Network


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

Hotspot


Dive into the research topics where Ashley Sample is active.

Publication


Featured researches published by Ashley Sample.


Autophagy | 2016

Autophagy positively regulates DNA damage recognition by nucleotide excision repair.

Lei Qiang; Baozhong Zhao; Palak Shah; Ashley Sample; Seungwon Yang; Yu-Ying He

ABSTRACT Macroautophagy (hereafter autophagy) is a cellular catabolic process that is essential for maintaining tissue homeostasis and regulating various normal and pathologic processes in human diseases including cancer. One cancer-driving process is accumulation of genetic mutations due to impaired DNA damage repair, including nucleotide excision repair. Here we show that autophagy positively regulates nucleotide excision repair through enhancing DNA damage recognition by the DNA damage sensor proteins XPC and DDB2 via 2 pathways. First, autophagy deficiency downregulates the transcription of XPC through TWIST1-dependent activation of the transcription repressor complex E2F4-RBL2. Second, autophagy deficiency impairs the recruitment of DDB2 to ultraviolet radiation (UV)-induced DNA damage sites through TWIST1-mediated inhibition of EP300. In mice, the pharmacological autophagy inhibitor Spautin-1 promotes UVB-induced tumorigenesis, whereas the autophagy inducer rapamycin reduces UVB-induced tumorigenesis. These findings demonstrate the crucial role of autophagy in maintaining proper nucleotide excision repair in mammalian cells and suggest a previously unrecognized tumor-suppressive mechanism of autophagy in cancer.


Journal of Biological Chemistry | 2017

NF-κB Signaling Activation Induced by Chloroquine Requires Autophagosome, p62 Protein, and c-Jun N-terminal Kinase (JNK) Signaling and Promotes Tumor Cell Resistance

Seungwon Yang; Lei Qiang; Ashley Sample; Palak Shah; Yu-Ying He

Macroautophagy (hereafter autophagy) is a catabolic cellular self-eating process by which unwanted organelles or proteins are delivered to lysosomes for degradation through autophagosomes. Although the role of autophagy in cancer has been shown to be context-dependent, the role of autophagy in tumor cell survival has attracted great interest in targeting autophagy for cancer therapy. One family of potential autophagy blockers is the quinoline-derived antimalarial family, including chloroquine (CQ). However, the molecular basis for tumor cell response to CQ remains poorly understood. We show here that in both squamous cell carcinoma cells and melanoma tumor cells, CQ induced NF-κB activation and the expression of its target genes HIF-1α, IL-8, BCL-2, and BCL-XL through the accumulation of autophagosomes, p62, and JNK signaling. The activation of NF-κB further increased p62 gene expression. Either genetic knockdown of p62 or inhibition of NF-κB sensitized tumor cells to CQ, resulting in increased apoptotic cell death following treatment. Our findings provide new molecular insights into the CQ response in tumor cells and CQ resistance in cancer therapy. These findings may facilitate development of improved therapeutic strategies by targeting the p62/NF-κB pathway.


Photodermatology, Photoimmunology and Photomedicine | 2018

Mechanisms and prevention of UV-induced melanoma

Ashley Sample; Yu-Ying He

Melanoma is the deadliest form of skin cancer and its incidence is rising, creating a costly and significant clinical problem. Exposure to ultraviolet (UV) radiation, namely UVA (315‐400 nm) and UVB (280‐315 nm), is a major risk factor for melanoma development. Cumulative UV radiation exposure from sunlight or tanning beds contributes to UV‐induced DNA damage, oxidative stress, and inflammation in the skin. A number of factors, including hair color, skin type, genetic background, location, and history of tanning, determine the skins response to UV radiation. In melanocytes, dysregulation of this UV radiation response can lead to melanoma. Given the complex origins of melanoma, it is difficult to develop curative therapies and universally effective preventative strategies. Here, we describe and discuss the mechanisms of UV‐induced skin damage responsible for inducing melanomagenesis, and explore options for therapeutic and preventative interventions.


Journal of Biological Chemistry | 2017

Adaptor protein p62 promotes skin tumor growth and metastasis and is induced by UVA radiation

Ashley Sample; Baozhong Zhao; Lei Qiang; Yu-Ying He

Skin cancer is the most common cancer, and exposure to ultraviolet (UV) radiation, namely UVA and UVB, is the major risk factor for skin cancer development. UVA is significantly less effective in causing direct DNA damage than UVB, but UVA has been shown to increase skin cancer risk. The mechanism by which UVA contributes to skin cancer remains unclear. Here, using RNA-Seq, we show that UVA induces autophagy and lysosomal gene expression, including the autophagy receptor and substrate p62. We found that UVA activates transcription factor EB (TFEB), a known regulator of autophagy and lysosomal gene expression, which, in turn, induces p62 transcription. Next, we identified a novel relationship between p62 and cyclooxygenase-2 (COX-2), a prostaglandin synthase critical for skin cancer development. COX-2 expression was up-regulated by UVA-induced p62, suggesting that p62 plays a role in UVA-induced skin cancer. Moreover, we found that p62 stabilizes COX-2 protein through the p62 ubiquitin-associated domain and that p62 regulates prostaglandin E2 production in vitro. In a syngeneic squamous cell carcinoma mouse model, p62 knockdown inhibited tumor growth and metastasis. Furthermore, p62-deficient tumors exhibited reduced immune cell infiltration and increased cell differentiation. Because prostaglandin E2 is known to promote pro-tumorigenic immune cell infiltration, increase proliferation, and inhibit keratinocyte differentiation in vivo, this work suggests that UVA-induced p62 acts through COX-2 to promote skin tumor growth and progression. These findings expand our understanding of UVA-induced skin tumorigenesis and tumor progression and suggest that targeting p62 can help prevent or treat UVA-associated skin cancer.


Molecular Cancer Research | 2017

EGFR Signals through a DOCK180-MLK3 Axis to Drive Glioblastoma Cell Invasion

Sean A. Misek; Jian Chen; Laura E. Schroeder; Chotirat Rattanasinchai; Ashley Sample; Jann N. Sarkaria; Kathleen A. Gallo

A hallmark of glioblastoma (GBM) tumors is their highly invasive behavior. Tumor dissemination into surrounding brain tissue is responsible for incomplete surgical resection, and subsequent tumor recurrence. Identification of targets that control GBM cell dissemination is critical for developing effective therapies to treat GBM. A majority of GBM tumors have dysregulated EGFR signaling, due most frequently to EGFR amplification or the presence of a constitutively active EGFRvIII mutant. Mixed lineage kinase 3 (MLK3) is a mitogen-activated protein kinase kinase kinase (MAP3K) that can activate multiple MAPK pathways. In this study, evidence is provided that MLK3 is essential for GBM cell migration and invasion, and that an MLK inhibitor blocks EGF-induced migration and invasion. MLK3 silencing or MLK inhibition blocks EGF-induced JNK activation, suggesting that MLK3-JNK signaling promotes invasion of GBM cells. Mechanistically, it is demonstrated that DOCK180, a RAC1 guanine nucleotide exchange factor (GEF) overexpressed in invasive GBM cells, activates the MLK3-JNK signaling axis in a RAC1-dependent manner. In summary, this investigation identifies an EGFR–DOCK180–RAC1–MLK3–JNK signaling axis that drives glioblastoma cell migration and dissemination. Implications: On the basis of these findings, MLK3 emerges as a potential therapeutic target for the treatment of glioblastoma. Mol Cancer Res; 15(8); 1085–95. ©2017 AACR.


Autophagy | 2017

Autophagy gene ATG7 regulates ultraviolet radiation-induced inflammation and skin tumorigenesis

Lei Qiang; Ashley Sample; Christopher R. Shea; Keyoumars Soltani; Kay F. Macleod; Yu-Ying He

ABSTRACT Macroautophagy (hereafter autophagy) is a cellular “self-eating” process that is implicated in many human cancers, where it can act to either promote or suppress tumorigenesis. However, the role of autophagy in regulation of inflammation during tumorigenesis remains unclear. Here we show that autophagy is induced in the epidermis by ultraviolet (UV) irradiation and autophagy gene Atg7 promoted UV-induced inflammation and skin tumorigenesis. Atg7 regulated UV-induced cytokine expression and secretion, and promoted Ptgs2/Cox-2 expression through both a CREB1/CREB-dependent cell autonomous mechanism and an IL1B/IL1β-dependent non-cell autonomous mechanism. Adding PGE2 increased UV-induced skin inflammation and tumorigenesis, reversing the epidermal phenotype in mice with Atg7 deletion in keratinocytes. Similar to ATG7 knockdown in human keratinocytes, ATG5 knockdown inhibited UVB-induced expression of PTGS2 and cytokines. Furthermore, ATG7 loss increased the activation of the AMPK pathway and the phosphorylation of CRTC1, and led to endoplasmic reticulum (ER) accumulation and reduction of ER stress. Inducing ER stress and inhibiting calcium influx into the ER by thapsigargin reverses the inflammation and tumorigenesis phenotype in mice with epidermal Atg7 deletion. Taken together, these findings demonstrate that deleting autophagy gene Atg7 leads to a suppression of carcinogen-induced protumorigenic inflammatory microenvironment and tumorigenesis of the epithelium.


Photochemistry and Photobiology | 2017

Autophagy in UV Damage Response

Ashley Sample; Yu-Ying He

UV radiation exposure from sunlight and artificial tanning beds is the major risk factor for the development of skin cancer and skin photoaging. UV‐induced skin damage can trigger a cascade of DNA damage response signaling pathways, including cell cycle arrest, DNA repair and, if damage is irreparable, apoptosis. Compensatory proliferation replaces the apoptotic cells to maintain skin barrier integrity. Disruption of these processes can be exploited to promote carcinogenesis by allowing the survival and proliferation of damaged cells. UV radiation also induces autophagy, a catabolic process that clears unwanted or damaged proteins, lipids and organelles. The mechanisms by which autophagy is activated following UV exposure, and the functions of autophagy in UV response, are only now being clarified. Here, we summarize the current understanding of the mechanisms governing autophagy regulation by UV, the roles of autophagy in regulating cellular response to UV‐induced photodamage and the implications of autophagy modulation in the treatment and prevention of photoaging and skin cancer.


Photochemistry and Photobiology | 2018

The Autophagy Receptor Adaptor p62 is Up-regulated by UVA Radiation in Melanocytes and in Melanoma Cells

Ashley Sample; Baozhong Zhao; Chunli Wu; Steven Y. Qian; Xianglin Shi; Andrew E. Aplin; Yu-Ying He

UVA (315–400 nm) is the most abundant form of UV radiation in sunlight and indoor tanning beds. However, much remains to be understood about the regulation of the UVA damage response in melanocytes and melanoma. Here, we show that UVA, but not the shorter waveband UVB (280–315 nm), up‐regulates adaptor protein p62 in an Nrf2‐ and reactive oxygen species (ROS)‐dependent manner, suggesting a UVA‐specific effect on p62 regulation. UVA‐induced p62 up‐regulation was inhibited by a mitochondria‐targeted antioxidant or Nrf2 knockdown. In addition, p62 knockdown inhibited UVA‐induced ROS production and Nrf2 up‐regulation. We also report here a novel regulatory feedback loop between p62 and PTEN in melanoma cells. PTEN overexpression reduced p62 protein levels, and p62 knockdown increased PTEN protein levels. As compared with normal human skin, p62 was up‐regulated in human nevus, malignant melanoma and metastatic melanoma. Furthermore, p62 was up‐regulated in melanoma cells relative to normal human epidermal melanocytes, independent of their BRAF or NRAS mutation status. Our results demonstrated that UVA up‐regulates p62 and induces a p62‐Nrf2 positive feedback loop to counteract oxidative stress. Additionally, p62 forms a feedback loop with PTEN in melanoma cells, suggesting p62 functions as an oncogene in UVA‐associated melanoma development and progression.


Scientific Reports | 2017

Epidermal SIRT1 regulates inflammation, cell migration, and wound healing

Lei Qiang; Ashley Sample; Han Liu; Xiaoyang Wu; Yu-Ying He

Sirtuins (SIRT1-7) are NAD-dependent proteins with the enzymatic activity of deacetylases and ADP ribosyltransferases. SIRT1 is the proto member of the proteins in the mammalian sirtuin family and plays multiple roles in aging and disease. Using mice with epidermis-specific SIRT1 deletion, we show that SIRT1 is required for efficient wound healing. SIRT1 deficiency in the epidermis inhibited the regeneration of both the epidermis and the dermal stroma. SIRT1 loss altered the production of many cytokines, inhibited the recruitment of macrophages, neutrophils, and mast cells, the recruitment and activation of fibroblasts, and angiogenesis in the granulation tissue. In keratinocytes, SIRT1 knockdown inhibited EMT, cell migration, and TGF-β signaling. For the first time, using skin-specific mouse model, we demonstrate that epidermal SIRT1 plays a crucial role in wound repair. These findings are novel in understanding how wound healing is regulated. Our findings provide in vivo and in vitro evidence that SIRT1 in the epidermis regulates cell migration, redox response, inflammation, epidermis re-epithelialization, granulation formation, and proper wound healing in mice.


Cancer Research | 2016

Abstract 4060: The role of p62-dependent regulation of COX-2 in UVA response and skin tumor progression

Ashley Sample; Lei Qiang; Baozhong Zhao; Yu-Ying He

Skin cancer is the most common type of cancer in the US, with an estimated 3.5 million cases diagnosed each year. Exposure to ultraviolet (UV) radiation, namely UVA (320-400 nm) and UVB (290-320 nm), is the major risk factor for the development of skin cancer. UVA is 20-fold more abundant in sunlight than UVB and is the major component of tanning beds. However, the mechanism of UVA9s contribution to skin cancer remains unclear. One putative effector of UVA in skin cancer is p62, a selective autophagy cargo protein and substrate. We have found that UVA upregulates p62 expression, independent of autophagy, by inducing transcription of p62. p62 is upregulated in many types of cancer, including squamous cell carcinoma (SCC) and melanoma, and can facilitate the activation of a number of pathways to promote cell proliferation, invasion, inflammation, and survival. We have previously shown that p62 stabilizes Twist1 to promote invasion and proliferation, indicating p62 can function independent of autophagy to promote tumor progression. However, the function of p62 in UVA-induced tumor progression is unknown. Using a candidate gene approach, we identified a novel interaction between p62 and cyclooxygenase-2 (COX-2), which suggests a putative function for p62 in UVA-induced skin cancer. COX-2 is a prostaglandin synthase often overexpressed in cancer, where it correlates with poor prognosis. COX-2 promotes cell proliferation and survival, and inhibition of COX-2 prevents skin cancer development. COX-2 protein expression is induced concomitantly with p62 in response to UVA and knockdown of p62 prevents induction of COX-2 by UVA. As COX-2 transcription is not induced by UVA in skin cancer cells along with p62, we assessed a possible protein-level regulatory mechanism. Co-immunoprecipitation of endogenous p62 and COX-2 in skin cancer cells reveals a novel physical interaction between these proteins. Therefore, we hypothesize that p62 transcription is induced by UVA to promote stability of cyclooxygenase-2 (COX-2) and consequently, tumor progression. This project aims to further understand how UVA regulates p62 and thus COX-2 availability, and the functional significance for skin tumor progression. As both p62 and COX-2 are critical for skin tumor progression, understanding the link between these proteins will unravel a signaling axis central to skin cancer progression. Citation Format: Ashley Sample, Lei Qiang, Baozhong Zhao, Yu-Ying He. The role of p62-dependent regulation of COX-2 in UVA response and skin tumor progression. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4060.

Collaboration


Dive into the Ashley Sample's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lei Qiang

University of Chicago

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
Top Co-Authors

Avatar

Andrew E. Aplin

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge