Network


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

Hotspot


Dive into the research topics where Elizabeth S. Henson is active.

Publication


Featured researches published by Elizabeth S. Henson.


Autophagy | 2008

HYPOXIA INDUCES AUTOPHAGIC CELL DEATH IN APOPTOSIS-COMPETENT CELLS THROUGH A MECHANISM INVOLVING BNIP3

Meghan B. Azad; Yongqiang Chen; Elizabeth S. Henson; Jeannick Cizeau; Eileen McMillan-Ward; Sara J. Israels; Spencer B. Gibson

Hypoxia (lack of oxygen) is a physiological stress often associated with solid tumors. Hypoxia correlates with poor prognosis since hypoxic regions within tumors are considered apoptosis-resistant. Autophagy (cellular “self digestion”) has been associated with hypoxia during cardiac ischemia and metabolic stress as a survival mechanism. However, although autophagy is best characterized as a survival response, it can also function as a mechanism of programmed cell death. Our results show that autophagic cell death is induced by hypoxia in cancer cells with intact apoptotic machinery. We have analyzed two glioma cell lines (U87, U373), two breast cancer cell lines (MDA-MB-231, ZR75) and one embryonic cell line (HEK293) for cell death response in hypoxia (


International Journal of Cancer | 2006

The pro-cell death Bcl-2 family member, BNIP3, is localized to the nucleus of human glial cells: Implications for glioblastoma multiforme tumor cell survival under hypoxia.

Teralee R. Burton; Elizabeth S. Henson; Priti K. Baijal; David D. Eisenstat; Spencer B. Gibson

The Bcl‐2 nineteen kilodalton interacting protein 3 (BNIP3) is a hypoxia‐inducible proapoptotic member of the Bcl‐2 family that induces cell death by associating with the mitochondria. Under normal conditions, BNIP3 is expressed in skeletal muscle and in the brain at low levels. In many human solid tumors, BNIP3 is upregulated in hypoxic regions but paradoxically, this BNIP3 expression fails to induce cell death. Herein, we have determined that BNIP3 is primarily localized to the nucleus of glial cells of the normal human brain, as well as in the malignant glioma cell line U251. Upon exposure of U251 cells to hypoxia, BNIP3 expression in the cytoplasm increases and localizes with the mitochondria, contributing to induction of cell death. In contrast, when BNIP3 is forcibly over expressed in the nucleus, it fails to induce cell death. Expression of N‐terminal BNIP3 (lacking the transmembrane and conserved domains) in U251 cells blocks hypoxia‐induced cell death acting as a dominant negative protein by binding to wild‐type BNIP3 and blocking its association with the mitochondria. In glioblastoma multiforme (GBM) tumors, BNIP3 expression is increased in hypoxic regions of the tumor and is primarily localized to the nucleus in ∼80% of tumors. Hence, BNIP3 is sequestered in the nucleus within the brain but under hypoxic conditions, BNIP3 becomes primarily cytoplasmic, promoting cell death. In GBMs, BNIP3 expression is increased but it remains sequestered in the nucleus in hypoxic regions, thereby blocking BNIP3s ability to associate with the mitochondria, providing tumor cells with a possible survival advantage.


Clinical Cancer Research | 2006

Herceptin Sensitizes ErbB2–Overexpressing Cells to Apoptosis by Reducing Antiapoptotic Mcl-1 Expression

Elizabeth S. Henson; Xiaojie Hu; Spencer B. Gibson

Purpose: Monoclonal antibodies, such as herceptin and trastuzumab, against the epidermal growth factor receptor ErbB2 (also known as HER2/neu) are an effective therapy for breast cancer patients with overexpression of ErbB2. Herceptin, in combination with standard chemotherapy, such as taxol or etoposide, gives a synergistically apoptotic response in breast tumors. Experimental Design: The mechanism underlying this synergy between chemotherapy and herceptin treatment is not well understood. Herein, we have determined that addition of herceptin, sensitized breast cancer cell lines MDA-MB-231 and MCF-7 to etoposide- or taxol-induced apoptosis. Results: This treatment resulted in reduced expression of ErbB2 and the antiapoptotic Bcl-2 family member Mcl-1 in MDA-MB-231 cells. Using antisense oligonucleotides against Mcl-1, MDA-MB-231 cells were rendered sensitive to etoposide-induced apoptosis similar to herceptin, but combined treatment of antisense against Mcl-1 and herceptin failed to give a significant increase in apoptosis. In 29 human breast tumors immunostained for ErbB2 and Mcl-1, we found that when ErbB2 was overexpressed, there was a corresponding increase in Mcl-1 expression. Discussion: Using murine fibroblasts that express human ErbB2, but no other ErbB family member (NE2), these cells showed resistance to both taxol- and etoposide-induced apoptosis compared with parental cells. In addition, NE2 cells preferentially express the antiapoptotic Bcl-2 family member Mcl-1 compared with parental cells, and treatment with herceptin reduces Mcl-1 expression. Taken together, these results suggest that herceptin sensitizes ErbB2-overexpressing cells to apoptosis by reducing antiapoptotic Mcl-1 protein levels.


Oncogene | 2011

Epidermal growth factor regulates Mcl-1 expression through the MAPK-Elk-1 signalling pathway contributing to cell survival in breast cancer

E P Booy; Elizabeth S. Henson; Spencer B. Gibson

Myeloid cell leukaemia-1 (Mcl-1) is an anti-apoptotic member of the Bcl-2 family that is elevated in a variety of tumour types including breast cancer. In breast tumours, increased Mcl-1 expression correlates with high tumour grade and poor patient survival. We have previously demonstrated that Her-2 levels correspond to increased Mcl-1 expression in breast tumours. Epidermal growth factor (EGF) receptor signalling is frequently deregulated in breast cancer and leads to increased proliferation and survival. Herein, we determined the critical downstream signals responsible for the EGF mediated increase of Mcl-1 and their role in cell survival. We found that both Mcl-1 mRNA and protein levels are rapidly induced upon stimulation with EGF. Promoter analysis revealed that an Elk-1 transcription factor-binding site is critical for EGF activation of the Mcl-1 promoter. Furthermore, we found that knockdown of Elk-1or inhibition of the Erk signalling pathway was sufficient to block EGF upregulation of Mcl-1 and EGF mediated cell survival. Using chromatin immunoprecipitation and biotin labelled probes of the Mcl-1 promoter, we found that Elk-1 and serum response factor are bound to the promoter after EGF stimulation. To determine whether Mcl-1 confers a survival advantage, we found that knockdown of Mcl-1 expression increased apoptosis whereas overexpression of Mcl-1 inhibited drug induced cell death. In human breast tumours, we found a correlation between phosphorylated Elk-1 and Mcl-1 protein levels. These results indicate that the EGF induced activation of Elk-1 is an important mediator of Mcl-1 expression and cell survival and therefore a potential therapeutic target in breast cancer.


Journal of Cellular Biochemistry | 2003

Increased expression of Mcl-1 is responsible for the blockage of TRAIL-induced apoptosis mediated by EGF/ErbB1 signaling pathway.

Elizabeth S. Henson; Erika M. Gibson; Jacylyn Villanueva; Nicolle Bristow; Neil Haney; Spencer B. Gibson

Epidermal growth factor (EGF) protects against death receptor induced apoptosis in epithelial cells. Herein, we demonstrate that EGF protection against tumor necrosis factor related apoptosis‐inducing ligand (TRAIL) induced apoptosis is mediated by increased expression of the Bcl‐2 family member myeloid cell leukemia 1 (Mcl‐1). EGF increased the mRNA and protein levels of Mcl‐1. Furthermore, expression of ErbB1 alone or in combination with ErbB2 in NIH3T3 cells up‐regulates Mcl‐1 following EGF treatment. In addition, up‐regulation of Mcl‐1 by EGF is mediated through AKT and NFκB activation since kinase inactive AKT and ΔIκB effectively blocks this up‐regulation. NFκB was also critical for the ability of EGF to prevent TRAIL induced apoptosis as a dominant negative IκB (ΔIκB) blocked NFκB activation, and relieved EGF protection against TRAIL mediated mitochondrial cytochrome‐c release and apoptosis. Finally, anti‐sense oligonucleotides directed against Mcl‐1 effectively reduced the protein levels of Mcl‐1 and blocked EGF protection against TRAIL induced mitochondrial cytochrome‐c release and apoptosis. Taken together, EGF signaling leads to increased Mcl‐1 expression that is required for blockage of TRAIL induced apoptosis.


Leukemia & Lymphoma | 2008

The role of TRAIL death receptors in the treatment of hematological malignancies

Elizabeth S. Henson; James B. Johnston; Spencer B. Gibson

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising new treatment for the hematological malignancies. TRAIL induces apoptosis by binding to its two death receptors DR4 (TRAIL-R1) and DR5 (TRAIL-R2). The extent of apoptosis by TRAIL is tightly regulated by the expression of these receptors and by downstream signaling. Chemotherapeutic agents increase the expressions of DR4 and DR5 on tumor cells through the activation of various transcription factors and there is enhanced killing on combining these agents with TRAIL. In this review, we will discuss the mechanism of TRAIL death receptor-induced apoptosis and the regulation of DR4 and DR5 expression. In particular, we will focus on the regulation of TRAIL death receptor signaling in hematological malignancies and the mechanisms responsible for the sensitization of leukemia and lymphoma cells to TRAIL-induced apoptosis by chemotherapy. Finally, we shall review the clinical data regarding the use of recombinant TRAIL and activating monoclonal antibodies against the TRAIL death receptors in the hematological malignancies.


Autophagy | 2016

Tyrosine kinase receptor EGFR regulates the switch in cancer cells between cell survival and cell death induced by autophagy in hypoxia

Yongqiang Chen; Elizabeth S. Henson; Wenyan Xiao; Daniel Huang; Eileen McMillan-Ward; Sara J. Israels; Spencer B. Gibson

ABSTRACT Autophagy is an intracellular lysosomal degradation pathway where its primary function is to allow cells to survive under stressful conditions. Autophagy is, however, a double-edge sword that can either promote cell survival or cell death. In cancer, hypoxic regions contribute to poor prognosis due to the ability of cancer cells to adapt to hypoxia in part through autophagy. In contrast, autophagy could contribute to hypoxia induced cell death in cancer cells. In this study, we showed that autophagy increased during hypoxia. At 4 h of hypoxia, autophagy promoted cell survival whereas, after 48 h of hypoxia, autophagy increased cell death. Furthermore, we found that the tyrosine phosphorylation of EGFR (epidermal growth factor receptor) decreased after 16 h in hypoxia. Furthermore, EGFR binding to BECN1 in hypoxia was significantly higher at 4 h compared to 72 h. Knocking down or inhibiting EGFR resulted in an increase in autophagy contributing to increased cell death under hypoxia. In contrast, when EGFR was reactivated by the addition of EGF, the level of autophagy was reduced which led to decreased cell death. Hypoxia led to autophagic degradation of the lipid raft protein CAV1 (caveolin 1) that is known to bind and activate EGFR in a ligand-independent manner during hypoxia. By knocking down CAV1, the amount of EGFR phosphorylation was decreased in hypoxia and amount of autophagy and cell death increased. This indicates that the activation of EGFR plays a critical role in the switch between cell survival and cell death induced by autophagy in hypoxia.


Journal of Biological Chemistry | 2002

MEK kinase 1 induces mitochondrial permeability transition leading to apoptosis independent of cytochrome c release

Erika M. Gibson; Elizabeth S. Henson; Jacylyn Villanueva; Spencer B. Gibson

Induction of apoptosis often converges on the mitochondria to induce permeability transition and release of apoptotic proteins into the cytoplasm resulting in the biochemical and morphological alteration of apoptosis. Activation of a serine threonine kinase MEK kinase 1 (MEKK1) is involved in the induction of apoptosis. Expression of a kinase-inactive MEKK1 blocks genotoxin-induced apoptosis. Upon apoptotic stimulation, MEKK1 is cleaved into a 91-kDa kinase fragment that further induces an apoptotic response. Mutation of a consensus caspase 3 site in MEKK1 prevents its induction of apoptosis. The mechanism of MEKK1-induced apoptosis downstream of its cleavage, however, is unknown. Herein we demonstrate that full-length and cleaved MEKK1 leads to permeability transition in the mitochondria. This permeability transition occurs through opening of the permeability transition (PT) pore. Inhibiting PT pore opening and reactive oxygen species production effectively reduced MEKK1-induced apoptosis. Overexpression of MEKK1, however, failed to release cytochrome c from the mitochondria or activate caspase 9. Since Bcl2 regulates changes in mitochondria and blocks MEKK1-induced apoptosis, we determined that Bcl2 blocks MEKK1-induced apoptosis when targeted to the mitochondria. This occurs downstream of MEKK1 cleavage, since Bcl2 fails to block cleavage of MEKK1. In mouse embryonic fibroblast cells lacking caspase 3, the cleaved but not full-length MEKK1 induces apoptosis and permeability transition in the mitochondria. Overall, this suggests that cleaved MEKK1 leads to permeability transition contributing to MEKK1-induced apoptosis independent of cytochrome c release from the mitochondria.


Cancers | 2017

EGFR Family Members’ Regulation of Autophagy Is at a Crossroads of Cell Survival and Death in Cancer

Elizabeth S. Henson; Yongqiang Chen; Spencer B. Gibson

The epidermal growth factor receptor (EGFR) signaling pathways are altered in many cancers contributing to increased cell survival. These alterations are caused mainly through increased expression or mutation of EGFR family members EGFR, ErbB2, ErbB3, and ErbB4. These receptors have been successfully targeted for cancer therapy. Specifically, a monoclonal antibody against ErbB2, trastuzumab, and a tyrosine kinase inhibitor against EGFR, gefitinib, have improved the survival of breast and lung cancer patients. Unfortunately, cancer patients frequently become resistant to these inhibitors. This has led to investigating how EGFR can contribute to cell survival and how cancer cells can overcome inhibition of its signaling. Indeed, it is coming into focus that EGFR signaling goes beyond a single signal triggering cell proliferation and survival and is a sensor that regulates the cell’s response to microenvironmental stresses such as hypoxia. It acts as a switch that modulates the ability of cancer cells to survive. Autophagy is a process of self-digestion that is inhibited by EGFR allowing cancer cells to survive under stresses that would normally cause death and become resistant to chemotherapy. Inhibiting EGFR signaling allows autophagy to contribute to cell death. This gives new opportunities to develop novel therapeutic strategies to treat cancers that rely on EGFR signaling networks and autophagy. In this review, we summarize the current understanding of EGFR family member regulation of autophagy in cancer cells and how new therapeutic strategies could be developed to overcome drug resistance.


Cell Death and Disease | 2013

BNIP3 acts as transcriptional repressor of death receptor-5 expression and prevents TRAIL-induced cell death in gliomas

Teralee R. Burton; Elizabeth S. Henson; Meghan B. Azad; M Brown; David D. Eisenstat; Spencer B. Gibson

Glioblastoma multiforme (GBM) is the most common and malignant brain tumor, and current treatment modalities such as surgical resection, adjuvant radiotherapy and temozolomide (TMZ) chemotherapy are ineffective. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a novel cancer therapeutic agent for GBM because of its capability of inducing apoptosis in glioma cells. Unfortunately, the majority of glioma cells are resistant to TRAIL-induced apoptosis. The Bcl-2 nineteen kilodalton interacting protein (BNIP3) is a pro-cell death BH3-only member of the Bcl-2 family that is one of the highest expressed genes in hypoxic regions of GBM tumors. We previously found that BNIP3 is localized to the nucleus in GBM tumors and suppresses cell death in glioma cells. Herein, we have discovered when BNIP3 nuclear expression is knockdown in glioma cell lines and in normal mouse astrocytes, TRAIL and its death receptor, death receptor-5 (DR5) expression is increased. In addition, when nuclear BNIP3 expression is increased, the amount of TRAIL-induced apoptosis is reduced. Using a streptavidin pull-down assay, we found that BNIP3 binds to the DR5 promoter and nuclear BNIP3 binds to the DR5 promoter. Furthermore, nuclear BNIP3 expression in GBM tumors correlates with decreased DR5 expression. Taken together, we have discovered a novel transcriptional repression function for BNIP3 conferring a TRAIL resistance in glioma cells.

Collaboration


Dive into the Elizabeth S. Henson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wenyan Xiao

University of Manitoba

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge