Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Seema S. Ahuja is active.

Publication


Featured researches published by Seema S. Ahuja.


Proceedings of the National Academy of Sciences of the United States of America | 2002

HIV-1 infection and AIDS dementia are influenced by a mutant MCP-1 allele linked to increased monocyte infiltration of tissues and MCP-1 levels

Enrique Gonzalez; Brad H. Rovin; Luisa Sen; Glen E. Cooke; Rahul Dhanda; Srinivas Mummidi; Hemant Kulkarni; Michael J. Bamshad; Vanessa Telles; Stephanie A. Anderson; Elizabeth A. Walter; Kevin T. Stephan; Michael F. Deucher; Andrea Mangano; Rosa Bologna; Seema S. Ahuja; Matthew J. Dolan; Sunil K. Ahuja

Studies in humans and in experimental models of HIV-1 infection indicate an important role for monocyte chemoattractant protein-1 (MCP-1; also known as CC chemokine ligand 2), a potent chemoattractant and activator of mononuclear phagocytes (MP) in the pathogenesis of HIV-associated dementia (HAD). We determined the influence of genetic variation in MCP-1 on HIV-1 pathogenesis in large cohorts of HIV-1-infected adults and children. In adults, homozygosity for the MCP-1 –2578G allele was associated with a 50% reduction in the risk of acquiring HIV-1. However, once HIV-1 infection was established, this same MCP-1 genotype was associated with accelerated disease progression and a 4.5-fold increased risk of HAD. We examined the molecular and cellular basis for these genotype–phenotype associations and found that the mutant MCP-1 –2578G allele conferred greater transcriptional activity via differential DNA–protein interactions, enhanced protein production in vitro, increased serum MCP-1 levels, as well as MP infiltration into tissues. Thus, MCP-1 expression had a two-edged role in HIV-1 infection: it afforded partial protection from viral infection, but during infection, its proinflammatory properties and ability to up-regulate HIV-1 replication collectively may contribute to accelerated disease progression and increased risk of dementia. Our findings suggest that MCP-1 antagonists may be useful in HIV-1 infection, especially for HAD, and that HIV+ individuals possessing the MCP-1 –2578G allele may benefit from early initiation of antiretroviral drugs that effectively cross the blood–brain barrier. In a broader context, the MCP-1 –2578G allele may serve as a genetic determinant of outcome of other disease states in which MP-mediated tissue injury is central to disease pathogenesis.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Global survey of genetic variation in CCR5, RANTES, and MIP-1α: Impact on the epidemiology of the HIV-1 pandemic

Enrique Gonzalez; Rahul Dhanda; Michael J. Bamshad; Srinivas Mummidi; Reni Geevarghese; Gabriel Catano; Stephanie A. Anderson; Elizabeth A. Walter; Kevin T. Stephan; Michael F. Hammer; Andrea Mangano; Luisa Sen; Robert A. Clark; Seema S. Ahuja; Matthew J. Dolan; Sunil K. Ahuja

Expression of CC chemokine receptor 5 (CCR5), the major coreceptor for HIV-1 cell entry, and its ligands (e.g., RANTES and MIP-1α) is widely regarded as central to the pathogenesis of HIV-1 infection. By surveying nearly 3,000 HIV+ and HIV− individuals from worldwide populations for polymorphisms in the genes encoding RANTES, MIP-1α, and CCR5, we show that the evolutionary histories of human populations have had a significant impact on the distribution of variation in these genes, and that this may be responsible, in part, for the heterogeneous nature of the epidemiology of the HIV-1 pandemic. The varied distribution of RANTES haplotypes (AC, GC, and AG) associated with population-specific HIV-1 transmission- and disease-modifying effects is a striking example. Homozygosity for the AC haplotype was associated with an increased risk of acquiring HIV-1 as well as accelerated disease progression in European Americans, but not in African Americans. Yet, the prevalence of the ancestral AC haplotype is high in individuals of African origin, but substantially lower in non-Africans. In a Japanese cohort, AG-containing RANTES haplotype pairs were associated with a delay in disease progression; however, we now show that their contribution to HIV-1 pathogenesis and epidemiology in other parts of the world is negligible because the AG haplotype is infrequent in non-Far East Asians. Thus, the varied distribution of RANTES, MIP-1α, and CCR5 haplotype pairs and their population-specific phenotypic effects on HIV-1 susceptibility and disease progression results in a complex pattern of biological determinants of HIV-1 epidemiology. These findings have important implications for the design, assessment, and implementation of effective HIV-1 intervention and prevention strategies.


Journal of Bone and Mineral Research | 2002

MLO-Y4 osteocyte-like cells support osteoclast formation and activation

Shujie Zhao; Y. Kato; Y. Zhang; S. E. Harris; Seema S. Ahuja; Lynda F. Bonewald

Osteocytes are terminally differentiated cells of the osteoblast lineage that have become embedded in mineralized matrix and may send signals that regulate bone modeling and remodeling. The hypothesis to be tested in this study is that osteocytes can stimulate and support osteoclast formation and activation. To test this hypothesis, an osteocyte‐like cell line called MLO‐Y4 and primary murine osteocytes were used in coculture with spleen or marrow cells. MLO‐Y4 cells support osteoclast formation in the absence of 1,25‐dihydroxyvitamin D3 [1,25(OD)2D3] or any other exogenous osteotropic factor. These cells alone stimulate osteoclast formation to the same extent or greater than adding 1,25(OH)2D3. Coaddition of 1,25(OH)2D3 with MLO‐Y4 cells synergistically increased osteoclast formation. Optimal osteoclast formation and pit formation on dentine was observed with 200–1000 MLO‐Y4 cells per 0.75‐cm2 well. No osteoclast formation was observed with 2T3, OCT‐1, or MC3T3‐E1 osteoblast cells (1000 cells/well). Conditioned media from the MLO‐Y4 cells had no effect on osteoclast formation, indicating that cell contact is necessary. Serial digestions of 2‐week‐old mouse calvaria yielded populations of cells that support osteoclast formation when cocultured with 1,25(OH)2D3 and marrow, but the population that remained in the bone particles supported the greatest number of osteoclasts with or without 1,25(OH)2D3. To examine the mechanism whereby these cells support osteoclast formation, the MLO‐Y4 cells were compared with a series of osteoblast and stromal cells for expression of macrophage colony‐stimulating factor (M‐CSF), RANKL, and osteoprotegerin (OPG). MLO‐Y4 cells express and secrete large amounts of M‐CSF. MLO‐Y4 cells express RANKL on their surface and their dendritic processes. The ratio of RANKL to OPG mRNA is greatest in the MLO‐Y4 cells compared with the other cell types. RANK‐Fc and OPG‐Fc blocked the formation of osteoclasts by MLO‐Y4 cells. These studies suggest that both RANKL and OPG may play a role in osteocyte signaling, OPG and M‐CSF as soluble factors and RANKL as a surface molecule that is functional in osteocytes or along their exposed dendritic processes.


Journal of Clinical Investigation | 1993

Negative transcriptional regulation of human interleukin 2 (IL-2) gene by glucocorticoids through interference with nuclear transcription factors AP-1 and NF-AT.

Fotini Paliogianni; Anastasios Raptis; Seema S. Ahuja; Sonia M. Najjar; Dimitrios T. Boumpas

IL-2 gene transcription is affected by several nuclear proteins. We asked whether dexamethasone (Dex) and cyclosporin A (CsA) inhibit IL-2 gene transcription by interfering with the activity of nuclear proteins that bind to the IL-2 promoter. Nuclear extracts from primary human T lymphocytes were analyzed by electrophoretic DNA mobility shift assays. Both Dex and CsA inhibited the binding of transcription factors AP-1 and NF-AT, but not of NF-kB and OCT-1/OAF, to their corresponding sites on the IL-2 gene promoter. To correlate changes in nuclear factor binding in vitro with transcriptional activity in vivo and define the structural requirements for IL-2 promoter repression, we used transient DNA transfections. Jurkat cells were transfected with plasmids containing either the intact IL-2 promoter or its AP-1, NF-AT, and NF-kB motifs. Dex inhibited the IL-2 promoter and the AP-1, but not the NF-AT and NF-kB plasmids. In contrast, CsA inhibited the IL-2 promoter and the NF-AT, but not the AP-1 and NF-kB plasmids. These results suggest that in human T lymphocytes both Dex and CsA inhibited IL-2 gene transcription through interference with transcription factors AP-1 and NF-AT. We propose that, while maximum inhibition may involve interaction with both transcription factors, AP-1 is the primary target of Dex.


Proceedings of the National Academy of Sciences of the United States of America | 2002

A strong signature of balancing selection in the 5' cis-regulatory region of CCR5.

Michael J. Bamshad; Srinivas Mummidi; Enrique Gonzalez; Seema S. Ahuja; Diane M. Dunn; W. Scott Watkins; Stephen Wooding; Anne C. Stone; Lynn B. Jorde; Robert B. Weiss; Sunil K. Ahuja

CCR5 encodes a cell surface chemokine receptor molecule that serves as the principal coreceptor, with CD4, for HIV-type 1 (HIV-1). Varied HIV-1 susceptibility and time to progression to AIDS have been associated with polymorphisms in CCR5. Many of these polymorphisms are located in the 5′ cis-regulatory region of CCR5, suggesting that it may have been a target of natural selection. We characterized CCR5 sequence variation in this region in 400 chromosomes from worldwide populations and compared it to a genome-wide analysis of 100 Alu polymorphisms typed in the same populations. Variation was substantially higher than expected and characterized by an excess of intermediate-frequency alleles. A genealogy of CCR5 haplotypes had deep branch lengths despite markedly little differentiation among populations. This finding suggested a deviation from neutrality not accounted for by population structure, which was confirmed by tests for natural selection. These results are strong evidence that balancing selection has shaped the pattern of variation in CCR5 and suggest that HIV-1 resistance afforded by CCR5 5′ cis-regulatory region haplotypes may be the consequence of adaptive changes to older pathogens.


Nature Immunology | 2007

CCL3L1 and CCR5 influence cell-mediated immunity and affect HIV-AIDS pathogenesis via viral entry-independent mechanisms

Matthew J. Dolan; Hemant Kulkarni; Jose F. Camargo; Weijing He; Alison Smith; Juan-Manuel Anaya; Toshiyuki Miura; Frederick Hecht; Manju Mamtani; Florencia Pereyra; Vincent C. Marconi; Andrea Mangano; Luisa Sen; Rosa Bologna; Robert A. Clark; Stephanie A. Anderson; Judith Delmar; Robert J. O'Connell; Andrew Lloyd; Jeffrey N. Martin; Seema S. Ahuja; Brian K. Agan; Bruce D. Walker; Steven G. Deeks; Sunil K. Ahuja

Although host defense against human immunodeficiency virus 1 (HIV-1) relies mainly on cell-mediated immunity (CMI), the determinants of CMI in humans are poorly understood. Here we demonstrate that variations in the genes encoding the chemokine CCL3L1 and HIV coreceptor CCR5 influence CMI in both healthy and HIV-infected individuals. CCL3L1-CCR5 genotypes associated with altered CMI in healthy subjects were similar to those that influence the risk of HIV transmission, viral burden and disease progression. However, CCL3L1-CCR5 genotypes also modify HIV clinical course independently of their effects on viral load and CMI. These results identify CCL3L1 and CCR5 as major determinants of CMI and demonstrate that these host factors influence HIV pathogenesis through their effects on both CMI and other viral entry–independent mechanisms.


Atherosclerosis | 2003

CCR5 deficiency is not protective in the early stages of atherogenesis in apoE knockout mice

William A. Kuziel; Tracey C. Dawson; Marlon P. Quinones; Edgar Garavito; George Chenaux; Seema S. Ahuja; Robert L. Reddick; Nobuyo Maeda

The accumulation of macrophages and T lymphocytes in vessel walls is a hallmark of atherogenesis. It has recently been demonstrated in mouse models of atherosclerosis that full disease potential is dependent on several regulators of leukocyte trafficking, including the chemokine monocyte chemotactic protein 1 (MCP-1) and the chemokine receptors CCR2 and CXCR2. A possible role for the chemokine receptor CCR5 in atherogenesis has been suggested by CCR5 expression on macrophages, T cells, coronary endothelial cells and aortic smooth muscle cells and by the presence of CCR5 ligands in atherosclerotic plaques. Moreover, individuals who are naturally deficient in CCR5 were reported to be at reduced risk for severe coronary artery disease (CAD) and early myocardial infarction (MI). To investigate whether CCR5 is pro-atherogenic in mice, we generated CCR5-deficient mice and crossed them with atherosclerosis-prone apoE-deficient mice. Although CCR5-deficient mice exhibit defects in induced macrophage trafficking, mean atherosclerotic lesion area did not differ significantly between apoE-deficient mice and apoE/CCR5-deficient mice after 16 weeks on a diet of normal chow. Ribonuclease protection assays (RPA) on RNA isolated from plaques from both apoE-deficient and apoE/CCR5-deficient animals showed strong signals for the macrophage marker F4/80 but no evidence for expression of prominent markers of T and B lymphocytes. These results indicate that the early stages of plaque formation in this model of lipid-mediated atherogenesis do not depend on CCR5.


Journal of Biological Chemistry | 1997

The human CC chemokine receptor 5 (CCR5) gene. Multiple transcripts with 5'-end heterogeneity, dual promoter usage, and evidence for polymorphisms within the regulatory regions and noncoding exons

Srinivas Mummidi; Seema S. Ahuja; Brent L. McDaniel; Sunil K. Ahuja

Human CC chemokine receptor 5 (CCR5), mediates the activation of cells by the chemokines macrophage inflammatory protein-1α, macrophage inflammatory protein-1β, and RANTES, and serves as a fusion cofactor for macrophage-tropic strains of human immunodeficiency virus type 1. To understand the molecular mechanisms that regulate human CCR5 gene expression, we initiated studies to determine its genomic and mRNA organization. Previous studies have identified a single CCR5 mRNA isoform whose open reading frame is intronless. We now report the following novel findings. 1) Complex alternative splicing and multiple transcription start sites give rise to several distinct CCR5 transcripts that differ in their 5′-untranslated regions (UTR). 2) The gene is organized into four exons and two introns. Exons 2 and 3 are not interrupted by an intron. Exon 4 and portions of exon 3 are shared by all isoforms. Exon 4 contains the open reading frame, 11 nucleotides of the 5′-UTR and the complete 3′-UTR. 3) The transcripts appear to be initiated from two distinct promoters: an upstream promoter (PU), upstream of exon 1, and a downstream promoter (PD), that includes the “intronic” region between exons 1 and 3. 4) PU and PD lacked the canonical TATA or CAAT motifs, and are AT-rich. 5) PD demonstrated strong constitutive promoter activity, whereas PU was a weak promoter in all three leukocyte cell environments tested (THP-1, Jurkat, and K562). 6) We provide evidence for polymorphisms in the noncoding sequences, including the regulatory regions and 5′-UTRs. The structure of CCR5 was strikingly reminiscent of the overall structure of other chemokine/chemoattractant receptors, underscoring an important evolutionarily conserved function for a prototypical gene structure. This is the first description of functional promoters for any CC chemokine receptor gene, and we speculate that the complex pattern of splicing events and dual promoter usage may function as a versatile mechanism to create diversity and flexibility in the regulation of CCR5 expression.


Journal of Clinical Investigation | 2004

Experimental arthritis in CC chemokine receptor 2–null mice closely mimics severe human rheumatoid arthritis

Marlon P. Quinones; Sunil K. Ahuja; Fabio Jimenez; Jason Schaefer; Edgar Garavito; Arun Rao; George Chenaux; Robert L. Reddick; William A. Kuziel; Seema S. Ahuja

The prevailing paradigm is that in human rheumatoid arthritis (RA), the accumulation of monocytes and T cells in the joint, mediated in part by such CC chemokine receptors (CCRs) as CCR2 and CCR5, respectively, plays a central role in disease pathogenesis. To further validate this paradigm, we conducted proof-of-principle studies and tested the hypothesis that gene inactivation of Ccr2 or Ccr5 will ameliorate experimental RA. Contrary to our expectations, we found that in two well-established murine models of experimental RA, CCR2 expression in the hematopoietic cell compartment served as a negative regulator of autoantibody production as well as arthritic disease onset, severity, and resolution. In contrast, the RA phenotype in Ccr5-null mice was similar to that of WT mice. Remarkably, the collagen-induced arthritis phenotype of Ccr2-/- mice mimicked closely that of severe human RA, including production of rheumatoid factor, enhanced T cell production, and monocyte/macrophage accumulation in the joints. Our findings demonstrate an essential protective role of CCR2 expression in RA, indicate the existence of alternative receptors responsible for monocyte/macrophage accumulation to inflamed joints, and emphasize the need to clarify carefully the complex effects of the chemokine system in RA before they can be considered as therapeutic targets.


Nature Medicine | 2008

CCL3L1-CCR5 genotype influences durability of immune recovery during antiretroviral therapy of HIV-1-infected individuals

Sunil K. Ahuja; Hemant Kulkarni; Gabriel Catano; Brian K. Agan; Jose F. Camargo; Weijing He; Robert J. O'Connell; Vincent C. Marconi; Judith Delmar; Joseph J. Eron; Robert A. Clark; Simon D. W. Frost; Jeffrey N. Martin; Seema S. Ahuja; Steven G. Deeks; Susan J. Little; Douglas D. Richman; Frederick Hecht; Matthew J. Dolan

The basis for the extensive variability seen in the reconstitution of CD4+ T cell counts in HIV-infected individuals receiving highly active antiretroviral therapy (HAART) is not fully known. Here, we show that variations in CCL3L1 gene dose and CCR5 genotype, but not major histocompatibility complex HLA alleles, influence immune reconstitution, especially when HAART is initiated at <350 CD4+ T cells/mm3. The CCL3L1-CCR5 genotypes favoring CD4+ T cell recovery are similar to those that blunted CD4+ T cell depletion during the time before HAART became available (pre-HAART era), suggesting that a common CCL3L1-CCR5 genetic pathway regulates the balance between pathogenic and reparative processes from early in the disease course. Hence, CCL3L1-CCR5 variations influence HIV pathogenesis even in the presence of HAART and, therefore, may prospectively identify subjects in whom earlier initiation of therapy is more likely to mitigate immunologic failure despite viral suppression by HAART. Furthermore, as reconstitution of CD4+ cells during HAART is more sensitive to CCL3L1 dose than to CCR5 genotypes, CCL3L1 analogs might be efficacious in supporting immunological reconstitution.

Collaboration


Dive into the Seema S. Ahuja's collaboration.

Top Co-Authors

Avatar

Sunil K. Ahuja

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Fabio Jimenez

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Marlon P. Quinones

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Hernan Martinez

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Robert A. Clark

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Srinivas Mummidi

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Carlos A. Estrada

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Robert L. Reddick

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Merry L. Lindsey

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Matthew J. Dolan

San Antonio Military Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge