1
|
Rao S, He Z, Wang Z, Yin H, Hu X, Tan Y, Wan T, Zhu H, Luo Y, Wang X, Li H, Wang Z, Hu X, Hong C, Wang Y, Luo M, Du W, Qian Y, Tang S, Xie H, Chen C. Extracellular vesicles from human urine-derived stem cells delay aging through the transfer of PLAU and TIMP1. Acta Pharm Sin B 2024; 14:1166-1186. [PMID: 38487008 PMCID: PMC10935484 DOI: 10.1016/j.apsb.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 03/17/2024] Open
Abstract
Aging increases the risks of various diseases and the vulnerability to death. Cellular senescence is a hallmark of aging that contributes greatly to aging and aging-related diseases. This study demonstrates that extracellular vesicles from human urine-derived stem cells (USC-EVs) efficiently inhibit cellular senescence in vitro and in vivo. The intravenous injection of USC-EVs improves cognitive function, increases physical fitness and bone quality, and alleviates aging-related structural changes in different organs of senescence-accelerated mice and natural aging mice. The anti-aging effects of USC-EVs are not obviously affected by the USC donors' ages, genders, or health status. Proteomic analysis reveals that USC-EVs are enriched with plasminogen activator urokinase (PLAU) and tissue inhibitor of metalloproteinases 1 (TIMP1). These two proteins contribute importantly to the anti-senescent effects of USC-EVs associated with the inhibition of matrix metalloproteinases, cyclin-dependent kinase inhibitor 2A (P16INK4a), and cyclin-dependent kinase inhibitor 1A (P21cip1). These findings suggest a great potential of autologous USC-EVs as a promising anti-aging agent by transferring PLAU and TIMP1 proteins.
Collapse
Affiliation(s)
- Shanshan Rao
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
| | - Zehui He
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
| | - Zun Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
- Xiangya School of Nursing, Central South University, Changsha 410013, China
| | - Hao Yin
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
| | - Xiongke Hu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
- Department of Pediatric Orthopedics, Hunan Children's Hospital, University of South China, Changsha 410007, China
| | - Yijuan Tan
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
| | - Tengfei Wan
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
| | - Hao Zhu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
| | - Yi Luo
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
| | - Xin Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
| | - Hongming Li
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
| | - Zhenxing Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
| | - Xinyue Hu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chungu Hong
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
| | - Yiyi Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
| | - Mingjie Luo
- Xiangya School of Nursing, Central South University, Changsha 410013, China
- School of Nursing, Xinjiang Medical University, Urumqi, Xinjiang 830000, China
| | - Wei Du
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Rehabilitation, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yuxuan Qian
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
| | - Siyuan Tang
- Xiangya School of Nursing, Central South University, Changsha 410013, China
| | - Hui Xie
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chunyuan Chen
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha 410008, China
| |
Collapse
|
2
|
Targeting PI3K/AKT/mTOR Signaling Pathway in Pancreatic Cancer: From Molecular to Clinical Aspects. Int J Mol Sci 2022; 23:ijms231710132. [PMID: 36077529 PMCID: PMC9456549 DOI: 10.3390/ijms231710132] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/01/2022] [Accepted: 09/01/2022] [Indexed: 02/06/2023] Open
Abstract
Although pancreatic cancer (PC) was considered in the past an orphan cancer type due to its low incidence, it may become in the future one of the leading causes of cancer death. Pancreatic ductal adenocarcinoma (PDAC) is the most frequent type of PC, being a highly aggressive malignancy and having a 5-year survival rate of less than 10%. Non-modifiable (family history, age, genetic susceptibility) and modifiable (smoking, alcohol, acute and chronic pancreatitis, diabetes mellitus, intestinal microbiota) risk factors are involved in PC pathogenesis. Chronic inflammation induced by various factors plays crucial roles in PC development from initiation to metastasis. In multiple malignant conditions such as PC, cytokines, chemokines, and growth factors activate the class I phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) (PI3K/AKT/mTOR) signaling pathway, which plays key roles in cell growth, survival, proliferation, metabolism, and motility. Currently, mTOR, AKT, and PI3K inhibitors are used in clinical studies. Moreover, PI3K/mTOR dual inhibitors are being tested in vitro and in vivo with promising results for PC patients. The main aim of this review is to present PC incidence, risk factors, tumor microenvironment development, and PI3K/AKT/mTOR dysregulation and inhibitors used in clinical, in vivo, and in vitro studies.
Collapse
|
3
|
Alshammari E, Zhang YX, Yang Z. Mechanistic and functional extrapolation of SET and MYND domain-containing protein 2 to pancreatic cancer. World J Gastroenterol 2022; 28:3753-3766. [PMID: 36157542 PMCID: PMC9367238 DOI: 10.3748/wjg.v28.i29.3753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/24/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal neoplasms worldwide and represents the vast majority of pancreatic cancer cases. Understanding the molecular pathogenesis and the underlying mechanisms involved in the initiation, maintenance, and progression of PDAC is an urgent need, which may lead to the development of novel therapeutic strategies against this deadly cancer. Here, we review the role of SET and MYND domain-containing protein 2 (SMYD2) in initiating and maintaining PDAC development through methylating multiple tumor suppressors and oncogenic proteins. Given the broad substrate specificity of SMYD2 and its involvement in diverse oncogenic signaling pathways in many other cancers, the mechanistic extrapolation of SMYD2 from these cancers to PDAC may allow for developing new hypotheses about the mechanisms driving PDAC tumor growth and metastasis, supporting a proposition that targeting SMYD2 could be a powerful strategy for the prevention and treatment of PDAC.
Collapse
Affiliation(s)
- Eid Alshammari
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University, Detroit, MI 48201, United States
| | - Ying-Xue Zhang
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University, Detroit, MI 48201, United States
| | - Zhe Yang
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, United States
| |
Collapse
|
4
|
Gillson J, Abd El-Aziz YS, Leck LYW, Jansson PJ, Pavlakis N, Samra JS, Mittal A, Sahni S. Autophagy: A Key Player in Pancreatic Cancer Progression and a Potential Drug Target. Cancers (Basel) 2022; 14:3528. [PMID: 35884592 PMCID: PMC9315706 DOI: 10.3390/cancers14143528] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 01/18/2023] Open
Abstract
Pancreatic cancer is known to have the lowest survival outcomes among all major cancers, and unfortunately, this has only been marginally improved over last four decades. The innate characteristics of pancreatic cancer include an aggressive and fast-growing nature from powerful driver mutations, a highly defensive tumor microenvironment and the upregulation of advantageous survival pathways such as autophagy. Autophagy involves targeted degradation of proteins and organelles to provide a secondary source of cellular supplies to maintain cell growth. Elevated autophagic activity in pancreatic cancer is recognized as a major survival pathway as it provides a plethora of support for tumors by supplying vital resources, maintaining tumour survival under the stressful microenvironment and promoting other pathways involved in tumour progression and metastasis. The combination of these features is unique to pancreatic cancer and present significant resistance to chemotherapeutic strategies, thus, indicating a need for further investigation into therapies targeting this crucial pathway. This review will outline the autophagy pathway and its regulation, in addition to the genetic landscape and tumor microenvironment that contribute to pancreatic cancer severity. Moreover, this review will also discuss the mechanisms of novel therapeutic strategies that inhibit autophagy and how they could be used to suppress tumor progression.
Collapse
Affiliation(s)
- Josef Gillson
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
| | - Yomna S. Abd El-Aziz
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
- Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta 31527, Egypt
| | - Lionel Y. W. Leck
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
- Cancer Drug Resistance and Stem Cell Program, University of Sydney, Sydney, NSW 2006, Australia
| | - Patric J. Jansson
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
- Cancer Drug Resistance and Stem Cell Program, University of Sydney, Sydney, NSW 2006, Australia
| | - Nick Pavlakis
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
| | - Jaswinder S. Samra
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Upper GI Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, St Leonards, Sydney, NSW 2065, Australia
- Australian Pancreatic Centre, St Leonards, Sydney, NSW 2065, Australia
| | - Anubhav Mittal
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Upper GI Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, St Leonards, Sydney, NSW 2065, Australia
- Australian Pancreatic Centre, St Leonards, Sydney, NSW 2065, Australia
- School of Medicine, University of Notre Dame, Darlinghurst, Sydney, NSW 2010, Australia
| | - Sumit Sahni
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
- Australian Pancreatic Centre, St Leonards, Sydney, NSW 2065, Australia
| |
Collapse
|
5
|
Huang H, Sun J, Li Z, Zhang X, Li Z, Zhu H, Yu X. Impact of the tumor immune microenvironment on the outcome of pancreatic cancer: a retrospective study based on clinical pathological analysis. Gland Surg 2022; 11:472-482. [PMID: 35284302 PMCID: PMC8899427 DOI: 10.21037/gs-22-45] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/16/2022] [Indexed: 07/07/2024]
Abstract
BACKGROUND The cancerous microenvironment, characterized by the infiltration of CD4+ and CD8+ T cells, play a critical role in regulating the progression of cancer and treating efficiency of immunotherapy. However, the distribution of these cells and their associated cytokines in the tumor microenvironment of pancreatic cancer (PC) are not yet fully understood. Our study aims to analyze the contents of CD4+IL-17+ and CD8+ T cells in PC and their relationship with the clinicopathological features and survival outcomes of patients. METHODS PC tissues and adjacent tissues were retrospectively collected from 40 patients in our hospital. The expression of CD4, IL-17, and CD8 in histological samples was measured by immunohistochemistry. The correlation between CD4, IL-17, and CD8 expression and clinical characteristics was analyzed using Kaplan-Meier survival analysis. The risk factors affecting the outcome of PC were examined by the Cox proportional hazards model, then a nomogram predicting the survival of PC using these risk factors was established. RESULTS The content of CD4+IL-17+ T cells in PC tissues was significantly higher than that in adjacent normal tissues, while the number of CD8+ T cells was significantly lower than that in adjacent normal tissues (P<0.01). CD4+ T cells in PC tissues was significantly associated with TNM stage and lymph node metastasis (P<0.05). IL-17 and CD8 were significantly associated with histological grade, TNM stage, local infiltration, and lymph node metastasis (P<0.05). The median survival times (MSTs) of CD4 positive and negative patients were 13.2 and 21.4 months, respectively. The MSTs of IL-17 positive and negative patients were 10.4 and 24.8 months, respectively. The MSTs were 21.9 and 11.8 months for CD8 positive and negative patients, respectively (P<0.05). The Cox regression model demonstrated that TNM staging, lymph node metastasis, and CD4+IL-17+ and CD8+ T cells affected PC prognosis (P<0.05). The nomogram showed that the survival probability was reduced in patients with TNM stage III to IV, lymph node metastasis, high CD4+IL-17+ level, and low CD8+ expression. CONCLUSIONS CD4+IL-17+ and CD8+ T cells in PC tissues are associated with TNM staging, lymph node metastasis, and MST, and can be used as new prognostic indicators for PC.
Collapse
Affiliation(s)
- Hui Huang
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jichun Sun
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhiqiang Li
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xianlin Zhang
- Department of General Surgery, Affiliated Renhe Hospital of China, Three Gorges University, Yichang, China
| | - Zheng Li
- Department of General Surgery, Affiliated Renhe Hospital of China, Three Gorges University, Yichang, China
| | - Hongwei Zhu
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Yu
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
6
|
Lu S, Louphrasitthiphol P, Goradia N, Lambert JP, Schmidt J, Chauhan J, Rughani MG, Larue L, Wilmanns M, Goding CR. TBX2 controls a proproliferative gene expression program in melanoma. Genes Dev 2021; 35:1657-1677. [PMID: 34819350 PMCID: PMC8653791 DOI: 10.1101/gad.348746.121] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/22/2021] [Indexed: 12/20/2022]
Abstract
Senescence shapes embryonic development, plays a key role in aging, and is a critical barrier to cancer initiation, yet how senescence is regulated remains incompletely understood. TBX2 is an antisenescence T-box family transcription repressor implicated in embryonic development and cancer. However, the repertoire of TBX2 target genes, its cooperating partners, and how TBX2 promotes proliferation and senescence bypass are poorly understood. Here, using melanoma as a model, we show that TBX2 lies downstream from PI3K signaling and that TBX2 binds and is required for expression of E2F1, a key antisenescence cell cycle regulator. Remarkably, TBX2 binding in vivo is associated with CACGTG E-boxes, present in genes down-regulated by TBX2 depletion, more frequently than the consensus T-element DNA binding motif that is restricted to Tbx2 repressed genes. TBX2 is revealed to interact with a wide range of transcription factors and cofactors, including key components of the BCOR/PRC1.1 complex that are recruited by TBX2 to the E2F1 locus. Our results provide key insights into how PI3K signaling modulates TBX2 function in cancer to drive proliferation.
Collapse
Affiliation(s)
- Sizhu Lu
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Pakavarin Louphrasitthiphol
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom.,Department of Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Nishit Goradia
- European Molecular Biology Laboratory, Hamburg Unit, 22607 Hamburg, Germany
| | - Jean-Philippe Lambert
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada.,Department of Molecular Medicine and Cancer Research Centre, Université Laval, Québec City, Québec G1R 3S3, Canada; CHU de Québec Research Center, Centre Hospitalier de l'Université Laval, Québec City, Québec G1V 4G2, Canada
| | - Johannes Schmidt
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Jagat Chauhan
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Milap G Rughani
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Lionel Larue
- Institut Curie, PSL Research University, U1021, Institut National de la Santé et de la Recherche Médicale, Normal and Pathological Development of Melanocytes, 91405 Orsay Cedex, France.,Université Paris-Sud, Université Paris-Saclay, UMR 3347 Centre National de la Recherche Scientifique, 91405 Orsay Cedex, France.,Equipe Labellisée Ligue Contre le Cancer, 91405 Orsay Cedex, France
| | - Matthias Wilmanns
- European Molecular Biology Laboratory, Hamburg Unit, 22607 Hamburg, Germany.,University Hamburg Clinical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Colin R Goding
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| |
Collapse
|
7
|
Chen X, An Y, Zhang Y, Xu D, Chen T, Yang Y, Chen W, Wu D, Zhang X. CCL26 is upregulated by nab-paclitaxel in pancreatic cancer-associated fibroblasts and promotes PDAC invasiveness through activation of the PI3K/AKT/mTOR pathway. Acta Biochim Biophys Sin (Shanghai) 2021; 53:612-619. [PMID: 33764366 DOI: 10.1093/abbs/gmab032] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Indexed: 01/05/2023] Open
Abstract
Recently, the combined use of FOLFIRINOX (leucovorin and fluorouracil plus irinotecan and oxaliplatin) and gemcitabine plus nab-paclitaxel has significantly improved the prognosis of patients with pancreatic cancer. However, there is still a high proportion of patients who develop metastatic pancreatic cancer in the course of chemotherapy or within a short period after chemotherapy. Previous reports have shown that chemotherapy-driven cytokine storms or the direct effects of certain chemotherapeutics on stromal and/or immune cells collectively change the microenvironment of the primary tumor, thus indirectly promoting metastasis. However, the mechanism underlying chemotherapy-induced metastasis in the course of chemotherapy, and afterwards, remains elusive in pancreatic cancer. In the present study, we aimed to determine the expression of CCL26 in the pancreatic cancer-associated fibroblasts (CAFs) after nab-paclitaxel treatment and to explore the role of CCL26 in the pancreatic adenocarcinoma (PDAC) invasion. Our results showed that nab-paclitaxel increased CCL26 mRNA and protein expression levels in a dose- and time-dependent manner. Subsequently, PDAC cell lines were treated with recombinant CCL26 for 48 h. The transwell migration assay showed that recombinant CCL26 enhanced the invasion of PDAC cells. Western blot analysis showed that the protein expression levels of phospho-(p-)PI3K, p-AKT, and p-mTOR were increased by CCL26 in PDAC cells. CCL26 expressions in 95 PDAC tissues and adjacent normal tissues were evaluated using reverse transcription-quantitative polymerase chain reaction and immunohistochemistry. CCL26 was found to be overexpressed in PDAC samples, and upregulated CCL26 expression was significantly associated with advanced perineural invasion, lymph node metastasis, and poor differentiation. In summary, our results showed that nab-paclitaxel increased the expression of CCL26 in CAFs, and CCL26 enhanced the invasive potential of pancreatic cancer cells by activating the PI3K/AKT/mTOR axis. Thus, CCL26 may be a potential prognostic biomarker for pancreatic cancer.
Collapse
Affiliation(s)
- Xuemin Chen
- Department of Hepato-Pancreato-Biliary Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213000, China
| | - Yong An
- Department of Hepato-Pancreato-Biliary Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213000, China
| | - Yue Zhang
- Department of Hepato-Pancreato-Biliary Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213000, China
| | - Dong Xu
- Department of General Surgery, Gaochun Branch, Drum Tower Hospital Affiliated to Nanjing University, Nanjing 211300, China
| | - Tongbing Chen
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou 213000, China
| | - Yue Yang
- Department of Hepato-Pancreato-Biliary Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213000, China
| | - Weibo Chen
- Department of Hepato-Pancreato-Biliary Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213000, China
| | - Di Wu
- Department of Hepato-Pancreato-Biliary Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213000, China
| | - Xiaoying Zhang
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213000, China
| |
Collapse
|
8
|
Wang ZX, Deng TX, Ma Z. Identification of a 4-miRNA signature as a potential prognostic biomarker for pancreatic adenocarcinoma. J Cell Biochem 2019; 120:16416-16426. [PMID: 31297864 DOI: 10.1002/jcb.28601] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/26/2022]
Abstract
An microRNA (miRNA) signature to predict the clinical outcome of pancreatic adenocarcinoma (PAAD) is still lacking. In the current study, we aimed at identifying and evaluating a prognostic miRNA signature for patients with PAAD. The miRNA expression profile and the clinical information regarding patients with PAAD were recruited from The Cancer Genome Atlas database. Differentially expressed miRNAs were identified between normal and tumor samples. By means of survival analysis, a 4-miRNA signature for predicting patients' with PAAD overall survival (OS) was constructed. Receiver operating characteristic (ROC) analysis was applied to determine the efficiency of survival prediction. Furthermore, the biological function of the predicted miRNAs was evaluated using a bioinformatics approach. Four (hsa-mir-126, hsa-mir-3613, hsa-mir-424, and hsa-mir-4772) out of 17 differentially expressed miRNAs were associated to the OS of patients with PAAD. Moreover, the area under the curve (AUC) of the constructed 4-miRNA signature associated to patients' with PAAD 2-year survival was 0.789. The multivariate Cox's proportional hazards regression model suggested that this 4-miRNA signature was an independent prognostic factor of other clinical parameters in patients with PAAD. Further pathway enrichment analyses revealed that the miRNAs in the 4-miRNA signature might regulate genes that affect focal adhesion, Wnt signaling pathway, and PI3K-Akt signaling pathway. Thus, these findings indicated that the 4-miRNA signature might be an effective independent prognostic biomarker in the prediction of PAAD patients' survival.
Collapse
Affiliation(s)
- Zhi-Xin Wang
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, Henan, China
| | - Tong-Xing Deng
- Department of Anatomy, Luohe Medical College, Luohe, Henan, China
| | - Zhao Ma
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
9
|
Ma J, Hou Y, Xia J, Zhu X, Wang ZP. Tumor suppressive role of rottlerin in cancer therapy. Am J Transl Res 2018; 10:3345-3356. [PMID: 30662591 PMCID: PMC6291697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 10/10/2018] [Indexed: 06/09/2023]
Abstract
Cancer as a major public health problem is a big trouble to be cured at present in the world. Thus, it is essential to discover better anticancer drugs to treat cancer patients. It has been reported that rottlerin, a natural polyphenolic compound from the mature fruits of Mallotus philippinensis, possesses multiple anti-cancer biological activities. Rottlerin exhibited its antitumor property in a variety of human cancers, suggesting that rottlerin could be a potential agent for treating cancers. In this review we discuss the recent literature regarding the biological functions and tumor suppressive mechanisms of rottlerin in cancers. We hope rottlerin will be further exploited for potential treatment of human cancers.
Collapse
Affiliation(s)
- Jia Ma
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical CollegeBengbu 233030, Anhui, China
| | - Yingying Hou
- Chinese Academy of Sciences Shanghai Institute of Materia MedicaShanghai 201203, China
| | - Jun Xia
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical CollegeBengbu 233030, Anhui, China
| | - Xueqiong Zhu
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325027, Zhejiang, China
| | - Z Peter Wang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical CollegeBengbu 233030, Anhui, China
- Center of Scientific Research, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325027, Zhejiang, China
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBoston, MA, USA
| |
Collapse
|
10
|
Liao X, Huang K, Huang R, Liu X, Han C, Yu L, Yu T, Yang C, Wang X, Peng T. Genome-scale analysis to identify prognostic markers in patients with early-stage pancreatic ductal adenocarcinoma after pancreaticoduodenectomy. Onco Targets Ther 2017; 10:4493-4506. [PMID: 28979141 PMCID: PMC5602474 DOI: 10.2147/ott.s142557] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Molecular analysis is a promising source of clinically useful prognostic biomarkers. The aim of this investigation was to identify prognostic biomarkers for patients with early-stage pancreatic ductal adenocarcinoma (PDAC) after pancreaticoduodenectomy. METHODS An RNA sequencing dataset of PDAC was obtained from The Cancer Genome Atlas. Survival analysis and weighted gene co-expression network analysis were used to investigate the prognostic markers of early-stage PDAC after pancreaticoduodenectomy. RESULTS Using whole genome expression level screening, we identified 1,238 markers that were related to the prognosis of PDAC after pancreaticoduodenectomy, and identified 9 hub genes (ARHGAP30, HCLS1, CD96, FAM78A, ARHGAP15, SLA2, CD247, GVINP1, and IL16) using the weighted gene co-expression network analysis approach. We also constructed a signature comprising the 9 hub genes and weighted by the regression coefficient derived from a multivariate Cox proportional hazards regression model to divide patients into a high-risk group, with increased risk of death, and a low-risk group, with significantly improved overall survival (adjusted P=0.026, adjusted HR =0.513, 95% CI =0.285-0.924). The prognostic signature of the 9 genes demonstrated good performance for predicting 1-year overall survival (area under the respective receiver operating characteristic curves =0.641). CONCLUSION Our results have provided a new prospect for prognostic biomarkers of PDAC after pancreaticoduodenectomy, and may have a value in clinical application.
Collapse
Affiliation(s)
- Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University
| | - Ketuan Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University
| | - Rui Huang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning
| | - Xiaoguang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University.,Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University
| | - Long Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University.,Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tingdong Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University
| | - Chengkun Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University
| | - Xiangkun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University
| |
Collapse
|
11
|
Liu W, Zhang B, Hu Q, Qin Y, Xu W, Shi S, Liang C, Meng Q, Xiang J, Liang D, Ji S, Liu J, Hu P, Liu L, Liu C, Long J, Ni Q, Yu X, Xu J. A new facet of NDRG1 in pancreatic ductal adenocarcinoma: Suppression of glycolytic metabolism. Int J Oncol 2017; 50:1792-1800. [PMID: 28350132 DOI: 10.3892/ijo.2017.3938] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/07/2017] [Indexed: 11/06/2022] Open
Abstract
N-myc downstream-regulated gene 1 (NDRG1) is known as tumor/metastasis suppressor in a variety of cancers including pancreas, being involved in angiogenesis, cancer growth and metastasis. However, the precise molecular mechanism how NDRG1 exerts its inhibitory function in pancreatic cancer remains unclear. In this investigation, we demonstrated that K-Ras plays a vital role in modulating NDRG1 protein level in PDAC cancer cells in vitro, which is mediated through ERK signaling. Noteworthy, K-Ras downstream Akt/mTOR signaling is inhibited upon NDRG1 overexpression, resulting in decease of HIF1α level. Moreover, NDRG1 has a unique role in modulating cancer metabolism of pancreatic ductal adenocarcinoma (PDAC). The mechanism accounting for NDRG1 in modulating aerobic glycolysis, at least partly, relied on its regulation of glycolysis genes including GLUT1, HK2, LDHA and PDK1. Additionally, NDRG1 is shown to suppress the activity of HIF1α, which is responsible for regulation of glycolysis enzymes. The current study is the first to elucidate a unique facet of the potent tumor/metastasis suppressor NDRG1 in the regulation of PDAC glycolysis, leading to important insights into the mechanism by which NDRG1 exert inhibitory function in PDAC.
Collapse
Affiliation(s)
- Wensheng Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Qiangsheng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Wenyan Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Chen Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Qingcai Meng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Jinfeng Xiang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Dingkong Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Jiang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Pengfei Hu
- Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Liang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Chen Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Jiang Long
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Quanxing Ni
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| |
Collapse
|
12
|
King H, Thillai K, Whale A, Arumugam P, Eldaly H, Kocher HM, Wells CM. PAK4 interacts with p85 alpha: implications for pancreatic cancer cell migration. Sci Rep 2017; 7:42575. [PMID: 28205613 PMCID: PMC5312077 DOI: 10.1038/srep42575] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 01/12/2017] [Indexed: 01/23/2023] Open
Abstract
It has been reported that p21-activated kinase 4 (PAK4) is amplified in pancreatic cancer tissue. PAK4 is a member of the PAK family of serine/threonine kinases, which act as effectors for several small GTPases, and has been specifically identified to function downstream of HGF-mediated c-Met activation in a PI3K dependent manner. However, the functionality of PAK4 in pancreatic cancer and the contribution made by HGF signalling to pancreatic cancer cell motility remain to be elucidated. We now find that elevated PAK4 expression is coincident with increased expression levels of c-Met and the p85α subunit of PI3K. Furthermore, we demonstrate that pancreatic cancer cells have a specific motility response to HGF both in 2D and 3D physiomimetic organotypic assays; which can be suppressed by inhibition of PI3K. Significantly, we report a specific interaction between PAK4 and p85α and find that PAK4 deficient cells exhibit a reduction in Akt phosphorylation downstream of HGF signalling. These results implicate a novel role for PAK4 within the PI3K pathway via interaction with p85α. Thus, PAK4 could be an essential player in PDAC progression representing an interesting therapeutic opportunity.
Collapse
Affiliation(s)
- Helen King
- Division of Cancer Studies, King's College London, UK
| | | | - Andrew Whale
- Division of Cancer Studies, King's College London, UK
| | - Prabhu Arumugam
- Barts Cancer Institute, a CRUK centre of Excellance, Queen Mary University of London, UK
| | - Hesham Eldaly
- Dept of Haematopathology Oncology Diagnostic Service, Addenbrooke's Hospital, Cambridge, UK
| | - Hemant M Kocher
- Barts Cancer Institute, a CRUK centre of Excellance, Queen Mary University of London, UK
| | | |
Collapse
|
13
|
Sharma N, Nanta R, Sharma J, Gunewardena S, Singh KP, Shankar S, Srivastava RK. PI3K/AKT/mTOR and sonic hedgehog pathways cooperate together to inhibit human pancreatic cancer stem cell characteristics and tumor growth. Oncotarget 2016; 6:32039-60. [PMID: 26451606 PMCID: PMC4741658 DOI: 10.18632/oncotarget.5055] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/22/2015] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) play major roles in cancer initiation, progression, and metastasis. It is evident from growing reports that PI3K/Akt/mTOR and Sonic Hedgehog (Shh) signaling pathways are aberrantly reactivated in pancreatic CSCs. Here, we examined the efficacy of combining NVP-LDE-225 (PI3K/mTOR inhibitor) and NVP-BEZ-235 (Smoothened inhibitor) on pancreatic CSCs characteristics, microRNA regulatory network, and tumor growth. NVP-LDE-225 co-operated with NVP-BEZ-235 in inhibiting pancreatic CSC's characteristics and tumor growth in mice by acting at the level of Gli. Combination of NVP-LDE-225 and NVP-BEZ-235 inhibited self-renewal capacity of CSCs by suppressing the expression of pluripotency maintaining factors Nanog, Oct-4, Sox-2 and c-Myc, and transcription of Gli. NVP-LDE-225 co-operated with NVP-BEZ-235 to inhibit Lin28/Let7a/Kras axis in pancreatic CSCs. Furthermore, a superior interaction of these drugs was observed on spheroid formation by pancreatic CSCs isolated from Pankras/p53 mice. The combination of these drugs also showed superior effects on the expression of proteins involved in cell proliferation, survival and apoptosis. In addition, NVP-LDE-225 co-operated with NVP-BEZ-235 in inhibiting EMT through modulation of cadherin, vimentin and transcription factors Snail, Slug and Zeb1. In conclusion, these data suggest that the combined inhibition of PI3K/Akt/mTOR and Shh pathways may be beneficial for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Narinder Sharma
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Rajesh Nanta
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | | | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Karan P Singh
- Division of Preventive Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Sharmila Shankar
- Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 66128, USA.,Department of Pathology, University of Missouri-School of Medicine, Kansas City, MO, 64108, USA
| | - Rakesh K Srivastava
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.,Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 66128, USA
| |
Collapse
|
14
|
Huang C, Wang WM, Gong JP, Yang K. Oncogenesis and the clinical significance of K-ras in pancreatic adenocarcinoma. Asian Pac J Cancer Prev 2015; 14:2699-701. [PMID: 23803017 DOI: 10.7314/apjcp.2013.14.5.2699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The RAS family genes encode small GTP-binding cytoplasmic proteins. Activated KRAS engages multiple effector pathways, notably the RAF-mitogen-activated protein kinase, phosphoinositide-3-kinase (PI3K) and RalGDS pathways. In the clinical field, K-ras oncogene activation is frequently found in human cancers and thus may serve as a potential diagnostic marker for cancer cells in circulation. This mini-review aims to summarise information on Ras-induced oncogenesis and the clinical significance of K-ras.
Collapse
Affiliation(s)
- Chun Huang
- Department of General Surgery, Three Gorges Medical College, Chongqing, China
| | | | | | | |
Collapse
|
15
|
Chen Y, Yang L, Cui T, Pacyna-Gengelbach M, Petersen I. HOPX is methylated and exerts tumour-suppressive function through Ras-induced senescence in human lung cancer. J Pathol 2014; 235:397-407. [DOI: 10.1002/path.4469] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 09/24/2014] [Accepted: 10/10/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Yuan Chen
- Institute of Pathology, University Hospital Jena; Friedrich-Schiller-University Jena; Ziegelmühlenweg 1 07743 Jena Germany
| | - Linlin Yang
- Institute of Pathology, University Hospital Jena; Friedrich-Schiller-University Jena; Ziegelmühlenweg 1 07743 Jena Germany
| | - Tiantian Cui
- Institute of Pathology, University Hospital Jena; Friedrich-Schiller-University Jena; Ziegelmühlenweg 1 07743 Jena Germany
| | | | - Iver Petersen
- Institute of Pathology, University Hospital Jena; Friedrich-Schiller-University Jena; Ziegelmühlenweg 1 07743 Jena Germany
| |
Collapse
|
16
|
Huang M, Tang SN, Upadhyay G, Marsh JL, Jackman CP, Srivastava RK, Shankar S. Rottlerin suppresses growth of human pancreatic tumors in nude mice, and pancreatic cancer cells isolated from Kras(G12D) mice. Cancer Lett 2014; 353:32-40. [PMID: 25050737 DOI: 10.1016/j.canlet.2014.06.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 05/12/2014] [Accepted: 06/09/2014] [Indexed: 01/29/2023]
Abstract
The purpose of the study was to examine the molecular mechanisms by which rottlerin inhibited growth of human pancreatic tumors in Balb C nude mice, and pancreatic cancer cells isolated from Kras(G12D) mice. AsPC-1 cells were injected subcutaneously into Balb c nude mice, and tumor-bearing mice were treated with rottlerin. Cell proliferation and apoptosis were measured by Ki67 and TUNEL staining, respectively. The expression of components of Akt, Notch, and Sonic Hedgehog (Shh) pathways were measured by the immunohistochemistry, Western blot analysis, and/or q-RT-PCR. The effects of rottlerin on pancreatic cancer cells isolated from Kras(G12D) mice were also examined. Rottlerin-treated mice showed a significant inhibition in tumor growth which was associated with suppression of cell proliferation, activation of capase-3 and cleavage of PARP. Rottlerin inhibited the expression of Bcl-2, cyclin D1, CDK2 and CDK6, and induced the expression of Bax in tumor tissues compared to untreated control. Rottlerin inhibited the markers of angiogenesis (Cox-2, VEGF, VEGFR, and IL-8), and metastasis (MMP-2 and MMP-9), thus blocking production of tumorigenic mediators in tumor microenvironment. Rottlerin also inhibited epithelial-mesenchymal transition by up-regulating E-cadherin and inhibiting the expression of Slug and Snail. Furthermore, rottlerin treatment of xenografted tumors or pancreatic cancer cells isolated from Kras(G12D) mice showed a significant inhibition in Akt, Shh and Notch pathways compared to control groups. These data suggest that rottlerin can inhibit pancreatic cancer growth by suppressing multiple signaling pathways which are constitutively active in pancreatic cancer. Taken together, our data show that the rottlerin induces apoptosis and inhibits pancreatic cancer growth by targeting Akt, Notch and Shh signaling pathways, and provide a new therapeutic approach with translational potential for humans.
Collapse
Affiliation(s)
- Minzhao Huang
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, The University of Kansas Cancer Center, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Su-Ni Tang
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, The University of Kansas Cancer Center, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Ghanshyam Upadhyay
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, The University of Kansas Cancer Center, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Justin L Marsh
- Department of Biochemistry, University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Christopher P Jackman
- Department of Biochemistry, University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Rakesh K Srivastava
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, The University of Kansas Cancer Center, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA.
| | - Sharmila Shankar
- Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO 64128, USA.
| |
Collapse
|
17
|
Zhao H, Wei R, Wang L, Tian Q, Tao M, Ke J, Liu Y, Hou W, Zhang L, Yang J, Hong T. Activation of glucagon-like peptide-1 receptor inhibits growth and promotes apoptosis of human pancreatic cancer cells in a cAMP-dependent manner. Am J Physiol Endocrinol Metab 2014; 306:E1431-41. [PMID: 24801389 DOI: 10.1152/ajpendo.00017.2014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) promotes pancreatic β-cell regeneration through GLP-1 receptor (GLP-1R) activation. However, whether it promotes exocrine pancreas growth and thereby increases the risk of pancreatic cancer has been a topic of debate in recent years. Clinical data and animal studies published so far have been controversial. In the present study, we report that GLP-1R activation with liraglutide inhibited growth and promoted apoptosis in human pancreatic cancer cell lines in vitro and attenuated pancreatic tumor growth in a mouse xenograft model in vivo. These effects of liraglutide were mediated through activation of cAMP production and consequent inhibition of Akt and ERK1/2 signaling pathways in a GLP-1R-dependent manner. Moreover, we examined GLP-1R expression in human pancreatic cancer tissues and found that 43.3% of tumor tissues were GLP-1R-null. In the GLP-1R-positive tumor tissues (56.7%), the level of GLP-1R was lower compared with that in tumor-adjacent normal pancreatic tissues. Furthermore, the GLP-1R-positive tumors were significantly smaller than the GLP-1R-null tumors. Our study shows for the first time that GLP-1R activation has a cytoreductive effect on human pancreatic cancer cells in vitro and in vivo, which may help address safety concerns of GLP-1-based therapies in the context of human pancreatic cancer.
Collapse
Affiliation(s)
- Hejun Zhao
- Department of Endocrinology and Metabolism and
| | - Rui Wei
- Department of Endocrinology and Metabolism and
| | - Liang Wang
- Department of Endocrinology and Metabolism and
| | - Qing Tian
- Department of Endocrinology and Metabolism and
| | - Ming Tao
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Jing Ke
- Department of Endocrinology and Metabolism and
| | - Ye Liu
- Department of Endocrinology and Metabolism and
| | - Wenfang Hou
- Department of Endocrinology and Metabolism and
| | - Lin Zhang
- Department of Endocrinology and Metabolism and
| | - Jin Yang
- Department of Endocrinology and Metabolism and
| | | |
Collapse
|
18
|
Huang M, Tang SN, Upadhyay G, Marsh JL, Jackman CP, Shankar S, Srivastava RK. Embelin suppresses growth of human pancreatic cancer xenografts, and pancreatic cancer cells isolated from KrasG12D mice by inhibiting Akt and Sonic hedgehog pathways. PLoS One 2014; 9:e92161. [PMID: 24694877 PMCID: PMC3973629 DOI: 10.1371/journal.pone.0092161] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Accepted: 02/19/2014] [Indexed: 12/23/2022] Open
Abstract
Pancreatic cancer is a deadly disease, and therefore effective treatment and/or prevention strategies are urgently needed. The objectives of this study were to examine the molecular mechanisms by which embelin inhibited human pancreatic cancer cell growth in vitro, and xenografts in Balb C nude mice, and pancreatic cancer cell growth isolated from KrasG12D transgenic mice. XTT assays were performed to measure cell viability. AsPC-1 cells were injected subcutaneously into Balb c nude mice and treated with embelin. Cell proliferation and apoptosis were measured by Ki67 and TUNEL staining, respectively. The expression of Akt, and Sonic Hedgehog (Shh) and their target gene products were measured by the immunohistochemistry, and Western blot analysis. The effects of embelin on pancreatic cancer cells isolated from 10-months old KrasG12D mice were also examined. Embelin inhibited cell viability in pancreatic cancer AsPC-1, PANC-1, MIA PaCa-2 and Hs 766T cell lines, and these inhibitory effects were blocked either by constitutively active Akt or Shh protein. Embelin-treated mice showed significant inhibition in tumor growth which was associated with reduced expression of markers of cell proliferation (Ki67, PCNA and Bcl-2) and cell cycle (cyclin D1, CDK2, and CDK6), and induction of apoptosis (activation of caspase-3 and cleavage of PARP, and increased expression of Bax). In addition, embelin inhibited the expression of markers of angiogenesis (COX-2, VEGF, VEGFR, and IL-8), and metastasis (MMP-2 and MMP-9) in tumor tissues. Antitumor activity of embelin was associated with inhibition of Akt and Shh pathways in xenografts, and pancreatic cancer cells isolated from KrasG12D mice. Furthermore, embelin also inhibited epithelial-to-mesenchymal transition (EMT) by up-regulating E-cadherin and inhibiting the expression of Snail, Slug, and ZEB1. These data suggest that embelin can inhibit pancreatic cancer growth, angiogenesis and metastasis by suppressing Akt and Shh pathways, and can be developed for the treatment and/or prevention of pancreatic cancer.
Collapse
Affiliation(s)
- Minzhao Huang
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, The University of Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Su-Ni Tang
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, The University of Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Ghanshyam Upadhyay
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, The University of Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Justin L. Marsh
- Department of Biochemistry, University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
| | - Christopher P. Jackman
- Department of Biochemistry, University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
| | - Sharmila Shankar
- Department of Pathology and Laboratory Medicine, The University of Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * E-mail: (SS); (RKS)
| | - Rakesh K. Srivastava
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, The University of Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * E-mail: (SS); (RKS)
| |
Collapse
|
19
|
Utomo WK, Narayanan V, Biermann K, van Eijck CHJ, Bruno MJ, Peppelenbosch MP, Braat H. mTOR is a promising therapeutical target in a subpopulation of pancreatic adenocarcinoma. Cancer Lett 2014; 346:309-17. [PMID: 24467966 DOI: 10.1016/j.canlet.2014.01.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 01/17/2014] [Accepted: 01/20/2014] [Indexed: 12/30/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a highly lethal disease, unusually resistant against therapy. It is generally felt that stratification of patients for personalized medicine is the way forward. Here, we report that a subpopulation of PDACs shows strong activation of the mTOR signaling cassette. Moreover, we show that inhibition of mTOR in pancreatic cancer cell lines showing high levels of mTOR signaling is associated with cancer cell death. Finally, we show using fine needle biopsies the existence of a subpopulation of PDAC patients with high activation of the mTOR signaling cassette and provide evidence that inhibition of mTOR might be clinically useful for this group. Thus, our results define an unrecognized subpopulation of PDACs, characterized by high activation of mTOR and show that identification of this specific patient group in the early phase of diagnosis is feasible.
Collapse
Affiliation(s)
- Wesley K Utomo
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, The Netherlands
| | - Vilvapathy Narayanan
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, The Netherlands
| | | | | | - Marco J Bruno
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Henri Braat
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
20
|
Palumbo S, Tsai TL, Li WJ. Macrophage migration inhibitory factor regulates AKT signaling in hypoxic culture to modulate senescence of human mesenchymal stem cells. Stem Cells Dev 2014; 23:852-65. [PMID: 24274936 DOI: 10.1089/scd.2013.0294] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hypoxic culture has been shown to delay premature senescence occurring during in vitro culture. Human mesenchymal stem cells (hMSCs) cultured under hypoxia have been reported to maintain their stemness properties and delay senescence compared to the cells cultured under normoxia. However, the molecular mechanism by which hypoxia regulates premature senescence has not been fully revealed. In this study, hMSCs were cultured under the conditions of 21% (normoxia) and 1% O2 (hypoxia) tension and analyzed for cell growth, expression of MSC surface markers, multilineage differentiation, and cellular senescence. Our results showed that more cells retained MSC surface markers in hypoxic culture than those in normoxic culture, and hypoxia was able to enhance multilineage differentiation of hMSCs. The hypoxic condition also delayed cellular senescence of hMSCs, increased activation of AKT signaling, and upregulated both intra- and extracellular levels of macrophage migration inhibitory factor (MIF) compared to the normoxic condition. Inhibition of AKT activity in hypoxic culture increased the number of cells with positive staining for senescence-associated β-galactosidase activity, upregulated expression levels of senescence-associated markers p16 and p21 mRNA transcripts, and decreased expression levels of potency-associated markers, including NANOG, OCT3/4, and SOX2. On the other hand, upregulated intra- and extracellular levels of MIF by stable MIF overexpression in normoxic culture increased the activation of AKT while decreasing mRNA expression of senescence-associated markers and increasing expression of potency-associated markers. Taken together, our findings suggest that hMSCs in hypoxic culture produce endogenous MIF to activate AKT signaling to delay the progression of cellular senescence.
Collapse
Affiliation(s)
- SunMi Palumbo
- 1 Laboratory of Musculoskeletal Biology and Regenerative Medicine, Department of Orthopedics and Rehabilitation, School of Medicine and Public Health, University of Wisconsin-Madison , Madison, Wisconsin
| | | | | |
Collapse
|
21
|
Matsubara S, Ding Q, Miyazaki Y, Kuwahata T, Tsukasa K, Takao S. mTOR plays critical roles in pancreatic cancer stem cells through specific and stemness-related functions. Sci Rep 2013; 3:3230. [PMID: 24231729 PMCID: PMC3828572 DOI: 10.1038/srep03230] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 10/31/2013] [Indexed: 12/23/2022] Open
Abstract
Pancreatic cancer is characterized by near-universal mutations in KRAS. The mammalian target of rapamycin (mTOR), which functions downstream of RAS, has divergent effects on stem cells. In the present study, we investigated the significance of the mTOR pathway in maintaining the properties of pancreatic cancer stem cells. The mTOR inhibitor, rapamycin, reduced the viability of CD133+ pancreatic cancer cells and sphere formation which is an index of self-renewal of stem-like cells, indicating that the mTOR pathway functions to maintain cancer stem-like cells. Further, rapamycin had different effects on CD133+ cells compared to cyclopamine which is an inhibitor of the Hedgehog pathway. Thus, the mTOR pathway has a distinct role although both pathways maintain pancreatic cancer stem cells. Therefore, mTOR might be a promising target to eliminate pancreatic cancer stem cells.
Collapse
Affiliation(s)
- Shyuichiro Matsubara
- Cancer and Regenerative Medicine, Frontier Biomedical Science and Swine Research Center, Sakuragaoka, Kagoshima, 890-8520, Japan
| | | | | | | | | | | |
Collapse
|
22
|
Zhao M, Tang SN, Marsh JL, Shankar S, Srivastava RK. Ellagic acid inhibits human pancreatic cancer growth in Balb c nude mice. Cancer Lett 2013; 337:210-7. [DOI: 10.1016/j.canlet.2013.05.009] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 04/23/2013] [Accepted: 05/09/2013] [Indexed: 12/30/2022]
|
23
|
Li N, Bu X, Tian X, Wu P, Yang L, Huang P. Fatty acid synthase regulates proliferation and migration of colorectal cancer cells via HER2-PI3K/Akt signaling pathway. Nutr Cancer 2012; 64:864-70. [PMID: 22860766 DOI: 10.1080/01635581.2012.701704] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recent evidence suggests that fatty acid synthase mediating de novo fatty acid synthesis plays a crucial role in the carcinogenesis process of various cancers. Moreover, HER2 and related PI3K/Akt signaling pathway, which links intimately with cellular metabolism, influence cancer biological behavior. However, it remains unknown whether malignant phenotype of colorectal cancer cells is regulated by the HER2-PI3K/Akt-FASN signaling pathway. In this study, Caco-2 cells were selected for functional characterization, and treated with ZSTK474, followed by RT-qPCR and Western blot assays examining PI3K, Akt, HER2, and FASN expression. The MTT and colony formation assays were used to assess proliferation. The migration was investigated by transwell, apoptosis, and cell-cycle analysis. We found that the blockade of PI3K/Akt signaling pathway by ZSTK474 treatment led to downregulation of PI3K, Akt, HER2, and FASN expression. The proliferation was decreased upon treatment which was consistent with an increased percentage of G(1) arrested cells instead of apoptosis. The migration of Caco-2 cells was also impaired by ZSTK474 treatment. Inhibition of HER2-PI3K/Akt signaling pathway suppresses FASN expression of Caco-2 cells, and inhibition of FASN expression changes malignant phenotype of Caco-2 cells.
Collapse
Affiliation(s)
- Nan Li
- Department of Internal Medicine, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | | | | | | | | | | |
Collapse
|