Network


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

Hotspot


Dive into the research topics where Rukhsana Aslam is active.

Publication


Featured researches published by Rukhsana Aslam.


Blood | 2010

A murine model of severe immune thrombocytopenia is induced by antibody- and CD8+ T cell-mediated responses that are differentially sensitive to therapy

Leola Chow; Rukhsana Aslam; Edwin R. Speck; Michael Kim; Norman Cridland; Michelle Lee Webster; Pingguo Chen; Kim Sahib; Heyu Ni; Alan H. Lazarus; M. Bernadette Garvey; John Freedman; John W. Semple

Immune thrombocytopenia (ITP) is a bleeding disorder characterized by antibody-opsonized platelets being prematurely destroyed in the spleen, although some patients with ITP may have a cell-mediated form of thrombocytopenia. Although several animal models of ITP have been developed, few mimic primary chronic ITP nor have any shown cell-mediated platelet destruction. To create this type of model, splenocytes from CD61 knockout mice immunized against CD61(+) platelets were transferred into severe combined immunodeficient (SCID) (CD61(+)) mouse recipients, and their platelet counts and phenotypes were observed. As few as 5 x 10(4) splenocytes induced a significant thrombocytopenia and bleeding mortality (80%) in recipients within 3 weeks after transfer. Depletion of lymphocyte subsets before transfer showed that the splenocytes ability to induce thrombocytopenia and bleeding completely depended on CD4(+) T helper cells and that both CD19(+) B cell (antibody)- and CD8(+) T cell (cell)-mediated effector mechanisms were responsible. Treatment of the SCID mouse recipients with intravenous gamma-globulins raised platelet counts and completely prevented bleeding mortality induced by antibody-mediated effector mechanisms but did not affect cell-mediated disease. This novel model not only shows both antibody- and cell-mediated ITP and bleeding but also suggests that these 2 effector mechanisms have a differential response to therapy.


Journal of Cell Biology | 2012

T granules in human platelets function in TLR9 organization and signaling

Jonathan N. Thon; Christopher Peters; Kellie R. Machlus; Rukhsana Aslam; Jesse W. Rowley; Hannah Macleod; Matthew T. Devine; Tobias A. Fuchs; Andrew S. Weyrich; John W. Semple; Robert Flaumenhaft; Joseph E. Italiano

TLR9 localizes to a novel intracellular compartment called the T granule to promote immune signaling by platelets.


Blood | 2012

Thymic retention of CD4+CD25+FoxP3+ T regulatory cells is associated with their peripheral deficiency and thrombocytopenia in a murine model of immune thrombocytopenia

Rukhsana Aslam; Yu Hu; Simon Gebremeskel; George B. Segel; Edwin R. Speck; Li Guo; Michael Kim; Heyu Ni; John Freedman; John W. Semple

Immune thrombocytopenia (ITP) is a bleeding disorder in which antibodies and/or T cells lead to enhanced peripheral platelet destruction and reduced bone marrow platelet production. Several reports have observed that ITP is associated with a peripheral deficiency of tolerance-inducing CD4+CD25+FoxP3+ T regulatory cells (Tregs). Using a murine model of ITP, we analyzed Tregs in the spleen and thymus. CD61 knockout mice were immunized against wild-type (CD61+) platelets, and their splenocytes were transferred into severe combined immunodeficient (SCID) mice. Compared with SCID mice receiving naive splenocytes, within 2 weeks after transfer, the ITP SCID mice became thrombocytopenic (< 200 × 10(9) platelets/L) and had increased serum anti-CD61 antibodies. The quantity of thymic Tregs by 2 weeks after transfer was significantly elevated, whereas Tregs in the spleens were significantly reduced. Treatment of the ITP mice with 2 g/kg intravenous immunoglobulin raised the platelet counts, reduced antibody production, and normalized the thymic and splenic Treg populations. Compared with thymocytes from ITP mice treated with intravenous immunoglobulin, thymocytes from untreated ITP mice delayed the onset of ITP when administered before engraftment with immune splenocytes. These results suggest that ITP in mice is associated with a peripheral Treg deficiency because of thymic retention and therapy normalizes the Tregs.


Transfusion | 2008

Transfusion-related immunomodulation by platelets is dependent on their expression of MHC Class I molecules and is independent of white cells

Rukhsana Aslam; Edwin R. Speck; Michael Kim; John Freedman; John W. Semple

BACKGROUND: Transfusion‐related immunomodulation (TRIM) has been correlated with the presence of white cells (WBCs) in blood transfusions, but the role of components such as platelets (PLTs) in mediating TRIM has not been extensively examined. We designed a murine PLT transfusion model to study whether leukoreduced PLTs mediate TRIM effects.


Blood | 2010

Recipient T lymphocytes modulate the severity of antibody-mediated transfusion-related acute lung injury

Yoke Lin Fung; Michael Kim; Arata Tabuchi; Rukhsana Aslam; Edwin R. Speck; Leola Chow; Wolfgang M. Kuebler; John Freedman; John W. Semple

Transfusion-related acute lung injury (TRALI) is a serious complication of transfusion and has been ranked as one of the leading causes of transfusion-related fatalities. Nonetheless, many details of the immunopathogenesis of TRALI, particularly with respect to recipient factors are unknown. We used a murine model of antibody-mediated TRALI in an attempt to understand the role that recipient lymphocytes might play in TRALI reactions. Intravenous injection of an IgG2a antimurine major histocompatibility complex class I antibody (34-1-2s) into BALB/c mice induced moderate hypothermia and pulmonary granulocyte accumulation but no pulmonary edema nor mortality. In contrast, 34-1-2s injections into mice with severe combined immunodeficiency caused severe hypothermia, severe pulmonary edema, and approximately 40% mortality indicating a critical role for T and B lymphocytes in suppressing TRALI reactions. Adoptive transfer of purified CD8(+) T lymphocytes or CD4(+) T cells but not CD19(+) B cells into the severe combined immunodeficiency mice alleviated the antibody-induced hypothermia, lung damage, and mortality, suggesting that T lymphocytes were responsible for the protective effect. Taken together, these results suggest that recipient T lymphocytes play a significant role in suppressing antibody-mediated TRALI reactions. They identify a potentially new recipient mechanism that controls the severity of TRALI reactions.


Transfusion | 2007

Platelet and red blood cell phagocytosis kinetics are differentially controlled by phosphatase activity within mononuclear cells

Rukhsana Aslam; Michael Kim; Edwin R. Speck; Arjuna Contram Seetanah; Steven Molinski; John Freedman; John W. Semple

BACKGROUND: Anti‐D treatment is effective in increasing platelet (PLT) counts in patients with autoimmune thrombocytopenic purpura (AITP); however, the exact mechanism of action is unknown. Previous results have suggested that anti‐D–coated red blood cells (RBCs) affect reticuloendothelial system phagocytosis by stimulating agents (e.g., reactive oxygen species) that alter signaling pathways within the phagocyte. To address this, a flow cytometric assay was used to compare the kinetics and signaling pathways responsible for opsonized PLT and RBC phagocytosis.


Blood | 2017

T regulatory cells and dendritic cells protect against transfusion-related acute lung injury via IL-10

Rick Kapur; Michael Kim; Rukhsana Aslam; Mark J. McVey; Arata Tabuchi; Jonathan Liu; Yuan Li; Shanjeevan Shanmugabhavananthan; Edwin R. Speck; Anne Zufferey; George M. Yousef; Haibo Zhang; Matthew T. Rondina; Andrew S. Weyrich; Leendert Porcelijn; Wolfgang M. Kuebler; Arthur S. Slutsky; John W. Semple

Transfusion-related acute lung injury (TRALI) is the leading cause of transfusion-related fatalities and is characterized by acute respiratory distress following blood transfusion. Donor antibodies are frequently involved; however, the pathogenesis and protective mechanisms in the recipient are poorly understood, and specific therapies are lacking. Using newly developed murine TRALI models based on injection of anti-major histocompatibility complex class I antibodies, we found CD4+CD25+FoxP3+ T regulatory cells (Tregs) and CD11c+ dendritic cells (DCs) to be critical effectors that protect against TRALI. Treg or DC depletion in vivo resulted in aggravated antibody-mediated acute lung injury within 90 minutes with 60% mortality upon DC depletion. In addition, resistance to antibody-mediated TRALI was associated with increased interleukin-10 (IL-10) levels, and IL-10 levels were found to be decreased in mice suffering from TRALI. Importantly, IL-10 injection completely prevented and rescued the development of TRALI in mice and may prove to be a promising new therapeutic approach for alleviating lung injury in this serious complication of transfusion.


Blood | 2014

Allogeneic platelet transfusions prevent murine T-cell-mediated immune thrombocytopenia

Li Guo; Lei Yang; Edwin R. Speck; Rukhsana Aslam; Michael Kim; Christopher G. J. McKenzie; Alan H. Lazarus; Heyu Ni; Ming Hou; John Freedman; John W. Semple

Platelet transfusions are life-saving treatments for many patients with thrombocytopenia; however, their use is generally discouraged in the autoimmune disorder known as immune thrombocytopenia (ITP). We examined whether allogeneic platelet major histocompatibility complex (MHC) class I transfusions affected antiplatelet CD61-induced ITP. BALB/c CD61 knockout mice (CD61(-)/H-2(d)) were immunized against platelets from wild-type syngeneic BALB/c (CD61(+)/H-2(d)), allogeneic C57BL/6 (CD61(+)/H-2(b)), or C57BL/6 CD61 KO (CD61(-)/H-2(b)) mice, and their splenocytes were transferred into severe combined immunodeficient (SCID) mice to induce ITP. When nondepleted splenocytes were transferred to induce antibody-mediated ITP, both CD61(+) platelet immunizations generated immunity that caused thrombocytopenia independently of allogeneic MHC molecules. In contrast, when B-cell-depleted splenocytes were transferred to induce T-cell-mediated ITP, transfer of allogeneic MHC-immunized splenocytes completely prevented CD61-induced ITP development. In addition, allogeneic platelet transfusions into SCID mice with established CD61-induced ITP rescued the thrombocytopenia. Compared with thrombocytopenic mice, bone marrow histology in the rescued mice showed normalized megakaryocyte morphology, and in vitro CD61-specific T-cell cytotoxicity was significantly suppressed. These results indicate that antibody-mediated ITP is resistant to allogeneic platelet transfusions, while the T-cell-mediated form of the disease is susceptible, suggesting that transfusion therapy may be beneficial in antibody-negative ITP.


Journal of Oral and Maxillofacial Surgery | 2008

Evaluation of platelet gel characteristics using thrombin produced by the thrombin processing device: a comparative study.

Elisabeth Semple; Edwin R. Speck; Rukhsana Aslam; Michael Kim; Vijay Kumar; John W. Semple

PURPOSE Autologous platelet gels can be prepared using the patients own platelet-rich plasma and thrombin produced by the Thrombin Processing Device (Thermogenesis Corp, Rancho Cordova, CA). As the Thrombin Processing Device thrombin contains a residual amount of ethanol, the purpose of this study was to investigate the effect of the Thrombin Processing Device thrombin on growth factor release from platelet gels, and the effect on cell viability and cell proliferation. MATERIALS AND METHODS Platelet gels were prepared using Thrombin Processing Device-produced human thrombin at platelet-rich plasma to thrombin ratios of 3.3 to 1 and 7 to 1. Commercially available bovine thrombin was used as control. The content of the growth factors, platelet-derived growth factor beta polypeptide, and transforming growth factor beta, were assessed in both the clot and supernatant. The influence of different concentrations of ethanol on cell viability was assessed by flow cytometry and cell proliferation was assessed by (3)H-thymidine incorporation. RESULTS Using a ratio of 3.3 to 1, the supernatant of the platelet gel produced with Thrombin Processing Device thrombin had a lower growth factor content compared with bovine thrombin but was similar when prepared using a ratio of platelet-rich plasma to thrombin of 7 to 1. Supernatants from the platelet gels did not affect the viability of human macrophage line cells or a fibroblast cell line. When the different platelet gels or their supernatants were tested for their ability to stimulate cell proliferation, similar rates of proliferation were observed. CONCLUSIONS These data suggest that residual ethanol in the Thrombin Processing Device-produced thrombin does not affect any of the tested parameters and has similar characteristics as commercially available bovine thrombin.


British Journal of Haematology | 2016

Splenic lymphocyte subtypes in immune thrombocytopenia: increased presence of a subtype of B‐regulatory cells

Rukhsana Aslam; George B. Segel; Richard Burack; Stephen A. Spence; Edward R. Speck; Li Guo; John W. Semple

in Japanese patients with Upshaw-Schulman syndrome. Blood, 103, 1305–1310. Park, H.W., Oh, D., Kim, N., Cho, H.Y., Moon, K.C., Chae, J.H., Ahn, H.S., Choi, Y. & Cheong, H.I. (2008) Congenital thrombotic thrombocytopenic purpura associated with unilateral moyamoya disease. Pediatric Nephrology, 23, 1555–1558. Perez-Rodriguez, A., Loures, E., Rodriguez-Trillo, A., Costa-Pinto, J., Garcia-Rivero, A., Batlle-Lopez, A., Batlle, J. & Lopez-Fernandez, M.F. (2014a) Inherited ADAMTS13 deficiency (Upshaw-Schulman syndrome): a short review. Thrombosis Research, 134, 1171–1175. Perez-Rodriguez, A., Batlle-Lopez, A., Blanco, R., Varela, I., Leon, J., Delgado, M.D., Loures, E., Rodriguez-Trillo, A., Garcia-Rivero, A., CostaPinto, J., Batlle, J. & Lopez-Fernandez, M.F. (2014b) A novel mutation in ADAMTS13 of a child with Upshaw-Schulman Syndrome. Thrombosis and Haemostasis, 112, 1065–1068. Zhang, P., Pan, W., Rux, A.H., Sachais, B.S. & Zheng, X.L. (2007) The cooperative activity between the carboxyl-terminal TSP1 repeats and the CUB domains of ADAMTS13 is crucial for recognition of von Willebrand factor under flow. Blood, 110, 1887–1894.

Collaboration


Dive into the Rukhsana Aslam's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael Kim

St. Michael's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heyu Ni

St. Michael's Hospital

View shared research outputs
Top Co-Authors

Avatar

Li Guo

St. Michael's Hospital

View shared research outputs
Top Co-Authors

Avatar

Rick Kapur

St. Michael's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge