101
|
|
102
|
Yoon D, Kim H, Suh-Kim H, Park RW, Lee K. Differentially co-expressed interacting protein pairs discriminate samples under distinct stages of HIV type 1 infection. BMC SYSTEMS BIOLOGY 2011; 5 Suppl 2:S1. [PMID: 22784566 PMCID: PMC3287475 DOI: 10.1186/1752-0509-5-s2-s1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background Microarray analyses based on differentially expressed genes (DEGs) have been widely used to distinguish samples across different cellular conditions. However, studies based on DEGs have not been able to clearly determine significant differences between samples of pathophysiologically similar HIV-1 stages, e.g., between acute and chronic progressive (or AIDS) or between uninfected and clinically latent stages. We here suggest a novel approach to allow such discrimination based on stage-specific genetic features of HIV-1 infection. Our approach is based on co-expression changes of genes known to interact. The method can identify a genetic signature for a single sample as contrasted with existing protein-protein-based analyses with correlational designs. Methods Our approach distinguishes each sample using differentially co-expressed interacting protein pairs (DEPs) based on co-expression scores of individual interacting pairs within a sample. The co-expression score has positive value if two genes in a sample are simultaneously up-regulated or down-regulated. And the score has higher absolute value if expression-changing ratios are similar between the two genes. We compared characteristics of DEPs with that of DEGs by evaluating their usefulness in separation of HIV-1 stage. And we identified DEP-based network-modules and their gene-ontology enrichment to find out the HIV-1 stage-specific gene signature. Results Based on the DEP approach, we observed clear separation among samples from distinct HIV-1 stages using clustering and principal component analyses. Moreover, the discrimination power of DEPs on the samples (70–100% accuracy) was much higher than that of DEGs (35–45%) using several well-known classifiers. DEP-based network analysis also revealed the HIV-1 stage-specific network modules; the main biological processes were related to “translation,” “RNA splicing,” “mRNA, RNA, and nucleic acid transport,” and “DNA metabolism.” Through the HIV-1 stage-related modules, changing stage-specific patterns of protein interactions could be observed. Conclusions DEP-based method discriminated the HIV-1 infection stages clearly, and revealed a HIV-1 stage-specific gene signature. The proposed DEP-based method might complement existing DEG-based approaches in various microarray expression analyses.
Collapse
Affiliation(s)
- Dukyong Yoon
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon 443-749, Korea
| | | | | | | | | |
Collapse
|
103
|
Abstract
Recent studies demonstrate that suppressive therapy can drive HIV-1 RNA levels to less than 50 copies mL(-1) in patient plasma. Yet, ultrasensitive assays show that most patients continue to harbour low-level persistent viremia. Treatment intensification studies indicate that low-level viremia could arise from several different sources. These sources include: (i) long-lived HIV-infected cells that replicate and produce virus; (ii) ongoing replication cycles in cells located in sanctuary sites where drug levels are suboptimal; and/or (iii) proliferation of latently infected cells with regeneration of a stable reservoir of slowly dividing infected cells. A well-defined latent reservoir of HIV is memory CD4+ T-cells where latency is established when an activated CD4+ T-cell becomes infected by HIV, but transitions to a terminally differentiated memory cell before it is eliminated. This review examines the dynamics and possible reservoirs of persistent HIV in patients on suppressive therapy, the mechanisms promoting viral latency and strategies to purge latent viral reservoirs. The promising research described here takes a number of steps forward to seriously address HIV remission and/or eradication.
Collapse
Affiliation(s)
- S Palmer
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden.
| | | | | |
Collapse
|
104
|
MacParland SA, Vali B, Ostrowski MA. Immunopathogenesis of HIV/hepatitis C virus coinfection. Future Virol 2011. [DOI: 10.2217/fvl.11.80] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As a result of shared infection routes, approximately 25% of individuals infected with HIV in North America are also infected with hepatitis C virus (HCV). In the setting of HIV coinfection, the course of HCV disease is more aggressive, resulting in higher HCV viral loads and a more rapid progression of liver pathology. With the success of HAART, HCV-related end-stage liver disease has become a leading cause of morbidity and mortality in HIV/HCV-coinfected patients. In this article, we will discuss recent studies examining the immune response during HIV and HCV coinfection, focusing on alterations or dysfunctions in virus-specific T-cell responses that may play a role in the immunopathogenesis of HIV/HCV coinfection. Summarizing the impact of HIV coinfection on HCV-specific T-cell immunity and highlighting some of the proposed mechanisms of T-cell dysfunction in HIV/HCV-coinfected individuals may uncover information that could lead to new treatment strategies for these patients experiencing accelerated liver disease and generally poorer outcomes than their HCV-monoinfected counterparts.
Collapse
Affiliation(s)
| | - Bahareh Vali
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Mario A Ostrowski
- Department of Immunology, University of Toronto, Toronto, ON, Canada; University of Toronto, Medical Sciences Building, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Clinical Sciences Division, University of Toronto, Toronto, ON, Canada
- Li Ka Shing Knowledge Institute at St Michael’s Hospital, Toronto, ON, Canada
| |
Collapse
|
105
|
Ibeh BO, Obidoa O, Nwuke C. Lipid Peroxidation Correlates with HIVmRNA in Serodiscordant Heterosexual HIVpartners of Nigerian Origin. Indian J Clin Biochem 2011; 26:249-56. [PMID: 22754188 PMCID: PMC3162947 DOI: 10.1007/s12291-011-0120-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 02/02/2011] [Indexed: 11/24/2022]
Abstract
We recruited 59 individuals of known HIV serostatus after informed consent however, 44 were serodiscordant heterosexual partners [serodiscordant seronegative (SSN group) and serodiscordant seropositive (SSP group)] while 15 were seronegative healthy individuals (SNH). In the case-control study we choose to determine Malondialdehyde (MDA) concentration as a marker of lipid peroxidation index (oxidative stress) spectrophotometrically and quantify HIV mRNA by Real Time-nucleic acid sequence based amplification assay (RT-NASBA). Here our result show for the first time a high concentration of lipid peroxidation product (MDA, 116.6%) with a significant (P < 0.05) increase in HIV serodiscordant seropositive subjects over their seronegative partners. However, Spearman rank correlation statistics of SSP group showed a positive correlation value (P < 0.01, r = 0.89) between MDA and mRNA and a negative correlation between MDA and T-cell ratio (P < 0.01, r = 0.96).The study may strongly indicate a possible lipid peroxidation product threshold for predicting HIV infection and progression in serodiscordant heterosexual partners.
Collapse
Affiliation(s)
- Bartholomew O. Ibeh
- Department of Biochemistry, University of Nigeria, Nsukka, Nigeria
- Department of Biochemistry, Michael Okpara University of Agriculture, Umudike, P.M.B. 7267, Umuahia, Abia State Nigeria
| | - Onyechi Obidoa
- Department of Biochemistry, University of Nigeria, Nsukka, Nigeria
| | - Chinedu Nwuke
- R & D Department, Shell Petroleum, PortHarcourt, Nigeria
| |
Collapse
|
106
|
Time to and Predictors of CD4+ T-Lymphocytes Recovery in HIV-Infected Children Initiating Highly Active Antiretroviral Therapy in Ghana. AIDS Res Treat 2011; 2011:896040. [PMID: 21738861 PMCID: PMC3124224 DOI: 10.1155/2011/896040] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 02/24/2011] [Accepted: 03/03/2011] [Indexed: 11/23/2022] Open
Abstract
Background. CD4+ T-lymphocyte monitoring is not routinely available in most resource-limited settings. We investigated predictors of time to CD4+ T-lymphocyte recovery in HIV-infected children on highly active antiretroviral (HAART) at Korle-Bu Teaching Hospital, Ghana. Methods. Time to CD4+ T-lymphocyte recovery was defined as achieving percent CD4+ T-lymphocytes of 25%. We used Cox proportional hazard models for identifying significant predictor variables. Results. Of the 233 children with complete CD4+ T-lymphocyte data, the mean age at HAART initiation was 5.5 (SD = 3.1) years. The median recovery time was 60 weeks (95% CL: 55–65). Evidence at baseline of severe suppression in CD4+ T-lymphocyte count adjusted for age, age at HAART initiation, gender, and having parents alive were statistically significant in predicting time to CD4+ T-lymphocyte recovery. Conclusions. A targeted approach based on predictors of CD4+ T-lymphocyte recovery can be a viable and cost-effective way of monitoring HAART in HIV-infected children in resource-limited settings.
Collapse
|
107
|
Marinda ET, Moulton LH, Humphrey JH, Hargrove JW, Ntozini R, Mutasa K, Levin J. In utero and intra-partum HIV-1 transmission and acute HIV-1 infection during pregnancy: using the BED capture enzyme-immunoassay as a surrogate marker for acute infection. Int J Epidemiol 2011; 40:945-54. [PMID: 21471020 DOI: 10.1093/ije/dyr055] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE The BED assay was developed to estimate the proportion of recent HIV infections in a population. We used the BED assay as a proxy for acute infection to quantify the associated risk of mother-to-child-transmission (MTCT) during pregnancy and delivery. Design A total of 3773 HIV-1 sero-positive women were tested within 96 h of delivery using the BED assay, and CD4 cell count measurements were taken. Mothers were classified according to their likelihood of having recently seroconverted. METHODS The risk of MTCT in utero and intra-partum was assessed comparing different groups defined by BED and CD4 cell count, adjusting for background factors using multinomial logistic models. RESULTS Compared with women with BED ≥ 0.8/CD4 ≥ 350 (typical of HIV-1 chronic patients) there was insufficient evidence to conclude that women presenting with BED < 0.8/CD4 ≥ 350 (typical of recent infections) were more likely to transmit in utero [adjusted odds ratio (aOR) = 1.37, 96% confidence interval (CI) 0.90-2.08, P = 0.14], whereas women with BED < 0.8/CD4 200-349 (possibly recently infected patients) had a 2.57 (95% CI 1.39-4.77, P-value < 0.01) odds of transmitting in utero. Women who had BED < 0.8/CD4 < 200 were most likely to transmit in utero (aOR 3.73, 95% CI 1.27-10.96, P = 0.02). BED and CD4 cell count were not predictive of intra-partum infections. CONCLUSIONS These data provide evidence that in utero transmission of HIV might be higher among women who seroconvert during pregnancy.
Collapse
|
108
|
Mahe E, Ross C, Sur M. Lymphoproliferative Lesions in the Setting of HIV Infection: A Five-Year Retrospective Case Series and Review. PATHOLOGY RESEARCH INTERNATIONAL 2011; 2011:618760. [PMID: 21559204 PMCID: PMC3090161 DOI: 10.4061/2011/618760] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 01/24/2011] [Accepted: 01/27/2011] [Indexed: 11/20/2022]
Abstract
A wide variety of noninfectious lesions have been identified in association with HIV infection. Many hematolymphoid lesions are possible in this patient group, both reactive and neoplastic. Epidemiologic data suggests that lymphoid malignancies are among the most common neoplasms in patients with HIV. We present a selective case series assembled over a 5-year period from the relatively low HIV-prevalence Hamilton Regional Laboratory Medicine Program (HRLMP), a tertiary care referral centre in Southern Ontario. This series serves to demonstrate the wide variety of lymphoid lesions that may be encountered in patients with HIV. In addition to outlining the pathologic work-up necessary in these cases, we discuss characteristics that distinguish the HIV-associated lesions from the pathobiologically similar non-HIV-associated lymphoid lesions.
Collapse
Affiliation(s)
- Etienne Mahe
- Division of Anatomical Pathology, Department of Pathology and Molecular Medicine, Juravinski Hospital and Cancer Centre, McMaster University, 711 Concession Street, Hamilton, ON, Canada L8V 1C
| | | | | |
Collapse
|
109
|
Elemans M, Thiébaut R, Kaur A, Asquith B. Quantification of the relative importance of CTL, B cell, NK cell, and target cell limitation in the control of primary SIV-infection. PLoS Comput Biol 2011; 7:e1001103. [PMID: 21408213 PMCID: PMC3048377 DOI: 10.1371/journal.pcbi.1001103] [Citation(s) in RCA: 19] [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: 06/10/2010] [Accepted: 01/28/2011] [Indexed: 01/22/2023] Open
Abstract
CD8+ cytotoxic T lymphocytes (CTLs), natural killer (NK) cells, B cells and target cell limitation have all been suggested to play a role in the control of SIV and HIV-1 infection. However, previous research typically studied each population in isolation leaving the magnitude, relative importance and in vivo relevance of each effect unclear. Here we quantify the relative importance of CTLs, NK cells, B cells and target cell limitation in controlling acute SIV infection in rhesus macaques. Using three different methods, we find that the availability of target cells and CD8+ T cells are important predictors of viral load dynamics. If CTL are assumed to mediate this anti-viral effect via a lytic mechanism then we estimate that CTL killing is responsible for approximately 40% of productively infected cell death, the remaining cell death being attributable to intrinsic, immune (CD8+ T cell, NK cell, B cell) -independent mechanisms. Furthermore, we find that NK cells have little impact on the death rate of infected CD4+ cells and that their net impact is to increase viral load. We hypothesize that NK cells play a detrimental role in SIV infection, possibly by increasing T cell activation.
Collapse
Affiliation(s)
- Marjet Elemans
- Department of Immunology, Imperial College London, London, United Kingdom.
| | | | | | | |
Collapse
|
110
|
Li L, Aiamkitsumrit B, Pirrone V, Nonnemacher MR, Wojno A, Passic S, Flaig K, Kilareski E, Blakey B, Ku J, Parikh N, Shah R, Martin-Garcia J, Moldover B, Servance L, Downie D, Lewis S, Jacobson JM, Kolson D, Wigdahl B. Development of co-selected single nucleotide polymorphisms in the viral promoter precedes the onset of human immunodeficiency virus type 1-associated neurocognitive impairment. J Neurovirol 2011; 17:92-109. [PMID: 21225391 PMCID: PMC3057211 DOI: 10.1007/s13365-010-0014-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 11/15/2010] [Accepted: 11/24/2010] [Indexed: 01/03/2023]
Abstract
The long terminal repeat (LTR) regulates gene expression of HIV-1 by interacting with multiple host and viral factors. Cross-sectional studies in the pre-HAART era demonstrated that single nucleotide polymorphisms (SNPs) in peripheral blood-derived LTRs (a C-to-T change at position 3 of C/EBP site I (3T) and at position 5 of Sp site III (5T)) increased in frequency as disease severity increased. Additionally, the 3T variant correlated with HIV-1-associated dementia. LTR sequences derived by longitudinal sampling of peripheral blood from a single patient in the DrexelMed HIV/AIDS Genetic Analysis Cohort resulted in the detection of the 3T and 5T co-selected SNPs before the onset of neurologic impairment, demonstrating that these SNPs may be useful in predicting HIV-associated neurological complications. The relative fitness of the LTRs containing the 3T and/or 5T co-selected SNPs as they evolve in their native patient-derived LTR backbone structure demonstrated a spectrum of basal and Tat-mediated transcriptional activities using the IIIB-derived Tat and colinear Tat derived from the same molecular clone containing the 3T/5T LTR SNP. In silico predictions utilizing colinear envelope sequence suggested that the patient's virus evolved from an X4 to an R5 swarm prior to the development of neurological complications and more advanced HIV disease. These results suggest that the HIV-1 genomic swarm may evolve during the course of disease in response to selective pressures that lead to changes in prevalence of specific polymorphisms in the LTR, env, and/or tat that could predict the onset of neurological disease and result in alterations in viral function.
Collapse
Affiliation(s)
- Luna Li
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA. Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Benjamas Aiamkitsumrit
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA. Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Vanessa Pirrone
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA. Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Michael R. Nonnemacher
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA. Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Adam Wojno
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA. Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Shendra Passic
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA. Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Katherine Flaig
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA. Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Evelyn Kilareski
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA. Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Brandon Blakey
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA. Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Jade Ku
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA. Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Nirzari Parikh
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA. Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Rushabh Shah
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA. Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Julio Martin-Garcia
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA. Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | | | - Laila Servance
- Division of Infectious Disease and HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - David Downie
- Division of Infectious Disease and HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Sharon Lewis
- Division of Infectious Disease and HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Jeffrey M. Jacobson
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA. Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA. Center for Clinical and Translational Medicine, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA. Division of Infectious Disease and HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Dennis Kolson
- Department of Neurology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA. Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA. Center for Clinical and Translational Medicine, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
111
|
Ragni MV, Moore CG, Soadwa K, Nalesnik MA, Zajko AB, Cortese-Hassett A, Whiteside TL, Hart S, Zeevi A, Li J, Shaikh OS, HHH Study Group. Impact of HIV on liver fibrosis in men with hepatitis C infection and haemophilia. Haemophilia 2011; 17:103-11. [PMID: 20722744 PMCID: PMC2990788 DOI: 10.1111/j.1365-2516.2010.02366.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Hepatitis C virus (HCV) is the major cause of liver disease in haemophilia. Few data exist on the proportion with liver fibrosis in this group after long-term HCV and HIV co-infection. We conducted a cross-sectional multi-centre study to determine the impact of HIV on the prevalence and risk factors for fibrosis in haemophilic men with chronic hepatitis C. Biopsies were independently scored by Ishak, Metavir and Knodell systems. Variables were tested for associations with fibrosis using logistic regression and receiver operating curves (ROC). Of 220 biopsied HCV(+) men, 23.6% had Metavir ≥ F3 fibrosis, with higher mean Metavir fibrosis scores among HIV/HCV co-infected than HCV mono-infected, 1.6 vs. 1.3 (P = 0.044). Variables significantly associated with fibrosis included AST, ALT, APRI score (AST/ULN × 100/platelet × 10(9) /L), alpha-fetoprotein (all P < 0.0001), platelets (P = 0.0003) and ferritin (P = 0.0008). In multiple logistic regression of serum markers, alpha-fetoprotein, APRI and ALT were significantly associated with ≥ F3 fibrosis [AUROC = 0.77 (95% CI 0.69, 0.86)]. Alpha-fetoprotein, APRI and ferritin were significant in HIV(-) [AUROC = 0.82 (95% CI 0.72, 0.92)], and alpha-fetoprotein and platelets in HIV(+) [AUROC = 0.77 (95% CI 0.65, 0.88]. In a multivariable model of demographic and clinical variables, transformed (natural logarithm) of alpha-fetoprotein (P = 0.0003), age (P = 0.006) and HCV treatment (P = 0.027) were significantly associated with fibrosis. Nearly one-fourth of haemophilic men have Metavir ≥ 3 fibrosis. The odds for developing fibrosis are increased in those with elevated alpha-fetoprotein, increasing age and past HCV treatment.
Collapse
Affiliation(s)
- M V Ragni
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh, Pittsburgh, PA 15213-4306, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
Collaborators
Ellis Neufeld, Joanna Hedstrom, Randi Stern, Joan Gill, Megan Gavin, Lisa Boggio, Sandy Harris, Doreen Brettler, Linnea Olson, Gilbert White, Alice Ma, Aime Grimsley, Keith Hoots, Kathryn Moynihan, Megan Ullman, Cynthia Rutherford, Patricia Dunnagan, Robert Bona, Ann Bartolomeo, Diana Beardsley, Judy C Sutton, Susan Marino, Alice Cohen, Ellen White, Peter Kouides, Laura Braggins, Chris Walsh, Johanna McCarthy, Zale Bernstein, Linda Belling, James Steinberg, Francie Lasseter, Philip Kuriakose, Angela Lambing, Roshni Kulkarni, Ajovi Scott Emuakpor, Laura Carlson, Carolyn Solomon, Ralph Gruppo, Ann P Green, Jeffrey Hord, Dawn Ali, Amy Shapiro, Karen Hieston, Renee Murry, Sarah May, Geoffrey Allen, Alexis Thompson, Nichele Willingham, Dena F Haddad, Leonard Valentino, Rosie Howard, Prasad Mathew, Marcia Schwartz, Hassan Yaish, Richard Lemons, Shirley Bleak, Jennifer Green, Diane Nugent, Marianne McDaniel, Michael Tarantino, Yvonne Lucas, Mary Brooks, Cindy Leissinger, Cecelia Schmidt, Joseph Palascak, Mary Galeano, Madeline Heffner, Mark Reding, Kerry Hansen, Joan Osip, Joachim Reimers, Judy Bagato, Patrick Fogarty, Patrycja Olszynski, Susan Karp, Donna DiMichele, Suchitra Acharya, Ilene Goldberg, Barbara Konkle, Karen Panckeri, Jerry Powell, Karen Scott, Marilyn Manco-Johnson, Sally Stabler, Carissa Smith, Brenda Riske,
Collapse
|
112
|
Montes M, Sanchez C, Lewis DE, Graviss EA, Seas C, Gotuzzo E, White AC. Normalization of FoxP3(+) regulatory T cells in response to effective antiretroviral therapy. J Infect Dis 2010; 203:496-9. [PMID: 21177309 DOI: 10.1093/infdis/jiq073] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Regulatory T cells (Tregs) blunt uncontrolled immune responses. In advanced human immunodeficiency virus (HIV) infection, the total number of Tregs is decreased, but the proportion of T cells with a regulatory phenotype is highly variable. We studied CD4(+)CD25(+)FoxP3(+) T cells from patients successfully treated with combination antiretroviral therapy (ART). The proportion of CD4(+)CD25(+)FoxP3(+) cells transiently increased and then decreased from a median of 13% at baseline to 5.1% at 48 weeks, similar to values in normal subjects. These data suggest that with effective therapy, the regulatory cell numbers normalize, and that the inflammatory signals driving their production may also abate.
Collapse
Affiliation(s)
- Martin Montes
- Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | | | | | | | | | | |
Collapse
|
113
|
Monitoring Antiretroviral Therapy in HIV-Infected Children in Resource-Limited Countries: A Tale of Two Epidemics. AIDS Res Treat 2010; 2011:280901. [PMID: 21490777 PMCID: PMC3066553 DOI: 10.1155/2011/280901] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Accepted: 10/30/2010] [Indexed: 11/30/2022] Open
Abstract
Twenty-nine years into the HIV epidemic, several advances have been made; however, there remain several challenges particularly with pediatric HIV in resource-limited countries. The obstacles facing pediatric antiretroviral therapy (ART) delivery in resource-limited countries are multifaceted: lack of health care infrastructure, limited availability of pediatric drug formulations, lack of early HIV diagnostic and monitoring techniques, limited manpower with expertise in pediatric HIV care, limited donor funding, and competing public health priorities with limited health care budget. In this paper, the challenges with various ART monitoring tools in resource-limited countries are discussed. Noninvasive (e.g., patient, clinical events outcome, and adherence) and invasive (e.g., immunologic and virologic) monitoring tools are discussed. Several cheap and technically less complex laboratory tests for monitoring are becoming available. Funding agencies and country programs should invest in validating the use of current technologies to optimize pediatric HIV care in resource-limited countries.
Collapse
|
114
|
Ohl M, Tate J, Duggal M, Skanderson M, Scotch M, Kaboli P, Vaughan-Sarrazin M, Justice A. Rural residence is associated with delayed care entry and increased mortality among veterans with human immunodeficiency virus infection. Med Care 2010; 48:1064-70. [PMID: 20966783 PMCID: PMC3138500 DOI: 10.1097/mlr.0b013e3181ef60c2] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
CONTEXT Rural persons with human immunodeficiency virus (HIV) face many barriers to care, but little is known about rural-urban variation in HIV outcomes. OBJECTIVE To determine the association between rural residence and HIV outcomes. DESIGN, SETTING, AND PATIENTS Retrospective cohort study of mortality among persons initiating HIV care in Veterans Administration (VA) during 1998-2006, with mortality follow-up through 2008. Rural residence was determined using Rural Urban Commuting Area codes. We identified 8489 persons initiating HIV care in VA with no evidence of combination antiretroviral therapy (cART) use at care entry, of whom 705 (8.3%) were rural. OUTCOME MEASURE All-cause mortality. RESULTS At care entry, rural persons were less likely than urban persons to have drug use problems (10.6% vs. 19.5%, P < 0.001) or hepatitis C (34.3% vs. 41.2%, P = 0.001), but had more advanced HIV infection (median CD4: 186 vs. 246, P < 0.001). By 2 years after care entry, 5874 persons had initiated cART (528 rural [74.9%] and 5346 urban [68.7%], P = 0.001), and there were 1022 deaths (108 rural [15.3%] and 914 urban [11.7%], P = 0.004). The mortality hazard ratio for rural persons compared with urban was 1.34 (95% confidence interval: 1.05-1.69). The hazard ratio decreased to 1.18 (95% confidence interval: 0.93-1.50) after adjustment for HIV severity (CD4 and AIDS-defining illnesses) at care entry, and was 1.17 (95% confidence interval: 0.92-1.50) in a model adjusting for age, HIV severity at care entry, substance use, hepatitis B or C diagnoses, and cART initiation. CONCLUSIONS Later entry into care drives increased mortality for rural compared with urban veterans with HIV. Future studies should explore the person, care system, and community-level determinants of late care entry for rural persons with HIV.
Collapse
Affiliation(s)
- Michael Ohl
- VA Office of Rural Health (ORH), Midwest Rural Health Resource Center, Iowa City VAMC, Iowa City, IA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
115
|
Dou Z, Chen RY, Wang Z, Ji G, Peng G, Qiao X, Fu J, Meng X, Bulterys M, Ma Y, Zhao Y, Wang N, Zhang F. HIV-infected former plasma donors in rural Central China: from infection to survival outcomes, 1985-2008. PLoS One 2010; 5:e13737. [PMID: 21060835 PMCID: PMC2966407 DOI: 10.1371/journal.pone.0013737] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 09/24/2010] [Indexed: 11/19/2022] Open
Abstract
Background The HIV epidemic among former plasma donors (FPDs) in rural Central China in the early-mid 1990s is likely the largest known HIV-infected cohort in the world related to commercial plasma donation but has never been fully described. The objectives of this study are to estimate the timing and geographic spread of HIV infection in this cohort and to demonstrate the impact of antiretroviral therapy on survival outcomes. Methodology/Principal Findings HIV-infected FPDs were identified using the national HIV epidemiology and treatment databases. Locations of subjects were mapped. Dates of infection and survival were estimated using the midpoint date between initial-final plasma donation dates from 1985–2008 among those with plasma donation windows ≤2 years. Among 37084 FPDs in the two databases, 36110 were included. 95% were located in focal areas of Henan Province and adjacent areas of surrounding provinces. Midpoint year between initial-final plasma donation dates was 1994 among FPDs with known donation dates. Median survival from infection to AIDS was 11.8 years and, among those not treated, 1.6 years from AIDS to death. Among those on treatment, 71% were still alive after five years. Using Cox proportional hazard modeling, untreated AIDS patients were 4.9 times (95% confidence interval 4.6–5.2) more likely to die than those on treatment. Conclusions/Significance The epidemic of HIV-infected FPD in China was not widespread throughout China but rather was centered in Henan Province and the adjacent areas of surrounding provinces. Even in these areas, infections were concentrated in focal locations. Overall, HIV infections in this cohort peaked in 1994, with median survival of 13.4 years from infection to death among those not treated. Among AIDS patients on treatment, 71% were still alive after five years.
Collapse
Affiliation(s)
- Zhihui Dou
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ray Y. Chen
- National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Beijing, China
| | - Zhe Wang
- Henan Center for Disease Control and Prevention, Zhengzhou, China
| | - Guoping Ji
- Anhui Center for Disease Control and Prevention, Hefei, China
| | - Guoping Peng
- Hubei Center for Disease Control and Prevention, Wuhan, China
| | - Xiaochun Qiao
- Shanxi Center for Disease Control and Prevention, Taiyuan, China
| | - Jihua Fu
- Shandong Center for Disease Control and Prevention, Jinan, China
| | - Xiangdong Meng
- Jilin Center for Disease Control and Prevention, Changchun, China
| | - Marc Bulterys
- Global AIDS Program, U.S. Centers for Disease Control and Prevention, Beijing, China
| | - Ye Ma
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yan Zhao
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ning Wang
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- * E-mail: (FZ); (NW)
| | - Fujie Zhang
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Beijing Ditan Hospital, Beijing, China
- * E-mail: (FZ); (NW)
| |
Collapse
|
116
|
Guilford T, Morris D, Gray D, Venketaraman V. Atherosclerosis: pathogenesis and increased occurrence in individuals with HIV and Mycobacterium tuberculosis infection. HIV AIDS-RESEARCH AND PALLIATIVE CARE 2010; 2:211-8. [PMID: 22096400 PMCID: PMC3218695 DOI: 10.2147/hiv.s11977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Atherosclerosis is a leading cause of coronary heart disease and stroke. Since 1981, more than 980,000 cases of AIDS have been reported in the United States. According to the Centers for Disease Control, more than 1 million Americans may be infected with HIV. By killing or damaging CD4+ T cells of the body’s immune system, HIV progressively destroys the body’s ability to fight infections. People diagnosed with AIDS often suffer from life-threatening diseases caused by opportunistic infections such as tuberculosis. HIV-infected individuals have increased risks for atherosclerosis. This review summarizes the effects of oxidized low density lipoproteins in impairing macrophage functions in individuals with atherosclerosis (with and without HIV infection) thereby enhancing the susceptibility to Mycobacterium tuberculosis infection.
Collapse
|
117
|
An integrated modelling approach for R5–X4 mutation and HAART therapy assessment. SWARM INTELLIGENCE 2010. [DOI: 10.1007/s11721-010-0046-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
118
|
Plasma Epstein-Barr viral load measurement as a diagnostic marker of lymphoma in HIV-infected patients. Med Clin (Barc) 2010; 135:485-90. [DOI: 10.1016/j.medcli.2010.02.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Revised: 02/05/2010] [Accepted: 02/09/2010] [Indexed: 11/20/2022]
|
119
|
Serpa J, Haque D, Valayam J, Breaux K, Rodriguez-Barradas MC. Effect of combination antiretroviral treatment on total protein and calculated globulin levels among HIV-infected patients. Int J Infect Dis 2010; 14 Suppl 3:e41-4. [DOI: 10.1016/j.ijid.2009.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 10/16/2009] [Accepted: 10/24/2009] [Indexed: 12/01/2022] Open
|
120
|
Pettersen FO, Taskén K, Kvale D. Combined Env- and Gag-specific T cell responses in relation to programmed death-1 receptor and CD4 T cell loss rates in human immunodeficiency virus-1 infection. Clin Exp Immunol 2010; 161:315-23. [PMID: 20491784 DOI: 10.1111/j.1365-2249.2010.04179.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Additional progression markers for human immunodeficiency virus (HIV) infection are warranted. In this study we related antigen-specific responses in CD4(+) and CD8(+) T cells to CD38, reflecting chronic immune activation, and to CD4(+) T cell loss rates. Clones transiently expressing CD107a (CD8(+)) or CD154 (CD4(+)) in response to Gag, Env and Nef overlapping peptide pools were identified, along with their expression of the inhibitory programmed death-1 receptor (PD-1) in fresh peripheral blood mononuclear cells (PBMC) from 31 patients off antiretroviral treatment (ART). HIV-specific CD8(+) T cell responses dominated over CD4(+) T cell responses, and among CD8(+) responses, Gag and Nef responses were higher than Env-responses (P < 0.01). PD-1 on CD8(+) HIV-specific subsets was higher than CMV-specific CD8(+) cells (P < 0.01), whereas PD-1 on HIV-specific CD4(+) cells was similar to PD-1 on CMV-specific CD4(+) cells. Gag and Env CD8(+) responses correlated oppositely to the CD4 loss rate. Env/Gag CD8(+) response ratios, independently of PD-1 levels, correlated more strongly to CD4 change rates (r = -0.50 to -0.77, P < 0.01) than the total number of Gag-specific CD8(+) cells (r = 0.44-0.85, P < or = 0.02). The Env/Gag ratio performed better than CD38 and HIV-RNA in logistic regression analysis predicting CD4 change rate as a measure of progression. In conclusion, HIV-specific CD8(+)CD107a(+) Env/Gag response ratio was a stronger predictor for progression than CD38 and HIV-RNA. The Env/Gag ratio may reflect the balance between possibly beneficial (Gag) and detrimental (Env) CD8(+) T cell responses and should be explored further as a progression marker.
Collapse
Affiliation(s)
- F O Pettersen
- Ullevål Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | | | | |
Collapse
|
121
|
Hapgood JP, Tomasicchio M. Modulation of HIV-1 virulence via the host glucocorticoid receptor: towards further understanding the molecular mechanisms of HIV-1 pathogenesis. Arch Virol 2010; 155:1009-19. [PMID: 20446002 DOI: 10.1007/s00705-010-0678-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 03/26/2010] [Indexed: 10/19/2022]
Abstract
The glucocorticoid receptor (GR) is a steroid receptor that regulates diverse functions, which include the immune response. In humans, the GR acts via binding to cortisol, resulting in the transcriptional modulation of key host genes. Several lines of evidence suggest that the host GR could be a key protein exploited by HIV at multiple levels to ensure its pathogenic success. Endogenous and therapeutic glucocorticoids play important roles in patients with HIV due to their well-established effects on immune function. AIDS patients develop glucocorticoid hypersensitivity, consistent with a mechanism involving an HIV-1-induced increase in expression or activity of the GR. Both the HIV-1 accessory protein Vpr and the host GR affect transcription of viral proteins from the long terminal repeat (LTR) region of the HIV-1 promoter. In addition, Vpr modulates host GR function to affect transcription of host genes, most likely via direct interaction with the GR. Vpr appears to regulate GR function by acting as a co-activator for the GR. Since both the GR and Vpr are involved in apoptosis in T cells and dendritic cells, crosstalk between these proteins may also regulate apoptosis in these and other cells. Given that cortisol is not the only ligand that activates the GR, other endogenous as well as synthetic GR ligands such as progestins may also modulate HIV pathogenesis, in particular in the cervicovaginal environment. Investigating the molecular determinants, ligand-selectivity and role in HIV pathogenesis of the GR-Vpr interaction may lead to new strategies for development of anti-HIV drugs.
Collapse
Affiliation(s)
- Janet Patricia Hapgood
- Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa.
| | | |
Collapse
|
122
|
Fujita H, Nishikori M, Takaori-Kondo A, Yoshinaga N, Ohara Y, Ishikawa T, Haga H, Uchiyama T. A case of HIV-associated lymphoproliferative disease that was successfully treated with highly active antiretroviral therapy. Int J Hematol 2010; 91:692-8. [PMID: 20217283 DOI: 10.1007/s12185-010-0542-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2009] [Revised: 02/09/2010] [Accepted: 02/14/2010] [Indexed: 12/29/2022]
Abstract
We report a case of a 41-year-old male with human immunodeficiency virus (HIV)-associated lymphoproliferative disease (LPD) who was successfully treated with highly active antiretroviral therapy (HAART). He presented with epigastralgia, and an upper endoscopic examination revealed submucosal tumors and ulcerations in his stomach. Histopathologic examination of a biopsy specimen resulted in a diagnosis of diffuse large B-cell lymphoma. He also showed systemic lymphadenopathy; whereas, a concurrent inguinal lymph node biopsy produced a diagnosis of follicular hyperplasia. He was treated with CHOP chemotherapy but the response was poor. He demonstrated several immunological abnormalities, such as eosinophilia and bone marrow insufficiency, and was suspected to be in an immunocompromised state. He was examined for HIV infection and turned out to be positive. The gastric and inguinal lymph node specimens were re-evaluated and diagnoses of HIV-LPD and HIV lymphadenitis were made, respectively. He was treated with HAART and achieved complete remission and has remained tumor-free for 20 months. To the best of our knowledge, there is no previous report in which HIV-LPD was successfully treated with antiretroviral therapy alone. It is assumed that HAART resulted in the restoration of anti-tumor immunity in this case, which led to the eradication of LPD cells.
Collapse
Affiliation(s)
- Haruyuki Fujita
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
123
|
Wasserstein-Robbins F. A mathematical model of HIV infection: Simulating T4, T8, macrophages, antibody, and virus via specific anti-HIV response in the presence of adaptation and tropism. Bull Math Biol 2010; 72:1208-53. [PMID: 20151219 DOI: 10.1007/s11538-009-9488-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Accepted: 11/12/2009] [Indexed: 12/11/2022]
Abstract
A mathematical model of the host's immune response to HIV infection is proposed. The model represents the dynamics of 13 subsets of T cells (HIV-specific and nonspecific, healthy and infected, T4 and T8 cells), infected macrophages, neutralizing antibodies, and virus. The results of simulation are in agreement with published data regarding T4 cell concentration and viral load, and exhibit the typical features of HIV infection, i.e. double viral peaks in the acute stage, sero conversion, inverted T cell ratio, establishment of set points, steady state, and decline into AIDS. This result is achieved by taking into account thymic aging, viral and infected cell stimulation of specific immune cells, background nonspecific antigens, infected cell proliferation, viral production by infected macrophages and T cells, tropism, viral, and immune adaptation. Starting from this paradigm, changes in the parameter values simulate observed differences in individual outcomes, and predict different scenarios, which can suggest new directions in therapy. In particular, large parameter changes highlight the potentially critical role of both very vigorous and extremely damped specific immune response, and of the elimination of virus release by macrophages. Finally, the time courses of virus, antibody and T cells production and removal are systematically investigated, and a comparison of T4 and T8 cell dynamics in a healthy and in a HIV infected host is offered.
Collapse
|
124
|
Tzitzivacos D, Tiemessen C, Stevens W, Papathanasopoulos M. Viral genetic determinants of nonprogressive HIV type 1 subtype C infection in antiretroviral drug-naive children. AIDS Res Hum Retroviruses 2009; 25:1141-8. [PMID: 19895210 DOI: 10.1089/aid.2009.0080] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Characterization of HIV-1 from slow progressors is important to facilitate vaccine and antiviral drug development. To identify virus attenuations that may contribute to slower rates of disease progression, the full-length viral genomes from primary isolates of six slow progressing HIV-positive children were sequenced. Proviral DNA was extracted from cocultured peripheral blood mononuclear cells and used to PCR amplify, sequence, and extensively analyze the near full-length genomes and LTR regions. All primary HIV-1 isolates were HIV-1 subtype C throughout their genome, and amino acid (AA) sequence analysis revealed open reading frames for all genes. However, all isolates had at least one unusual gene/protein. For example, isolate LT5 had a 2AA insertion in the Vpr mitochondriotoxic domain. Isolate LT21 contained an additional 5AA in the C-terminus of tat exon 2, while integrase in isolate LT39 had an additional 4AA at the C-terminus. Rev from isolates LT45 and LT46 did not have the characteristic subtype C 16AA truncation, and in addition, had a further 3AA. Furthermore, altered functional domains were noted in several isolates, such as the cAMP-dependent kinase PKA phosphorylation site in Nef (LT5), a Vpr mutation involved in decreased proapoptotic activity (all isolates), and the Nef ExxxLL motif involved in the interaction with AP-1 and AP-2 (LT46). The slower HIV-1 disease progression in these six children may be attributed to altered protein functions. For example, LT46 Nef is unable to bind AP-1 and AP-2 and therefore is inactive on CD4 endocytosis. The biological relevance of these findings requires further investigation.
Collapse
Affiliation(s)
- D.B. Tzitzivacos
- HIV Pathogenesis Research Laboratory, Department of Molecular Medicine and Haematology, University of the Witwatersrand Medical School, Parktown, Johannesburg 2193, South Africa
| | - C.T. Tiemessen
- Cell Biology, AIDS Virus Research Unit, National Institute for Communicable Diseases, Johannesburg 2131, South Africa
| | - W.S. Stevens
- HIV Pathogenesis Research Laboratory, Department of Molecular Medicine and Haematology, University of the Witwatersrand Medical School, Parktown, Johannesburg 2193, South Africa
| | - M.A. Papathanasopoulos
- HIV Pathogenesis Research Laboratory, Department of Molecular Medicine and Haematology, University of the Witwatersrand Medical School, Parktown, Johannesburg 2193, South Africa
| |
Collapse
|
125
|
Development and assessment of a multiplex real-time PCR assay for quantification of human immunodeficiency virus type 1 DNA. J Clin Microbiol 2009; 47:2194-9. [PMID: 19420173 DOI: 10.1128/jcm.01264-08] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previous studies showed that high levels of human immunodeficiency virus type 1 (HIV-1) DNA are associated with a faster progression to AIDS, an increased risk of death, and a higher risk of HIV RNA rebound in patients on highly active antiretroviral therapy. Our objective was to develop and assess a highly sensitive real-time multiplex PCR assay for the quantification of HIV-1 DNA (RTMP-HIV) based on molecular beacons. HIV-1 DNA quantification was carried out by RTMP in a LightCycler 2.0 apparatus. HIV-1 DNA was quantified in parallel with CCR5 as a reference gene, and reported values are numbers of HIV-1 DNA copies/10(6) peripheral blood mononuclear cells (PBMCs). The clinical sensitivity of the assay was assessed for 115 newly diagnosed HIV-1-infected individuals. The analytical sensitivity was estimated to be 12.5 copies of HIV-1 DNA per 10(6) PBMCs, while the clinical sensitivity was 100%, with levels ranging from 1.23 to 4.25 log(10) HIV-1 DNA copies/10(6) PBMCs. In conclusion, we developed and assessed a new RTMP-HIV assay based on molecular beacons, using a LightCycler 2.0 instrument. This multiplex assay has comparable sensitivity, reproducibility, and accuracy to single real-time PCR assays.
Collapse
|
126
|
Vázquez-Guillén JM, García-Jacobo PJ, Zapata-Benavides P, Rosas-Taraco AG, Ordaz-Sánchez MI, López-Guillén P, Trejo-Avila L, Alcocer-González JM, Rodríguez-Padilla C, Rivera-Morales LG. Expression of DC-SIGN in peripheral blood dendritic cells of patients with typical, slow, and rapid progression to AIDS. Arch Med Res 2009; 40:132-5. [PMID: 19237024 DOI: 10.1016/j.arcmed.2008.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2008] [Accepted: 11/14/2008] [Indexed: 11/25/2022]
Abstract
The main access route for human immunodeficiency virus (HIV) into the lymph nodes is through the mucosa. Once there, dendritic cells (DCs) are the first cells to interact with the virus. Then, DCs can uptake and transport to the lymph nodes, beginning a disseminated infection. Interaction between the virus and DCs is mediated by the receptor DC-SIGN. This study seeks to determine any relationship between HIV-AIDS immunopathology and DC-SIGN expression levels in DCs from typical, rapid, and slow progressors. A DC separation system was implemented using peripheral blood mononuclear cells from infected subjects. The study included 27 patients classified as typical, rapid, and slow progressors according to their clinical and epidemiological files. Finally, quantification of DC-SIGN was achieved by real-time PCR and by applying the Relative Quantification Scheme (DeltaDeltaCt). We isolated DCs from peripheral blood of 27 HIV-infected patients. Nineteen were considered as typical progressors, five as slow progressors, and three as rapid progressors. No significant differences were observed on the expression levels of DC-SIGN among the three groups of patients. Even if there are differences in expression levels among the analyzed patients, we did not find any significant differences in DC-SIGN expression among the three included groups. We therefore cannot conclude that the expression level of the receptor is related with the progression to AIDS.
Collapse
|
127
|
Kondo M, Sudo K, Tanaka R, Sano T, Sagara H, Iwamuro S, Takebe Y, Imai M, Kato S. Quantitation of HIV-1 group M proviral DNA using TaqMan MGB real-time PCR. J Virol Methods 2009; 157:141-6. [PMID: 19126416 DOI: 10.1016/j.jviromet.2008.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Revised: 12/05/2008] [Accepted: 12/09/2008] [Indexed: 10/21/2022]
Abstract
The level of human immunodeficiency virus type 1 (HIV-1) proviral DNA is likely to be an important marker of the long-term effectiveness of highly active antiretroviral therapy. A new method was developed for quantifying HIV-1 group M proviral DNA using TaqMan real-time PCR, in which degenerate primers and an MGB probe were used to resolve the difference in amplification efficiencies among different subtypes. The present assay provided good linearity and accuracy in the range of 4-5000 copies of proviral DNA in 0.5microg of cellular DNA. The intra-assay and inter-assay coefficients were <31.6% and <30.1%, respectively. In 19 HIV-1 clinical isolates of six subtypes (A, B, C, CRF01_AE, F, and G), quantitation values by the real-time PCR assay matched closely those by Poisson distribution analysis of PCR results at endpoint dilution (R(2)=0.988). This assay is characterized by the use of degenerate primers and having been validated by comparing with a Poisson distribution-based assay. The present real-time PCR assay is highly sensitive, linear, reproducible, accurate, and independent of group M subtypes. The assay will be useful for studying the relationship between HIV-1 proviral loads and the long-term efficacy of antiretroviral therapy for subtype B as well as non-B subtype strains.
Collapse
Affiliation(s)
- Makiko Kondo
- Kanagawa Prefectural Institute of Public Health, Shimomachiya, Chigasaki, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Gurunathan S, Habib RE, Baglyos L, Meric C, Plotkin S, Dodet B, Corey L, Tartaglia J. Use of predictive markers of HIV disease progression in vaccine trials. Vaccine 2009; 27:1997-2015. [DOI: 10.1016/j.vaccine.2009.01.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 12/19/2008] [Accepted: 01/08/2009] [Indexed: 10/21/2022]
|
129
|
|
130
|
Burkhead EG, Hawkins JM, Molinek DK. A Dynamical Study of a Cellular Automata Model of the Spread of HIV in a Lymph Node. Bull Math Biol 2008; 71:25-74. [DOI: 10.1007/s11538-008-9351-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Accepted: 08/01/2008] [Indexed: 10/21/2022]
|
131
|
Chenine AL, Shai-Kobiler E, Steele LN, Ong H, Augostini P, Song R, Lee SJ, Autissier P, Ruprecht RM, Secor WE. Acute Schistosoma mansoni infection increases susceptibility to systemic SHIV clade C infection in rhesus macaques after mucosal virus exposure. PLoS Negl Trop Dis 2008; 2:e265. [PMID: 18648516 PMCID: PMC2447882 DOI: 10.1371/journal.pntd.0000265] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2007] [Accepted: 06/24/2008] [Indexed: 11/22/2022] Open
Abstract
Background Individuals living in sub-Saharan Africa represent 10% of the world's population but almost 2/3 of all HIV-1/AIDS cases. The disproportionate HIV-1 infection rates in this region may be linked to helminthic parasite infections that affect many individuals in the developing world. However, the hypothesis that parasite infection increases an individual's susceptibility to HIV-1 has never been prospectively tested in a relevant in vivo model. Methodology/Principal Findings We measured whether pre-existing infection of rhesus monkeys with a parasitic worm would facilitate systemic infection after mucosal AIDS virus exposure. Two groups of animals, one consisting of normal monkeys and the other harboring Schistosoma mansoni, were challenged intrarectally with decreasing doses of R5-tropic clade C simian-human immunodeficiency virus (SHIV-C). Systemic infection occurred in parasitized monkeys at viral doses that remained sub-infectious in normal hosts. In fact, the 50% animal infectious (AID50) SHIV-C dose was 17-fold lower in parasitized animals compared to controls (P<0.001). Coinfected animals also had significantly higher peak viral RNA loads than controls (P<0.001), as well as increased viral replication in CD4+ central memory cells (P = 0.03). Conclusions/Significance Our data provide the first direct evidence that acute schistosomiasis significantly increases the risk of de novo AIDS virus acquisition, and the magnitude of the effect suggests that control of helminth infections may be a useful public health intervention to help decrease the spread of HIV-1. To test the hypothesis that infection with helmiths may increase host susceptibility to infection with HIV-1, we quantified the amount of a clade C simian-human immunodeficiency virus needed to infect rhesus macaques that had acute Schistosoma mansoni infections. Compared to control animals exposed to virus alone, monkeys with schistosomiasis required exposure to 17-fold lower levels of virus to become infected. The schistosome-infected monkeys also had significantly higher levels of initial virus replication and loss of a certain subset of memory T cells, both predictors of a more rapid progression to immune dysfunction. These results suggest that worm infections may increase the risk of becoming infected with HIV-1 among individuals with viral exposures. Furthermore, they support the idea that control programs for schistosomiasis and perhaps other parasitic worm infections may also be useful in helping to reduce the spread of HIV/AIDS in developing countries where helminths are endemic.
Collapse
Affiliation(s)
- Agnès-Laurence Chenine
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ela Shai-Kobiler
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lisa N. Steele
- Division of Parasitic Diseases, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Helena Ong
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Peter Augostini
- Division of Parasitic Diseases, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Ruijiang Song
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sandra J. Lee
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Patrick Autissier
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Ruth M. Ruprecht
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (RMR); (WES)
| | - W. Evan Secor
- Division of Parasitic Diseases, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail: (RMR); (WES)
| |
Collapse
|
132
|
Paintsil E, Ghebremichael M, Romano S, Andiman WA. Absolute CD4+ T-lymphocyte count as a surrogate marker of pediatric human immunodeficiency virus disease progression. Pediatr Infect Dis J 2008; 27:629-35. [PMID: 18520446 PMCID: PMC2924626 DOI: 10.1097/inf.0b013e3181693892] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Traditionally in pediatric HIV, the CD4+ T-lymphocyte percent is used to monitor disease progression because of the variability in absolute CD4+ T-lymphocyte numbers. Because of the high cost of equipment, sophisticated and delicate technology, most laboratories in resource-limited settings use simple protocols that enumerate only the absolute CD4+ T-lymphocyte counts. We assessed the use of absolute CD4+ T-lymphocyte count as a surrogate marker of pediatric HIV disease progression. METHODS We analyzed the CD4+ T-lymphocytes and HIV viral load over a 10-year period (1996-2006) of 97 HIV-infected children enrolled in the Yale Prospective Longitudinal Pediatric HIV Cohort using generalized linear mixed models. Both CD4+ T-lymphocytes and HIV viral load were assessed at baseline and every 2-3 months. The modeling approach used in this study allows the intercept and the rate at which outcome variables change over time to vary across participants. RESULTS We determined that absolute CD4+ T-lymphocytes count was just as reliable at monitoring pediatric HIV as CD4+ T-lymphocyte percentage. Antiretroviral treatment, regardless of the regimen used, was associated with higher CD+ T-lymphocytes count (P < 0.01). Race was significantly associated with CD4+ T-lymphocytes counts (with lower values for blacks compared with nonblacks; P < 0.01). The presence of other infections was associated with lower CD4+ T-lymphocyte count (P = 0.01) and higher viral load (P < 0.01), respectively. CONCLUSIONS In situations where determination of CD4+ T-lymphocyte percentages is not readily available, the absolute count may provide an affordable and accessible laboratory surrogate marker of HIV disease progression in children.
Collapse
Affiliation(s)
- Elijah Paintsil
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | | | | | |
Collapse
|
133
|
NANDWANI R, GAZZARD B, BARTON S, HAWKINS DA, ZEMELMAN V, STAUGHTON R. Does HIV disease progression influence epidermal Langerhans cell density? Br J Dermatol 2008. [DOI: 10.1046/j.1365-2133.1996.d01-907.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
134
|
D'Cruz OJ, Uckun FM. Evaluation of local tolerance of the antiretroviral spermicide (WHI-07)-loaded gel-microemulsion in the porcine female reproductive tract. J Appl Toxicol 2008; 28:303-14. [PMID: 17624922 DOI: 10.1002/jat.1280] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The local tolerance of the antiretroviral spermicide, WHI-07 (5-bromo-6-methoxy-5,6-dihydro-3'-azidothymidine-5'-(p-bromophenyl)-methoxyalaninyl phosphate)-loaded gel-microemulsion was evaluated in a physiologically relevant and sensitive porcine model. Gilts (Duroc) in nonestrus stages of the reproductive cycle received either a single or a daily intravaginal application of 2.0% WHI-07 via a gel-microemulsion for 6 days. Cervicovaginal lavage (CVL) fluid was obtained for up to 72 h after a single exposure and the cellular profile and levels of inflammatory cytokines (IL-1beta, IL-8, IFN-gamma and TNF-alpha) were quantitated by flow cytometry and chemiluminescence-based multiplex immunoassay, respectively. The reproductive tract (vagina, cervix, uteri and Fallopian tubes) harvested on day 7 was scored histologically for evidence of mucosal irritation using a new scoring criterion for ten histological endpoints that reflect pathological changes in the epithelial/ subepithelial and vascular/perivascular compartments. When compared with irritant reactions caused by the detergent-type spermicide, benzalkonium chloride (BZK), the scatter profile of CVL immune cells and basal levels of proinflammatory cytokines (IL-1beta, IL-8, IFN-gamma and TNF-alpha) in CVL fluid were unaffected by intravaginal exposure to 2% WHI-07. Unlike BZK, endpoint histology of the proximal and distal regions of the reproductive tract from gilts treated with 2.0% WHI-07 via gel-microemulsion for 6 days did not result in mucosal irritation or alteration in the epithelium, subepithelium/lamina propria, vessels/perivascular tissues and underlying/surrounding muscles. Based on surrogate markers for inflammation, leukocyte profile and histologic data for local tolerance, repeated intravaginal administration of WHI-07 via gel-microemulsion as a prophylactic contraceptive is unlikely to cause vaginal irritation.
Collapse
Affiliation(s)
- Osmond J D'Cruz
- Paradigm Pharmaceuticals, LLC, St Paul, Minnesota 55113, USA.
| | | |
Collapse
|
135
|
Estes JD, Haase AT, Schacker TW. The role of collagen deposition in depleting CD4+ T cells and limiting reconstitution in HIV-1 and SIV infections through damage to the secondary lymphoid organ niche. Semin Immunol 2008; 20:181-6. [PMID: 18595731 PMCID: PMC2650387 DOI: 10.1016/j.smim.2008.04.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Accepted: 04/16/2008] [Indexed: 10/21/2022]
Abstract
The hallmark of HIV/SIV infections is the progressive depletion of CD4+ T cells that ultimately renders the host incapable of defending against AIDS defining opportunistic infections and malignancies. Although many potential mechanisms have been proposed to explain CD4+ T cell loss, we review here the growing evidence that fibrotic 'scarring' and consequent damage to the lymphatic tissue niche contributes to CD4+ T cell decline and limits CD4+ T cell re-population with retroviral therapy.
Collapse
Affiliation(s)
- Jacob D Estes
- AIDS and Cancer Virus Program, Science Applications International Corporation-Frederick, Inc., National Cancer Institute, Frederick, MD, USA
| | | | | |
Collapse
|
136
|
Mitchell CM, Balkus J, Agnew KJ, Cohn S, Luque A, Lawler R, Coombs RW, Hitti JE. Bacterial vaginosis, not HIV, is primarily responsible for increased vaginal concentrations of proinflammatory cytokines. AIDS Res Hum Retroviruses 2008; 24:667-71. [PMID: 18462081 DOI: 10.1089/aid.2007.0268] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The relative effect of HIV-1 infection compared with vaginal infections on vaginal cytokine concentrations is not well characterized. We compared vaginal fluid samples from HIV-1-infected women with those from HIV-negative women, to assess the effect of HIV-1 infection on concentrations of vaginal proinflammatory cytokines and the mucosal defense molecule secretory leukocyte protease inhibitor (SLPI). Twenty-seven HIV-1-infected women and 54 HIV-negative controls, matched for bacterial vaginosis (BV) status, had proinflammatory cytokine [interleukin (IL)-1beta, IL-6, IL-8] and SLPI concentrations measured from archived cervicovaginal lavage and vaginal swab samples using an enzyme-linked immunosorbent assay (ELISA). Log-transformed concentrations were compared by BV and HIV status in univariate analysis using Student's t-test, and in multivariate analysis using a linear regression model. In univariate analysis there were no significant differences in cytokine concentrations among HIV-1-infected and HIV-negative women. In a multivariable linear regression model, BV was significantly associated with an increase in IL-1 beta (p = 0.003). HIV infection was associated with an increased concentration of SLPI (p = 0.008), while BV status was significantly associated with a decrease in SLPI concentrations (p = 0.005). Neither HIV nor BV was associated with changes in IL-6 or IL-8. HIV does not have a major impact on vaginal concentrations of proinflammatory cytokines when controlling for the presence of bacterial vaginosis.
Collapse
Affiliation(s)
- Caroline M. Mitchell
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington 98195
| | - Jennifer Balkus
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington 98195
| | - Kathy J. Agnew
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington 98195
| | - Susan Cohn
- Department of Medicine, University of Rochester, Rochester, New York 14642
| | - Amneris Luque
- Department of Medicine, University of Rochester, Rochester, New York 14642
| | - Richard Lawler
- Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Robert W. Coombs
- Department of Medicine, University of Washington, Seattle, Washington 98195
| | - Jane E. Hitti
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington 98195
| |
Collapse
|
137
|
|
138
|
Park KH, Yu HS, Jung SI, Shin DH, Shin JH. Acute human immunodeficiency virus syndrome presenting with hemophagocytic lymphohistiocytosis. Yonsei Med J 2008; 49:341-2; author reply 342-3. [PMID: 18452272 PMCID: PMC2615318 DOI: 10.3349/ymj.2008.49.2.325] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Hemophagocytic lymphohistiocytosis (HLH) has been described in patients with advanced stages of human immunodeficiency virus (HIV) infection, but rarely occurs during the seroconversion stage of acute HIV infection. We report a case of acute HIV syndrome that presented with virus-associated HLH. The patient recovered spontaneously without any immunomodulating therapy. This case suggests that acute HIV infection should be included in the differential diagnosis of HLH and indicates that HLH associated with acute HIV infection can have a favorable outcome.
Collapse
Affiliation(s)
- Kyung-Hwa Park
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Ho-Sung Yu
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Sook-In Jung
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Dong-Hyeon Shin
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Jong-Hee Shin
- Department of Laboratory Medicine, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
139
|
Dhillon NK, Williams R, Peng F, Tsai YJ, Dhillon S, Nicolay B, Gadgil M, Kumar A, Buch SJ. Cocaine-mediated enhancement of virus replication in macrophages: implications for human immunodeficiency virus-associated dementia. J Neurovirol 2008; 13:483-95. [PMID: 18097880 DOI: 10.1080/13550280701528684] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Injection drug use has been recognized as a major risk factor for acquired immunodeficiency syndrome (AIDS) from the outset of the epidemic. Cocaine, one of the most widely abused drugs in the United States, can both impair the functions of macrophages and CD4(+) lymphocytes and also activate human immunodeficiency virus (HIV)-1 expression in these cells. Because the brain is the target organ for both cocaine and HIV, the objective of the present study was to explore the effects of cocaine on virus replication in macrophages, the target cells for the virus in the central nervous system (CNS). Cocaine markedly enhanced virus production in simian human immunodeficiency virus (SHIV)-infected monocyte-derived macrophages (MDMs) and in U1 cells, a chronically infected promonocytic cell line as monitored by enzyme-linked immunosorbent assay (ELISA) and immunocytochemistry. Cocaine treatment also resulted in the activation of nuclear factor (NF)-kappa B and transcriptional activation of the HIV-LTR (long terminal repeat) gag-GFP (green fluorescent protein). Analyses of chemokines in cocaine-treated macrophages by real-time reverse transcriptase-polymerase chain reaction (RT-PCR) and Luminex assays suggested increased expression of interleukin (IL)-10, a cytokine that is known to promote HIV replication in MDMs. In addition to enhancing IL-10 expression, cocaine also caused an up-regulation of the macrophage activation marker, human leukocyte antigen (HLA)-DR, in MDMs. The synergistic effect of cocaine on virus replication and its enhancement of host activation markers suggest that cocaine functions at multiple pathways to accelerate HIV-associated dementia (HAD).
Collapse
Affiliation(s)
- Navneet K Dhillon
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
140
|
D'Cruz OJ, Uckun FM. Preclinical evaluation of a dual-acting microbicidal prodrug WHI-07 in combination with vanadocene dithiocarbamate in the female reproductive tract of rabbit, pig, and cat. Toxicol Pathol 2008; 35:910-27. [PMID: 18098038 DOI: 10.1080/01926230701748115] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The mucosal safety of the combination antiretroviral spermicide,WHI-07 [5-bromo-6-methoxy-5,6-dihydro-3'-azidothymidine-5'-(p-bromophenyl)-methoxy alaninyl phosphate] and vanadocene dithiocarbamate (VDDTC), was evaluated in 3 different animal models. Twenty-seven NZW rabbits in four subgroups were exposed intravaginally to a gel-microemulsion (GM) with and without three dose levels of WHI-07 plus VDDTC (0.5+0.06%, 1.0+0.12% and 2.0+0.25%) or 4% nonoxynol-9 (N-9; Conceptrol) for 14 consecutive days. Ten nonestrus gilts (Duroc) in three subgroups received either a single or daily intravaginal application of GM with and without 2.0% WHI-07 plus 0.25% VDDTC or 2.0% benzalkonium chloride (BZK)-containing gel for 6 and 4 consecutive days, respectively. Five cats received a single intravaginal application of GM incorporating 2.0% WHI-07 plus 0.25% VDDTC. Genital tract histopathology was performed in the pig and rabbit at the end of dosing period but after 18 weeks post-dosing in the cat. Porcine cervicovaginal lavage (CVL) fluid was obtained for up to 72 hours after a single exposure and changes in the levels of inflammatory cytokines (IL-1beta, IL-8, IFN-gamma, and TNF-alpha) were quantitated by a multiplexed chemiluminescence-based immunoassay. Rabbit vaginal tissues were evaluated for localized cellular inflammation and in situ apoptosis by immunohistochemical staining for CD45, nuclear factor (NF)-kappa B, and terminal deoxynucleotidyl transferase-mediated FITC-deoxyuridine triphosphate nick-end labeling (TUNEL) using confocal laser scanning microscopy (CLSM), respectively. Vanadium content in selected organs and body fluids from rabbits and pigs was determined by atomic absorption spectroscopy. When compared with 4% N-9 (total irritation score 13-14 out of a possible 16), none of the rabbits given WHI-07 plus VDDTC intravaginally, developed histological alterations such as epithelial erosion, edema, leukocyte influx or vascular congestion characteristic of inflammation (total irritation score 4-6). CD45 and NF-kappa B immunoreactivity was limited to cells within the vascular lumen of both control and WHI-07 plus VDDTC-treated vaginal tissues. TUNEL assay revealed lack of increased apoptotic cells in vaginal mucosa exposed to increasing concentrations of WHI-07 plus VDDTC. Basal levels of proinflammatory cytokines (IL-1beta, IL-8, IFN-gamma and TNF-alpha) in porcine CVL were unaffected by intravaginal exposure to WHI-07 plus VDDTC when compared with BZK used as a positive control. Endpoint histology of the reproductive tract from cats and pigs after a single or repeated intravaginal exposure to WHI-07 plus VDDTC, respectively, revealed lack of irritation/inflammation in the epithelium, subepithelium/lamina propria, vessels/perivascular tissues, and underlying/surrounding muscles. Vanadium was not preferentially incorporated into rabbit or porcine tissues and body fluids at levels above 1 microg/g. Based on comparative histologic data and surrogate markers for inflammation, repeated intravaginal administration of WHI-07 plus VDDTC via a gel-microemulsion did not result in vaginal irritation, mucosal toxicity, or systemic absorption of vanadium. Therefore, the combined use of WHI-07 and VDDTC via gel-microemulsion appears safe for topical use as a prophylactic anti-HIV microbicide.
Collapse
Affiliation(s)
- Osmond J D'Cruz
- Drug Discovery Program, Paradigm Pharmaceuticals, LLC, St. Paul, MN 55113, USA.
| | | |
Collapse
|
141
|
Chakravarti A, Marceau AA, Flamand L, Poubelle PE. Normal human primary CD4+ T lymphocytes synthesize and release functional osteoprotegerin in vitro. J Transl Med 2008; 88:171-84. [PMID: 18040268 DOI: 10.1038/labinvest.3700701] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Osteoprotegerin (OPG) acts as a decoy receptor for receptor activator of nuclear factor-kappaB ligand (RANKL) and TNF-related apoptosis-inducing ligand (TRAIL). OPG regulates bone remodeling and the immune response. The primary objective was to decipher, among human peripheral blood mononuclear leukocytes (PBML) that produce OPG, the subset(s) responsible for this synthesis and its regulation. To this end, normal human PBML and CD4-, 8-, 19-, 14-enriched subpopulations were studied in vitro for OPG synthesis. PBML were subjected to adherence and immunomagnetic separation, and OPG expression was analyzed by PCR, northern and western blotting, and ELISA. The antiapoptotic effects of OPG were studied on TRAIL-stimulated RPMI 8226 myeloma cells. OPG was time-dependently produced by primary CD4+ T lymphocytes exclusively. OPG secretion was upregulated by anti-CD3 antibody stimulation or incubation with interleukin (IL)-4, IL-1beta, TNF-alpha, GM-CSF, and vitamin D(3). In contrast, IL-10 inhibited the basal and IL-4-induced production of OPG by T cells. Conditioned media from activated T lymphocytes decreased TRAIL-induced apoptosis of RPMI 8226 cells. This effect was reversed by addition of RANKL to the T-cell conditioned media. As human immunodeficiency virus-1 (HIV-1) targets CD4+ T cells, we evaluated the effects of recombinant HIV-1 gp120 proteins on OPG synthesis. The gp120 from three different HIV-1 strains significantly reduced the basal output of OPG from T cells. Furthermore, all four protease inhibitors (PIs) used in highly active antiretroviral therapy decreased OPG synthesis by human blood T cells, nelfinavir being the most efficient PI. The simultaneous presence of an HIV-1 gp120 and a PI abrogated the basal output of OPG. In conclusion, these results highlight a new role for T lymphocytes involved in pathologies. Activated CD4+ T cells could, through OPG release, have a paracrine effect on adjacent cells and contribute to reduce the local process of bone remodeling and cellular apoptosis.
Collapse
|
142
|
Garrido M, Mozos A, Martínez A, García F, Serafín A, Morente V, Caballero M, Gil C, Fumero E, Miró JM, Climent N, Gatell JM, Alos L. HIV-1 upregulates intercellular adhesion molecule-1 gene expression in lymphoid tissue of patients with chronic HIV-1 infection. J Acquir Immune Defic Syndr 2008; 46:268-74. [PMID: 17786132 DOI: 10.1097/qai.0b013e318142c74c] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Intercellular adhesion molecule (ICAM)-1 is an adhesion molecule that plays an important role in the transmission of HIV-1 to CD4+ target cells and in the decrease of these cells in lymphoid tissue (LT). Our main objective was to study ICAM-1 expression in LT from HIV-1-infected persons and to correlate this expression with LT viral load and the immunoarchitecture alteration before and after highly active antiretroviral therapy (HAART). METHODS Tonsillar LT samples from 16 patients with chronic asymptomatic HIV-1 infection were studied before initiating treatment and after 12 months of HAART. ICAM-1 protein expression was studied by immunohistochemistry in all cases, and ICAM-1 messenger RNA (mRNA) was quantified from frozen tissue in 6 patients using quantitative real-time polymerase chain reaction (PCR). LT viral load was determined by PCR. The LT immunoarchitecture, p24 immunoexpression, and CD4+ cell count were assessed from tissue sections. RESULTS Before initiating HAART, there was high immunohistochemical ICAM-1 expression in follicular dendritic and endothelial cells and high ICAM-1 mRNA quantification. These findings correlated with a high LT viral load, strong p24 expression, and an effacement of LT immunoarchitecture with a low number of CD4+ cells. After HAART, there was a significant decrease of immunohistochemical and gene ICAM-1 expression. These results correlated with a significant decrease of LT viral load and p24 immunoexpression, a recovery of LT architecture, and a significant increase of CD4+ cells. CONCLUSIONS HIV-1 upregulates ICAM-1 expression in LT. This finding is associated with a marked effacement of LT architecture. HAART produces downregulation of ICAM-1 expression and recovery of LT architecture by reducing LT viral load significantly.
Collapse
Affiliation(s)
- Marta Garrido
- Department of Pathology, Hospital Clínic, Institut d'Investigacions Biomédiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
An examination of the effect of intestinal first pass extraction on intestinal lymphatic transport of saquinavir in the rat. Pharm Res 2007; 25:1125-33. [PMID: 17975709 DOI: 10.1007/s11095-007-9473-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Accepted: 10/05/2007] [Indexed: 12/15/2022]
Abstract
PURPOSE To assess the impact of intestinally based efflux/elimination processes on the extent of intestinal lymphatic transport of saquinavir. To compare the relative effects of co-administration of P-gp/CYP modulators on intestinal lymphatic transport versus systemic bioavailability of saquinavir. METHODS A cremophor mixed micelle formulation of saquinavir alone, or co-administered with P-gp/CYP modulators, verapamil, ketoconazole or cyclosporine, was dosed intraduodenally in the mesenteric lymph duct cannulated anaesthetized rat model. RESULTS Co-administration of P-gp/CYP modulators resulted in significant increases in the extent of intestinal lymphatic transport of saquinavir. A comparison of the relative enhancement of lymphatic transport and plasma bioavailability compared to control (i.e. saquinavir alone) reveals a greater effect of verapamil and ketoconazole on the amount of drug transported by the lymphatic route, an observation consistent with a preferential targeting of saquinavir via the intestinal lymphatics. In contrast co-administration of cyclosporine increased both the extent of lymphatic transport (5.5-fold), and systemic bioavailability (4.1-fold). CONCLUSIONS Intestinal P-gp/CYP efflux/elimination restricts saquinavir transport via the intestinal lymphatics in the rat. Targeted increases in intestinal lymphatic levels of saquinavir may be achieved by selective inhibition of intestinal P-gp and/or CYP.
Collapse
|
144
|
Gandhi NR, Skanderson M, Gordon KS, Concato J, Justice AC. Delayed presentation for human immunodeficiency virus (HIV) care among veterans: a problem of access or screening? Med Care 2007; 45:1105-9. [PMID: 18049352 PMCID: PMC3460382 DOI: 10.1097/mlr.0b013e3181271476] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Despite the effectiveness of antiretroviral therapy, nearly half of patients entering human immunodeficiency virus (HIV) care have advanced disease. Many attribute this delay to poor access to healthcare. Others argue that delays will persist until routine screening is adopted. The Veterans Health Administration (VA) is a unique laboratory to examine whether access to comprehensive health benefits results in earlier entry into HIV care. METHODS Retrospective observational study of 4368 HIV-positive patients entering HIV care during 1998-2002 at VA medical centers nationwide. OUTCOMES OF INTEREST rates of acquired immune deficiency syndrome in year of presentation; duration of VA utilization before HIV presentation; presence of "clinical triggers," signaling greater risk of HIV infection, before presentation. RESULTS Fifty-one percent (n = 2211) of all patients presented with CD4 counts of < 200 cells/mm. Thirty-nine percent (n = 1697) of all patients used other VA services before presentation for HIV care, with median duration of 3.6 years (interquartile range 25-75: 2.2-5.1 year) and 6 physician visits [interquartile range (IQR), 25-75: 2-18 visits] between first utilization and HIV presentation. No difference existed in the percentage presenting with CD4 counts <200 cells/mm among those with and without prior VA healthcare (50% vs. 51%, P = 0.76). Only 13% of those with prior VA healthcare demonstrated a clinical trigger before HIV presentation. CONCLUSIONS More than half of veterans entered HIV care with an acquired immune deficiency syndrome diagnosis at presentation irrespective of whether they had previously established healthcare in the VA. Access to care does not seem to be the primary cause of delayed HIV presentation. Widespread HIV screening is needed to improve rates of early detection.
Collapse
Affiliation(s)
- Neel R Gandhi
- Robert Wood Johnson Clinical Scholars Program and Section of General Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | | | | | | | | |
Collapse
|
145
|
Interaction of SIV/SHIV infection and morphine on plasma oxidant/antioxidant balance in macaque. Mol Cell Biochem 2007; 308:169-75. [PMID: 17934700 DOI: 10.1007/s11010-007-9625-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Accepted: 10/03/2007] [Indexed: 01/26/2023]
Abstract
A homeostatic balance exists between the cellular generation of oxidant species and endogenous antioxidants under normal physiological conditions. Human Immunodeficiency Virus (HIV) infection is known to affect this balance causing oxidative stress. However, the interaction of HIV infection with a substance abuse on cellular oxidant/antioxidant system is sparse. This study was designed in order to investigate the interactive effect of morphine abuse and Simian Immunodeficiency Virus/ Simian Human Immunodeficiency Virus (SIV/SHIV) infection on plasma oxidant/antioxidant balance in rhesus macaques. Six rhesus macaques adapted to morphine dependence (20 weeks) along with three controls were infected with mixture of SHIV(KU-1B), SHIV(89.6P), and SIV(17E-Fr). Plasma samples from morphine-dependent and control macaques were analyzed for an array of oxidative stress indices after 16 weeks of infection. Morphine-dependence significantly increased plasma malondialdehyde (MDA) and 8-isoprostane levels (8-fold and 2-fold), but these animals showed higher MDA and 8-isoprostane levels after viral infection (18-fold and 4-fold) which was directly correlated with increase in viral load and decline in CD4+ cells. Plasma glutathione (GSH) level depleted (55%) with morphine dependence that was further depleted (25%) by the infection. Activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) were increased by 30% and 110%, respectively with morphine dependence, but that was decreased by the infection. Catalase (CAT) activity declined (25%) with morphine dependence that was further declined by infection. Our results clearly suggest that morphine interaction with SIV/SHIV infection causes higher oxidative tissue injury that might have implication in the pathogenesis of AIDS in morphine-dependent macaques.
Collapse
|
146
|
Kilsztajn S, Lopes ES, do Carmo MSN, Rocha PAF. Improvement in survival among symptomatic AIDS patients by exposure category in Sao Paulo. J Acquir Immune Defic Syndr 2007; 45:342-7. [PMID: 17496563 DOI: 10.1097/qai.0b013e31806910ff] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE The study assesses AIDS survival time per AIDS-case definition and exposure category in Sao Paulo State, Brazil during the periods 1992 to 1995 and 1998 to 2001. METHODS Case-fatality rate per AIDS case, Cox proportional hazards analysis, and Kaplan-Meier survival time. RESULTS The case-fatality rate per AIDS case in 1998 to 2001 was 37.6% for symptomatic (Centers for Disease Control and Prevention/modified and/or Paho/Caracas) and 9.7% for the Brazilian asymptomatic CD4 count<350 cells/mm3 AIDS-case definitions. Heterosexual/female patients were diagnosed earlier and presented the lowest case-fatality rate, followed by homosexual/male patients, heterosexual/male patients, and injecting drug users. In the multivariate Cox proportional hazards model, the period of diagnosis (hazard ratio=2.66; 95% confidence interval [CI]: 2.58 to 2.74) and AIDS-case definition (hazard ratio=4.48; 95% CI: 4.53 to 5.16) were strong predictors of survival. For the total AIDS cases, excluding death definition and undetermined exposure category, the estimated first quarter survival time improved from 4 months in 1992 to 1995 to 50 months in 1998 to 2001. Considering only the symptomatic AIDS-case definition, however, the improvement was from 4 months in the period 1992 to 1995 to 14 months in the period 1998 to 2001. CONCLUSION The survival improvement in Sao Paulo State was attributable to the introduction of antiretroviral therapy with free universal access in 1996 and to earlier diagnosis associated with the introduction in 1998 of the Brazilian asymptomatic CD4 count<350 cells/mm3 AIDS-case definition with superior sensitivity compared with the symptomatic AIDS-case definitions.
Collapse
Affiliation(s)
- Samuel Kilsztajn
- Laboratório de Economia Social, and Secretaria de Economia e Planejamento, Estado de São Paulo, São Paulo, Brazil
| | | | | | | |
Collapse
|
147
|
Thibault S, Tardif MR, Barat C, Tremblay MJ. TLR2 Signaling Renders Quiescent Naive and Memory CD4+T Cells More Susceptible to Productive Infection with X4 and R5 HIV-Type 1. THE JOURNAL OF IMMUNOLOGY 2007; 179:4357-66. [PMID: 17878330 DOI: 10.4049/jimmunol.179.7.4357] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It has been recently demonstrated that circulating microbial products are responsible for a systemic immune activation in individuals infected with HIV-type 1. Bacterial products carry structural conserved motifs recognized by TLRs. Some TLR members are expressed in primary human CD4+ T cells but the precise functional role played by these pattern recognition receptors is still imprecise. In this study, we report that engagement of TLR2 in quiescent naive and memory CD4+ T cells leads to the acquisition of an effector-like phenotype. Interestingly, engagement of TLR2 renders both cell subsets more susceptible to productive infection with X4 virions and a higher virus production was seen with R5 viruses. It can be proposed that exposure of resting CD4+ T cells to pathogen-derived products that can engage TLR2 induces the acquisition of an effector-like phenotype in naive and memory CD4+ T lymphocytes, a phenomenon that might result in an acceleration of virus replication, immune dysregulation, and HIV-type 1-mediated disease progression.
Collapse
Affiliation(s)
- Sandra Thibault
- Centre de Recherche en Infectiologie, Centre Hospitalier de l'Université Laval, and Faculté de Médecine, Université Laval, Québec, Canada
| | | | | | | |
Collapse
|
148
|
Ciuffreda D, Pantaleo G, Pascual M. Effects of immunosuppressive drugs on HIV infection: implications for solid-organ transplantation. Transpl Int 2007; 20:649-58. [PMID: 17425723 DOI: 10.1111/j.1432-2277.2007.00483.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
With the advent of highly active antiretroviral therapy (HAART), HIV infection has become a chronic disease. Various end-stage organ failures have now become common co-morbidities and are primary causes of mortality in HIV-infected patients. Solid-organ transplantation therefore has been proposed to these patients, as HIV infection is not anymore considered an absolute contraindication. The initial results of organ transplantation in HIV-infected patients are encouraging with no differences in patient and graft survival compared with non-HIV-infected patients. The use of immunosuppressive drug therapy in HIV-infected patients has so far not shown major detrimental effects, and some drugs in combination with HAART have even demonstrated possible beneficial effects for specific HIV settings. Nevertheless, organ transplantation in HIV-infected patients remains a complex intervention, and more studies will be required to clarify open questions such as long-term effects of drug interactions between antiretroviral and immunosuppressive drugs, outcome of recurrent HCV infection in HIV-infected patients, incidence of graft rejection, or long-term graft and patient survival. In this article, we first review the immunological pathogenesis of HIV infection and the rationale for using immunosuppression combined with HAART. We then discuss the most recent results of solid-organ transplantation in HIV-infected patients.
Collapse
Affiliation(s)
- Donatella Ciuffreda
- Division of Immunology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| | | | | |
Collapse
|
149
|
Nasr SH, Share DS, Vargas MT, D'Agati VD, Markowitz GS. Acute poststaphylococcal glomerulonephritis superimposed on diabetic glomerulosclerosis. Kidney Int 2007; 71:1317-21. [PMID: 17311069 DOI: 10.1038/sj.ki.5002135] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- S H Nasr
- Department of Pathology, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA.
| | | | | | | | | |
Collapse
|
150
|
|