1
|
Son B, Lee W, Kim H, Shin H, Park HH. Targeted therapy of cancer stem cells: inhibition of mTOR in pre-clinical and clinical research. Cell Death Dis 2024; 15:696. [PMID: 39349424 PMCID: PMC11442590 DOI: 10.1038/s41419-024-07077-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 10/02/2024]
Abstract
Cancer stem cells (CSCs) are a type of stem cell that possesses not only the intrinsic abilities of stem cells but also the properties of cancer cells. Therefore, CSCs are known to have self-renewal and outstanding proliferation capacity, along with the potential to differentiate into specific types of tumor cells. Cancers typically originate from CSCs, making them a significant target for tumor treatment. Among the related cascades of the CSCs, mammalian target of rapamycin (mTOR) pathway is regarded as one of the most important signaling pathways because of its association with significant upstream signaling: phosphatidylinositol 3‑kinase/protein kinase B (PI3K/AKT) pathway and mitogen‑activated protein kinase (MAPK) cascade, which influence various activities of stem cells, including CSCs. Recent studies have shown that the mTOR pathway not only affects generation of CSCs but also the maintenance of their pluripotency. Furthermore, the maintenance of pluripotency or differentiation into specific types of cancer cells depends on the regulation of the mTOR signal in CSCs. Consequently, the clinical potential and importance of mTOR in effective cancer therapy are increasing. In this review, we demonstrate the association between the mTOR pathway and cancer, including CSCs. Additionally, we discuss a new concept for anti-cancer drug development aimed at overcoming existing drawbacks, such as drug resistance, by targeting CSCs through mTOR inhibition.
Collapse
Affiliation(s)
- Boram Son
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul, 02707, Republic of Korea
| | - Wonhwa Lee
- Department of Chemistry, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyeonjeong Kim
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea.
| | - Hee Ho Park
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea.
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
2
|
Gómez-de Castro C, Santos-Juanes R, Nuñez-Gómez B, Fernández-Vega I, Vivanco B, Fernández-Velasco A, Reyes-García S, Carrero-Martín J, García-Pedrero JM, Rodrigo JP, González-Vela MDC, Santos-Juanes J, Galache C. Low-Level Expression of p-S6 Is Associated with Nodal Metastasis in Patients with Head and Neck Cutaneous Squamous Cell Carcinoma. Int J Mol Sci 2024; 25:4304. [PMID: 38673889 PMCID: PMC11049968 DOI: 10.3390/ijms25084304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the second most common form of skin cancer. The incidence of metastasis for cSCC is estimated to be around 1.2-5%. Ribosomal protein S6 (p-S6) and the p21 protein (p21) are two proteins that play central roles in other cancers. These proteins may be equally important in cSCC, and together, these could constitute a good candidate for metastasis risk assessment of these patients. We investigate the relationship of p-S6 and p21 expression with the impact on the prognosis of head and neck cSCC (cSCCHN). p-S6 and p21 expression was analyzed by immunohistochemistry on paraffin-embedded tissue samples from 116 patients with cSCCHN and associations sought with clinical characteristics. Kaplan-Meier estimators and Cox proportional hazard regression models were also used. The expression of p-S6 was significantly inversely associated with tumor thickness, tumor size, desmoplastic growth, pathological stage, perineural invasion and tumor buds. p21 expression was significantly inversely correlated with >6 mm tumor thickness, desmoplastic growth, and perineural invasion. p-S6-negative expression significantly predicted an increased risk of nodal metastasis (HR = 2.63, 95% CI 1.51-4.54; p < 0.001). p21 expression was not found to be a significant risk factor for nodal metastasis. These findings demonstrate that p-S6-negative expression is an independent predictor of nodal metastasis. The immunohistochemical expression of p-S6 might aid in better risk stratification and management of patients with cSCCHN.
Collapse
Affiliation(s)
- Celia Gómez-de Castro
- Grider, Grupo de Investigación en Dermatología, Universidad de Oviedo, 33006 Oviedo, Spain; (C.G.-d.C.); (B.V.); (A.F.-V.); (C.G.)
- Dermatology Division, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (S.R.-G.); (J.C.-M.)
| | - Raquel Santos-Juanes
- Dermatology Area, Department of Medicine, University of Oviedo, 33006 Oviedo, Spain; (R.S.-J.); (B.N.-G.)
| | - Borja Nuñez-Gómez
- Dermatology Area, Department of Medicine, University of Oviedo, 33006 Oviedo, Spain; (R.S.-J.); (B.N.-G.)
| | - Iván Fernández-Vega
- Department of Pathology, Hospital Universitario Central de Asturias, Biobank of the Principality of Asturias (BioPA), 33011 Oviedo, Spain;
- Department of Pathology, University of Oviedo, 33006 Oviedo, Spain
| | - Blanca Vivanco
- Grider, Grupo de Investigación en Dermatología, Universidad de Oviedo, 33006 Oviedo, Spain; (C.G.-d.C.); (B.V.); (A.F.-V.); (C.G.)
- Department of Pathology, Hospital Universitario Central de Asturias, Biobank of the Principality of Asturias (BioPA), 33011 Oviedo, Spain;
- Department of Pathology, University of Oviedo, 33006 Oviedo, Spain
| | - Adela Fernández-Velasco
- Grider, Grupo de Investigación en Dermatología, Universidad de Oviedo, 33006 Oviedo, Spain; (C.G.-d.C.); (B.V.); (A.F.-V.); (C.G.)
- Department of Pathology, Hospital Universitario Central de Asturias, Biobank of the Principality of Asturias (BioPA), 33011 Oviedo, Spain;
- Department of Pathology, University of Oviedo, 33006 Oviedo, Spain
| | - Sebastián Reyes-García
- Dermatology Division, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (S.R.-G.); (J.C.-M.)
| | - Jimena Carrero-Martín
- Dermatology Division, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (S.R.-G.); (J.C.-M.)
| | - Juana M. García-Pedrero
- Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (J.M.G.-P.); (J.P.R.)
- Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33006 Oviedo, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Juan P. Rodrigo
- Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (J.M.G.-P.); (J.P.R.)
- Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33006 Oviedo, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Otolaryngology-Head and Neck Surgery, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | | | - Jorge Santos-Juanes
- Grider, Grupo de Investigación en Dermatología, Universidad de Oviedo, 33006 Oviedo, Spain; (C.G.-d.C.); (B.V.); (A.F.-V.); (C.G.)
- Dermatology Division, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (S.R.-G.); (J.C.-M.)
- Dermatology Area, Department of Medicine, University of Oviedo, 33006 Oviedo, Spain; (R.S.-J.); (B.N.-G.)
- Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (J.M.G.-P.); (J.P.R.)
- Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33006 Oviedo, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Cristina Galache
- Grider, Grupo de Investigación en Dermatología, Universidad de Oviedo, 33006 Oviedo, Spain; (C.G.-d.C.); (B.V.); (A.F.-V.); (C.G.)
- Dermatology Division, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (S.R.-G.); (J.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (J.M.G.-P.); (J.P.R.)
- Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33006 Oviedo, Spain
| |
Collapse
|
3
|
Interaction of Arsenic Exposure and Transcriptomic Profile in Basal Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14225598. [PMID: 36428691 PMCID: PMC9688807 DOI: 10.3390/cancers14225598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/08/2022] [Accepted: 11/12/2022] [Indexed: 11/16/2022] Open
Abstract
Exposure to inorganic arsenic (As) is recognized as risk factor for basal cell carcinoma (BCC). We have followed-up 7000 adults for 6 years who were exposed to As and had manifest As skin toxicity. Of them, 1.7% developed BCC (males = 2.2%, females = 1.3%). In this study, we compared transcriptome-wide RNA sequencing data from the very first 26 BCC cases and healthy skin tissue from independent 16 individuals. Genes in “ cell carcinoma pathway”, “Hedgehog signaling pathway”, and “Notch signaling pathway” were overexpressed in BCC, confirming the findings from earlier studies in BCC in other populations known to be exposed to As. However, we found that the overexpression of these known pathways was less pronounced in patients with high As exposure (urinary As creatinine ratio (UACR) > 192 µg/gm creatinine) than patients with low UACR. We also found that high UACR was associated with impaired DNA replication pathway, cellular response to different DNA damage repair mechanisms, and immune response. Transcriptomic data were not strongly suggestive of great potential for immune checkpoint inhibitors; however, it suggested lower chance of platinum drug resistance in BCC patients with high UACR compared high platinum drug resistance potential in patients with lower UACR.
Collapse
|
4
|
Wang J, Cui B, Chen Z, Ding X. The regulation of skin homeostasis, repair and the pathogenesis of skin diseases by spatiotemporal activation of epidermal mTOR signaling. Front Cell Dev Biol 2022; 10:950973. [PMID: 35938153 PMCID: PMC9355246 DOI: 10.3389/fcell.2022.950973] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
The epidermis, the outmost layer of the skin, is a stratified squamous epithelium that protects the body from the external world. The epidermis and its appendages need constantly renew themselves and replace the damaged tissues caused by environmental assaults. The mechanistic target of rapamycin (mTOR) signaling is a central controller of cell growth and metabolism that plays a critical role in development, homeostasis and diseases. Recent findings suggest that mTOR signaling is activated in a spatiotemporal and context-dependent manner in the epidermis, coordinating diverse skin homeostatic processes. Dysregulation of mTOR signaling underlies the pathogenesis of skin diseases, including psoriasis and skin cancer. In this review, we discuss the role of epidermal mTOR signaling activity and function in skin, with a focus on skin barrier formation, hair regeneration, wound repair, as well as skin pathological disorders. We propose that fine-tuned control of mTOR signaling is essential for epidermal structural and functional integrity.
Collapse
Affiliation(s)
- Juan Wang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Baiping Cui
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Zhongjian Chen
- School of Medicine, Shanghai University, Shanghai, China
- Shanghai Engineering Research Center for External Chinese Medicine, Shanghai, China
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaolei Ding
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
- *Correspondence: Xiaolei Ding,
| |
Collapse
|
5
|
Remenár É, Dóczi R, Dirner A, Sipos A, Perjési A, Tihanyi D, Vodicska B, Lakatos D, Horváth K, Kajáry K, Schwáb R, Déri J, Lengyel CG, Várkondi E, Vályi-Nagy I, Peták I. Lasting Complete Clinical Response of a Recurring Cutaneous Squamous Cell Carcinoma With MEK Mutation and PIK3CA Amplification Achieved by Dual Trametinib and Metformin Therapy. JCO Precis Oncol 2022; 6:e2100344. [PMID: 35005996 DOI: 10.1200/po.21.00344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Éva Remenár
- Buda Hospitaller Order of St John of God, Budapest, Hungary
| | - Róbert Dóczi
- Oncompass Medicine Hungary Ltd, Budapest, Hungary
| | - Anna Dirner
- Oncompass Medicine Hungary Ltd, Budapest, Hungary
| | - Anna Sipos
- Oncompass Medicine Hungary Ltd, Budapest, Hungary
| | | | - Dóra Tihanyi
- Oncompass Medicine Hungary Ltd, Budapest, Hungary
| | | | - Dóra Lakatos
- Oncompass Medicine Hungary Ltd, Budapest, Hungary
| | | | | | - Richárd Schwáb
- Oncompass Medicine Hungary Ltd, Budapest, Hungary.,MiND Klinika Kft, Budapest, Hungary
| | - Júlia Déri
- Oncompass Medicine Hungary Ltd, Budapest, Hungary
| | | | | | - István Vályi-Nagy
- Centrum Hospital of Southern Pest, National Hematology and Infectology Institute, Budapest, Hungary
| | - István Peták
- Oncompass Medicine Hungary Ltd, Budapest, Hungary.,Department of Pharmacology, Semmelweis University, Budapest, Hungary.,Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
6
|
The effects of UVB irradiance on aberrant epidermal proliferation: Novel insights on how to improve currently available sunscreens. Life Sci 2022; 288:120181. [PMID: 34843737 DOI: 10.1016/j.lfs.2021.120181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 11/12/2021] [Accepted: 11/21/2021] [Indexed: 11/23/2022]
Abstract
AIMS Sunscreen use, which prolonged the time required to develop sunburn by reducing the irradiance (mW/cm2) of the UVB radiation, is thought to protect the skin from developing cancers. Recently, in addition to fluence (mJ/cm2), irradiance of the UVB radiation was demonstrated to play an important role leading to photocarcinogenesis of the skin. After equivalent fluence of UVB exposure, enhanced aberrant keratinocyte proliferation contributes significantly to the photocarcinogenic capacity of low irradiance (LI) UVB as compared to its high irradiance (HI) UVB counterpart. However, the mechanism involved remains unclear. MAIN METHODS Relevant cell and animal models were employed to investigate the effects of equivalent UVB fluence administered at HI or LI on keratinocyte proliferation. Additionally, the mechanisms involved were also explored. KEY FINDINGS We found that at equivalent fluence, LIUVB induces significantly higher reactive oxidative species (ROS) production, cell proliferation, as well as phosphorylated AKT (pAKT) expression in both cell and animal models as compare to its HIUVB counterpart. Pretreating cultured keratinocytes with antioxidant or AKT inhibitor significantly reduced the UVB-induced ROS, cell proliferation, and pAKT expression. Additionally, these pretreatments abrogate the difference between the LI and HIUVB treated keratinocytes. Similar findings were noted using animal model treated with AKT inhibitor. SIGNIFICANCE In summary, at equivalent fluence, LIUVB induces significantly more aberrant epidermal proliferation via enhanced ROS and pAKT signaling. Reducing UVB-induced AKT phosphorylation presents a novel strategy to improve the protective capacity of the currently available sunscreens.
Collapse
|
7
|
Sanz Ressel BL, Massone AR, Barbeito CG. Persistent activation of the mammalian target of rapamycin signalling pathway in cutaneous squamous cell carcinomas in cats. Vet Dermatol 2021; 32:675-e180. [PMID: 34240493 DOI: 10.1111/vde.13001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 04/05/2021] [Accepted: 04/09/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Cutaneous squamous cell carcinoma (CSCC) represents the most common malignant tumour of the feline skin. Emerging evidence suggests that the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) signalling pathway may represent a potential target for pharmacological intervention in human and canine CSCC. HYPOTHESIS/OBJECTIVES The present study aimed to explore the expression pattern and status of activation of relevant signalling proteins of the PI3K/Akt/mTOR signalling pathway in feline CSCC. METHODS AND MATERIALS The expression of pEGFRTyr1068 , pAktSer473 , pS6Ser235/236 combined with Ki-67, and the tumour suppressor protein PTEN was evaluated by immunohistochemical analysis in 45 samples of feline CSCC, using a tissue microarray. RESULTS The immunodetection using phosphospecific antibodies to detect the activated forms of signalling proteins showed that the PI3K/Akt/mTOR signalling pathway is frequently activated in feline CSCCs, and may be independent of the activation of EGFR. The results also showed that PTEN expression is not significantly altered in feline CSCCs. CONCLUSIONS AND CLINICAL IMPORTANCE Our study shows that the persistent activation of the PI3K/Akt/mTOR signalling pathway represents a key event in feline CSCC, pointing to this signalling pathway being a potential therapeutic target in feline patients with CSCC.
Collapse
Affiliation(s)
- Berenice L Sanz Ressel
- Laboratorio de Histología y Embriología Descriptiva, Experimental y Comparada (LHYEDEC), Facultad de Ciencias Veterinarias (FCV), Calle 60 y 118, La Plata, Buenos Aires, CP 1900, Argentina.,FCV - Consejo Nacional de Investigaciones Científicas y Tecnicas (CONICET), Calle 60 y 118, La Plata, Buenos Aires, CP 1900, Argentina
| | - Adriana R Massone
- FCV - Laboratorio de Patología Especial Veterinaria Dr. Bernardo Epstein, Universidad Nacional de La Plata (UNLP), Calle 60 y 118, CP 1900, La Plata, Buenos Aires, Argentina
| | - Claudio G Barbeito
- Laboratorio de Histología y Embriología Descriptiva, Experimental y Comparada (LHYEDEC), Facultad de Ciencias Veterinarias (FCV), Calle 60 y 118, La Plata, Buenos Aires, CP 1900, Argentina.,FCV - Consejo Nacional de Investigaciones Científicas y Tecnicas (CONICET), Calle 60 y 118, La Plata, Buenos Aires, CP 1900, Argentina
| |
Collapse
|
8
|
Mercurio L, Albanesi C, Madonna S. Recent Updates on the Involvement of PI3K/AKT/mTOR Molecular Cascade in the Pathogenesis of Hyperproliferative Skin Disorders. Front Med (Lausanne) 2021; 8:665647. [PMID: 33996865 PMCID: PMC8119789 DOI: 10.3389/fmed.2021.665647] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
PhosphoInositide-3 Kinase (PI3K) represents a family of different classes of kinases which control multiple biological processes in mammalian cells, such as cell growth, proliferation, and survival. Class IA PI3Ks, the main regulators of proliferative signals, consists of a catalytic subunit (α, β, δ) that binds p85 regulatory subunit and mediates activation of AKT and mammalian Target Of Rapamycin (mTOR) pathways and regulation of downstream effectors. Dysregulation of PI3K/AKT/mTOR pathway in skin contributes to several pathological conditions characterized by uncontrolled proliferation, including skin cancers, psoriasis, and atopic dermatitis (AD). Among cutaneous cancers, basal cell carcinoma (BCC) and cutaneous squamous cell carcinoma (cSCC) display PI3K/AKT/mTOR signaling hyperactivation, implicated in hyperproliferation, and tumorigenesis, as well as in resistance to apoptosis. Upregulation of mTOR signaling proteins has also been reported in psoriasis, in association with enhanced proliferation, defective keratinocyte differentiation, senescence-like growth arrest, and resistance to apoptosis, accounting for major parts of the overall disease phenotypes. On the contrary, PI3K/AKT/mTOR role in AD is less characterized, even though recent evidence demonstrates the relevant function for mTOR pathway in the regulation of epidermal barrier formation and stratification. In this review, we provide the most recent updates on the role and function of PI3K/AKT/mTOR molecular axis in the pathogenesis of different hyperproliferative skin disorders, and highlights on the current status of preclinical and clinical studies on PI3K-targeted therapies.
Collapse
Affiliation(s)
- Laura Mercurio
- Laboratory of Experimental Immunology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Rome, Italy
| | - Cristina Albanesi
- Laboratory of Experimental Immunology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Rome, Italy
| | - Stefania Madonna
- Laboratory of Experimental Immunology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Rome, Italy
| |
Collapse
|
9
|
Growth and Viability of Cutaneous Squamous Cell Carcinoma Cell Lines Display Different Sensitivities to Isoform-Specific Phosphoinositide 3-Kinase Inhibitors. Int J Mol Sci 2021; 22:ijms22073567. [PMID: 33808215 PMCID: PMC8036316 DOI: 10.3390/ijms22073567] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/19/2021] [Indexed: 12/20/2022] Open
Abstract
Cutaneous squamous cell carcinomas (cSCCs) account for about 20% of keratinocyte carcinomas, the most common cancer in the UK. Therapeutic options for cSCC patients who develop metastasis are limited and a better understanding of the biochemical pathways involved in cSCC development/progression is crucial to identify novel therapeutic targets. Evidence indicates that the phosphoinositide 3-kinases (PI3Ks)/Akt pathway plays an important role, in particular in advanced cSCC. Questions remain of whether all four PI3K isoforms able to activate Akt are involved and whether selective inhibition of specific isoform(s) might represent a more targeted strategy. Here we determined the sensitivity of four patient-derived cSCC cell lines to isoform-specific PI3K inhibitors to start investigating their potential therapeutic value in cSCC. Parallel experiments were performed in immortalized keratinocyte cell lines. We observed that pan PI3Ks inhibition reduced the growth/viability of all tested cell lines, confirming the crucial role of this pathway. Selective inhibition of the PI3K isoform p110α reduced growth/viability of keratinocytes and of two cSCC cell lines while affecting the other two only slightly. Importantly, p110α inhibition reduced Akt phosphorylation in all cSCC cell lines. These data indicate that growth and viability of the investigated cSCC cells display differential sensitivity to isoform-specific PI3K inhibitors.
Collapse
|
10
|
Chamcheu JC, Esnault S, Adhami VM, Noll AL, Banang-Mbeumi S, Roy T, Singh SS, Huang S, Kousoulas KG, Mukhtar H. Fisetin, a 3,7,3',4'-Tetrahydroxyflavone Inhibits the PI3K/Akt/mTOR and MAPK Pathways and Ameliorates Psoriasis Pathology in 2D and 3D Organotypic Human Inflammatory Skin Models. Cells 2019; 8:E1089. [PMID: 31540162 PMCID: PMC6770767 DOI: 10.3390/cells8091089] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/07/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023] Open
Abstract
Psoriasis is a chronic immune-mediated skin disease that involves the interaction of immune and skin cells, and is characterized by cytokine-driven epidermal hyperplasia, deviant differentiation, inflammation, and angiogenesis. Because the available treatments for psoriasis have significant limitations, dietary products are potential natural sources of therapeutic molecules, which can repair the molecular defects associated with psoriasis and could possibly be developed for its management. Fisetin (3,7,3',4'-tetrahydroxyflavone), a phytochemical naturally found in pigmented fruits and vegetables, has demonstrated proapoptotic and antioxidant effects in several malignancies. This study utilized biochemical, cellular, pharmacological, and tissue engineering tools to characterize the effects of fisetin on normal human epidermal keratinocytes (NHEKs), peripheral blood mononuclear cells (PBMC), and CD4+ T lymphocytes in 2D and 3D psoriasis-like disease models. Fisetin treatment of NHEKs dose- and time-dependently induced differentiation and inhibited interleukin-22-induced proliferation, as well as activation of the PI3K/Akt/mTOR pathway. Fisetin treatment of TNF-α stimulated NHEKs also significantly inhibited the activation of p38 and JNK, but had enhanced effect on ERK1/2 (MAPK). In addition, fisetin treatment significantly decreased the secretion of Th1/Th-17 pro-inflammatory cytokines, particularly IFN-γ and IL-17A by 12-O-tetradecanolylphorbol 13-acetate (TPA)-stimulated NHEKs and anti-CD3/CD28-activated human PBMCs. Furthermore, we established the in vivo relevance of fisetin functions, using a 3D full-thickness human skin model of psoriasis (FTRHSP) that closely mimics in vivo human psoriatic skin lesions. Herein, fisetin significantly ameliorated psoriasis-like disease features, and decreased the production of IL-17 by CD4+ T lymphocytes co-cultured with FTRHSP. Collectively, our data identify the prodifferentiative, antiproliferative, and anti-inflammatory effects of fisetin, via modulation of the PI3K-Akt-mTOR and p38/JNK pathways and the production of cytokines in 2D and 3D human skin models of psoriasis. These results suggest that fisetin has a great potential to be developed as an effective and inexpensive agent for the treatment of psoriasis and other related inflammatory skin disorders.
Collapse
Affiliation(s)
- Jean Christopher Chamcheu
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA.
| | - Stephane Esnault
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, Madison, WI 53706, USA.
| | - Vaqar M Adhami
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA.
| | - Andrea L Noll
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, Madison, WI 53706, USA.
| | - Sergette Banang-Mbeumi
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA.
| | - Tithi Roy
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA.
| | - Sitanshu S Singh
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA.
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA.
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA.
| | - Konstantin G Kousoulas
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Hasan Mukhtar
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, Madison, WI 53706, USA.
| |
Collapse
|
11
|
Chamcheu JC, Roy T, Uddin MB, Banang-Mbeumi S, Chamcheu RCN, Walker AL, Liu YY, Huang S. Role and Therapeutic Targeting of the PI3K/Akt/mTOR Signaling Pathway in Skin Cancer: A Review of Current Status and Future Trends on Natural and Synthetic Agents Therapy. Cells 2019; 8:cells8080803. [PMID: 31370278 PMCID: PMC6721560 DOI: 10.3390/cells8080803] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/16/2022] Open
Abstract
The mammalian or mechanistic target of rapamycin (mTOR) and associated phosphatidyl-inositiol 3-kinase (PI3K)/protein kinase B (Akt) pathways regulate cell growth, differentiation, migration, and survival, as well as angiogenesis and metabolism. Dysregulation of these pathways is frequently associated with genetic/epigenetic alterations and predicts poor treatment outcomes in a variety of human cancers including cutaneous malignancies like melanoma and non-melanoma skin cancers. Recently, the enhanced understanding of the molecular and genetic basis of skin dysfunction in patients with skin cancers has provided a strong basis for the development of novel therapeutic strategies for these obdurate groups of skin cancers. This review summarizes recent advances in the roles of PI3K/Akt/mTOR and their targets in the development and progression of a broad spectrum of cutaneous cancers and discusses the current progress in preclinical and clinical studies for the development of PI3K/Akt/mTOR targeted therapies with nutraceuticals and synthetic small molecule inhibitors.
Collapse
Affiliation(s)
| | - Tithi Roy
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | | | - Sergette Banang-Mbeumi
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
- Division for Research and Innovation, POHOFI Inc., P.O. Box 44067, Madison, WI 53744, USA
- School of Nursing and Allied Health Sciences, Louisiana Delta Community College, Monroe, LA 71203, USA
| | | | - Anthony L Walker
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | - Yong-Yu Liu
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
| |
Collapse
|
12
|
Khandelwal AR, Kent B, Hillary S, Alam MM, Ma X, Gu X, DiGiovanni J, Nathan CAO. Fibroblast growth factor receptor promotes progression of cutaneous squamous cell carcinoma. Mol Carcinog 2019; 58:1715-1725. [PMID: 31254372 DOI: 10.1002/mc.23012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/15/2019] [Accepted: 03/19/2019] [Indexed: 12/14/2022]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is a keratinocyte-derived invasive and metastatic tumor of the skin. It is the second-most commonly diagnosed form of skin cancer striking 200 000 Americans annually. Further, in organ transplant patients, there is a 65- to 100-fold increased incidence of cSCC compared to the general population. Excision of cSCC of the head and neck results in significant facial disfigurement. Therefore, increased understanding of the mechanisms involved in the pathogeneses of cSCC could identify means to prevent, inhibit, and reverse this process. In our previous studies, inhibition of fibroblast growth factor receptor (FGFR) significantly decreased ultraviolet B-induced epidermal hyperplasia and hyperproliferation in SKH-1 mice, suggesting an important role for FGFR signaling in skin cancer development. However, the role of FGFR signaling in the progression of cSCC is not yet elucidated. Analysis of the expression of FGFR in cSCC cells and normal epidermal keratinocytes revealed protein overexpression and increased FGFR2 activation in cSCC cells compared to normal keratinocytes. Further, tumor cell-specific overexpression of FGFR2 was detected in human cSCCs, whereas the expression of FGFR2 was low in premalignant lesions and normal skin. Pretreatment with the pan-FGFR inhibitor; AZD4547 significantly decreased cSCC cell-cycle traverse, proliferation, migration, and motility. Interestingly, AZD4547 also significantly downregulated mammalian target of rapamycin complex 1 and AKT activation in cSCC cells, suggesting an important role of these signaling pathways in FGFR-mediated effects. To further bolster the in vitro studies, NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice with SCC12A tumor xenografts treated with AZD4547 (15 mg/kg/bw, twice weekly oral gavage) exhibited significantly decreased tumor volume compared to the vehicle-only treatment group. The current studies provide mechanistic evidence for the role of FGFR and selectively FGFR2 in the early progression of cSCC and identifies FGFR as a putative therapeutic target in the treatment of skin cancer.
Collapse
Affiliation(s)
- Alok R Khandelwal
- Department of Otolaryngology, Head and Neck Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana.,Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Burton Kent
- Department of Otolaryngology, Head and Neck Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Savage Hillary
- Department of Otolaryngology, Head and Neck Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Md Maksudul Alam
- Department of Otolaryngology, Head and Neck Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Xiaohua Ma
- Department of Otolaryngology, Head and Neck Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Xin Gu
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - John DiGiovanni
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Cherie-Ann O Nathan
- Department of Otolaryngology, Head and Neck Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana.,Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, Louisiana.,Department of Surgery, Overton Brooks Veterans Affairs Hospital, Shreveport, Louisiana
| |
Collapse
|
13
|
Stump CL, Feehan RP, Jordan T, Shantz LM, Nowotarski SL. Knocking down raptor in human keratinocytes affects ornithine decarboxylase in a post-transcriptional Manner following ultraviolet B exposure. Amino Acids 2019; 52:141-149. [PMID: 30972602 DOI: 10.1007/s00726-019-02732-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/04/2019] [Indexed: 12/19/2022]
Abstract
Non-melanoma skin cancer (NMSC) is the most common form of cancer. Ultraviolet-B (UVB) radiation has been shown to be a complete carcinogen in the development of NMSC. The mammalian target of rapamycin complex 1 (mTORC1) is upregulated by UVB. Ornithine decarboxylase (ODC), the first enzyme of the polyamine biosynthetic pathway, is also upregulated in response to UVB. However, the interplay between these two pathways after UVB exposure remains unclear. The studies described here compare mRNA stability between normal human keratinocytes (HaCaT cells) and HaCaT cells with low levels of raptor to investigate whether the induction of ODC by UVB is dependent on mTORC1. We show that the knockdown of mTORC1 activity led to decreased levels of ODC protein both before and after exposure to 20 mJ/cm2 UVB. ODC mRNA was less stable in cells with decreased mTORC1 activity. Polysome profiles revealed that the initiation of ODC mRNA translation did not change in UVB-treated cells. We have shown that the ODC transcript is stabilized by the RNA-binding protein human antigen R (HuR). To expand these studies, we investigated whether HuR functions to regulate ODC mRNA stability in human keratinocytes exposed to UVB. We show an increased cytoplasmic localization of HuR after UVB exposure in wild-type cells. The ablation of HuR via CRISPR/Cas9 did not alter the stability of the ODC message, suggesting the involvement of other trans-acting factors. These data suggest that in human keratinocytes, ODC mRNA stability is regulated, in part, by an mTORC1-dependent mechanism after UVB exposure.
Collapse
Affiliation(s)
- Coryn L Stump
- Division of Science, The Pennsylvania State University Berks Campus, Reading, PA, 19610, USA
| | - Robert P Feehan
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Torey Jordan
- Division of Science, The Pennsylvania State University Berks Campus, Reading, PA, 19610, USA
| | - Lisa M Shantz
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Shannon L Nowotarski
- Division of Science, The Pennsylvania State University Berks Campus, Reading, PA, 19610, USA.
| |
Collapse
|
14
|
Hu SCS, Lai YC, Lin CL, Tzeng WS, Yen FL. Inclusion complex of saikosaponin-d with hydroxypropyl-β-cyclodextrin: Improved physicochemical properties and anti-skin cancer activity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 57:174-182. [PMID: 30776588 DOI: 10.1016/j.phymed.2018.11.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 11/05/2018] [Accepted: 11/07/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Saikosaponin-d (SSD) is a triterpene saponin isolated from Bupleurum plants. It has been shown to exhibit antioxidant, anti-inflammatory, and anticancer activities. However, its biomedical applications are limited by its poor water solubility. Cyclodextrins are highly water soluble oligosaccharide compounds which can form inclusion complexes with lipophilic drugs. PURPOSE We complexed SSD with hydroxypropyl-β-cyclodextrin (HPBCD) in various ratios to form SSD-HPBCD inclusion complexes. The inclusion complexes were evaluated for their solubility, physicochemical properties and cytotoxic effects in cutaneous squamous cell carcinoma cells. METHODS Surface morphology of pure SSD and SSD-HPBCD inclusion complexes was evaluated by scanning electron microscopy. Crystalline structure was determined by X-ray diffractometry. Intermolecular hydrogen bond formation between SSD and HPBCD was investigated by Fourier transform infrared spectroscopy. Human cutaneous squamous cell carcinoma HSC-1 cell viability was determined by the MTS assay, and cell apoptosis by the caspase 3/7 assay. Signal transduction pathways were investigated by Western blotting. RESULTS SSD-HPBCD inclusion complexes showed greatly increased water solubility. This was associated with an improvement in physicochemical properties, including transformation of crystalline structure to amorphous form, and formation of hydrogen bonds between SSD and HPBCD. In addition, SSD-HPBCD inclusion complexes induced apoptosis in HSC-1 cells, and this was mediated through activation of MAPK and suppression of Akt-mTOR signaling pathways. CONCLUSION SSD-HPBCD inclusion complex shows improvement in water solubility and physicochemical properties, and exhibits anticancer effects against cutaneous squamous cell carcinoma cells. Therefore, it may be a potential drug formulation for the treatment of skin cancer.
Collapse
Affiliation(s)
- Stephen Chu-Sung Hu
- Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Chien Lai
- Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chi-Ling Lin
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Dermatology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung, Taiwan
| | - Wen-Sheng Tzeng
- Department of Radiology, Pingtung Christian Hospital, Pingtung, Taiwan.
| | - Feng-Lin Yen
- Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan.
| |
Collapse
|
15
|
Sanz Ressel B, Massone A, Barbeito C. Immunohistochemical expression of selected phosphoproteins of the mTOR signalling pathway in canine cutaneous squamous cell carcinoma. Vet J 2019; 245:41-48. [DOI: 10.1016/j.tvjl.2018.12.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 11/15/2018] [Accepted: 12/21/2018] [Indexed: 10/27/2022]
|
16
|
Zhang M, Zhang X. The role of PI3K/AKT/FOXO signaling in psoriasis. Arch Dermatol Res 2018; 311:83-91. [PMID: 30483877 DOI: 10.1007/s00403-018-1879-8] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/26/2018] [Accepted: 11/18/2018] [Indexed: 12/12/2022]
Abstract
Phosphatidylinositol 3-kinase (PI3K) and protein kinase B (AKT) signaling pathway play a central role in multiple cellular functions such as cell proliferation and survival. The forkhead box O (FOXO) transcription factors are negatively regulated by the PI3K/AKT signaling pathway and considered to have inhibitory effect on cell proliferation. Psoriasis is a multifactorial disease with a strong genetic background and characterized by hyperproliferative keratinocyte. PI3K signaling regulates proliferation of keratinocyte by activating AKT and other targets, and by inducing FOXO downregulation. The amplification of PI3K and AKT and the loss of the FOXO are gradually being recognized in psoriatic lesions. The upstream and downstream of PI3K/AKT signaling molecules such as tumor suppressor phosphatase and tensin homolog (PTEN) and mammalian target of Rapamycin (mTOR), respectively, are also frequently altered in psoriasis. In this review, we highlight the recent studies on the roles and mechanisms of PI3K and AKT in regulating hyperproliferation of keratinocyte, and the roles of the downstream targets FOXO in psoriasis. Finally, we summarized that PI3K/AKT/FOXO signaling and its upstream and downstream molecule which could be underlying therapeutic target for psoriasis. This article is part of a special issue entitled: PI3K-AKT-FOXO axis in psoriasis.
Collapse
Affiliation(s)
- Miao Zhang
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaoyan Zhang
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
17
|
Protein activation mapping of human sun-protected epidermis after an acute dose of erythemic solar simulated light. NPJ Precis Oncol 2017; 1. [PMID: 29167824 PMCID: PMC5695572 DOI: 10.1038/s41698-017-0037-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Ultraviolet radiation is an important etiologic factor in skin cancer and a better understanding of how solar stimulated light (SSL) affects signal transduction pathways in human skin which is needed in further understanding activated networks that could be targeted for skin cancer prevention. We utilized Reverse Phase Protein Microarray Analysis (RPPA), a powerful technology that allows for broad-scale and quantitative measurement of the activation/phosphorylation state of hundreds of key signaling proteins and protein pathways in sun-protected skin after an acute dose of two minimal erythema dose (MED) of SSL. RPPA analysis was used to map the altered cell signaling networks resulting from acute doses of solar simulated radiation (SSL). To that end, we exposed sun-protected skin in volunteers to acute doses of two MED of SSL and collected biopsies pre-SSL and post-SSL irradiation. Frozen biopsies were subjected to laser capture microdissection (LCM) and then assessed by RPPA. The activation/phosphorylation or total levels of 128 key signaling proteins and drug targets were selected for statistical analysis. Coordinate network-based analysis was performed on specific signaling pathways that included the PI3k/Akt/mTOR and Ras/Raf/MEK/ERK pathways. Overall, we found early and sustained activation of the PI3K-AKT-mTOR and MAPK pathways. Cell death and apoptosis-related proteins were activated at 5 and 24 h. Ultimately, expression profile patterns of phosphorylated proteins in the epidermal growth factor receptor(EGFR), AKT, mTOR, and other relevant pathways may be used to determine pharmacodynamic activity of new and selective topical chemoprevention agents administered in a test area exposed to SSL to determine drug-induced attenuation or reversal of skin carcinogenesis pathways. Skin exposure to ultraviolet radiation leads to the activation of proteins involved in carcinogenic pathways. Janine Einspahr and Clara Curiel-Lewandrowski of the Arizona Cancer Center and colleagues in the US exposed normally ultraviolet protected skin of 12 individuals to two times the dose of solar-simulated light needed to induce redness. Skin biopsies were taken before and after exposure and 128 proteins known to be involved in key cancer signaling pathways were examined using ‘reverse phase protein microarray analysis’. They found early and sustained activation of multiple signaling pathways, in addition to activation of cell death and apoptosis-related proteins. The study may serve as a model for investigating the pathways involved in chronic or ultraviolet-induced carcinogenesis, which may ultimately lead to the development of targeted therapies to attenuate or reverse skin cancer pathways.
Collapse
|
18
|
Phosphoinositide 3-Kinase-Dependent Signalling Pathways in Cutaneous Squamous Cell Carcinomas. Cancers (Basel) 2017; 9:cancers9070086. [PMID: 28696382 PMCID: PMC5532622 DOI: 10.3390/cancers9070086] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/26/2017] [Accepted: 07/03/2017] [Indexed: 01/11/2023] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) derives from keratinocytes in the epidermis and accounts for 15–20% of all cutaneous malignancies. Although it is usually curable by surgery, 5% of these tumours metastasise leading to poor prognosis mostly because of a lack of therapies and validated biomarkers. As the incidence rate is rising worldwide it has become increasingly important to better understand the mechanisms involved in cSCC development and progression in order to develop therapeutic strategies. Here we discuss some of the evidence indicating that activation of phosphoinositide 3-kinases (PI3Ks)-dependent signalling pathways (in particular the PI3Ks targets Akt and mTOR) has a key role in cSCC. We further discuss available data suggesting that inhibition of these pathways can be beneficial to counteract the disease. With the growing number of different inhibitors currently available, it would be important to further investigate the specific contribution of distinct components of the PI3Ks/Akt/mTOR pathways in order to identify the most promising molecular targets and the best strategy to inhibit cSCC.
Collapse
|
19
|
Inflammation dependent mTORC1 signaling interferes with the switch from keratinocyte proliferation to differentiation. PLoS One 2017; 12:e0180853. [PMID: 28700632 PMCID: PMC5507280 DOI: 10.1371/journal.pone.0180853] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 06/06/2017] [Indexed: 12/13/2022] Open
Abstract
Psoriasis is a frequent and often severe inflammatory skin disease, characterized by altered epidermal homeostasis. Since we found previously that Akt/mTOR signaling is hyperactivated in psoriatic skin, we aimed at elucidating the role of aberrant mTORC1 signaling in this disease. We found that under healthy conditions mTOR signaling was shut off when keratinocytes switch from proliferation to terminal differentiation. Inflammatory cytokines (IL-1β, IL-17A, TNF-α) induced aberrant mTOR activity which led to enhanced proliferation and reduced expression of differentiation markers. Conversely, regular differentiation could be restored if mTORC1 signaling was blocked. In mice, activation of mTOR through the agonist MHY1485 also led to aberrant epidermal organization and involucrin distribution. In summary, these results not only identify mTORC1 as an important signal integrator pivotal for the cells fate to either proliferate or differentiate, but emphasize the role of inflammation-dependent mTOR activation as a psoriatic pathomechanism.
Collapse
|
20
|
Chamcheu JC, Adhami VM, Esnault S, Sechi M, Siddiqui IA, Satyshur KA, Syed DN, Dodwad SJM, Chaves-Rodriquez MI, Longley BJ, Wood GS, Mukhtar H. Dual Inhibition of PI3K/Akt and mTOR by the Dietary Antioxidant, Delphinidin, Ameliorates Psoriatic Features In Vitro and in an Imiquimod-Induced Psoriasis-Like Disease in Mice. Antioxid Redox Signal 2017; 26:49-69. [PMID: 27393705 PMCID: PMC5206770 DOI: 10.1089/ars.2016.6769] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AIM The treatment of psoriasis remains elusive, underscoring the need for identifying novel disease targets and mechanism-based therapeutic approaches. We recently reported that the PI3K/Akt/mTOR pathway that is frequently deregulated in many malignancies is also clinically relevant for psoriasis. We also provided rationale for developing delphinidin (Del), a dietary antioxidant for the management of psoriasis. This study utilized high-throughput biophysical and biochemical approaches and in vitro and in vivo models to identify molecular targets regulated by Del in psoriasis. RESULTS A kinome-level screen and Kds analyses against a panel of 102 human kinase targets showed that Del binds to three lipid (PIK3CG, PIK3C2B, and PIK3CA) and six serine/threonine (PIM1, PIM3, mTOR, S6K1, PLK2, and AURKB) kinases, five of which belong to the PI3K/Akt/mTOR pathway. Surface plasmon resonance and in silico molecular modeling corroborated Del's direct interactions with three PI3Ks (α/c2β/γ), mTOR, and p70S6K. Del treatment of interleukin-22 or TPA-stimulated normal human epidermal keratinocytes (NHEKs) significantly inhibited proliferation, activation of PI3K/Akt/mTOR components, and secretion of proinflammatory cytokines and chemokines. To establish the in vivo relevance of these findings, an imiquimod (IMQ)-induced Balb/c mouse psoriasis-like skin model was employed. Topical treatment of Del significantly decreased (i) hyperproliferation and epidermal thickness, (ii) skin infiltration by immune cells, (iii) psoriasis-related cytokines/chemokines, (iv) PI3K/Akt/mTOR pathway activation, and (v) increased differentiation when compared with controls. Innovation and Conclusion: Our observation that Del inhibits key kinases involved in psoriasis pathogenesis and alleviates IMQ-induced murine psoriasis-like disease suggests a novel PI3K/AKT/mTOR pathway modulator that could be developed to treat psoriasis. Antioxid. Redox Signal. 26, 49-69.
Collapse
Affiliation(s)
- Jean Christopher Chamcheu
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Vaqar M Adhami
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Stephane Esnault
- 2 Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Mario Sechi
- 3 Department of Chemistry and Pharmacy, University of Sassari , Sassari, Italy
| | - Imtiaz A Siddiqui
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Kenneth A Satyshur
- 4 Small Molecule Screening Facility, Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin.,5 Middleton VA Medical Center , Madison, Wisconsin
| | - Deeba N Syed
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Shah-Jahan M Dodwad
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Maria-Ines Chaves-Rodriquez
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin.,6 Centro de Investigación en Biotecnología Instituto Tecnológico de Costa Rica , Cartago, Republica de Costa Rica
| | - B Jack Longley
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Gary S Wood
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| | - Hasan Mukhtar
- 1 Department of Dermatology, School of Medicine and Public Health, University of Wisconsin , Madison, Wisconsin
| |
Collapse
|
21
|
Voiculescu V, Calenic B, Ghita M, Lupu M, Caruntu A, Moraru L, Voiculescu S, Ion A, Greabu M, Ishkitiev N, Caruntu C. From Normal Skin to Squamous Cell Carcinoma: A Quest for Novel Biomarkers. DISEASE MARKERS 2016; 2016:4517492. [PMID: 27642215 PMCID: PMC5011506 DOI: 10.1155/2016/4517492] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/25/2016] [Indexed: 12/14/2022]
Abstract
Squamous cells carcinoma (SCC) is the second most frequent of the keratinocyte-derived malignancies after basal cell carcinoma and is associated with a significant psychosocial and economic burden for both the patient himself and society. Reported risk factors for the malignant transformation of keratinocytes and development of SCC include ultraviolet light exposure, followed by chronic scarring and inflammation, exposure to chemical compounds (arsenic, insecticides, and pesticides), and immune-suppression. Despite various available treatment methods and recent advances in noninvasive or minimal invasive diagnostic techniques, the risk recurrence and metastasis are far from being negligible, even in patients with negative histological margins and lymph nodes. Analyzing normal, dysplastic, and malignant keratinocyte proteome holds special promise for novel biomarker discovery in SCC that could be used in the future for early detection, risk assessment, tumor monitoring, and development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Vlad Voiculescu
- Department of Dermatology and Allergology, Elias Emergency University Hospital, Bucharest, Romania
| | - Bogdan Calenic
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - Mihaela Ghita
- Dermatology Research Laboratory, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - Mihai Lupu
- Department of Dermatology and Allergology, Elias Emergency University Hospital, Bucharest, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, Bucharest, Romania
| | - Liliana Moraru
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, Bucharest, Romania
| | - Suzana Voiculescu
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - Alexandra Ion
- Department of Dermatology and Allergology, Elias Emergency University Hospital, Bucharest, Romania
| | - Maria Greabu
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - Nikolay Ishkitiev
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine, Medical University, Sofia, Bulgaria
| | - Constantin Caruntu
- Dermatology Research Laboratory, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
22
|
Bridgeman BB, Wang P, Ye B, Pelling JC, Volpert OV, Tong X. Inhibition of mTOR by apigenin in UVB-irradiated keratinocytes: A new implication of skin cancer prevention. Cell Signal 2016; 28:460-468. [PMID: 26876613 DOI: 10.1016/j.cellsig.2016.02.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 02/10/2016] [Accepted: 02/10/2016] [Indexed: 12/18/2022]
Abstract
Ultraviolet B (UVB) radiation is the major environmental risk factor for developing skin cancer, the most common cancer worldwide, which is characterized by aberrant activation of Akt/mTOR (mammalian target of rapamycin). Importantly, the link between UV irradiation and mTOR signaling has not been fully established. Apigenin is a naturally occurring flavonoid that has been shown to inhibit UV-induced skin cancer. Previously, we have demonstrated that apigenin activates AMP-activated protein kinase (AMPK), which leads to suppression of basal mTOR activity in cultured keratinocytes. Here, we demonstrated that apigenin inhibited UVB-induced mTOR activation, cell proliferation and cell cycle progression in mouse skin and in mouse epidermal keratinocytes. Interestingly, UVB induced mTOR signaling via PI3K/Akt pathway, however, the inhibition of UVB-induced mTOR signaling by apigenin was not Akt-dependent. Instead, it was driven by AMPK activation. In addition, mTOR inhibition by apigenin in keratinocytes enhanced autophagy, which was responsible, at least in part, for the decreased proliferation in keratinocytes. In contrast, apigenin did not alter UVB-induced apoptosis. Taken together, our results indicate the important role of mTOR inhibition in UVB protection by apigenin, and provide a new target and strategy for better prevention of UV-induced skin cancer.
Collapse
Affiliation(s)
- Bryan B Bridgeman
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Pu Wang
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Boping Ye
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jill C Pelling
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Olga V Volpert
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Xin Tong
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
23
|
Eukaryotic initiation factor 4E-binding protein 1 (4E-BP1): a master regulator of mRNA translation involved in tumorigenesis. Oncogene 2016; 35:4675-88. [DOI: 10.1038/onc.2015.515] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 12/11/2015] [Accepted: 12/11/2015] [Indexed: 01/17/2023]
|
24
|
Dickinson SE, Janda J, Criswell J, Blohm-Mangone K, Olson ER, Liu Z, Barber C, Petricoin EF, Calvert VS, Einspahr J, Dickinson JE, Stratton SP, Curiel-Lewandrowski C, Saboda K, Hu C, Bode AM, Dong Z, Alberts DS, Timothy Bowden G. Inhibition of Akt Enhances the Chemopreventive Effects of Topical Rapamycin in Mouse Skin. Cancer Prev Res (Phila) 2016; 9:215-24. [PMID: 26801880 DOI: 10.1158/1940-6207.capr-15-0419] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/01/2016] [Indexed: 11/16/2022]
Abstract
The PI3Kinase/Akt/mTOR pathway has important roles in cancer development for multiple tumor types, including UV-induced nonmelanoma skin cancer. Immunosuppressed populations are at increased risk of aggressive cutaneous squamous cell carcinoma (SCC). Individuals who are treated with rapamycin (sirolimus, a classical mTOR inhibitor) have significantly decreased rates of developing new cutaneous SCCs compared with those that receive traditional immunosuppression. However, systemic rapamycin use can lead to significant adverse events. Here, we explored the use of topical rapamycin as a chemopreventive agent in the context of solar-simulated light (SSL)-induced skin carcinogenesis. In SKH-1 mice, topical rapamycin treatment decreased tumor yields when applied after completion of 15 weeks of SSL exposure compared with controls. However, applying rapamycin during SSL exposure for 15 weeks, and continuing for 10 weeks after UV treatment, increased tumor yields. We also examined whether a combinatorial approach might result in more significant tumor suppression by rapamycin. We validated that rapamycin causes increased Akt (S473) phosphorylation in the epidermis after SSL, and show for the first time that this dysregulation can be inhibited in vivo by a selective PDK1/Akt inhibitor, PHT-427. Combining rapamycin with PHT-427 on tumor prone skin additively caused a significant reduction of tumor multiplicity compared with vehicle controls. Our findings indicate that patients taking rapamycin should avoid sun exposure, and that combining topical mTOR inhibitors and Akt inhibitors may be a viable chemoprevention option for individuals at high risk for cutaneous SCC.
Collapse
Affiliation(s)
- Sally E Dickinson
- The University of Arizona Cancer Center, Tucson, Arizona. Department of Pharmacology, The University of Arizona, Tucson, Arizona.
| | - Jaroslav Janda
- The University of Arizona Cancer Center, Tucson, Arizona
| | - Jane Criswell
- The University of Arizona Cancer Center, Tucson, Arizona
| | | | - Erik R Olson
- The University of Arizona Cancer Center, Tucson, Arizona
| | - Zhonglin Liu
- Department of Medical Imaging, The University of Arizona, Tucson, Arizona
| | - Christy Barber
- Department of Medical Imaging, The University of Arizona, Tucson, Arizona
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Valerie S Calvert
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Janine Einspahr
- The University of Arizona Cancer Center, Tucson, Arizona. Department of Medicine, The University of Arizona, Tucson, Arizona
| | - Jesse E Dickinson
- Arizona Water Science Center, U.S. Geological Survey, Tucson, Arizona
| | - Steven P Stratton
- The University of Arizona Cancer Center, Tucson, Arizona. Department of Medicine, The University of Arizona, Tucson, Arizona
| | - Clara Curiel-Lewandrowski
- The University of Arizona Cancer Center, Tucson, Arizona. Department of Medicine, The University of Arizona, Tucson, Arizona
| | | | - Chengcheng Hu
- The University of Arizona Cancer Center, Tucson, Arizona
| | - Ann M Bode
- Department of Molecular Medicine and Biopharmaceutical Sciences, The Hormel Institute, The University of Minnesota, Austin, Minnesota
| | - Zigang Dong
- Department of Molecular Medicine and Biopharmaceutical Sciences, The Hormel Institute, The University of Minnesota, Austin, Minnesota
| | - David S Alberts
- The University of Arizona Cancer Center, Tucson, Arizona. Department of Medicine, The University of Arizona, Tucson, Arizona
| | - G Timothy Bowden
- The University of Arizona Cancer Center, Tucson, Arizona. Department of Medicine, The University of Arizona, Tucson, Arizona. Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona
| |
Collapse
|
25
|
Dillenburg CS, Martins MD, Meurer L, Castilho RM, Squarize CH. Keratoacanthoma of the Lip: Activation of the mTOR Pathway, Tumor Suppressor Proteins, and Tumor Senescence. Medicine (Baltimore) 2015; 94:e1552. [PMID: 26402814 PMCID: PMC4635754 DOI: 10.1097/md.0000000000001552] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The PI3K-PTEN-mTOR is one of the most important pathways involved in cancer development and progression; however, its role in keratoacanthoma (KA) is poorly understood. In this study, we investigated the activation of key proteins in the PI3K-mTOR pathway in lip KA. We analyzed the activation of the PI3K-PTEN-mTOR pathway using human tumor samples stained for well-established protein markers in this pathway, including pS6 and pAKT phosphoproteins. We assessed proliferation using Ki-67 and performed additional morphological and immunohistochemical analysis using anti-PTEN and anti-p16 antibodies.We found that the majority of KA labeled to pS6 and not pAKT. PTEN expression was inversely correlated with Ki-67 expression. In addition to PTEN expression, KA cells were positive for p16 senescence marker. PI3K-PTEN-mTOR pathway is activated in lip KA, leading to downstream activation of mTORC1, but not mTORC2. This pathway plays an important role in KA progression by promoting proliferation and activation of oncogenic-induced senescence.
Collapse
Affiliation(s)
- Caroline Siviero Dillenburg
- From the Department of Periodontics and Oral Medicine, Laboratory of Epithelial Biology, University of Michigan School of Dentistry, Ann Arbor, MI (CSD, MDM, RMC, CHS); Department of Oral Pathology, School of Dentistry (CSD, MDM); and Department of Pathology, Hospital de Clínicas de Porto Alegre and Medical School, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil (LM)
| | | | | | | | | |
Collapse
|
26
|
Bermudez Y, Stratton SP, Curiel-Lewandrowski C, Warneke J, Hu C, Bowden GT, Dickinson SE, Dong Z, Bode AM, Saboda K, Brooks CA, Petricoin EF, Hurst CA, Alberts DS, Einspahr JG. Activation of the PI3K/Akt/mTOR and MAPK Signaling Pathways in Response to Acute Solar-Simulated Light Exposure of Human Skin. Cancer Prev Res (Phila) 2015; 8:720-8. [PMID: 26031292 DOI: 10.1158/1940-6207.capr-14-0407] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 05/06/2015] [Indexed: 12/14/2022]
Abstract
The incidence of skin cancer is higher than all other cancers and continues to increase, with an average annual cost over $8 billion in the United States. As a result, identifying molecular pathway alterations that occur with UV exposure to strategize more effective preventive and therapeutic approaches is essential. To that end, we evaluated phosphorylation of proteins within the PI3K/Akt and MAPK pathways by immunohistochemistry in sun-protected skin after acute doses of physiologically relevant solar-simulated ultraviolet light (SSL) in 24 volunteers. Biopsies were performed at baseline, 5 minutes, 1, 5, and 24 hours after SSL irradiation. Within the PI3K/Akt pathway, we found activation of Akt (serine 473) to be significantly increased at 5 hours while mTOR (serine 2448) was strongly activated early and was sustained over 24 hours after SSL. Downstream, we observed a marked and sustained increase in phospho-S6 (serine 235/S236), whereas phospho-4E-BP1 (threonines 37/46) was increased only at 24 hours. Within the MAPK pathway, SSL-induced expression of phospho-p38 (threonine 180/tyrosine 182) peaked at 1 to 5 hours. ERK 1/2 was observed to be immediate and sustained after SSL irradiation. Phosphorylation of histone H3 (serine 10), a core structural protein of the nucleosome, peaked at 5 hours after SSL irradiation. The expression of both p53 and COX-2 was increased at 5 hours and was maximal at 24 hours after SSL irradiation. Apoptosis was significantly increased at 24 hours as expected and indicative of a sunburn-type response to SSL. Understanding the timing of key protein expression changes in response to SSL will aid in development of mechanistic-based approaches for the prevention and control of skin cancers.
Collapse
Affiliation(s)
- Yira Bermudez
- College of Medicine, University of Arizona, Tucson, Arizona. The University of Arizona Cancer Center, Tucson, Arizona
| | - Steven P Stratton
- College of Medicine, University of Arizona, Tucson, Arizona. The University of Arizona Cancer Center, Tucson, Arizona
| | - Clara Curiel-Lewandrowski
- College of Medicine, University of Arizona, Tucson, Arizona. The University of Arizona Cancer Center, Tucson, Arizona
| | - James Warneke
- College of Medicine, University of Arizona, Tucson, Arizona. The University of Arizona Cancer Center, Tucson, Arizona. Department of Surgery, University of Arizona, Tucson, Arizona
| | - Chengcheng Hu
- The University of Arizona Cancer Center, Tucson, Arizona
| | | | | | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | | | | | | | - Craig A Hurst
- College of Medicine, University of Arizona, Tucson, Arizona. Department of Surgery, University of Arizona, Tucson, Arizona
| | - David S Alberts
- College of Medicine, University of Arizona, Tucson, Arizona. The University of Arizona Cancer Center, Tucson, Arizona
| | - Janine G Einspahr
- College of Medicine, University of Arizona, Tucson, Arizona. The University of Arizona Cancer Center, Tucson, Arizona.
| |
Collapse
|
27
|
Carr TD, Feehan RP, Hall MN, Rüegg MA, Shantz LM. Conditional disruption of rictor demonstrates a direct requirement for mTORC2 in skin tumor development and continued growth of established tumors. Carcinogenesis 2015; 36:487-97. [PMID: 25740823 DOI: 10.1093/carcin/bgv012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 02/15/2015] [Indexed: 12/31/2022] Open
Abstract
Activation of signaling dependent on the mammalian target of rapamycin (mTOR) has been demonstrated in a variety of human malignancies, and our previous work suggests that mTOR complex (mTORC) 1 and mTORC2 may play unique roles in skin tumorigenesis. The purpose of these studies was to investigate the function of mTORC2-dependent pathways in skin tumor development and the maintenance of established tumors. Using mice that allow spatial and temporal control of mTORC2 in epidermis by conditional knockout of its essential component Rictor, we studied the effect of mTORC2 loss on both epidermal proliferation and chemical carcinogenesis. The results demonstrate that mTORC2 is dispensable for both normal epidermal proliferation and the hyperproliferative response to treatment with tetradecanoyl phorbol acetate (TPA). In contrast, deletion of epidermal Rictor prior to initiation in DMBA/TPA chemical carcinogenesis was sufficient to dramatically delay tumor development and resulted in reduced tumor number and size compared with control groups. Silencing of Rictor expression in tumor-bearing animals triggered regression of established tumors and increased caspase-3 cleavage without changes in proliferation. In vitro experiments demonstrate an increased sensitivity to caspase-dependent apoptosis in the absence of rictor, which is dependent on mTORC2 signaling. These studies demonstrate that mTORC2 activation is essential for keratinocyte survival, and suggest that inhibition of mTORC2 has value in chemoprevention by eliminating carcinogen-damaged cells during the early stages of tumorigenesis, and in therapy of existing tumors by restricting critical pro-survival pathways.
Collapse
Affiliation(s)
- Theresa D Carr
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA and
| | - Robert P Feehan
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA and
| | - Michael N Hall
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | - Markus A Rüegg
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | - Lisa M Shantz
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA and
| |
Collapse
|
28
|
Comparison of Akt/mTOR/4E-BP1 pathway signal activation and mutations of PIK3CA in Merkel cell polyomavirus-positive and Merkel cell polyomavirus-negative carcinomas. Hum Pathol 2014; 46:210-6. [PMID: 25466966 DOI: 10.1016/j.humpath.2014.07.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 07/03/2014] [Accepted: 07/09/2014] [Indexed: 01/19/2023]
Abstract
Merkel cell polyomavirus (MCPyV) integrates monoclonally into the genomes of approximately 80% of Merkel cell carcinomas (MCCs), affecting their clinicopathological features. The molecular mechanisms underlying MCC development after MCPyV infection remain unclear. We investigated the association of MCPyV infection with activation of the Akt/mammalian target of rapamycin (mTOR)/4E-binding protein 1 (4E-BP1) signaling pathway in MCCs to elucidate the role of these signal transductions and to identify molecular targets for treatment. We analyzed the molecular and pathological characteristics of 41 MCPyV-positive and 27 MCPyV-negative MCCs. Expression of mTOR, TSC1, and TSC2 messenger RNA was significantly higher in MCPyV-negative MCCs, and Akt (T308) phosphorylation also was significantly higher (92% vs 66%; P = .019), whereas 4E-BP1 (S65 and T70) phosphorylation was common in both MCC types (92%-100%). The expression rates of most other tested signals were high (60%-100%) and not significantly correlated with MCPyV large T antigen expression. PIK3CA mutations were observed more frequently in MCPyV-positive MCCs (6/36 [17%] vs 2/20 [10%]). These results suggest that protein expression (activation) of most Akt/mTOR/4E-BP1 pathway signals was not significantly different in MCPyV-positive and MCPyV-negative MCCs, although these 2 types may differ in tumorigenesis, and MCPyV-negative MCCs showed significantly more frequent p-Akt (T308) activation. Therefore, certain Akt/mTOR/4E-BP1 pathway signals could be novel therapeutic targets for MCC regardless of MCPyV infection status.
Collapse
|
29
|
Brinkhuizen T, Weijzen CAH, Eben J, Thissen MR, van Marion AM, Lohman BG, Winnepenninckx VJL, Nelemans PJ, van Steensel MAM. Immunohistochemical analysis of the mechanistic target of rapamycin and hypoxia signalling pathways in basal cell carcinoma and trichoepithelioma. PLoS One 2014; 9:e106427. [PMID: 25181405 PMCID: PMC4152244 DOI: 10.1371/journal.pone.0106427] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 07/30/2014] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Basal cell carcinoma (BCC) is the most common cancer in Caucasians. Trichoepithelioma (TE) is a benign neoplasm that strongly resembles BCC. Both are hair follicle (HF) tumours. HFs are hypoxic microenvironments, therefore we hypothesized that hypoxia-induced signalling pathways could be involved in BCC and TE as they are in other human malignancies. Hypoxia-inducible factor 1 (HIF1) and mechanistic/mammalian target of rapamycin (mTOR) are key players in these pathways. OBJECTIVES To determine whether HIF1/mTOR signalling is involved in BCC and TE. METHODS We used immunohistochemical staining of formalin-fixed paraffin-embedded BCC (n = 45) and TE (n = 35) samples to assess activity of HIF1, mTORC1 and their most important target genes. The percentage positive tumour cells was assessed manually in a semi-quantitative manner and categorized (0%, <30%, 30-80% and >80%). RESULTS Among 45 BCC and 35 TE examined, expression levels were respectively 81% and 57% (BNIP3), 73% and 75% (CAIX), 79% and 86% (GLUT1), 50% and 19% (HIF1α), 89% and 88% (pAKT), 55% and 61% (pS6), 15% and 25% (pMTOR), 44% and 63% (PHD2) and 44% and 49% (VEGF-A). CAIX, Glut1 and PHD2 expression levels were significantly higher in TE when only samples with at least 80% expression were included. CONCLUSIONS HIF and mTORC1 signalling seems active in both BCC and TE. There are no appreciable differences between the two with respect to pathway activity. At this moment immunohistochemical analyses of HIF, mTORC1 and their target genes does not provide a reliable diagnostic tool for the discrimination of BCC and TE.
Collapse
Affiliation(s)
- Tjinta Brinkhuizen
- Department of Dermatology, Maastricht University Medical Centre, Maastricht, the Netherlands
- GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
- * E-mail:
| | - Chantal A. H. Weijzen
- Department of Dermatology, Maastricht University Medical Centre, Maastricht, the Netherlands
- GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Jonathan Eben
- Department of Pathology, Maastricht University Medical Centre, Maastricht, the Netherlands
- GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Monique R. Thissen
- Department of Dermatology, Maastricht University Medical Centre, Maastricht, the Netherlands
- GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | | | - Björn G. Lohman
- Department of Pathology, Laurentius Hospital, Roermond, the Netherlands
| | - Véronique J. L. Winnepenninckx
- Department of Pathology, Maastricht University Medical Centre, Maastricht, the Netherlands
- GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Patty J. Nelemans
- Department of Epidemiology, Maastricht University Medical Centre, Maastricht, the Netherlands
- GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Maurice A. M. van Steensel
- Department of Dermatology, Maastricht University Medical Centre, Maastricht, the Netherlands
- Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, the Netherlands
- GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
- Institute of Medical Biology, Immunos, Singapore, Singapore
| |
Collapse
|
30
|
Phytochemical modulation of the Akt/mTOR pathway and its potential use in cutaneous disease. Arch Dermatol Res 2014; 306:861-71. [DOI: 10.1007/s00403-014-1480-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 05/04/2014] [Accepted: 05/10/2014] [Indexed: 10/25/2022]
|
31
|
Buerger C, Malisiewicz B, Eiser A, Hardt K, Boehncke WH. Mammalian target of rapamycin and its downstream signalling components are activated in psoriatic skin. Br J Dermatol 2014; 169:156-9. [PMID: 23398394 DOI: 10.1111/bjd.12271] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2013] [Indexed: 12/26/2022]
Abstract
BACKGROUND Mammalian target of rapamycin (mTOR) signalling integrates signals leading to cellular growth, proliferation and differentiation. Disturbance of this tightly regulated interplay leads to malignancies, as reflected by altered mTOR signalling in epidermal tumours. As psoriatic keratinocytes also show features of perturbed cell growth and differentiation, the question arises as to whether mTOR signalling also plays a role in the pathogenesis of psoriasis. OBJECTIVES To investigate the activation status of mTOR signalling components in psoriasis. METHODS Biopsies from lesional and nonlesional skin of patients with psoriasis (n = 10), as well as samples from healthy donors (n = 3), were analysed by immunohistochemistry and Western blot, utilizing antibodies detecting phosphorylated mTOR, phospho-S6 kinase and phospho-S6 ribosomal protein. RESULTS We found mTOR and its downstream signalling molecule, the ribosomal protein S6, to be activated in lesional psoriatic skin. While mTOR is activated throughout the whole epidermis, with particularly strong activation in the basal layer, S6 is active in suprabasal layers of differentiating keratinocytes. CONCLUSIONS Altogether these results suggest a role for mTOR signalling in the epidermal changes leading to the psoriatic phenotype. mTOR inhibition might be a mode of action to explore in developing innovative antipsoriatic drugs.
Collapse
Affiliation(s)
- C Buerger
- Department of Dermatology, Clinic of the Goethe University, Frankfurt am Main, Germany.
| | | | | | | | | |
Collapse
|
32
|
Degen M, Barron P, Natarajan E, Widlund HR, Rheinwald JG. RSK activation of translation factor eIF4B drives abnormal increases of laminin γ2 and MYC protein during neoplastic progression to squamous cell carcinoma. PLoS One 2013; 8:e78979. [PMID: 24205356 PMCID: PMC3810258 DOI: 10.1371/journal.pone.0078979] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 09/26/2013] [Indexed: 12/17/2022] Open
Abstract
Overexpression of the basement membrane protein Laminin γ2 (Lamγ2) is a feature of many epidermal and oral dysplasias and all invasive squamous cell carcinomas (SCCs). This abnormality has potential value as an immunohistochemical biomarker of premalignancy but its mechanism has remained unknown. We recently reported that Lamγ2 overexpression in culture is the result of deregulated translation controls and depends on the MAPK-RSK signaling cascade. Here we identify eIF4B as the RSK downstream effector responsible for elevated Lamγ2 as well as MYC protein in neoplastic epithelial cells. Premalignant dysplastic keratinocytes, SCC cells, and keratinocytes expressing the E6 oncoprotein of human papillomavirus (HPV) type 16 displayed MAPK-RSK and mTOR-S6K1 activation and overexpressed Lamγ2 and MYC in culture. Immunohistochemical staining of oral dysplasias and SCCs for distinct, RSK- and S6K1-specific S6 phosphorylation events revealed that their respective upstream pathways become hyperactive at the same time during neoplastic progression. However, pharmacologic kinase inhibitor studies in culture revealed that Lamγ2 and MYC overexpression depends on MAPK-RSK activity, independent of PI3K-mTOR-S6K1. eIF4B knockdown reduced Lamγ2 and MYC protein expression, consistent with the known requirement for eIF4B to translate mRNAs with long, complex 5′ untranslated regions (5′-UTRs). Accordingly, expression of a luciferase reporter construct preceded by the Lamγ2 5′-UTR proved to be RSK-dependent and mTOR-independent. These results demonstrate that RSK activation of eIF4B is causally linked to elevated Lamγ2 and MYC protein levels during neoplastic progression to invasive SCC. These findings have potential clinical significance for identifying premalignant lesions and for developing targeted drugs to treat SCC.
Collapse
Affiliation(s)
- Martin Degen
- Department of Dermatology, Brigham and Women's Hospital and Harvard Skin Disease Research Center, Boston, Massachusetts, United States of America
| | - Patricia Barron
- Department of Dermatology, Brigham and Women's Hospital and Harvard Skin Disease Research Center, Boston, Massachusetts, United States of America
| | - Easwar Natarajan
- Section of Oral and Maxillofacial Pathology, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Hans R. Widlund
- Department of Dermatology, Brigham and Women's Hospital and Harvard Skin Disease Research Center, Boston, Massachusetts, United States of America
| | - James G. Rheinwald
- Department of Dermatology, Brigham and Women's Hospital and Harvard Skin Disease Research Center, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
33
|
Strozyk E, Kulms D. The role of AKT/mTOR pathway in stress response to UV-irradiation: implication in skin carcinogenesis by regulation of apoptosis, autophagy and senescence. Int J Mol Sci 2013; 14:15260-85. [PMID: 23887651 PMCID: PMC3759859 DOI: 10.3390/ijms140815260] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 06/27/2013] [Accepted: 07/01/2013] [Indexed: 02/06/2023] Open
Abstract
Induction of DNA damage by UVB and UVA radiation may generate mutations and genomic instability leading to carcinogenesis. Therefore, skin cells being repeatedly exposed to ultraviolet (UV) light have acquired multilayered protective mechanisms to avoid malignant transformation. Besides extensive DNA repair mechanisms, the damaged skin cells can be eliminated by induction of apoptosis, which is mediated through the action of tumor suppressor p53. In order to prevent the excessive loss of skin cells and to maintain the skin barrier function, apoptotic pathways are counteracted by anti-apoptotic signaling including the AKT/mTOR pathway. However, AKT/mTOR not only prevents cell death, but is also active in cell cycle transition and hyper-proliferation, thereby also counteracting p53. In turn, AKT/mTOR is tuned down by the negative regulators being controlled by the p53. This inhibition of AKT/mTOR, in combination with transactivation of damage-regulated autophagy modulators, guides the p53-mediated elimination of damaged cellular components by autophagic clearance. Alternatively, p53 irreversibly blocks cell cycle progression to prevent AKT/mTOR-driven proliferation, thereby inducing premature senescence. Conclusively, AKT/mTOR via an extensive cross talk with p53 influences the UV response in the skin with no black and white scenario deciding over death or survival.
Collapse
Affiliation(s)
- Elwira Strozyk
- Experimental Dermatology, Department of Dermatology, TU Dresden, 01307 Dresden, Germany; E-Mail:
| | - Dagmar Kulms
- Experimental Dermatology, Department of Dermatology, TU Dresden, 01307 Dresden, Germany; E-Mail:
| |
Collapse
|
34
|
Carr TD, DiGiovanni J, Lynch CJ, Shantz LM. Inhibition of mTOR suppresses UVB-induced keratinocyte proliferation and survival. Cancer Prev Res (Phila) 2012; 5:1394-404. [PMID: 23129577 DOI: 10.1158/1940-6207.capr-12-0272-t] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
UV radiation is the major risk factor for developing skin cancer, the most prevalent cancer worldwide. Several studies indicate that mTOR signaling is activated by UVB and may play an important role in skin tumorigenesis. mTOR exists in two functionally and compositionally distinct protein complexes: the rapamycin-sensitive mTOR complex 1 (mTORC1) and the rapamycin-resistant mTOR complex 2 (mTORC2). The purpose of these studies was to investigate the roles of the two mTOR complexes in UVB-mediated proliferation and apoptosis in the skin. We used rapamycin, a pharmacologic inhibitor of mTORC1, and an inducible mTOR-deficient (K5-CreER(T2);mTOR(fl/fl)) mouse model that allows epidermal-specific disruption of mTOR following topical treatment with 4-hydroxytamoxifen (4OHT). Rapamycin blocked UVB-induced phosphorylation of S6K, the downstream target of mTORC1, and significantly reduced UVB-stimulated epidermal proliferation and cell-cycle progression, but had no effect on cell death. In contrast, mTOR deletion, which attenuated UVB-induced phosphorylation of both S6K and the mTORC2 target AKT(Ser473), significantly increased apoptosis both in vivo and in keratinocyte cultures, in addition to reducing hyperproliferation following UVB irradiation. The role of mTORC2 in UVB-induced prosurvival signaling was verified in Rictor(-/-) mouse embryo fibroblasts, which lack functional mTORC2 and were more sensitive to UVB-induced apoptosis than controls. These studies show that mTORC1 and mTORC2 play unique but complementary roles in controlling proliferation and apoptosis in the skin. Our findings underscore the importance of both mTOR complexes in mediating UVB-induced signaling in keratinocytes and provide new insight into the pathogenesis of skin cancer.
Collapse
Affiliation(s)
- Theresa D Carr
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
35
|
Chaudhary SC, Kurundkar D, Elmets CA, Kopelovich L, Athar M. Metformin, an antidiabetic agent reduces growth of cutaneous squamous cell carcinoma by targeting mTOR signaling pathway. Photochem Photobiol 2012; 88:1149-56. [PMID: 22540890 PMCID: PMC3476735 DOI: 10.1111/j.1751-1097.2012.01165.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The biguanide metformin is widely used for the treatment of Type-II diabetes. Its antiproliferative and pro-apoptotic effects in various tumor cells suggest its potential candidacy for cancer chemoprevention. Herein, we report that metformin significantly inhibited human epidermoid A431 tumor xenograft growth in nu/nu mice, which was associated with a significant reduction in proliferative biomarkers PCNA and cyclins D1/B1. This tumor growth reduction was accompanied by the enhanced apoptotic cell death and an increase in Bax:Bcl2 ratio. The mechanism by which metformin manifests antitumor effects appears to be dependent on the inhibition of nuclear factor kappa B (NFkB) and mTOR signaling pathways. Decreased phosphorylation of NFkB inhibitory protein IKBα together with reduced enhancement of NFkB transcriptional target proteins, iNOS/COX-2 were observed. In addition, a decrease in the activation of ERK/p38-driven MAP kinase signaling was seen. Similarly, AKT signaling activation as assessed by the diminished phosphorylation at Ser473 with a concomitant decrease in mTOR signaling pathway was also noted as phosphorylation of mTOR regulatory proteins p70S6K and 4E-BP-1 was significantly reduced. Consistently, decreased phosphorylation of GSK3β, which is carried out by AKT kinases was also observed. These results suggest that metformin blocks SCC growth by dampening NFkB and mTOR signaling pathways.
Collapse
Affiliation(s)
- Sandeep C. Chaudhary
- Department of Dermatology, University of Alabama at Birmingham, 1530 3 Avenue South, VH 509, Birmingham, AL 35294-0019
| | - Deepali Kurundkar
- Department of Dermatology, University of Alabama at Birmingham, 1530 3 Avenue South, VH 509, Birmingham, AL 35294-0019
| | - Craig A. Elmets
- Department of Dermatology, University of Alabama at Birmingham, 1530 3 Avenue South, VH 509, Birmingham, AL 35294-0019
| | - Levy Kopelovich
- Division of Cancer Prevention, National Cancer Institute, 6130 Executive Blvd, Suite 2114, Bethesda, MD 20892
| | - Mohammad Athar
- Department of Dermatology, University of Alabama at Birmingham, 1530 3 Avenue South, VH 509, Birmingham, AL 35294-0019
| |
Collapse
|
36
|
Einspahr JG, Calvert V, Alberts DS, Curiel-Lewandrowski C, Warneke J, Krouse R, Stratton SP, Liotta L, Longo C, Pellacani G, Pellicani G, Prasad A, Sagerman P, Bermudez Y, Deng J, Bowden GT, Petricoin EF. Functional protein pathway activation mapping of the progression of normal skin to squamous cell carcinoma. Cancer Prev Res (Phila) 2012; 5:403-13. [PMID: 22389437 PMCID: PMC3297971 DOI: 10.1158/1940-6207.capr-11-0427] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Reverse phase protein microarray analysis was used to identify cell signaling derangements in squamous cell carcinoma (SCC) compared with actinic keratosis (AK) and upper inner arm (UIA). We analyzed two independent tissue sets with isolation and enrichment of epithelial cells by laser capture microdissection. Set 1 served as a pilot and a means to identify protein pathway activation alterations that could be further validated in a second independent set. Set 1 was comprised of 4 AK, 13 SCC, and 20 UIA. Set 2 included 15 AK, 9 SCCs, and 20 UIAs. Activation of 51 signaling proteins, known to be involved in tumorigenesis, were assessed for set 1 and showed that the MEK-ERK [mitogen-activated protein (MAP)/extracellular signal-regulated (ERK; MEK)] pathway was activated in SCC compared with AK and UIA, and that epidermal growth factor receptor (EGFR) and mTOR pathways were aberrantly activated in SCC. Unsupervised two-way hierarchical clustering revealed that AK and UIA shared a common signaling network activation architecture while SCC was dramatically different. Statistical analysis found that prosurvival signaling through phosphorylation of ASK and 4EBP1 as well as increased Bax and Bak expression was higher in AK compared with UIA. We expanded pathway network activation mapping in set 2 to 101 key signaling proteins, which corroborated activation of MEK-ERK, EGFR, and mTOR pathways through discovery of a number of upstream and downstream signaling molecules within these pathways to conclude that SCC is indeed a pathway activation-driven disease. Pathway activation mapping of SCC compared with AK revealed several interconnected networks that could be targeted with drug therapy for potential chemoprevention and therapeutic applications.
Collapse
Affiliation(s)
- Janine G Einspahr
- University of Arizona Cancer Center, 1515 North Campbell Avenue, Tucson, AZ 85724, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Yadav V, Denning MF. Fyn is induced by Ras/PI3K/Akt signaling and is required for enhanced invasion/migration. Mol Carcinog 2010; 50:346-52. [PMID: 21480388 DOI: 10.1002/mc.20716] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 10/15/2010] [Accepted: 11/01/2010] [Indexed: 12/12/2022]
Abstract
Src family kinases (SFKs) are frequently over-expressed and/or activated in human cancers, and play key roles in cancer cell invasion, metastasis, proliferation, survival, and angiogenesis. Allosteric activation of SFKs occurs through well-defined post-translational mechanisms, however the SFK member Fyn is over-expressed in multiple human cancers (prostate, melanoma, pancreatic, glioma, chronic myelogenous leukemia) and the mechanism of increased Fyn expression is unclear. Since activation of Ras oncogenes is a common oncogenic event leading to the activation of multiple effector pathways, we explored if Ras could induce Fyn expression. Retroviral transduction of the human keratinocyte cell line HaCaT with oncogenic H-Ras dramatically up-regulated Fyn mRNA (>100-fold, P < 0.001), protein, and kinase activity without affecting Src levels or activity. Activation of Akt, but not MAPK or EGFR, was necessary and sufficient for induction of Fyn by H-Ras. Expression of active Fyn was sufficient to increase HaCaT cell migration and invasion, and the enhanced migration and invasion induced by H-Ras could be significantly blocked (70% reduction, P < 0.01) by knockdown of Fyn with a specific siRNA or inhibition of SFKs with PP2. In addition, expression of Fyn in MDA-MB-231 breast cancer cells was dependent on PI3K activity and was involved in their invasive phenotype. Thus, the Ras/PI3K/Akt pathway can account for Fyn over-expression in cancers, and Fyn is a critical mediator of the Ras-stimulated invasive cell phenotype. These results support the development of therapeutic strategies targeting Akt/Fyn pathway to block migration and invasion of tumor cells.
Collapse
Affiliation(s)
- Vipin Yadav
- Molecular Biology Program, Loyola University Chicago, Maywood, Illinois, USA
| | | |
Collapse
|
38
|
Cyclin-dependent kinase 2 (CDK-2) expression in nonmelanocytic human cutaneous lesions. Appl Immunohistochem Mol Morphol 2010; 18:357-64. [PMID: 20216405 DOI: 10.1097/pai.0b013e3181d4069c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Lesions originating from different types of skin cells differ significantly with respect to their pathologic importance. The aim of this work was to examine as to what extent the differences in the origin are reflected in expression levels of CDK-2 and to investigate whether CDK-2 expression might be considered as potential marker useful for diagnostics and assessment of invasiveness of human nonmelanocytic lesions. We conducted comparative immunohistochemical studies of expression of cyclin-dependent kinase 2 (CDK-2) in 16 benign epithelial skin lesions, 11 precancerous lesions, 19 cases of basal cell carcinoma (first such study), 14 squamous cell carcinomas (SCCs), and 7 fibromas. Development of benign epithelial skin lesions was not associated with considerable increase of the CDK-2 expression. Increase of the CDK-2 level was observed in precancerous lesions, and the expression was strongest in SCCs. The level of CDK-2 may be related to invasiveness of skin cancers, as squamous cell carcinomas expressed the enzyme significantly stronger than basal cell carcinomas. Higher percentage fraction of CDK-2 positive cells observed in SCC compared with precancerous lesions may be useful for histopathologic diagnostics of this cancer. Moreover, strong immunohistochemical CDK-2 staining of the cancer cells present deep in dermis may facilitate their detection in histopathologic examinations.
Collapse
|
39
|
Alexandroff A, Flohr C, Johnston G. Updates from the British Association of Dermatologists 89th Annual Meeting, 7-10 July 2009, Glasgow, U.K. Br J Dermatol 2010; 163:27-37. [DOI: 10.1111/j.1365-2133.2010.09814.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
40
|
Therapeutic role of sirolimus in non-transplant kidney disease. Pharmacol Ther 2009; 123:187-206. [DOI: 10.1016/j.pharmthera.2009.03.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2009] [Accepted: 03/24/2009] [Indexed: 12/20/2022]
|