Network


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

Hotspot


Dive into the research topics where Jocelyn A. Schroeder is active.

Publication


Featured researches published by Jocelyn A. Schroeder.


Journal of Thrombosis and Haemostasis | 2012

Lentivirus‐mediated platelet gene therapy of murine hemophilia A with pre‐existing anti‐factor VIII immunity

Erin L. Kuether; Jocelyn A. Schroeder; Scot A. Fahs; Brian C. Cooley; Yingyu Chen; Robert R. Montgomery; David A. Wilcox; Qizhen Shi

Summary.  Background:  The development of inhibitory antibodies, referred to as inhibitors, against exogenous factor VIII in a significant subset of patients with hemophilia A remains a persistent challenge to the efficacy of protein replacement therapy. Our previous studies using the transgenic approach provided proof‐of‐principle that platelet‐specific expression could be successful in treating hemophilia A in the presence of inhibitory antibodies.


Molecular Therapy | 2014

Platelet Gene Therapy by Lentiviral Gene Delivery to Hematopoietic Stem Cells Restores Hemostasis and Induces Humoral Immune Tolerance in FIXnull Mice

Yingyu Chen; Jocelyn A. Schroeder; Erin L. Kuether; Guowei Zhang; Qizhen Shi

Here, we developed a clinically translatable platelet gene therapy approach for hemophilia B. Platelet-targeted FIX (2bF9) expression was introduced by transplantation of hematopoietic stem cells (HSCs) transduced with 2bF9 lentivirus (LV). Sustained therapeutic levels of platelet-FIX expression were obtained in FIX(null) mice that received 2bF9 LV-transduced HSCs. Approximately 6-39% of the platelets expressed FIX in the transduced recipients, which was sufficient to rescue the bleeding diathesis in FIX(null) mice in tail clipping models. Sequential bone marrow transplantation demonstrated that platelet-FIX expression in the secondary recipients was sustained, leading to phenotypic correction. Notably, none of the transduced recipients developed anti-FIX antibodies after platelet gene therapy. Only one of the nine recipients developed a low titer of inhibitory antibodies (1.6 BU/ml) after challenge with rhFIX. These data suggest that platelet gene therapy can not only restore hemostasis but also induce immune tolerance in hemophilia B mice, indicating that this approach may be a promising strategy for gene therapy of hemophilia B in humans.Here, we developed a clinically translatable platelet gene therapy approach for hemophilia B. Platelet-targeted FIX (2bF9) expression was introduced by transplantation of hematopoietic stem cells (HSCs) transduced with 2bF9 lentivirus (LV). Sustained therapeutic levels of platelet-FIX expression were obtained in FIXnull mice that received 2bF9 LV-transduced HSCs. Approximately 6-39% of the platelets expressed FIX in the transduced recipients, which was sufficient to rescue the bleeding diathesis in FIXnull mice in tail clipping models. Sequential bone marrow transplantation demonstrated that platelet-FIX expression in the secondary recipients was sustained, leading to phenotypic correction. Notably, none of the transduced recipients developed anti-FIX antibodies after platelet gene therapy. Only one of the nine recipients developed a low titer of inhibitory antibodies (1.6 BU/ml) after challenge with rhFIX. These data suggest that platelet gene therapy can not only restore hemostasis but also induce immune tolerance in hemophilia B mice, indicating that this approach may be a promising strategy for gene therapy of hemophilia B in humans.


Blood | 2014

Platelet gene therapy corrects the hemophilic phenotype in immunocompromised hemophilia A mice transplanted with genetically manipulated human cord blood stem cells

Qizhen Shi; Erin L. Kuether; Yingyu Chen; Jocelyn A. Schroeder; Scot A. Fahs; Robert R. Montgomery

Our previous studies have demonstrated that platelet FVIII (2bF8) gene therapy can improve hemostasis in hemophilia A mice, even in the presence of inhibitory antibodies, but none of our studies has targeted human cells. Here, we evaluated the feasibility for lentivirus (LV)-mediated human platelet gene therapy of hemophilia A. Human platelet FVIII expression was introduced by 2bF8LV-mediated transduction of human cord blood (hCB) CD34(+) cells followed by xenotransplantation into immunocompromised NSG mice or NSG mice in an FVIII(null) background (NSGF8KO). Platelet FVIII was detected in all recipients that received 2bF8LV-transduced hCB cells as long as human platelet chimerism persisted. All NSGF8KO recipients (n = 7) that received 2bF8LV-transduced hCB cells survived tail clipping if animals had greater than 2% of platelets derived from 2bF8LV-transduced hCB cells, whereas 5 of 7 survived when human platelets were 0.3% to 2%. Whole blood clotting time analysis confirmed that hemostasis was improved in NSGF8KO mice that received 2bF8LV-transduced hCB cells. We demonstrate, for the first time, the feasibility of 2bF8LV gene delivery to human hematopoietic stem cells to introduce FVIII expression in human platelets and that human platelet-derived FVIII can improve hemostasis in hemophilia A.


Journal of Thrombosis and Haemostasis | 2014

In vivo enrichment of genetically manipulated platelets corrects the murine hemophilic phenotype and induces immune tolerance even using a low multiplicity of infection

Jocelyn A. Schroeder; Yingyu Chen; Juan Fang; David A. Wilcox; Qizhen Shi

Our previous studies have demonstrated that platelet‐specific gene delivery to hematopoietic stem cells can induce sustained therapeutic levels of platelet factor VIII (FVIII) expression in mice with hemophilia A.


Molecular Therapy | 2012

Correction of Murine Bernard–Soulier Syndrome by Lentivirus-mediated Gene Therapy

Sachiko Kanaji; Erin L. Kuether; Scot A. Fahs; Jocelyn A. Schroeder; Jerry Ware; Robert R. Montgomery; Qizhen Shi

Bernard-Soulier syndrome (BSS) is an inherited bleeding disorder caused by a defect in the platelet glycoprotein (GP) Ib-IX-V complex. The main treatment for BSS is platelet transfusion but it is often limited to severe bleeding episodes or surgical interventions due to the risk of alloimmunization. We have previously reported successful expression of human GPIbα (hGPIbα) in human megakaryocytes using a lentiviral vector (LV) encoding human GP1BA under control of the platelet-specific integrin αIIb promoter (2bIbα). In this study, we examined the efficacy of this strategy for the gene therapy of BSS using GPIbα(null) as a murine model of BSS. GPIbα(null) hematopoietic stem cells (HSC) transduced with 2bIbα LV were transplanted into lethally irradiated GPIbα(null) littermates. Therapeutic levels of hGPIbα expression were achieved that corrected the tail bleeding time and improved the macrothrombocytopenia. Sequential bone marrow (BM) transplants showed sustained expression of hGPIbα with similar phenotypic correction. Antibody response to hGPIbα was documented in 1 of 17 total recipient mice but was tolerated without any further treatment. These results demonstrate that lentivirus-mediated gene transfer can provide sustained phenotypic correction of murine BSS, indicating that this approach may be a promising strategy for gene therapy of BSS patients.


Journal of Thrombosis and Haemostasis | 2012

Factor VIII inhibitors: von Willebrand factor makes a difference in vitro and in vivo

Qizhen Shi; Erin L. Kuether; Jocelyn A. Schroeder; Crystal L. Perry; Scot A. Fahs; J. Cox Gill; Robert R. Montgomery

Summary.  Background:  The important association between von Willebrand factor (VWF) and factor VIII (FVIII) has been investigated for decades, but the effect of VWF on the reactivity of FVIII inhibitory antibodies, referred to as inhibitors, is still controversial.


Haemophilia | 2012

Intravascular recovery of VWF and FVIII following intraperitoneal injection and differences from intravenous and subcutaneous injection in mice.

Qizhen Shi; Erin L. Kuether; Jocelyn A. Schroeder; Scot A. Fahs; Robert R. Montgomery

Summary.  Intravenous infusion studies in humans suggest that both von Willebrand factor (VWF) and factor VIII (FVIII) remain intravascular in contrast to other coagulation proteins. We explored whether infusion of VWF and FVIII by either intraperitoneal (i.p.) or subcutaneous (s.c.) injection would result in efficient absorption of these large proteins into the vascular circulation. FVIIInull or VWFnull mice were infused with plasma‐derived or recombinant VWF and/or FVIII by i.p., s.c., or intravenous (i.v.) injection. Both VWF and FVIII were absorbed into the blood circulation after i.p. injection with a peak between 2 and 4 h at levels similar to those observed in mice infused intravenously. In contrast, neither VWF nor FVIII was detected in the plasma following s.c. injection. Although i.v. injection achieved peak plasma levels quickly, both human VWF and FVIII rapidly decreased during the first 2 h following i.v. injection. Following both i.v. and i.p. infusion of VWF, the multimeric structure of circulating VWF was similar to that observed in the infusate. These results demonstrate that both VWF and FVIII can be efficiently absorbed into the blood circulation following i.p., but not s.c. injection, indicating that i.p. administration could be an alternative route for VWF or FVIII infusion.


Journal of Thrombosis and Haemostasis | 2015

The important role of von Willebrand factor in platelet‐derived FVIII gene therapy for murine hemophilia A in the presence of inhibitory antibodies

Qizhen Shi; Jocelyn A. Schroeder; Erin L. Kuether; Robert R. Montgomery

Our previous studies have demonstrated that targeting FVIII expression to platelets results in FVIII storage together with von Willebrand factor (VWF) in platelet α‐granules and that platelet‐derived FVIII (2bF8) corrects the murine hemophilia A phenotype even in the presence of high‐titer anti‐FVIII inhibitory antibodies (inhibitors).


Blood | 2016

The immunogenicity of platelet-derived FVIII in hemophilia A mice with or without preexisting anti-FVIII immunity

Yingyu Chen; Jocelyn A. Schroeder; Juan Chen; Xiaofeng Luo; Christina K. Baumgartner; Robert R. Montgomery; Jianda Hu; Qizhen Shi

Evidence shows that factor VIII (FVIII) ectopically expressed in platelets (2bF8) is therapeutic in FVIII(null) mice even with anti-FVIII inhibitory antibodies (inhibitors). If current efforts to generate platelets in vitro succeed, genetically manipulated platelets containing FVIII may be used therapeutically in hemophilia A patients with inhibitors. One important concern is the immunogenicity of platelet-derived FVIII. To address this concern, we infused 2bF8 transgenic (2bF8(Tg)) platelets into naïve FVIII(null) mice weekly for 8 weeks. No anti-FVIII antibodies were detected in the infused animals during the study course. We then explored whether platelet-derived FVIII is immunogenic in FVIII(null) mice with inhibitors. The 2bF8(Tg) platelets were transfused into rhF8-primed FVIII(null) mice, resulting in no augmentation of anti-FVIII antibodies. To investigate whether preconditioning affects the immune response, animals were sublethally irradiated and subsequently transfused with 2bF8(Tg) platelets. No anti-FVIII antibodies were detected in the recipients after platelet infusions. Following further challenge with rhF8, the inhibitor titer in this group was significantly lower than in naïve FVIII(null) mice utilizing the same immunization protocol. Thus, our data demonstrate that infusion of platelets containing FVIII triggers neither primary nor memory anti-FVIII immune response in FVIII(null) mice and that sublethal irradiation plus 2bF8(Tg) platelet infusion suppresses anti-FVIII immune response in FVIII(null) mice.


Journal of Thrombosis and Haemostasis | 2017

Immune tolerance induced by platelet‐targeted factor VIII gene therapy in hemophilia A mice is CD4 T cell mediated

Yingyu Chen; Xiaofeng Luo; Jocelyn A. Schroeder; Juan Chen; Christina K. Baumgartner; Jianda Hu; Qizhen Shi

Essentials The immune response is a significant concern in gene therapy. Platelet‐targeted gene therapy can restore hemostasis and induce immune tolerance. CD4 T cell compartment is tolerized after platelet gene therapy. Preconditioning regimen affects immune tolerance induction in platelet gene therapy.

Collaboration


Dive into the Jocelyn A. Schroeder's collaboration.

Top Co-Authors

Avatar

Qizhen Shi

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Robert R. Montgomery

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Erin L. Kuether

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Juan Chen

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Scot A. Fahs

Gulf Coast Regional Blood Center

View shared research outputs
Top Co-Authors

Avatar

Yingyu Chen

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Jianda Hu

Fujian Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiaofeng Luo

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

David A. Wilcox

Medical College of Wisconsin

View shared research outputs
Researchain Logo
Decentralizing Knowledge