Xiaoyan Jiang

Professor

Relevant Thesis-Based Degree Programs

 
 

Graduate Student Supervision

Doctoral Student Supervision

Dissertations completed in 2010 or later are listed below. Please note that there is a 6-12 month delay to add the latest dissertations.

Identification of new predictive biomarkers and characterization of molecular mechanisms of drug-resistance in chronic myeloid leukemia (2022)

Treatment of BCR-ABL1⁺ human leukemia, especially for early phase chronic myeloid leukemia (CML) patients, has been greatly improved by ABL tyrosine kinase inhibitor (TKI) therapies. However, early relapses and acquired drug resistance remain problems. Thus, identification of new biomarkers and therapeutic targets are needed to predict patients’ responses for providing alternative treatment strategy and to overcome drug resistance by developing more effective therapies in CML.To identify new biomarkers that can predict a patient’s response to TKI therapies in CML, the expression of 47 microRNAs (miRNAs) that were differentially expressed between normal bone marrow and CML or in Imatinib (IM)-responders versus nonresponders was evaluated in CD34⁺ CML cells pre- and post-nilotinib (NL) therapy from a cohort of 58 patients enrolled in a clinical trial. Using Cox Proportional Hazard analysis and machine learning algorithms, miR-145 and miR-708 were identified as predictors for NL nonresponse in treatment-naïve cells, while miR-150 and miR-185 were predictors at 1-month and 3-month post-NL treatment. Interestingly, incorporation of in vitro colony formation data into either panel improved the predictive power at each time point. Thus, this new predictive model may be developed into a prognostic tool for use in the clinic. To investigate the molecular functions of the Ahi-1 oncogene and its SH3 domain in regulation of TKI resistance, a high-content antibody microarray was performed in BCR-ABL1⁺ cells expressing different constructs of Ahi-1. This analysis uncovered that the eIF4F complex, the key regulator of the mRNA-ribosome recruitment phase of translation initiation, was differentially expressed in wildtype Ahi-1 and IM-resistant cells. Interestingly, increased expression of several eIF4F complex members was demonstrated in CD34⁺ CML patient cells compared to normal bone marrow, particularly eIF4G1, the scaffolding protein of the complex. Strikingly, inhibition of eIF4G1 by shRNA or a selective inhibitor, SBI-756 impaired survival, increased IM sensitivity and reduced eIF4F complex activity significantly in IM-resistant cells. Additionally, inhibition of eIF4G1 resulted in a significant reduction of BCR-ABL1 protein expression in resistant cells, which may provide a novel strategy of targeting BCR-ABL1. Thus, understanding the mechanism of drug resistance mediated by eIF4G1 could lead to novel strategies to overcome these challenges.

View record

Functional and structural study of the AHI-1 SH3 domain, characterization of the BCR-ABL-AHI-1-Dynamin-2 protein complex and investigation of oncogenic roles of dynamin-2 in chronic myeloid leukemia (2017)

Tyrosine kinase inhibitor (TKI) therapies have been introduced into clinical practice with remarkable effects on chronic myeloid leukemia (CML). However, early relapse, acquired drug resistance and persistence of leukemic stem cells (LSCs) remain problematic. Improved treatments specifically targeting key molecular elements active in CML LSCs are needed. One candidate is the oncoprotein AHI-1 (Abelson helper integration site-1), which is highly deregulated in LSCs. It harbors two key domains, SH3 and WD40-repeat, which are known important mediators of protein-protein interactions. An AHI-1-mediated protein complex containing BCR-ABL and JAK2 has been shown to modulate transforming activity and TKI-response/resistance of CML LSCs. In this study, I investigated the functional roles of the AHI-1 SH3 domain in regulation of cellular resistance of primitive CML cells to TKIs. I showed that deletion of the SH3 domain of Ahi-1 significantly enhanced apoptotic response of BCR-ABL⁺ cells to TKIs compared to cells expressing full-length Ahi-1. I solved the crystal structure of the AHI-1 SH3 domain and identified several unique features, providing potential target sites for designing specific drugs. Using immunoprecipitation/mass spectrometry, I identified a novel protein interaction between AHI-1 and Dynamin-2 (DNM2), a GTPase, through the AHI-1 SH3 domain. I showed that DNM2 expression was significantly upregulated in CML stem/progenitor cells compared to normal bone marrow cells. I also determined that the AHI-1 SH3 domain and the proline rich domain of DNM2 were mainly responsible for their interaction. Most importantly, I identified a novel protein complex in CML cells, containing BCR-ABL, AHI-1 and DNM2. Furthermore, I demonstrated an oncogenic role of DNM2 in primitive CML cells by showing that knockdown of DNM2 greatly impaired the survival of CML stem/progenitor cells and sensitized them to TKI treatments. Lastly, I illustrated that DNM2 might be involved in deregulation of endocytosis, ROS production and autophagy in TKI-insensitive CML stem/progenitor cells. This study detailed the identification and characterization of the newly-identified BCR-ABL-AHI-1-DNM2 protein complex and described the oncogenic functions of DNM2 in primitive CML cells. It further suggested that targeting DNM2 may facilitate eradication of LSCs as a new treatment option in CML.

View record

Characterization of Novel Therapeutic Targets in Chronic Myeloid Leukemia (2016)

The identification of BCR-ABL1 as the key molecular event in chronic myeloid leukemia (CML) has revolutionized treatment opportunities for early phase patients. Imatinib mesylate (IM) and other ABL1 tyrosine kinase inhibitors (TKIs) have been introduced into the clinic with remarkable effects. However, initial and acquired resistance, relapse and in particular, the persistence of CML stem cells upon TKI therapy represent critical challenges and warrant the identification of predictive biomarkers and novel, distinct targets for improved treatment strategies. In this work, I investigated how CML stem and progenitor cells survive TKI therapy through intrinsic and bone marrow (BM) niche-associated mechanisms. I revealed that the core autophagy protease ATG4B, and the focal adhesion protein and serine/threonine kinase Integrin-linked kinase (ILK) play crucial roles in CML, and that they can be successfully targeted with small molecule inhibitors. By comparing the expression of various core autophagy genes and proteins, ATG4B was identified as potential biomarker in CML to predict IM-responders versus IM-nonresponders prior to the initiation of therapy. Furthermore, my studies illustrated that deregulation of ATG4B is critical to autophagy, survival and growth of CML stem and progenitor cells. Inhibition or suppression of ATG4B decreased CML cell viability significantly and sensitized leukemic cells to TKI treatment highlighting ATG4B as a novel target in CML. ILK was identified as a differentially expressed gene between CD34⁺ CML patient cells and healthy donors by RNA-sequencing (RNA-seq) analysis, and the importance of the ILK protein and its kinase functions in mediating TKI responses and resistance in CML stem and progenitor cells was demonstrated by ILK inhibitor (QLT0267) and ILK suppression studies. Moreover, various in vitro and in vivo assays showed that the simultaneous kinase inhibition of ILK and BCR-ABL1 is effective in targeting both leukemic stem and progenitor cells, including quiescent CML cells, and in the presence of stromal cells of the BM microenvironment that make TKI monotherapies ineffective. Overall, these studies provide the first evidence of the importance of ATG4B and ILK in CML, and their potential as novel therapeutic targets for improved combination treatments with TKIs to specifically eliminate CML stem and progenitor cells.

View record

Clinical Application and Functional Characterization of TOX in Cutaneous T-Cell Lymphoma (2016)

Cutaneous T-cell lymphoma (CTCL) is a group of lymphoproliferative disorders consisting of two main subtypes: mycosis fungoides (MF) and Sézary syndrome (SS). Due to the lack of robust histological markers, it remains a challenge to establish an accurate diagnosis and offer long term prognostication for CTCL. In addition, the molecular pathogenesis of CTCL is only partially understood. Previously our group discovered that early stage MF skin biopsies contained ectopic expression of TOX gene, which is essential for the early development of CD4⁺ T cells but normally is switched off in mature CD4⁺ T cells in the peripheral tissues. The objectives of my thesis research are to evaluate if TOX can be used to improve CTCL diagnosis and prognostication, and to characterize the functional role of TOX in the pathogenesis of CTCL.Using skin biopsies and clinical databases from Vancouver, Beijing and Boston, I confirmed that TOX expression levels were significantly upregulated in the full spectrum of MF and in SS. In addition, as a diagnostic marker, high TOX expression levels differentiated CTCL from non-CTCL controls with good sensitivity and specificity. Furthermore, as a prognostic marker, high TOX mRNA levels correlated with increased risks of disease progression and disease-specific mortality in MF, and increased risks of disease-specific mortality in SS.I also investigated the functional role of TOX in CTCL pathogenesis using multiple CTCL cell lines and a mouse xenograft model. TOX knockdown in three CTCL cell lines led to markedly increased apoptosis, reduced cell proliferation, and impaired tumorigenic ability. These effects were partially mediated by increased expression of two cell cycle regulators, CDKN1B and CDKN1C. In addition, transcriptome analysis between TOX-suppressed cells and control CTCL cells uncovered additional potential molecules downstream of TOX, such as tumor suppressors FOXO3 and HBP1.Our results provide strong evidence that aberrant activation of TOX can serve as a diagnostic and prognostic biomarker for CTCL. Further, we demonstrated that TOX plays a crucial oncogenic role in CTCL pathogenesis, partially through regulating transcription of CDKN1B, CDKN1C and other downstream genes. Therefore TOX and/or its downstream genes may be promising therapeutic targets for CTCL.

View record

Identification and Characterization of Novel Therapeutic Targets and Biomarkers in Chronic Myeloid Leukemia (2016)

Chronic myeloid leukemia (CML) has long served as a paradigm for new insights into the cellular origin, pathogenesis and treatment of human cancers. ABL tyrosine kinase inhibitor (TKI) therapies have had remarkable effects on treatment of early phase CML. However, TKI monotherapies are not curative, and initial and acquired TKI resistance remain clinically challenging. Particularly, CML stem/progenitor cells are insensitive to TKIs. Therefore, novel treatments and predictive biomarkers are clearly needed. In this work, I studied the biological effects of dual BCR-ABL and JAK2 suppressions on TKI-nonresponder stem/progenitor cells, and identified and characterized novel microRNA (miRNA) biomarkers in these cells. I examined the biological effects of a new JAK2 inhibitor, BMS-911543, in combination with TKIs on CD34⁺ CML cells from IM-nonresponders. I demonstrated that combination therapy significantly reduces JAK2/STAT5 and CRKL activities, induces apoptosis, inhibits colony growth, and eliminates leukemic stem cells in vitro, while sparing healthy counterparts. I further showed that oral BMS-911543 combined with dasatinib is more effective in eliminating leukemic cells in an aggressive mouse model of BCR-ABL⁺ human leukemia. Next, I identified differentially expressed miRNAs in CD34⁺ CML cells using RNA-seq analysis, and validated the results in additional samples using high-throughput qPCR. Potential miRNA target genes were also identified by integrating miRNA expression profiles with gene expression profiles using strand-specific RNA-seq. These studies revealed that expression of miR-185 is significantly reduced in CD34⁺ CML cells from TKI-nonresponders compared to TKI-responders. Restoration of miR-185 expression by lentiviral transduction in CD34⁺ TKI-nonresponder cells significantly impairs survival of these cells and sensitizes them to TKI treatment in vitro and in vivo. Additionally, I validated the target genes of miR-185 to rationalize its roles in CML. Lastly, I demonstrated that the expression levels of several miRNAs, including miR185, were restored in patients treated with nilotinib, suggesting their potential as biomarkers to predict clinical response to TKI therapies.These studies have uncovered the biological significance of JAK2 and miR-185 in regulation of the properties of drug-insensitive CML stem/progenitor cells, and their potential as therapeutic targets for improved treatments with TKIs especially in patients at risk of developing TKI resistance.

View record

The Role of BIN1 in the Regulation of Cell Proliferation, Apoptosis and Tumor Formation in Cutaneous T-Cell Lymphoma (2014)

Cutaneous T-cell lymphomas (CTCLs) represent a group of lymphoproliferative disorders characterized by homing of malignant T-cells to the skin’s surface. There are two main types of CTCL: Mycosis Fungoides (MF) and Sezary Syndrome (SS). We have demonstrated that expression of the Abelson helper integration site-1 (AHI-1) oncogene is significantly increased in CD4⁺CD7- cells from SS patients. Bridging integrator 1 (BIN1) has been identified by microarray analysis of CTCL cells as a candidate gene involved in AHI-1-mediated lymphomagenesis. Interestingly, BIN1expression is significantly reduced in SS patient samples. However, the role of BIN1 and its molecular connection to AHI-1 in lymphomagenesis remains unexplored. I extensively investigated the role of key BIN1 isoforms in primary and CTCL cell line model systems both in vitro and in vivo. I demonstrated that overexpression/restored expression of BIN1 isoforms has strong anti-proliferative and pro-apoptotic roles in CTCL cells in vitro, and significantly inhibits the tumorigenic activity of these cells in vivo. The pro-apoptotic role of BIN1 in CTCL cells occurs through downregulation of c-FLIP, a critical inhibitor of Fas/FasL-mediated apoptosis. I also observed significant reduction and increase in BIN1 and c-FLIP transcripts in primary CTCL samples, respectively. Interestingly, high BIN1 and low c-FLIP transcripts correlated with better survival rate in SS patients. Thus, BIN1 deficiency may play an important role in CTCL pathogenesis by causing apoptosis resistance.Furthermore, I explored potential mechanisms by which AHI-1 leads to downregulation of BIN1, by (1) examining if AHI-1 physically interacts with BIN1; and (2) determining if AHI-1 alters transcription of BIN1 by changing the methylation status of the BIN1 promoter. These experiments did not yield direct evidence of these two potential mechanisms of AHI-1’s role in BIN1 suppression. Thus, the mechanism by which AHI-1 regulates BIN1 remains unknown. Nevertheless, several potential BIN1 interacting proteins were uncovered in CTCL cells, including α/β-tubulin and β-actin. Overall, this study provides the first evidence of strong tumor suppressor activity of BIN1 in CTCL. It points to the loss of BIN1 and subsequent upregulation of c-FLIP as an important mechanism to induce apoptosis resistance in CTCL cells, and identifies BIN1 and c-FLIP as potential CTCL therapeutic targets.

View record

Master's Student Supervision

Theses completed in 2010 or later are listed below. Please note that there is a 6-12 month delay to add the latest theses.

Characterization of the biologic and molecular effects of newly developed oncolytic herpes simplex viruses in acute myeloid leukemia (2022)

Acute myeloid leukemia (AML) is a rapidly progressing, often fatal hematopoietic malignancy characterized by clonal expansion of leukemic stem cells and differentiation block in the myeloid lineage, with accumulation of blasts. Progress has been made in identifying therapeutic targets and several approved therapies, but resistance to frontline chemotherapy remains a major cause of treatment failure, highlighting the need for new therapies. Oncolytic viruses are a promising class of therapeutics that rely on tumor specific oncolysis and the generation of a potent adaptive anti-tumor immune response for efficacy. To investigate if newly developed oncolytic herpes simplex viruses (oHSVs), designed to potentiate anti-leukemia immunity, effectively target primitive AML cells, I evaluated oHSV-VG161, which is engineered to express IL-12, IL-15 and the IL-15 receptor alpha subunit, along with a peptide fusion protein capable of disrupting PD-1/PD-L1 interaction. After screening several AML cell lines that expressed relatively high levels of a common HSV entry receptor (TNFRSF14), I demonstrated that VG161-infected OCIAML3 and MOLM13 cells had significantly enhanced cell killing, 2-3 fold higher than control VG160-infected cells. VG161-infected AML cells also induced apoptosis in a timely, dose-dependent manner, with increased protein expression of cleaved PARP, Caspase-3 and Caspase-8. Both VG160 and VG161 viruses replicated efficiently in AML cells, but IL-12 protein expression was only detected in VG161-infected cells. ELISA analysis confirmed the production of IL-12 and IL-15/IL-15RA. Interestingly, the secreted IL-12 could be neutralized by a human IL-12 neutralizing antibody, ivsuggesting that functional IL-12 is indeed produced from VG161-infected AML cells. Mechanistically, transcript levels of several immune response genes were highly increased in VG161-infected AML cells, including the type I interferon and other interferon regulating genes such as IRF7, IRF9, ISG54. Moreover, increased phosphorylation of STAT1 and its total protein expression were also found in VG161-infected cells as compared to VG160 control cells. In addition, VG160 or VG161-infected OCIAML3 and MOLM13 cells showed enhanced cell killing when co-cultured with healthy peripheral blood mononuclear cells and increased PD-L1 expression in these cells. Thus, I have demonstrated that newly developed oHSVs engineered with immunomodulatory transgenes effectively target AML cells, suggesting a potential treatment strategy for AML.

View record

Characterization of the miR-185-PAK6-mediated survival and cell cycle controls in chronic myeloid leukemia (2021)

Overcoming drug resistance and targeting leukemic stem cells (LSCs) remain major challenges for curative treatment of human leukemia, including chronic myeloid leukemia (CML). Thus, there is a need to identify novel biomarkers and therapeutic strategies to target LSCs. Increasing evidence indicates that LSCs are susceptible to disturbances in cellular metabolism and cell cycle regulation. Previously, through global transcriptome profiling, our lab identified a key microRNA (miRNA), miR-185 as a predictive biomarker which had significantly reduced expression levels in CD34⁺ (LSC/progenitor) treatment-naïve CML cells. Through RNA-seq analysis, PAK6, a serine/threonine-protein kinase, was identified as a target gene of miR-185; it is upregulated in CD34⁺ TKI-nonresponder cells vs. TKI-responders, correlating with reduced miR-185 expression. Gene set enrichment analysis (GSEA) in the same CD34⁺ patient cells where miR-185 and PAK6 were identified as being differentially expressed revealed a significant gene set enrichment of oxidative phosphorylation (OXPHOS) and reactive oxygen species (ROS) in CD34⁺ CML cells compared to healthy CD34⁺ cells. These levels were also significantly higher in TKI-nonresponder cells than in TKI-responders. Thus, the miR-185-PAK6 axis may contribute to the perturbation of specific metabolic pathways in TKI-nonresponder LSC/progenitor cells and confer therapy-resistance to these cells. Indeed, a pre-clinically validated pan-PAK inhibitor (PF-3758309) alone, or in combination with a TKI, greatly reduced mitochondrial quantity (MitoTracker) and ROS production (CellROX) in TKI-nonresponder cells, an effect that was not seen in the same cells treated with a TKI. Notably, PF-3758309 also significantly reduced the growth of TKI-resistant cell lines and CD34⁺ TKI-nonresponder cells and increased their apoptosis; these effects were greatly enhanced by TKIs. These results were further confirmed in TKI-resistant cells using a lentiviral shRNA knockdown system that specifically targets PAK6. Interestingly, cell cycle analysis demonstrated an accumulation of cells in G2/M phase with increased senescence associated (SA) β-galactosidase staining in TKI-resistant cells following PAK6 knockdown. These observations were accompanied by increases in cell cycle and senescence protein markers p21 and p27. Taken together, these findings indicate that dual targeting of miR-185-PAK6-mediated survival, cell cycle and metabolic pathways, along with BCR-ABL, selectively eradicates therapy-resistant LSC/progenitors, providing a valuable therapeutic strategy for improved treatment and care.

View record

Targeting autophagy in chronic myeloid leukemia through inhibition of the core autophagy protein ATG4B (2018)

Treatment of chronic myeloid leukemia (CML) targets the BCR-ABL1 fusion oncoprotein that characterizes its pathogenesis using tyrosine kinase inhibitors (TKIs); however, drug resistance and relapse can occur when BCR-ABL1-independant survival pathways such as autophagy are activated. Our lab found that the key autophagy enzyme ATG4B is upregulated in CML stem/progenitor cells from patients that clinically do not respond to TKIs vs. patients that do. Knockdown of ATG4B was found to suppress autophagy and sensitize CML cells to TKIs. This study investigates if combined suppression of BCR-ABL1 and ATG4B by novel ABL1 and ATG4B inhibitors in autophagy-inducing conditions may present a novel therapeutic approach to overcome TKI-resistance in CML. I found that inhibition of ATG4B by DB2 significantly inhibits growth and induces apoptosis in CML cell lines alone and in combination with TKIs when autophagy is induced during serum deprivation. There also is a decrease in colony forming cells after DB2+TKI treatment compared to TKIs alone in non-responding CML cells (p
View record

Assessment of a potential therapeutic target in the Hedgehog pathway for the eradication of primitive chronic myeloid leukemia cells (2017)

Chronic myeloid leukemia (CML) is a hematological malignancy characterized by the presence of a novel fusion oncoprotein called BCR-ABL1 in a hematopoietic stem cell. BCR-ABL1 has constitutively active tyrosine kinase activity and deregulates many intracellular signaling pathways contributing to cancer formation, maintenance, and progression. The consistent genetic aberration of BCR-ABL1 in CML led to the development of the first molecularly-targeted cancer therapy called imatinib (IM), which revolutionized the treatment of early phase CML. However, IM is not curative, and 40% of patients with advanced CML experience primary intolerance or acquire resistance to IM. In addition, leukemic stem cells (LSCs) are relatively insensitive to IM and do not exclusively rely on BCR-ABL1 for survival. Therefore, it is important to investigate alternative pathways that are critical for LSC maintenance, to develop a strategy to eradicate them. The Hedgehog (HH) pathway, and particularly the protein Smoothened (SMO), has recently been described to be essential for CML LSCs in a mouse model. I hypothesized that the HH pathway was critical for the survival of CML stem/progenitor cells, and that dual inhibition of BCR-ABL1 and SMO would be superior to either alone in killing CML LSCs. I used a variety of biological and molecular assays to investigate expression changes of several HH pathway-associated genes and the functionality of different leukemic cell subsets from primary CML patient samples. I observed that HH pathway genes SMO and GLI2 were upregulated in CML compared with healthy bone marrow controls, and were more highly expressed in CD34⁺ cells from IM non-responders as compared with responders. In addition, these genes were most highly expressed in the stem cell-enriched Lin-CD34⁺CD38ˉ subpopulation in IM non-responders compared with progenitors and mature leukemic cells in the same patients. I also observed that CD34⁺ IM non-responder cells were more sensitive to SMO inhibition compared with responders in terms of viability, apoptosis, re-plating potential, and colony-forming ability following long-term culture.Taken together, my results support the hypothesis that the HH pathway is more critical for primitive CML cells from IM non-responders, and may represent a mechanism by which drug-resistant cells evade eradication by TKIs.

View record

Targeting tyrosine kinase inhibitor-insensitive chronic myeloid leukemia stem/progenitor cells by effective inhibition of a novel PP2A-AHI-1-BCR-ABL-JAK2-complex (2015)

Imatinib Mesylate (IM) and other tyrosine kinase inhibitors (TKIs) have had a major impact on treatment of early phase Chronic Myeloid Leukemia (CML) patients. However, TKI monotherapies are not curative and initial and acquired resistance remain challenges. Particularly, CML stem cells are less responsive to TKIs and are a critical target population for TKI resistance. Thus, improved treatments targeting key elements active in CML stem cells are needed. One candidate is Abelson helper integration site-1 (AHI-1), an oncogene that is highly upregulated in CML stem cells and interacts with multiple kinases, including BCR-ABL and JAK2. AHI-1-mediated complexes regulate TKI response/resistance of CML stem/progenitor cells, indicating that AHI-1 is a new therapeutic target in CML. By screening the Prestwick Chemical Library, a specific growth inhibitory compound that potentially targets AHI-1 was identified: Cantharidin (CAN), an inhibitor of protein phosphatase 2A (PP2A). CAN is toxic however, so two new PP2A inhibitors, LB100 and LB102, were identified for this study. These new inhibitors specifically inhibit PP2A activity and suppress growth of CML cell lines. Importantly, these new PP2A inhibitors selectively target CML stem/progenitor cells while sparing healthy stem/progenitor cells. When combined with TKIs there is significant further suppression of growth in cell lines and in CD34+ treatment-naïve IM-nonresponder cells. Furthermore, this combination effect was determined to be synergistic. Cell cycle analysis showed that treatment with PP2A inhibitors alone induced a shift from G1 to G2/M phase. Confocal microscopy confirmed that the G2/M arrest led to mitotic catastrophe. However a similar shift in cell population was observed after combination with IM, suggesting that the G2/M phase arrest is solely due to PP2A inhibition. Mechanistically, the PP2A-PR55α subunit was identified as a new AHI-1 interacting protein. Western blot analysis showed that, compared to single agents, the combination treatment greatly suppresses protein expression of AHI-1, BCR-ABL, JAK2, STAT5, AKT, β-catenin, P-38 and JNK. The combination treatment also affected PP2A and BCR-ABL-mediated β-catenin dephosphorylation/phosphorylation. These results indicate that simultaneously targeting both BCR-ABL and PP2A activities in CML stem/progenitor cells may provide a novel treatment option for CML patients, through destabilization of the protein-protein interactions mediated by AHI-1.

View record

An investigation into the role of C-terminal tensin-like protein (Cten) in melanomagenesis (2013)

C-terminal tensin-like protein (Cten) is a focal adhesion protein with no or limited protein expression in normal tissues, which has recently been reported to be overexpressed and act as an oncoprotein in numerous cancers. Since its expression status in human cutaneous melanoma is currently unknown, I used tissue microarrays and immunohistochemical staining to examine the protein expression of Cten throughout melanoma progression. I found that Cten was significantly up-regulated in dysplastic nevi (DN) compared to normal nevi (NN), and in primary melanoma (PM) compared to both DN and NN. Strong Cten staining was associated with a poorer 5- and 10-year overall and disease-specific survival for PM patients, and was an adverse independent prognostic factor for the 5-year survival of the same patients. In vitro studies using two melanoma cell lines supported these findings and indicated that Cten functions as an oncogene in melanoma.Since relatively little is known about how Cten contributes to tumorigenesis, I next investigated the expression profile of the RhoGAP Deleted in Liver Cancer-1 (DLC1), the only protein known to bind to Cten, in melanomas. Both cytoplasmic and nuclear DLC1 were detected, and both were down-regulated in metastatic melanoma (MM) compared to PM and nevi, with nuclear DLC1 expression additionally being reduced in PM compared to nevi. Both cytoplasmic and nuclear DLC1 were associated with the 5-year overall and disease-specific survival of all melanoma and MM patients, and with the disease-specific 10-year survival of all melanoma patients. Combined analysis of cytoplasmic and nuclear DLC1 revealed that for MM patients, concurrent loss of both cytoplasmic and nuclear DLC1 was associated with the worst survival outcome, with loss of either or both forms being a significant adverse independent prognostic factor for the 5-year survival of all melanoma and MM patients. A preliminary investigation into the relationship between Cten and DLC1 indicated that the effects of Cten on patient survival were dependent on the levels of DLC1, as expected.In summary, I here provide an initial characterization of the expression status and role of Cten in melanomagenesis, and speculate that it functions partly via interactions with the tumour suppressor DLC1.

View record

A novel oncogene AHI-1 interacts with BCR-ABL and JAK2 and mediates cellular resistance to tyrosine kinase inhibitors in CML (2010)

Chronic myeloid leukemia is a myeloproliferative disorder characterized by the presence of the Philadelphia chromosome, encoding a unique fusion gene BCR-ABL. The current first line treatment for patients diagnosed with CML involves administration of the ABL kinase inhibitor imatinib mesylate (IM). However, early relapses and acquired drug resistance remain a current impediment to successful treatment for many patients. This suggests the necessity for alternate treatment options which may include combination therapy targeting multiple vital proteins involved in the malignancy of the leukemia. AHI-1 (Abelson helper integration site 1) is a recently discovered oncogene that is highly deregulated in murine lymphomas and leukemias. AHI-1 displays a significant pattern of overexpression in a Philadelphia chromosome positive cell line K562 cells. To investigate AHI-1’s involvement in CML, AHI-1 was either stably overexpressed or suppressed in K562 cells. Interestingly, an increase in cellular proliferation and colony formation and a decrease in apoptosis were observed in the presence of IM when AHI-1 was overexpressed, while suppression of AHI-1 had the opposite effects. Phosphorylation and total protein expression levels of several proteins known to be involved in BCR-ABL signalling were quantified. Interestingly, elevated phosphorylation and total gene/protein expression levels of several of these proteins were observed when AHI-1 was overexpresessed, in particular NF-κB and JAK2/STAT5 displayed increased expression. Due to the strong effects AHI-1 had on the JAK2/STAT5 signalling cascade, we then inhibited JAK2 activity using a new JAK2 inhibitor, TG101209. AHI-1 overexpression led to a reduction in the cellular response to the inhibitor while suppression of AHI-1 caused an increase in sensitivity in viability, apoptosis, and colony forming cell assays. Finally, a combination of IM and TG101209 was examined in the same K562 cells lines. Results from suggest that using a combination treatment approach was more effective at inhibiting cellular viability and colony formation than either treatment alone. These findings together suggest that AHI-1 may play an important role in mediating cellular resistance to IM and TG101209 and activates several BCR-ABL signalling pathways, and that it may be a vital target in eradicating the malignant leukemic cells arising in CML.

View record

 
 

If this is your researcher profile you can log in to the Faculty & Staff portal to update your details and provide recruitment preferences.

 
 

Sign up for an information session to connect with students, advisors and faculty from across UBC and gain application advice and insight.