Network


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

Hotspot


Dive into the research topics where Christopher L. Morris is active.

Publication


Featured researches published by Christopher L. Morris.


Journal of Biological Chemistry | 2016

Transcriptional Regulation of JARID1B/KDM5B Histone Demethylase by Ikaros, Histone Deacetylase 1 (HDAC1), and Casein Kinase 2 (CK2) in B-cell Acute Lymphoblastic Leukemia

Haijun Wang; Chunhua Song; Yali Ding; Xiaokang Pan; Zheng Ge; Bi-Hua Tan; Chandrika Gowda; Mansi Sachdev; Sunil Muthusami; Hongsheng Ouyang; Liangxue Lai; Olivia L. Francis; Christopher L. Morris; Hisham Abdel-Azim; Glenn Dorsam; Meixian Xiang; Kimberly J. Payne; Sinisa Dovat

Impaired function of the Ikaros (IKZF1) protein is associated with the development of high-risk B-cell precursor acute lymphoblastic leukemia (B-ALL). The mechanisms of Ikaros tumor suppressor activity in leukemia are unknown. Ikaros binds to the upstream regulatory elements of its target genes and regulates their transcription via chromatin remodeling. Here, we report that Ikaros represses transcription of the histone H3K4 demethylase, JARID1B (KDM5B). Transcriptional repression of JARID1B is associated with increased global levels of H3K4 trimethylation. Ikaros-mediated repression of JARID1B is dependent on the activity of the histone deacetylase, HDAC1, which binds to the upstream regulatory element of JARID1B in complex with Ikaros. In leukemia, JARID1B is overexpressed, and its inhibition results in cellular growth arrest. Ikaros-mediated repression of JARID1B in leukemia is impaired by pro-oncogenic casein kinase 2 (CK2). Inhibition of CK2 results in increased binding of the Ikaros-HDAC1 complex to the promoter of JARID1B, with increased formation of trimethylated histone H3 lysine 27 and decreased histone H3 Lys-9 acetylation. In cases of high-risk B-ALL that carry deletion of one Ikaros (IKZF1) allele, targeted inhibition of CK2 restores Ikaros binding to the JARID1B promoter and repression of JARID1B. In summary, the presented data suggest a mechanism through which Ikaros and HDAC1 regulate the epigenetic signature in leukemia: via regulation of JARID1B transcription. The presented data identify JARID1B as a novel therapeutic target in B-ALL and provide a rationale for the use of CK2 inhibitors in the treatment of high-risk B-ALL.


Haematologica | 2016

A novel xenograft model to study the role of TSLP-induced CRLF2 signals in normal and malignant human B lymphopoiesis

Olivia L. Francis; Terry-Ann Milford; Shannalee R. Martinez; Ineavely Baez; Jacqueline S. Coats; Karina Mayagoitia; Katherine Concepcion; Elizabeth Ginelli; Cornelia Beldiman; Abigail Benitez; Abby J. Weldon; Keshav Arogyaswamy; Parveen Shiraz; Ross Fisher; Christopher L. Morris; Xiao-Bing Zhang; Valeri Filippov; Ben Van Handel; Zheng Ge; Chunhua Song; Sinisa Dovat; Ruijun Jeanna Su; Kimberly J. Payne

Thymic stromal lymphopoietin (TSLP) stimulates in vitro proliferation of human fetal B-cell precursors. However, its in vivo role during normal human B lymphopoiesis is unknown. Genetic alterations that cause overexpression of its receptor component, cytokine receptor-like factor 2 (CRLF2), lead to high-risk B-cell acute lymphoblastic leukemia implicating this signaling pathway in leukemogenesis. We show that mouse thymic stromal lymphopoietin does not stimulate the downstream pathways (JAK/STAT5 and PI3K/AKT/mTOR) activated by the human cytokine in primary high-risk leukemia with overexpression of the receptor component. Thus, the utility of classic patient-derived xenografts for in vivo studies of this pathway is limited. We engineered xenograft mice to produce human thymic stromal lymphopoietin (+T mice) by injection with stromal cells transduced to express the cytokine. Control (−T) mice were produced using stroma transduced with control vector. Normal levels of human thymic stromal lymphopoietin were achieved in sera of +T mice, but were undetectable in −T mice. Patient-derived xenografts generated from +T as compared to −T mice showed a 3–6-fold increase in normal human B-cell precursors that was maintained through later stages of B-cell development. Gene expression profiles in high-risk B-cell acute lymphoblastic leukemia expanded in +T mice indicate increased mTOR pathway activation and are more similar to the original patient sample than those from −T mice. +T/−T xenografts provide a novel pre-clinical model for understanding this pathway in B lymphopoiesis and identifying treatments for high-risk B-cell acute lymphoblastic leukemia with overexpression of cytokine-like factor receptor 2.


European Journal of Immunology | 2016

TSLP or IL‐7 provide an IL‐7Rα signal that is critical for human B lymphopoiesis

Terry-Ann Milford; Ruijun Jeanna Su; Olivia L. Francis; Ineavely Baez; Shannalee R. Martinez; Jacqueline S. Coats; Abby J. Weldon; Milcris N. Calderon; Michael C. Nwosu; Allen R. Botimer; Batul T. Suterwala; Xiao-Bing Zhang; Christopher L. Morris; David J. Weldon; Sinisa Dovat; Kimberly J. Payne

Thymic stromal lymphopoietin (TSLP) and IL‐7 are cytokines that signal via the IL‐7 receptor alpha (IL‐7Rα) to exert both overlapping and unique functions during early stages of mouse B‐cell development. In human B lymphopoiesis, the requirement for IL‐7Rα signaling is controversial and the roles of IL‐7 and TSLP are less clear. Here, we evaluated human B‐cell production using novel in vitro and xenograft models of human B‐cell development that provide selective IL‐7 and human TSLP (hTSLP) stimulation. We show that in vitro human B‐cell production is almost completely blocked in the absence of IL‐7Rα stimulation, and that either TSLP or IL‐7 can provide a signal critical for the production and proliferation of human CD19+ PAX5+ pro‐B cells. Analysis of primary human bone marrow stromal cells shows that they express both IL‐7 and TSLP, providing an in vivo source of these cytokines. We further show that the in vivo production of human pro‐B cells under the influence of mouse IL‐7 in a xenograft scenario is reduced by anti‐IL‐7 neutralizing antibodies, and that this loss can be restored by hTSLP at physiological levels. These data establish the importance of IL‐7Rα mediated signals for normal human B‐cell production.


Cancer Research | 2016

Abstract A07: A novel patient-derived xenograft model to define the role of TSLP-induced CRLF2 signals and identify therapies for Ph-like B-ALL

Olivia L. Francis; Terry-Ann Milford; Ineavely Baez; Jacqueline S. Coats; Christopher L. Morris; Ross Fisher; Ben Van Handel; Ruijun Su; Batul T. Suterwala; Muhammad Omair Kamal; Shadi Farzin Gohar; Sinisa Dovat; Kimberly J. Payne

A subset of high-risk B cell acute lymphoblastic leukemia (ALL) shows a gene expression profile similar to Philadelphia chromosome positive (Ph+) ALL and has been described as Ph-like ALL. Approximately 50% of Ph-like B-ALL is characterized by genetic alterations leading to overexpression of CRLF2 (CRLF2 B-ALL). CRLF2 B-ALL occurs 5 times more often in Hispanic and Native American children than others and is prevalent in adolescents and young adults. Biologically, CRLF2 acts as a receptor component for the cytokine, TSLP, which induces JAK2-STAT5 and PI3/AKT/mTOR pathway activation downstream of binding to CRLF2. While activating JAK mutations are associated with CRLF2 B-ALL, over half of CRLF2 B-ALL lack such mutations. Our data show that primary human bone marrow (BM) stromal cells express TSLP. Thus TSLP is present in the tumor microenvironment to provide TSLP-induced CRLF2 signals that could play a role in the initiation, maintenance and/or progression of CRLF2 B-ALL. Consistent with this, TSLP has been reported to increase in vitro production of human fetal B cell precursors. However studies of TSLP in B lymphopoiesis have been conducted almost exclusively in mice which show low homology (~40%) with respect to human TSLP and CRLF2. Further, phospho flow cytometry assays show that human, but not mouse TSLP activates CRLF2 signals in primary human CRLF2 B-ALL cells and cell lines as indicated by increased pSTAT5, pAKT and pS6. These data indicate that the mouse TSLP present in classic patient derived xenograft models (PDX) does not produce the TSLP-induced CRLF2 signals present in the patient. To address this challenge we engineered PDX mice to produce human TSLP (hTSLP) by transplanting them with stromal cells transduced to express hTSLP (+T mice). Control (T) mice were produced by transplantation with stroma transduced with a control vector. Supernatant from engineered +T stroma, but not T stroma, induced JAK/STAT5 and PI3K/AKT/mTOR pathway activation in human CRLF2 B-ALL cells. ELISA assays showed that serum levels of hTSLP in mice was proportional to numbers of stromal cells injected at weekly time points. Normal human serum levels of hTSLP (12-32 pg/ml) could be achieved in +T mice, while hTSLP was undetectable in T mice. Because TSLP has been shown to increase in vitro production of human B cell precursors, we evaluated the in vivo functionality of our model by comparing the production of normal B cell precursors in the BM of +T and T PDX mice generated with human umbilical cord blood CD34+ cells. Data from 3 different cord blood donors showed that production of B cell precursors is 3-5 fold increased in +T as compared to T mice. TSLP-induced increases were specific to B lineage cells, initiated in the earliest CD19+ B cell precursors, and maintained through later stages of B cell development. Next we evaluate the in vivo functionality of our model using primary CRLF2 B-ALL leukemia cells. Human CRLF2 B-ALL cells were isolated from the BM of PDX mice and whole genome microarray was performed. Evaluation of microarray data by Gene Set Enrichment Analysis (GSEA) and Ingenuity Pathway Analysis showed that genes downstream of mTOR pathway activation were upregulated in +T as compared to T PDX mice, confirming hTSLP activity in the +T PDX mice. To determine whether +T PDX mice provide a preclinical model of B-ALL that more closely mirrors patients than T PDX mice, we compared RNAseq gene expression profiles of leukemia cells from +T and T PDX mice to that from original patient sample. The gene expression pattern in +T mice was significantly closer to primary patient sample than that from T mice. The +T and T PDX mice described here provide a novel preclinical model for studying the role of TSLP in the initiation, progression and maintenance of CRLF2 B-ALL and for evaluating drug efficacy in an in vivo model that more closely mirrors the in vivo environment present in patients. Citation Format: Olivia L. Francis, Terry-Ann Milford, Ineavely Baez, Jacqueline S. Coats, Christopher L. Morris, Ross Fisher, Ben Van Handel, Ruijun Su, Batul Suterwala, Muhammad Kamal, Shadi Farzin Gohar, Sinisa Dovat, Kimberly J. Payne. A novel patient-derived xenograft model to define the role of TSLP-induced CRLF2 signals and identify therapies for Ph-like B-ALL. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Pediatric Cancer Research: From Mechanisms and Models to Treatment and Survivorship; 2015 Nov 9-12; Fort Lauderdale, FL. Philadelphia (PA): AACR; Cancer Res 2016;76(5 Suppl):Abstract nr A07.


Cancer Research | 2015

Abstract 3295: A novel patient-derived xenograft model for evaluating the role of TSLP in CRLF2 B-ALL

Olivia L. Francis; Parveen Shiraz; Terry-Ann Milford; Ineavely Baez; Jacqueline S. Coats; Karina Mayagoitia; Elizabeth Ginelli; Katherine R. Salcedo-Concepcion; Shannalee R. Martinez; Xiao-Bing Zhang; Valeri Filippov; Ruijun Jeanna Su; Ross Fisher; Christopher L. Morris; Sinisa Dovat; Kimberly J. Payne

B-cell acute lymphoblastic leukemia (B-ALL) with genetic alterations leading to overexpression of CRLF2 (CRLF2 B-ALL) is associated with poor outcomes. CRLF2 B-ALL is 5 times more common in Hispanic children than others making it a significant biological component of pediatric cancer health disparities. CRLF2 is a component of the receptor complex that is activated by the cytokine, TSLP. Receptor signaling induces Jak/STAT5 and PI3/AKT/mTOR pathway activation and plays a role in the proliferation and differentiation B cell precursors. We found that primary human bone marrow (BM) stroma express TSLP providing an in vivo source of TSLP to stimulate CRLF2 B-ALL cells. Our goal was to develop patient-derived xenograft (PDX) models of CRLF2 B-ALL for studies to understand disease mechanisms and identify therapies to treat CRLF2 B-ALL and reduce the health disparities for Hispanic children with this disease. PDX models are possible because many cytokines produced in the mouse show cross species activity on human cells. However, available data suggests that mouse TSLP does not activate human CRLF2-mediated signals. Using phospho flow cytometry we show that mouse TSLP was unable to induce increases in pSTAT5, pAKT and pS6 observed in CRLF2 B-ALL cells stimulated with human TSLP. We developed a human TSLP +/- PDX model system by transplanting immune deficient NSG mice with HS-27 stroma transduced to express human hTSLP (hTSLP+ mice) or with control vector (hTSLP- mice). Human TSLP was present at normal human serum levels in hTSLP+ mice but undetectable in hTSLP- mice. To identify genes targeted by TSLP in CRLF2 B-ALL and verify pathway activation, we transplanted primary leukemia cells from a Hispanic patient into hTSLP+ and hTSLP- mice. Whole genome microarray was performed on CRLF2 B-ALL cells isolated from the BM of the hTSLP+ and hTSLP- PDX mice. Microarray identified 280 genes that are upregulated and 281 genes that are downregulated in vivo in leukemia cells from hTSLP+ as compared to hTSLP- PDX mice. Evaluation of microarray data by Gene Set Enrichment Analysis (GSEA) and Ingenuity Pathway Analysis showed that genes downstream of mTOR pathway activation were upregulated in hTSLP+ as compared to hTSLP- mice, confirming hTSLP activity in the hTSLP+ PDX mice. Our next question was whether cells expanded in hTSLP+ vs. hTSLP- mice would exhibit changes in their ability to respond to TSLP. When we subjected PDX-expanded primary CRLF2 B-ALL cells to ex vivo TSLP stimulation ∼1/3 fewer gene targets were up- and downregulated in the leukemia cells expanded in hTSLP- mice as compared to cells from hTSLP+ mice. This suggests that CRLF2 B-ALL cells expanded in xenograft without TSLP lose some of their ability to respond to TSLP. The hTSLP+ CRLF2 B-ALL PDX mice described here provide a novel preclinical model for studying disease mechanisms and identifying therapies to target signaling pathways activated by TSLP in CRLF2 B-ALL and reduce cancer health disparities for this disease. Citation Format: Olivia L. Francis, Parveen Shiraz, Terry-Ann Milford, Ineavely Baez, Jacqueline S. Coats, Karina Mayagoitia, Elizabeth Ginelli, Katherine R. Salcedo-Concepcion, Shannalee Martinez, Xiaobing Zhang, Valeri Filippov, Ruijun J. Su, Ross Fisher, Christopher L. Morris, Sinisa Dovat, Kimberly J. Payne. A novel patient-derived xenograft model for evaluating the role of TSLP in CRLF2 B-ALL. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 3295. doi:10.1158/1538-7445.AM2015-3295


Molecular Cancer Research | 2014

Abstract B25: A human-mouse xenograft model to evaluate therapies and study the role of TSLP-induced signals in Ph-like ALL

Ruijun Su; Francis L. Olivia; Shannalee R. Martinez; Ineavely Baez; Terry Ann Milford; Terrence Bennett; Ross Fisher; Christopher L. Morris; Sinisa Dovat; Kimberly J. Payne

While the overall survival rate for children with B cell precursor acute lymphoblastic leukemia (B-ALL) is high, a subset of children with this disease are at high risk for relapse and death. Genome-wide analysis has shown that gene expression profiles in these high-risk B-ALLs is similar to that of Philadelphia chromosome–positive ALL and these are designated Ph-like ALL. Approximately half of Ph-like ALL are characterized by genetic defects resulting in overexpression of CRLF2. CRLF2, together with the IL-7Rα, forms a receptor complex that is activated by the cytokine, TSLP. The JAK-STAT5 pathway is phosphorylated downstream of this receptor complex activation. The activating JAK mutations found in some CRLF2 B-ALL have led to speculation that TSLP stimulation is not a factor in CRLF B-ALL. In preliminary studies to address this question we evaluated the effect of TSLP on a CRLF2 B-ALL cell lines with JAK defects and which have been reported to exhibit constitutive JAK-STAT5 activation. Our data show that TSLP increases STAT5 phosphorylation in these cell lines and also in primary CRLF2 B-ALL cells. Our next step was to evaluate the role of TSLP-CRLF2 interactions in vivo in the human-mouse xenograft model. However, mouse TSLP is different from most other cytokines produced in the xenograft in that it is species-specific and does not activate the human TSLP receptor complex that includes CRLF2. Thus, traditional xenograft models do not provide the TSLP-CRLF2 interactions that we believe to be a major factor in CRLF2 B-ALL. To overcome this obstacle we engineered immune-deficient NOD/SCID IL-2Rγ null (NSG) mice to express human TSLP (hTSLP+ mice) as well as control mice that lack the TSLP cytokine (hTSLP– mice). ELISA assays show serum hTSLP levels in the hTSLP+ mice that approximate the normal range in human serum. We used this hTSLP+/- xenograft model system to study the in vivo effects of TSLP on mice transplanted with a CRLF2 B-ALL. We used this hTSLP+/– xenograft model system to evaluate the in vivo effects of TSLP on survival and proliferation of transplanted CRLF2 B-ALL cells harboring a JAK defect (MUTZ5 cell line). Mice were euthanized at 5 weeks and BM was harvested. Evaluation of BM disease by flow cytometry showed that the percentage of viable human leukemia cells in hTSLP+ mice was twice that observed in hTSLP– mice. Evaluation of cell cycle progression in human CRLF2 B-ALL cells isolated from xenograft BM showed that the percentage of cycling cells in hTSLP+ mice was 2.5 fold higher than in hTSLP– mice. When primary Ph-like ALL cells were transplanted to produce hTSLP+/– xenografts, the viable pre-B ALL cells present in the BM of hTSLP+ mice showed higher expression levels of the TSLPR components (CRLF2 and IL-7Rα) than those in the hTSLP- mice. These data provide evidence that the TSLP produced in this model is active and that it impacts primary pre-B ALL cells. Preliminary data obtained from this model suggests that TSLP provides a signal that promotes in vivo survival of CRLF2 B-ALL cells and that it may play a role in selection of leukemia clones during in vivo leukemogenesis. Microarray analysis comparing gene expression in primary CRLF2 B-ALL cells isolated from hTSLP+ and hTSLP– xenograft mice identified 565 that genes are differentially regulated (> 2 fold up or downregulated; p Citation Format: Ruijun Su, Francis L. Olivia, Shannalee R. Martinez, Ineavely Baez, Terry Ann Milford, Terrence Bennett, Ross Fisher, Christopher L. Morris, Sinisa Dovat, Kimberly J. Payne. A human-mouse xenograft model to evaluate therapies and study the role of TSLP-induced signals in Ph-like ALL. [abstract]. In: Proceedings of the AACR Special Conference: The Translational Impact of Model Organisms in Cancer; Nov 5-8, 2013; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Res 2014;12(11 Suppl):Abstract nr B25.


Cancer Research | 2014

Abstract B21: A human-mouse xenograft model to evaluate therapies and study the role of TSLP-induced signals in Ph-like ALL

Olivia L. Francis; Ruijun Su; Shannalee R. Martinez; Ineavely Baez; Terry-Ann Milford; Terrence Bennett; Ross Fisher; Christopher L. Morris; Sinisa Dovat; Kimberly J. Payne

While the overall survival rate for children with B cell precursor acute lymphoblastic leukemia (B-ALL) is high, a subset of children with this disease are at high risk for relapse and death. Genome-wide analysis has shown that gene expression profiles in these high-risk B-ALLs is similar to that of Philadelphia chromosome–positive ALL and these are designated Ph-like ALL. Approximately half of Ph-like ALL are characterized by genetic defects resulting in overexpression of CRLF2. CRLF2, together with the IL-7Rα, forms a receptor complex that is activated by the cytokine, TSLP. The JAK-STAT5 pathway is phosphorylated downstream of this receptor complex activation. The activating JAK mutations found in some CRLF2 B-ALL have led to speculation that TSLP stimulation is not a factor in CRLF B-ALL. In preliminary studies to address this question we evaluated the effect of TSLP on a CRLF2 B-ALL cell lines with JAK defects and which have been reported to exhibit constitutive JAK-STAT5 activation. Our data show that TSLP increases STAT5 phosphorylation in these cell lines and also in primary CRLF2 B-ALL cells. Our next step was to evaluate the role of TSLP-CRLF2 interactions in vivo in the human-mouse xenograft model. However, mouse TSLP is different from most other cytokines produced in the xenograft in that it is species-specific and does not activate the human TSLP receptor complex that includes CRLF2. Thus, traditional xenograft models do not provide the TSLP-CRLF2 interactions that we believe to be a major factor in CRLF2 B-ALL. To overcome this obstacle we engineered immune-deficient NOD/SCID IL-2Rγ null (NSG) mice to express human TSLP (hTSLP+ mice) as well as control mice that lack the TSLP cytokine (hTSLP– mice). ELISA assays show serum hTSLP levels in the hTSLP+ mice that approximate the normal range in human serum. We used this hTSLP+/- xenograft model system to study the in vivo effects of TSLP on mice transplanted with Ph-like B-ALL. First, we used the hTSLP+/– xenograft model system to evaluate the in vivo effects of TSLP on survival and proliferation of transplanted CRLF2 B-ALL cells harboring a JAK defect (MUTZ5 cell line). Mice were euthanized at 5 weeks and BM was harvested. Evaluation of BM disease by flow cytometry showed that the percentage of viable human leukemia cells in hTSLP+ mice was twice that observed in hTSLP– mice. Evaluation of cell cycle progression in human CRLF2 B-ALL cells isolated from xenograft BM showed that the percentage of cycling cells in hTSLP+ mice was 2.5 fold higher than in hTSLP– mice. When primary Ph-like ALL cells were transplanted to produce hTSLP+/– xenografts, the viable pre-B ALL cells present in the BM of hTSLP+ mice showed higher expression levels of the TSLPR components (CRLF2 and IL-7Rα) than those in the hTSLP- mice. These data provide evidence that the TSLP produced in this model is active and that it impacts primary pre-B ALL cells. Preliminary data obtained from this model suggests that TSLP provides a signal that promotes in vivo survival of CRLF2 B-ALL cells and that it may play a role in selection of leukemia clones during in vivo leukemogenesis. Microarray analysis comparing gene expression in primary CRLF2 B-ALL cells isolated from hTSLP+ and hTSLP– xenograft mice identified 565 that genes are differentially regulated (> 2 fold up or downregulated; p Citation Format: Olivia Francis, Ruijun Su, Shannalee Martinez, Ineavely Baez, Terry-Ann Milford, Terrence Bennett, Ross Fisher, Christopher L. Morris, Sinisa Dovat, Kimberly J. Payne. A human-mouse xenograft model to evaluate therapies and study the role of TSLP-induced signals in Ph-like ALL. [abstract]. In: Proceedings of the AACR Special Conference on Pediatric Cancer at the Crossroads: Translating Discovery into Improved Outcomes; Nov 3-6, 2013; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2013;74(20 Suppl):Abstract nr B21.


Cancer Research | 2014

Abstract 3097: TSLP regulates expression of genes involved in cell survival in a preclinical xenograft model of CRLF2 B-ALL

Olivia L. Francis; Ruijun Su; Shannalee R. Martinez; Ineavely Baez; Terry-Ann Milford; Ross Fisher; Christopher L. Morris; Xiao-Bing Zhang; Valeri Filippov; Sinisa Dovat; Kimberly J. Payne

A subset of children with B cell precursor acute lymphoblastic leukemia (B-ALL) are at high risk for relapse and death. Gene expression profiles in these high-risk B-ALLs is similar to that of Philadelphia chromosome-positive ALL. Approximately half of these Ph-like B-ALL are characterized by genetic defects resulting in overexpression of CRLF2. This defect occurs 5 times more commonly in Hispanic children than others and thus is a significant biological component of pediatric cancer health disparities. CRLF2, together with the IL-7Rα, forms a receptor complex that is activated by the cytokine, TSLP. Receptor complex activation leads to JAK-STAT5 phosphorylation. The activating JAK mutations found in some CRLF2 B-ALL led to speculation that TSLP stimulation is not a factor in CRLF B-ALL. However, we and others have found that TSLP increases STAT5 phosphorylation in CRLF2 B-ALL cells, including those with JAK defects. Our next step was to evaluate the role of TSLP-CRLF2 interactions in vivo in the human-mouse xenograft model. However, mouse TSLP is species-specific and does not activate the human TSLP receptor complex that includes CRLF2. Thus, traditional xenograft models do not provide the TSLP-CRLF2 interactions that may contribute to high risk CRLF2 B-ALL. We engineered immune-deficient NOD/SCID IL-2Rγ null (NSG) mice to express human TSLP (hTSLP+ mice), as well as control mice that lack the TSLP cytokine (hTSLP- mice). Then we used this hTSLP+/- xenograft model system to evaluate the in vivo effects of TSLP on transplanted CRLF2 B-ALL cells harboring a JAK defect (MUTZ5 cell line). Transplanted mice were euthanized at 5 weeks and bone marrow (BM) was harvested. Evaluation of BM by flow cytometry showed that approximately half of the human leukemia cells were apoptotic in mice without TSLP while apoptosis was virtually absent in CRLF2 B-ALL cells harvested from hTSLP+ mice. Next we used Ingenuity Pathway Analysis to identify functions and pathways regulated by TSLP. Primary CRLF2 B-ALL cells were transplanted into hTSLP+ and hTSLP- mice. Whole genome microarray performed on primary human leukemia cells isolated from BM of xenograft mice identified 280 genes that were upregulated and 281 genes that were downregulated (> 2 fold up or downregulated; p Citation Format: Olivia L. Francis, Ruijun Su, Shannalee R. Martinez, Ineavely Baez, Terry-Ann Milford, Ross Fisher, Christopher L. Morris, Xiaobing Zhang, Valeri Filippov, Sinisa Dovat, Kimberly J. Payne. TSLP regulates expression of genes involved in cell survival in a preclinical xenograft model of CRLF2 B-ALL. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 3097. doi:10.1158/1538-7445.AM2014-3097


Cancer Research | 2013

Abstract 5027: In vivo effects of TSLP in a human-mouse xenograft model of CRLF2 B-ALL.

Ruijun Su; Olivia L. Francis; Shannalee R. Martinez; Ineavely Baez; Terry-Ann Milford; Christopher L. Morris; Ross Fisher; Xiao-Bing Zhang; Sinisa Dovat; Kimberly J. Payne

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Pediatric CRLF2 B-cell Acute Lymphoblastic Leukemia (CRLF2 B-ALL) is a high-risk form of leukemia that is associated with relapse and poor patient survival outcomes. This leukemia occurs five times more frequently among children of Hispanic/Latino ethnicity than others and represents the most significant biological component of childhood cancer health disparities identified to date. CRLF2 B-ALL arises from genetic alterations that result in overexpression of the CRLF2 gene. CRLF2, together with the IL-7 receptor α, forms a receptor complex that is activated by the cytokine, TSLP. The JAK-STAT5 pathway is phosphorylated downstream of receptor activation. The activating JAK mutations found in some CRLF2 B-ALL have led to speculation that TSLP stimulation is not a factor in CRLF B-ALL. However, we found that TSLP increases STAT5 phosphorylation, even in CRLF2 B-ALL cells with JAK mutations. Our studies of normal B cell development show that TSLP stimulation increases cellular proliferation in early human B lineage cells. We also find that pediatric bone marrow (BM) stromal cells express TSLP and thus provide an in vivo source of TSLP that can act on CRLF2 B-ALL cells. We proposed to evaluate the role of TSLP in human CRLF2 B-ALL using a human-mouse xenograft model. However, TSLP is different from most other cytokines produced in the mouse in that it is species-specific and therefore does not activate the human TSLP receptor complex that contains CRLF2. Thus, traditional xenograft models do not provide the TSLP-CRLF2 interactions that we believe to be a contributing factor in CRLF2 B-ALL. To overcome this hurdle we engineered immune deficient mice to express human TSLP (hTSLP+ mice) as well as control mice that lack the human TSLP cytokine (hTSLP- mice). We used this hTSLP+/- xenograft model system to evaluate the in vivo effects of TSLP on survival and proliferation of transplanted CRLF2 B-ALL cells harboring a JAK defect (MUTZ5 cell line). Mice were euthanized at 5 weeks and BM was harvested. Evaluation of BM disease by flow cytometry showed that the percentage of viable human leukemia cells in hTSLP+ mice was twice that observed in hTSLP-mice. Evaluation of cell cycle progression in human CRLF2 B-ALL cells isolated from xenograft BM showed that the percentage of cycling cells in hTSLP+ mice was 2.5 fold higher than in hTSLP- mice. These preliminary data suggest that TSLP may contribute to CRLF2 B-ALL by increasing the survival and proliferation of CRLF2-B-ALL cells. Ongoing studies are focused on evaluating in vivo effects of TSLP in primary CRLF2 B-ALL from Hispanic patients. The identification of genes downstream of TSLP-CRLF2 signaling has the potential of providing drug targets for combination therapy to effectively treat CRLF2 B-ALL and reduce cancer health disparities in Hispanic childhood B-ALL. This work is supported by NIH R21R21CA162259, a St. Baldricks Foundation Research Grant, and a LLU GCAT award (KJP) Citation Format: Rui-jun Su, Olivia L. Francis, Shannalee R. Martinez, Ineavely Baez, Terry-Ann Milford, Christopher L. Morris, Ross O. Fisher, Xiao-Bing Zhang, Sinisa Dovat, Kimberly J. Payne. In vivo effects of TSLP in a human-mouse xenograft model of CRLF2 B-ALL. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5027. doi:10.1158/1538-7445.AM2013-5027


Cancer Research | 2012

Abstract 1347: A human-mouse xenograft model to study the role of TSLP in CRLF2d B-ALL

Kimberly J. Payne; Ruijun Su; Olivia L. Francis; Shannalee R. Martinez; Terrence Bennett; Keshav Arogyaswamy; Christopher L. Morris; Sinisa Dovat

B-cell precursor ALL where genetic defects lead to overexpression of CRLF2 (CRLF2d B-ALL) are high-risk with poor prognosis. CRLFd B-ALL occurs 5 times more frequently among children of Hispanic/Latino ethnicity and is a major contributor to the health disparity in survival of Hispanic children with ALL. CRLF2, together with the IL-7Rα, forms a receptor complex that is activated by the cytokine, TSLP. The JAK-STAT5 pathway is phosphorylated downstream of receptor activation. The activating JAK mutations found in some CRLF2 B-ALL have led to speculation that TSLP stimulation is not a factor in CRLF B-ALL. In preliminary studies to address this question we evaluated the effect of TSLP on two CRLF2d B-ALL cell lines with JAK defects and which have been reported to exhibit constitutive JAK-STAT5 activation. Our data show that TSLP increases STAT5 phosphorylation in both of these cell lines and in primary B-ALL cells that overexpress CRLF2. Our next step was to evaluate the role of TSLP-CRLF2 interaction in vivo in the human-mouse xenograft model. However, mouse TSLP is different from most other cytokines produced in the xenograft in that it is species-specific and does not activate the human TSLP receptor complex that contains CRLF2. Thus, traditional xenograft models do not provide the TSLP-CRLF2 interactions that we believe to be a major factor in CRLF2 B-ALL. To overcome this obstacle we have engineered immune deficient NOD/SCID IL-2Rγ null (NSG) mice to express human TSLP (hTSLP+ mice) as well as control mice that lack the TSLP cytokine (hTSLP- mice). ELISA assays show plasma hTSLP levels in the hTSLP+ mice that approximate the normal range in human plasma. We have used this hTSLP+/− system to expand a sample of primary pre-B ALL cells from a patient that includes clones of CRLF2-HI and CRLF2- B-ALL cells. Preliminary data indicate that the pre-B ALL cells expanded in hTSLP+ mice show higher expression levels of the TSLPR components (CRLF2 and IL-7Rα) than cells expanded in the hTSLP- mice. This data provide evidence that the TSLP produced in this model is active and that it impacts primary pre-B ALL cells. The hTSLP+ mice that we produce will allow for the first time the study of normal and malignant B lymphopoiesis in a model that provides the complex bone marrow architecture of the xenograft while providing the full range of cytokines that are known to act on early B lineage cells (IL-7, FL and TSLP). This model will be particularly important for identifying therapies that can effectively target CRLF2-d B-ALL and reduce cancer health disparities in Hispanic childhood B-ALL. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 1347. doi:1538-7445.AM2012-1347

Collaboration


Dive into the Christopher L. Morris's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sinisa Dovat

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ruijun Su

Loma Linda University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge