Ann Pendergast

Overview:

Research Overview:

Tyrosine Kinase-regulated Transcription Networks in tumor progression to metastasis and the regeneration response to injury.

The long-term goal of our research is to define the role of protein tyrosine kinase-regulated transcription networks in the regulation of cell polarity, growth, survival, differentiation, adhesion, and migration during cancer metastasis and the response to tissue injury. We have a long-standing research interest on the role of protein tyrosine phosphorylation in tumorigenesis. Our early research led to seminal discoveries that defined the critical pathways employed by the BCR-ABL tyrosine kinase to induce human leukemia. We employ animal models and state-of-the art transcriptomic technologies to investigate the role of tyrosine kinase-dependent transcription factor networks during tumor metastasis as well as the regeneration response following lung injury. In particular, we are dissecting the pathways that modulate the crosstalk among multiple cell types during metastasis to the brain. Brain metastases represent the most common adult intracranial malignancy with more than 200,000 patients diagnosed in the U.S. annually. Approximately, 20 to 40% of patients with solid tumors will develop brain metastases and lung cancer patients exhibit the highest prevalence of brain metastasis (40-60%) among all cancer types.  Current therapies to treat brain metastases have proven ineffective due to variable, transient and incomplete responses, as well as inability for drugs to cross the blood-brain-barrier (BBB) to reach therapeutic doses to treat brain metastasis. We have recently reported that ABL tyrosine kinase-driven transcriptional networks promote brain metastasis in mouse models, and found that treatment with ABL allosteric inhibitors impairs brain metastasis in pre-clinical models. Among the research areas currently being pursued in our laboratory are defining the mechanisms that regulate the cross-talk between brain metastatic cells and associated cells in the brain tumor microenvironment. High-level expression of ABL1, ABL2 and a subset of ABL-dependent target genes correlates with shortened survival of lung adenocarcinoma patients. Thus, ABL-specific allosteric inhibitors might be effective to treat metastatic lung cancer with an activated ABL pathway signature. The ultimate goal of our studies is to develop novel therapies for the treatment of metastatic solid tumors by targeting not only cancer cells but also associated stromal cells in the tumor microenvironment.

Repair following injury requires dynamic intercellular signaling to promote the proper balance of proliferation and differentiation of specialized epithelial progenitor cell populations required to restore normal lung epithelial architecture and barrier function. Absence or imbalance of these processes may result in death or long-term pulmonary disease among survivors. Currently little is known regarding the identity of signaling networks that might be effectively targeted to promote recovery from lung injury. Unexpectedly we found that inhibition of the ABL kinases promotes lung epithelial regeneration in mice after bacterial pneumonia challenge. Further, pathogen exposure elicits a dramatic increase in Abl1 expression in bronchial epithelial cells. Our exciting data demonstrate that inactivation of ABL kinases in mouse models of bacterial and viral pneumonia promotes alveolar epithelial cell regeneration.

Mentoring Philosophy:

My goal is to train the next generation of scientists and leaders by providing essential skills to develop into independent and creative thinkers. I have extensive experience in training and mentoring students, postdoctoral fellows and junior faculty.  My laboratory provides a collegial and highly interactive environment to promote collaboration and engagement among lab members and colleagues across the University. We conduct weekly laboratory research and journal club meetings, and weekly one-on-one meetings with trainees to discuss research progress, trouble shooting, planning future research, and writing publications and grants. Lab trainees have gone to successful academic careers and are currently Professors, Associate and Assistant Professors at various academic institutions. I have also trained outstanding post-doctoral fellows who have gone to successful research careers in industry.

 

Positions:

Anthony R. Means Cancer Biology Distinguished Professor

Pharmacology & Cancer Biology
School of Medicine

Professor of Pharmacology and Cancer Biology

Pharmacology & Cancer Biology
School of Medicine

Professor of Cell Biology

Cell Biology
School of Medicine

Member of the Duke Cancer Institute

Duke Cancer Institute
School of Medicine

Education:

Ph.D. 1986

University of California - Riverside

Grants:

Exploring the role of polyploidy in tumor progression

Administered By
Pharmacology & Cancer Biology
Awarded By
National Institutes of Health
Role
Co-Sponsor
Start Date
End Date

Erythropoietin Receptor Regulation of Erythorpoiesis

Administered By
Medicine, Hematology
Awarded By
National Institutes of Health
Role
Mentor
Start Date
End Date

Identification of actionable networks promoting breast cancer progression and brain metastasis

Administered By
Pharmacology & Cancer Biology
Awarded By
Department of Defense
Role
Principal Investigator
Start Date
End Date

Identification of a novel target for treating breast cancer metastasis

Administered By
Pharmacology & Cancer Biology
Awarded By
American Cancer Society, Inc.
Role
Principal Investigator
Start Date
End Date

Investigation of an ABL kinase-dependent signaling axis required for lung cancer brain metastasis

Administered By
Pharmacology & Cancer Biology
Awarded By
National Institutes of Health
Role
Principal Investigator
Start Date
End Date

Publications:

ABL kinases regulate the stabilization of HIF-1α and MYC through CPSF1.

The hypoxia-inducible factor 1-α (HIF-1α) enables cells to adapt and respond to hypoxia (Hx), and the activity of this transcription factor is regulated by several oncogenic signals and cellular stressors. While the pathways controlling normoxic degradation of HIF-1α are well understood, the mechanisms supporting the sustained stabilization and activity of HIF-1α under Hx are less clear. We report that ABL kinase activity protects HIF-1α from proteasomal degradation during Hx. Using a fluorescence-activated cell sorting (FACS)-based CRISPR/Cas9 screen, we identified HIF-1α as a substrate of the cleavage and polyadenylation specificity factor-1 (CPSF1), an E3-ligase which targets HIF-1α for degradation in the presence of an ABL kinase inhibitor in Hx. We show that ABL kinases phosphorylate and interact with CUL4A, a cullin ring ligase adaptor, and compete with CPSF1 for CUL4A binding, leading to increased HIF-1α protein levels. Further, we identified the MYC proto-oncogene protein as a second CPSF1 substrate and show that active ABL kinase protects MYC from CPSF1-mediated degradation. These studies uncover a role for CPSF1 in cancer pathobiology as an E3-ligase antagonizing the expression of the oncogenic transcription factors, HIF-1α and MYC.
Authors
Mayro, B; Hoj, JP; Cerda-Smith, CG; Hutchinson, HM; Caminear, MW; Thrash, HL; Winter, PS; Wardell, SE; McDonnell, DP; Wu, C; Wood, KC; Pendergast, AM
MLA Citation
Mayro, Benjamin, et al. “ABL kinases regulate the stabilization of HIF-1α and MYC through CPSF1.Proc Natl Acad Sci U S A, vol. 120, no. 16, Apr. 2023, p. e2210418120. Pubmed, doi:10.1073/pnas.2210418120.
URI
https://scholars.duke.edu/individual/pub1571506
PMID
37040401
Source
pubmed
Published In
Proc Natl Acad Sci U S A
Volume
120
Published Date
Start Page
e2210418120
DOI
10.1073/pnas.2210418120

Multi-apical polarity of alveolar stem cells and their dynamics during lung development and regeneration.

Epithelial cells of diverse tissues are characterized by the presence of a single apical domain. In the lung, electron microscopy studies have suggested that alveolar type-2 epithelial cells (AT2s) en face multiple alveolar sacs. However, apical and basolateral organization of the AT2s and their establishment during development and remodeling after injury repair remain unknown. Thick tissue imaging and electron microscopy revealed that a single AT2 can have multiple apical domains that enface multiple alveoli. AT2s gradually establish multi-apical domains post-natally, and they are maintained throughout life. Lineage tracing, live imaging, and selective cell ablation revealed that AT2s dynamically reorganize multi-apical domains during injury repair. Single-cell transcriptome signatures of residual AT2s revealed changes in cytoskeleton and cell migration. Significantly, cigarette smoke and oncogene activation lead to dysregulation of multi-apical domains. We propose that the multi-apical domains of AT2s enable them to be poised to support the regeneration of a large array of alveolar sacs.
Authors
Konkimalla, A; Konishi, S; Kobayashi, Y; Kadur Lakshminarasimha Murthy, P; Macadlo, L; Mukherjee, A; Elmore, Z; Kim, S-J; Pendergast, AM; Lee, PJ; Asokan, A; Knudsen, L; Bravo-Cordero, JJ; Tata, A; Tata, PR
MLA Citation
Konkimalla, Arvind, et al. “Multi-apical polarity of alveolar stem cells and their dynamics during lung development and regeneration.Iscience, vol. 25, no. 10, Oct. 2022, p. 105114. Pubmed, doi:10.1016/j.isci.2022.105114.
URI
https://scholars.duke.edu/individual/pub1553012
PMID
36185377
Source
pubmed
Published In
Iscience
Volume
25
Published Date
Start Page
105114
DOI
10.1016/j.isci.2022.105114

ABL kinases regulate translation in HER2+ cells through Y-box-binding protein 1 to facilitate colonization of the brain.

Patients with human epidermal growth factor receptor 2-positive (HER2+/ERBB2) breast cancer often present with brain metastasis. HER2-targeted therapies have not been successful to treat brain metastases in part due to poor blood-brain barrier (BBB) penetrance and emergence of resistance. Here, we report that Abelson (ABL) kinase allosteric inhibitors improve overall survival and impair HER2+ brain metastatic outgrowth in vivo. Mechanistically, ABL kinases phosphorylate the RNA-binding protein Y-box-binding protein 1 (YB-1). ABL kinase inhibition disrupts binding of YB-1 to the ERBB2 mRNA and impairs translation, leading to a profound decrease in HER2 protein levels. ABL-dependent tyrosine phosphorylation of YB-1 promotes HER2 translation. Notably, loss of YB-1 inhibits brain metastatic outgrowth and impairs expression of a subset of ABL-dependent brain metastatic targets. These data support a role for ABL kinases in the translational regulation of brain metastatic targets through YB-1 and offer a therapeutic target for HER2+ brain metastasis patients.
Authors
McKernan, CM; Khatri, A; Hannigan, M; Child, J; Chen, Q; Mayro, B; Snyder, D; Nicchitta, CV; Pendergast, AM
MLA Citation
McKernan, Courtney M., et al. “ABL kinases regulate translation in HER2+ cells through Y-box-binding protein 1 to facilitate colonization of the brain.Cell Rep, vol. 40, no. 9, Aug. 2022, p. 111268. Pubmed, doi:10.1016/j.celrep.2022.111268.
URI
https://scholars.duke.edu/individual/pub1534962
PMID
36044842
Source
pubmed
Published In
Cell Reports
Volume
40
Published Date
Start Page
111268
DOI
10.1016/j.celrep.2022.111268

ABL allosteric inhibitors synergize with statins to enhance apoptosis of metastatic lung cancer cells.

Targeting mitochondrial metabolism has emerged as a treatment option for cancer patients. The ABL tyrosine kinases promote metastasis, and enhanced ABL signaling is associated with a poor prognosis in lung adenocarcinoma patients. Here we show that ABL kinase allosteric inhibitors impair mitochondrial integrity and decrease oxidative phosphorylation. To identify metabolic vulnerabilities that enhance this phenotype, we utilized a CRISPR/Cas9 loss-of-function screen and identified HMG-CoA reductase, the rate-limiting enzyme of the mevalonate pathway and target of statin therapies, as a top-scoring sensitizer to ABL inhibition. Combination treatment with ABL allosteric inhibitors and statins decreases metastatic lung cancer cell survival in vitro in a synergistic manner. Notably, combination therapy in mouse models of lung cancer brain metastasis and therapy resistance impairs metastatic colonization with a concomitant increase in animal survival. Thus, metabolic combination therapy might be effective to decrease metastatic outgrowth, leading to increased survival for lung cancer patients with advanced disease.
Authors
Luttman, JH; Hoj, JP; Lin, KH; Lin, J; Gu, JJ; Rouse, C; Nichols, AG; MacIver, NJ; Wood, KC; Pendergast, AM
MLA Citation
Luttman, Jillian Hattaway, et al. “ABL allosteric inhibitors synergize with statins to enhance apoptosis of metastatic lung cancer cells.Cell Reports, vol. 37, no. 4, Oct. 2021, p. 109880. Epmc, doi:10.1016/j.celrep.2021.109880.
URI
https://scholars.duke.edu/individual/pub1500503
PMID
34706244
Source
epmc
Published In
Cell Reports
Volume
37
Published Date
Start Page
109880
DOI
10.1016/j.celrep.2021.109880

Role of the ABL tyrosine kinases in the epithelial-mesenchymal transition and the metastatic cascade.

The ABL kinases, ABL1 and ABL2, promote tumor progression and metastasis in various solid tumors. Recent reports have shown that ABL kinases have increased expression and/or activity in solid tumors and that ABL inactivation impairs metastasis. The therapeutic effects of ABL inactivation are due in part to ABL-dependent regulation of diverse cellular processes related to the epithelial to mesenchymal transition and subsequent steps in the metastatic cascade. ABL kinases target multiple signaling pathways required for promoting one or more steps in the metastatic cascade. These findings highlight the potential utility of specific ABL kinase inhibitors as a novel treatment paradigm for patients with advanced metastatic disease. Video abstract.
Authors
Luttman, JH; Colemon, A; Mayro, B; Pendergast, AM
MLA Citation
Luttman, Jillian Hattaway, et al. “Role of the ABL tyrosine kinases in the epithelial-mesenchymal transition and the metastatic cascade.Cell Commun Signal, vol. 19, no. 1, May 2021, p. 59. Pubmed, doi:10.1186/s12964-021-00739-6.
URI
https://scholars.duke.edu/individual/pub1483299
PMID
34022881
Source
pubmed
Published In
Cell Communication and Signaling : Ccs
Volume
19
Published Date
Start Page
59
DOI
10.1186/s12964-021-00739-6