Center of Excellence for Infectious Diseases, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA
| | | | | | | |
Collapse
|
41
|
Zhang J, Fu Y, Li G, Zhao RY, Lakowicz JR. Direct observation of chemokine receptors 5 on T-lymphocyte cell surfaces using fluorescent metal nanoprobes 2: Approximation of CCR5 populations. Biochem Biophys Res Commun 2011; 407:63-7. [PMID: 21356199 PMCID: PMC3077572 DOI: 10.1016/j.bbrc.2011.02.104] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Accepted: 02/21/2011] [Indexed: 11/20/2022]
Abstract
Metal nanoparticle probes were used as molecular imaging agents to detect the expression levels and spatial distributions of the CCR5 receptors on the cell surfaces. Alexa Fluor 647-labeled anti-CCR5 monoclonal antibodies (mAbs) were covalently bound to 20 nm silver nanoparticles to synthesize the mAb-metal complexes. We measured the single nanoparticle emission of the mAb-metal complexes, showing that the complexes displayed enhanced intensities and reduced lifetimes in comparison with the metal-free mAbs. Six HeLa cell lines with various CCR5 expressions were incubated with the mAb-metal complexes for the target-specific binding to the cell surfaces. Fluorescence cell images were recorded on a time-resolved confocal microscope. The collected images expressed clear CCR5 expression-dependent optical properties. Two regression curves were obtained on the basis of the emission intensity and lifetime over the entire cell images against the number of the CCR5 expression on the cells. The emission from the single mAb-metal complexes could be distinctly identified from the cellular autofluorescence on the cell images. The CCR5 spatial distributions on the cells were analyzed on the cell images and showed that the low-expression cells have the CCR5 receptors as individuals or small clusters but the high expression cells have them as the dense and discrete clusters on the cell surfaces.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Biochemistry and Molecular Biology, Center for Fluorescence Spectroscopy, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD 21201, United States.
| | | | | | | | | |
Collapse
|
42
|
Zhang J, Fu Y, Li G, Zhao RY, Lakowicz JR. Detection of CXCR4 receptors on cell surface using a fluorescent metal nanoshell. JOURNAL OF BIOMEDICAL OPTICS 2011; 16:016011. [PMID: 21280917 PMCID: PMC3041243 DOI: 10.1117/1.3528623] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 11/17/2010] [Accepted: 11/24/2010] [Indexed: 05/30/2023]
Abstract
Fluorescence cell imaging can be used for disease diagnosis and cellular signal transduction. Using a metal nanoshell as molecular imaging agent, we develop a cellular model system to detect CXCR4 chemokine receptor on T-lymphatic cell surface. These metal nanoshells are observed to express enhanced emission intensity and shortened lifetimes due to the near-field interactions. They are covalently bound with anti-CXCR4 monoclonal antibodies for immunoreactions with the target sites of the CXCR4 receptors on the CEM-SS cells. The fluorescence intensity and lifetime cell images are recorded with a time-resolved confocal microscopy. As expected, the emission signals from the metal nanoshells are clearly isolated from the cellular autofluorescence due to strong intensities and distinctive lifetimes. The number of emission spots on the single cell image is estimated by direct count to the emission signals. Analyzing a pool of cell images, a maximal count number is obtained in a range of 200±50. Because there is an average of ̃approximately 6000 binding sites on the cell surface, we estimate that one emission spot from the metal nanoshell may represent ̃approximately 30 CXCR5 receptors. In addition, the CXCR4 receptors are estimated to distribute on ̃approximately 70% area of the cell surface.
Collapse
Affiliation(s)
- Jian Zhang
- University of Maryland School of Medicine, Center for Fluorescence Spectroscopy, Department of Biochemistry and Molecular Biology, 725 West Lombard Street, Baltimore, Maryland 21201, USA.
| | | | | | | | | |
Collapse
|
43
|
Abstract
HIV entry into CD4(+) cells requires interaction with a cellular receptor, generally either CCR5 or CXCR4. We have previously reported the case of an HIV-infected patient in whom viral replication remained absent despite discontinuation of antiretroviral therapy after transplantation with CCR5Δ32/Δ32 stem cells. However, it was expected that the long-lived viral reservoir would lead to HIV rebound and disease progression during the process of immune reconstitution. In the present study, we demonstrate successful reconstitution of CD4(+) T cells at the systemic level as well as in the gut mucosal immune system after CCR5Δ32/Δ32 stem cell transplantation, while the patient remains without any sign of HIV infection. This was observed although recovered CD4(+) T cells contain a high proportion of activated memory CD4(+) T cells, ie, the preferential targets of HIV, and are susceptible to productive infection with CXCR4-tropic HIV. Furthermore, during the process of immune reconstitution, we found evidence for the replacement of long-lived host tissue cells with donor-derived cells, indicating that the size of the viral reservoir has been reduced over time. In conclusion, our results strongly suggest that cure of HIV has been achieved in this patient.
Collapse
|
44
|
Unstimulated primary CD4+ T cells from HIV-1-positive elite suppressors are fully susceptible to HIV-1 entry and productive infection. J Virol 2010; 85:979-86. [PMID: 21068257 DOI: 10.1128/jvi.01721-10] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Elite controllers or suppressors (ES) are a group of HIV-1-infected individuals who maintain viral loads below the limit of detection of commercial assays for many years. The mechanisms responsible for this remarkable control are under intense study, with the hope of developing therapeutic vaccines effective against HIV-1. In this study, we addressed the question of the intrinsic susceptibility of ES CD4(+) T cells to infection. While we and others have previously shown that CD4(+) T cells from ES can be infected by HIV-1 isolates in vitro, these studies were confounded by exogenous activation and in vitro culture of CD4(+) T cells prior to infection. In order to avoid the changes in chemokine receptor expression that have been associated with such exogenous activation, we infected purified CD4(+) T cells directly after isolation from the peripheral blood of ES, viremic patients, and uninfected donors. We utilized a green fluorescent protein (GFP)-expressing proviral construct pseudotyped with CCR5-tropic or CXCR4-tropic envelope to compare viral entry using a fluorescence resonance energy transfer-based, single-round virus-cell fusion assay. The frequency of productive infection was also compared by assessing GFP expression. CD4(+) T cells from ES were as susceptible as or more susceptible than cells from viremic patients and uninfected donors to HIV-1 entry and productive infection. The results of this physiological study strongly suggest that differences in HIV-1 entry and infection of CD4(+) T cells alone cannot explain the elite control of viral replication.
Collapse
|
45
|
Hammad MM, Kuang YQ, Yan R, Allen H, Dupré DJ. Na+/H+ exchanger regulatory factor-1 is involved in chemokine receptor homodimer CCR5 internalization and signal transduction but does not affect CXCR4 homodimer or CXCR4-CCR5 heterodimer. J Biol Chem 2010; 285:34653-64. [PMID: 20801883 PMCID: PMC2966081 DOI: 10.1074/jbc.m110.106591] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 08/25/2010] [Indexed: 11/06/2022] Open
Abstract
Chemokine receptors are members of the G protein-coupled receptor (GPCR) family. CCR5 is also the principal co-receptor for macrophage-tropic strains of human immunodeficiency virus, type 1 (HIV-1), and efforts have been made to develop ligands to inhibit HIV-1 infection by promoting CCR5 receptor endocytosis. Given the nature of GPCRs and their propensity to form oligomers, one can consider ligand-based therapies as unselective in terms of the oligomeric composition of complexes. For example, a ligand targeting a CCR5 homomer could likely induce signal transduction on a heteromeric CCR5-CXCR4. Other avenues could therefore be explored. We identified a receptor adaptor interacting specifically with one receptor complex but not others. NHERF1, an adaptor known for its role in desensitization, internalization, and regulation of the ERK signaling cascade for several GPCRs, interacts via its PDZ2 domain with the CCR5 homodimer but not with the CXCR4-CCR5 heterodimer or CXCR4 homodimer. To further characterize this interaction, we also show that NHERF1 increases the CCR5 recruitment of arrestin2 following stimulation. NHERF1 is also involved in CCR5 internalization, as we demonstrate that co-expression of constructs bearing the PDZ2 domain can block CCR5 internalization. We also show that NHERF1 potentiates RANTES (regulated on activation normal T cell expressed and secreted)-induced ERK1/2 phosphorylation via CCR5 activation and that this activation requires NHERF1 but not arrestin2. Taken together, our results suggest that oligomeric receptor complexes can associate specifically with partners and that in this case NHERF1 could represent an interesting new target for the regulation of CCR5 internalization and potentially HIV infection.
Collapse
Affiliation(s)
- Maha M. Hammad
- From the Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| | - Yi-Qun Kuang
- From the Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| | - Ronald Yan
- From the Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| | - Heather Allen
- From the Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| | - Denis J. Dupré
- From the Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| |
Collapse
|
46
|
Alkhatib G. Counterpoint: Cord blood stem cell therapy for acquired immune deficiency syndrome. Stem Cells Dev 2010; 18:5-6. [PMID: 19196115 DOI: 10.1089/scd.2008.0304.cp2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Ghalib Alkhatib
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| |
Collapse
|
47
|
Gonzalez G, Shpall EJ, Gathe JC, Behringer RR. Cord blood stem cell therapy for acquired immune deficiency syndrome. Stem Cells Dev 2010; 18:1-2. [PMID: 19018697 DOI: 10.1089/scd.2008.0304] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cord blood stem cell transplantation is routinely used to treat hematopoietic diseases. Individuals who are homozygous for the 32 polymorphism of the CCR5 locus, encoding a co-receptor for HIV-1, are normal and are resistant to HIV infection. Here we suggest that public cord blood repositories are likely to contain CCR5 homozygous units that could be used as a therapy for HIV-infected individuals.
Collapse
Affiliation(s)
- Gabriel Gonzalez
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
48
|
|
49
|
Psomas KC, Corbeau P, Reynes J. [CCR5 antagonists and HIV-1 infection: Bases and consequences of this therapeutic approach]. ACTA ACUST UNITED AC 2010; 12:27-41. [PMID: 32288525 PMCID: PMC7146793 DOI: 10.1016/j.antib.2010.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
La molécule CCR5 est un récepteur de chimiokines qui joue un rôle important en pathologie infectieuse : corécepteur des souches du VIH-1 à tropisme R5, il est également impliqué dans la défense immunitaire contre certains agents transmissibles. Les antagonistes de CCR5 constituent une nouvelle approche thérapeutique antirétrovirale. Trois inhibiteurs du CCR5 ont atteint les phases IIb et III de développement clinique : aplaviroc (GlaxoSmithKine), vicriviroc (Schering-Plough) et maraviroc (Pfizer). Le développement de l’aplaviroc a été interrompu pour toxicité hépatique. Les essais ACTG 5211 et Motivate ont démontré une amélioration de la réponse antirétrovirale par l’addition respectivement de vicriviroc (actuellement en phase III) et de maraviroc (ayant déjà obtenu l’Autorisation de Mise sur le Marché) à un traitement optimisé chez des patients en échec thérapeutique. Le rôle de cette nouvelle cible thérapeutique dans les stratégies de traitement initial, de substitution ou de sauvetage reste à préciser, de même que leur intérêt chez des patients ayant une réponse immunovirologique dissociée, en immunodépresssion sévère ou infectés par des souches à tropisme non-R5. Plusieurs points sont également à éclaircir comme la tolérance à long terme, le risque d’induire une commutation R5-X4, en particulier dans les tissus, le risque d’interférer avec les réponses immunitaires, ainsi que l’impact d’une discordance de tropisme entre le plasma et les autres compartiments de l’organisme.
Collapse
Affiliation(s)
- K C Psomas
- Institut de génétique humaine, CNRS, 142, rue de la Cardonille, 34396 Montpellier cedex 5, France.,Service des maladies infectieuses et tropicales, CHU Gui-de-Chauliac, 80, avenue Augustin-Fliche, 34295 Montpellier cedex 5, France
| | - P Corbeau
- Institut de génétique humaine, CNRS, 142, rue de la Cardonille, 34396 Montpellier cedex 5, France.,Fonctionnelle d'immunologie, hôpital Carémeau, place du Pr-Robert-Debré, 30029 Nîmes cedex, France.,Faculté de médecine, université Montpellier 1, 2, rue École-de-Médecine, 34060 Montpellier cedex 2, France
| | - J Reynes
- Faculté de médecine, université Montpellier 1, 2, rue École-de-Médecine, 34060 Montpellier cedex 2, France.,Service des maladies infectieuses et tropicales, CHU Gui-de-Chauliac, 80, avenue Augustin-Fliche, 34295 Montpellier cedex 5, France.,UMR 145, 911, avenue Agropolis, 34394 Montpellier cedex 5, France
| |
Collapse
|
50
|
Lim JK, McDermott DH, Lisco A, Foster GA, Krysztof D, Follmann D, Stramer SL, Murphy PM. CCR5 deficiency is a risk factor for early clinical manifestations of West Nile virus infection but not for viral transmission. J Infect Dis 2010; 201:178-85. [PMID: 20025530 DOI: 10.1086/649426] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND West Nile virus (WNV) is a neurotropic flavivirus transmitted to humans by mosquito vectors. Homozygosity for CCR5Delta32, a complete loss-of-function mutation in CC chemokine receptor 5 (CCR5), has been previously associated with severe symptomatic WNV infection in patients who present with clinical disease; however, whether it acts at the level of initial infection or in promoting clinical progression is unknown. METHODS Here, we address this gap in knowledge by comparing CCR5Delta32 distribution among US blood donors identified through a comprehensive blood supply screening program (34,766,863 donations from 2003 through 2008) as either WNV true positive (634 WNV-positive cases) or false positive (422 WNV-negative control participants). All subjects self-reported symptoms occurring during the 2 weeks following blood donation using a standardized questionnaire. RESULTS No difference was observed in CCR5Delta32 homozygous frequency between the WNV-positive cases and WNV-negative control participants. However, CCR5Delta32 homozygosity was associated in cases but not controls with clinical symptoms consistent with WNV infection (P = .002). CONCLUSIONS CCR5 deficiency is not a risk factor for WNV infection per se, but it is a risk factor for both early and late clinical manifestations after infection. Thus, CCR5 may function normally to limit disease due to WNV infection in humans.
Collapse
Affiliation(s)
- Jean K Lim
- Molecular Signaling Section, Laboratory of Molecular Immunology, Laboratory of Cellular and Molecular Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|