Network


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

Hotspot


Dive into the research topics where Rehana Z. Hussain is active.

Publication


Featured researches published by Rehana Z. Hussain.


Journal of Immunology | 2002

Peroxisome Proliferator-Activated Receptor-γ Agonist 15-Deoxy-Δ12,1412,14-Prostaglandin J2 Ameliorates Experimental Autoimmune Encephalomyelitis

Asim Diab; Caishu Deng; Jeff D. Smith; Rehana Z. Hussain; Bounleut Phanavanh; Amy E. Lovett-Racke; Paul D. Drew; Michael K. Racke

Peroxisome proliferator-activated receptors (PPAR) are members of a nuclear hormone receptor superfamily that includes receptors for steroids, retinoids, and thyroid hormone, all of which are known to affect the immune response. Previous studies dealing with PPAR-γ expression in the immune system have been limited. Recently, PPAR-γ was identified in monocyte/macrophage cells. In this study we examined the role of PPAR-γ in experimental autoimmune encephalomyelitis (EAE), an animal model for the human disease multiple sclerosis. The hypothesis we are testing is whether PPAR-γ plays an important role in EAE pathogenesis and whether PPAR-γ ligands can inhibit the clinical expression of EAE. Initial studies have shown that the presence of the PPAR-γ ligand 15-deoxy-Δ12,14-PGJ2 (15d-PGJ2) inhibits the proliferation of Ag-specific T cells from the spleen of myelin basic protein Ac1–11 TCR-transgenic mice. 15d-PGJ2 suppressed IFN-γ, ΙL-10, and IL-4 production by both Con A- and myelin basic protein Ac1–11 peptide-stimulated lymphocytes as determined by ELISA and ELISPOT assay. Culture of encephalitogenic T cells with 15d-PGJ2 in the presence of Ag reduced the ability of these cells to adoptively transfer EAE. Examination of the target organ, the CNS, during the course of EAE revealed expression of PPAR-γ in the spinal cord inflammatory infiltrate. Administration of 15d-PGJ2 before and at the onset of clinical signs of EAE significantly reduced the severity of disease. These results suggest that PPAR-γ ligands may be a novel therapeutic agent for diseases such as multiple sclerosis.


Journal of Experimental Medicine | 2011

Fumarates improve psoriasis and multiple sclerosis by inducing type II dendritic cells

Kamran Ghoreschi; Jürgen Brück; Christina Kellerer; Caishu Deng; Haiyan Peng; Oliver Rothfuss; Rehana Z. Hussain; Anne R. Gocke; Annedore Respa; Ivana Glocova; Nadejda Valtcheva; Eva Alexander; Susanne Feil; Robert Feil; Klaus Schulze-Osthoff; Rudolf A. Rupec; Amy E. Lovett-Racke; Ralf Dringen; Michael K. Racke; Martin Röcken

Fumarates suppress Th1 responses by blocking IL-12 and IL-23 production by dendritic cells via distinct pathways.


Journal of Immunology | 2007

T-bet Regulates the Fate of Th1 and Th17 Lymphocytes in Autoimmunity

Anne R. Gocke; Petra D. Cravens; Li-Hong Ben; Rehana Z. Hussain; Sara C. Northrop; Michael K. Racke; Amy E. Lovett-Racke

IL-17-producing T cells (Th17) have recently been implicated in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model for the human disease multiple sclerosis. However, little is known about the transcription factors that regulate these cells. Although it is clear that the transcription factor T-bet plays an essential role in the differentiation of IFN-γ-producing CD4+ Th1 lymphocytes, the potential role of T-bet in the differentiation of Th17 cells is not completely understood. In this study, therapeutic administration of a small interfering RNA specific for T-bet significantly improved the clinical course of established EAE. The improved clinical course was associated with suppression of newly differentiated T cells that express IL-17 in the CNS as well as suppression of myelin basic protein-specific Th1 autoreactive T cells. Moreover, T-bet was found to directly regulate transcription of the IL-23R, and, in doing so, influenced the fate of Th17 cells, which depend on optimal IL-23 production for survival. We now show for the first time that suppression of T-bet ameliorates EAE by limiting the differentiation of autoreactive Th1 cells, as well as inhibiting pathogenic Th17 cells via regulation of IL-23R.


Journal of Immunology | 2004

Peroxisome Proliferator-Activated Receptor α Agonists as Therapy for Autoimmune Disease

Amy E. Lovett-Racke; Rehana Z. Hussain; Sara C. Northrop; Judy Choy; Anne Rocchini; Lela Matthes; Janet A. Chavis; Asim Diab; Paul D. Drew; Michael K. Racke

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily. PPARγ ligands, which include the naturally occurring PG metabolite 15-deoxy-Δ12,14-PGJ2 (15d-PGJ2), as well as thiazolidinediones, have been shown to have anti-inflammatory activity. The PPARα agonists, gemfibrozil, ciprofibrate, and fenofibrate, have an excellent track history as oral agents used to treat hypertriglyceridemia. In the present study, we demonstrate that these PPARα agonists can increase the production of the Th2 cytokine, IL-4, and suppress proliferation by TCR transgenic T cells specific for the myelin basic protein Ac1–11, as well as reduce NO production by microglia. Oral administration of gemfibrozil and fenofibrate inhibited clinical signs of experimental autoimmune encephalomyelitis. More importantly, gemfibrozil was shown to shift the cytokine secretion of human T cell lines by inhibiting IFN-γ and promoting IL-4 secretion. These results suggest that PPARα agonists such as gemfibrozil and fenofibrate, may be attractive candidates for use in human inflammatory conditions such as multiple sclerosis.


Journal of Neuroimmunology | 2004

Ligands for the peroxisome proliferator-activated receptor-γ and the retinoid X receptor exert additive anti-inflammatory effects on experimental autoimmune encephalomyelitis

Asim Diab; Rehana Z. Hussain; Amy E. Lovett-Racke; Janet A. Chavis; Paul D. Drew; Michael K. Racke

Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) is a member of the nuclear-receptor superfamily that binds to DNA with retinoid X receptors (RXRs) as PPAR-RXR heterodimers. In experimental autoimmune encephalomyelitis (EAE), the gene expression of PPAR-gamma was demonstrated in spinal cord during the course of EAE. Administration of 15-deoxy-(12,14)-prostaglandin J2 (15d-PGJ2) or 9-cis-retinoic acid (RA) alone at the onset of clinical signs of EAE reduced the severity of disease, however, their combination resulted in enhanced amelioration of disease. These results suggest that use of RXR specific ligands may be highly effective when combined with PPAR-gamma agonists in the treatment of autoimmune demyelinating diseases such as multiple sclerosis (MS).


Journal of Immunology | 2003

IL-1 Receptor-Associated Kinase 1 Regulates Susceptibility to Organ-Specific Autoimmunity

Caishu Deng; Caius G. Radu; Asim Diab; May F. Tsen; Rehana Z. Hussain; John S. Cowdery; Michael K. Racke; James A. Thomas

Infections often precede the development of autoimmunity. Correlation between infection with a specific pathogen and a particular autoimmune disease ranges from moderately strong to quite weak. This lack of correspondence suggests that autoimmunity may result from microbial activation of a generic, as opposed to pathogen-specific host-defense response. The Toll-like receptors, essential to host recognition of microbial invasion, signal through a common, highly conserved pathway, activate innate immunity, and control adaptive immune responses. To determine the influence of Toll/IL-1 signaling on the development of autoimmunity, the responses of wild-type (WT) mice and IL-1R-associated kinase 1 (IRAK1)-deficient mice to induction of experimental autoimmune encephalomyelitis were compared. C57BL/6 and B6.IRAK1-deficient mice were immunized with MOG 35–55/CFA or MOG 35–55/CpG DNA/IFA. WT animals developed severe disease, whereas IRAK1-deficient mice were resistant to experimental autoimmune encephalomyelitis, exhibiting little or no CNS inflammation. IRAK1-deficient T cells also displayed impaired Th1 development, particularly during disease induction, despite normal TCR signaling. These results suggest that IRAK1 and the Toll/IL-1 pathway play an essential role in T cell priming, and demonstrate one means through which innate immunity can control subsequent development of autoimmunity. These findings may also help explain the association between antecedent infection and the development or exacerbations of some autoimmune diseases.


Journal of Neuroimmunology | 2006

Multiple toll-like receptor agonists act as potent adjuvants in the induction of autoimmunity

Baranda S. Hansen; Rehana Z. Hussain; Amy E. Lovett-Racke; James A. Thomas; Michael K. Racke

Infections can trigger or exacerbate the course of Multiple Sclerosis, and both bacterial and viral agents have been implicated. These agents are recognized by host cells via pathogen-associated molecular patterns activating TLRs. We investigated the role that PAMPs play in the animal model Experimental Autoimmune Encephalomyelitis, and found various MyD88-dependent PAMPs can participate as the adjuvant to induce EAE. Studies with IRAK1-deficient mice suggest that signaling through TLRs is not required in the target organ to develop disease. This suggests that PAMPs play an important role in priming of autoreactive T cells in EAE and potentially MS.


Annals of Neurology | 2010

Silencing Nogo-A Promotes Functional Recovery in Demyelinating Disease

Yuhong Yang; Yue Liu; Ping Wei; Haiyan Peng; Ryan Winger; Rehana Z. Hussain; Li-Hong Ben; Petra D. Cravens; Anne R. Gocke; Krishna Puttaparthi; Michael K. Racke; Dana M. McTigue; Amy E. Lovett-Racke

To determine if suppressing Nogo‐A, an axonal inhibitory protein, will promote functional recovery in a murine model of multiple sclerosis (MS).


PLOS ONE | 2011

Rituximab Therapy Reduces Organ-Specific T Cell Responses and Ameliorates Experimental Autoimmune Encephalomyelitis

Nancy L. Monson; Petra D. Cravens; Rehana Z. Hussain; Christopher T. Harp; Matthew Cummings; Maria de Pilar Martin; Li Hong Ben; Julie Do; Jeri-Anne Lyons; Amy Lovette-Racke; Anne H. Cross; Michael K. Racke; Olaf Stüve; Mark J. Shlomchik; Todd N. Eagar

Recent clinical trials have established B cell depletion by the anti-CD20 chimeric antibody Rituximab as a beneficial therapy for patients with relapsing-remitting multiple sclerosis (MS). The impact of Rituximab on T cell responses remains largely unexplored. In the experimental autoimmune encephalomyelitis (EAE) model of MS in mice that express human CD20, Rituximab administration rapidly depleted peripheral B cells and strongly reduced EAE severity. B cell depletion was also associated with diminished Delayed Type Hypersensitivity (DTH) and a reduction in T cell proliferation and IL-17 production during recall immune response experiments. While Rituximab is not considered a broad immunosuppressant, our results indicate a role for B cells as a therapeutic cellular target in regulating encephalitogenic T cell responses in specific tissues.


Journal of Immunology | 2009

Transcriptional Modulation of the Immune Response by Peroxisome Proliferator-Activated Receptor-α Agonists in Autoimmune Disease

Anne R. Gocke; Rehana Z. Hussain; Yuhong Yang; Haiyan Peng; Jeffrey Weiner; Li Hong Ben; Paul D. Drew; Olaf Stüve; Amy E. Lovett-Racke; Michael K. Racke

Peroxisome proliferator-activated receptor-α (PPARα) agonists have been shown to have a therapeutic benefit in experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis (MS). In this study, we investigated the mechanism by which the PPARα agonist gemfibrozil induces immune deviation and protects mice from EAE. We demonstrated that treatment with gemfibrozil increases expression of the Th2 transcription factor GATA-3 and decreases expression of the Th1 transcription factor T-bet in vitro and directly ex vivo. These changes correlated with an increase in nuclear PPARα expression. Moreover, the protective effects of PPARα agonists in EAE were shown to be partially dependent on IL-4 and to occur in a receptor-dependent manner. PPARα was demonstrated, for the first time, to regulate the IL-4 and IL-5 genes and to bind the IL-4 promoter in the presence of steroid receptor coactivator-1, indicating that PPARα can directly transactivate the IL-4 gene. Finally, therapeutic administration of PPARα agonists ameliorated clinically established EAE, suggesting that PPARα agonists may provide a treatment option for immune-mediated inflammatory diseases.

Collaboration


Dive into the Rehana Z. Hussain's collaboration.

Top Co-Authors

Avatar

Michael K. Racke

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Olaf Stüve

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Petra D. Cravens

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Todd N. Eagar

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Li Hong Ben

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sara C. Northrop

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Anne R. Gocke

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Benjamine Arellano

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Caishu Deng

University of Texas Medical Branch

View shared research outputs
Researchain Logo
Decentralizing Knowledge