1
|
Chu YD, Chen CW, Lai MW, Lim SN, Lin WR. Bioenergetic alteration in gastrointestinal cancers: The good, the bad and the ugly. World J Gastroenterol 2023; 29:4499-4527. [PMID: 37621758 PMCID: PMC10445009 DOI: 10.3748/wjg.v29.i29.4499] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/23/2023] [Accepted: 07/03/2023] [Indexed: 08/02/2023] Open
Abstract
Cancer cells exhibit metabolic reprogramming and bioenergetic alteration, utilizing glucose fermentation for energy production, known as the Warburg effect. However, there are a lack of comprehensive reviews summarizing the metabolic reprogramming, bioenergetic alteration, and their oncogenetic links in gastrointestinal (GI) cancers. Furthermore, the efficacy and treatment potential of emerging anticancer drugs targeting these alterations in GI cancers require further evaluation. This review highlights the interplay between aerobic glycolysis, the tricarboxylic acid (TCA) cycle, and oxidative phosphorylation (OXPHOS) in cancer cells, as well as hypotheses on the molecular mechanisms that trigger this alteration. The role of hypoxia-inducible transcription factors, tumor suppressors, and the oncogenetic link between hypoxia-related enzymes, bioenergetic changes, and GI cancer are also discussed. This review emphasizes the potential of targeting bioenergetic regulators for anti-cancer therapy, particularly for GI cancers. Emphasizing the potential of targeting bioenergetic regulators for GI cancer therapy, the review categorizes these regulators into aerobic glycolysis/ lactate biosynthesis/transportation and TCA cycle/coupled OXPHOS. We also detail various anti-cancer drugs and strategies that have produced pre-clinical and/or clinical evidence in treating GI cancers, as well as the challenges posed by these drugs. Here we highlight that understanding dysregulated cancer cell bioenergetics is critical for effective treatments, although the diverse metabolic patterns present challenges for targeted therapies. Further research is needed to comprehend the specific mechanisms of inhibiting bioenergetic enzymes, address side effects, and leverage high-throughput multi-omics and spatial omics to gain insights into cancer cell heterogeneity for targeted bioenergetic therapies.
Collapse
Affiliation(s)
- Yu-De Chu
- Liver Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Chun-Wei Chen
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Ming-Wei Lai
- Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Siew-Na Lim
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Wey-Ran Lin
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Department of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
2
|
Ayala-Calvillo E, Rodríguez-Fragoso L, Álvarez-Ayala E, Leija-Salas A. EGF-receptor phosphorylation and downstream signaling are activated by genistein during subacute liver damage. J Mol Histol 2023:10.1007/s10735-023-10127-8. [PMID: 37227557 DOI: 10.1007/s10735-023-10127-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/05/2023] [Indexed: 05/26/2023]
Abstract
The epidermal growth factor receptor (EGFR) plays an important role on hepatic protection in acute and chronic liver injury. The aim of this study was to investigate the role of genistein on EGFR expression, phosphorylation and signaling pathways in experimental subacute liver damage induced by carbon tetrachloride (CCl4). We used male Wistar rats that were randomly divided into four groups: (1) Control; (2) Genistein 5 mg/kg per oral; (3) Subacute liver damage induced by CCl4 4 mg/kg subcutaneously; and (4) Animals received CCl4 and genistein at the dosage indicated. The effect of genistein on EGFR expression, phosphorylation and signaling pathways were investigated by western blot and densitometric analyses. Histological changes were evaluated on slices stained with Hematoxylin-Eosin and Masson´s trichromic, as well as an immunohistochemical analysis for proliferating cell nuclear antigen (PCNA). Additionally, pro-inflammatory cytokines and liver enzymes were quantified. Our study showed that genistein increased EGFR expression, EGFR-specific tyrosine residues phosphorylation (pY1068-EGFR and pY84-EGFR), signal transducer and activator of transcription phosphorylation (pSTAT5), protein kinase B phosphorylation (pAKT) and PCNA in animals with CCl4-induced subacute liver damage. It was found a significant reduction of pro-inflammatory cytokines in serum from animals with subacute liver damage treated with genistein. Those effects were reflected in an improvement in the architecture and liver function. In conclusion, genistein can induce a transactivation of EGFR leading to downstream cell signaling pathways as early events associated with regeneration and hepatoprotection following subacute liver damage.
Collapse
Affiliation(s)
- Erick Ayala-Calvillo
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos. Av Universidad, 1001 Col. Chamilpa CP 62210, Cuernavaca, Morelos, México
| | - Lourdes Rodríguez-Fragoso
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos. Av Universidad, 1001 Col. Chamilpa CP 62210, Cuernavaca, Morelos, México
| | - Elizabeth Álvarez-Ayala
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos. Av Universidad, 1001 Col. Chamilpa CP 62210, Cuernavaca, Morelos, México
| | - Alfonso Leija-Salas
- Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Av. Universidad 2001, CP62210 Col. Chamilpa, Morelos, Cuernavaca, Mexico.
| |
Collapse
|
3
|
Molecular and Cellular Mechanisms of Propolis and Its Polyphenolic Compounds against Cancer. Int J Mol Sci 2022; 23:ijms231810479. [PMID: 36142391 PMCID: PMC9499605 DOI: 10.3390/ijms231810479] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 12/12/2022] Open
Abstract
In recent years, interest in natural products such as alternative sources of pharmaceuticals for numerous chronic diseases, including tumors, has been renewed. Propolis, a natural product collected by honeybees, and polyphenolic/flavonoid propolis-related components modulate all steps of the cancer progression process. Anticancer activity of propolis and its compounds relies on various mechanisms: cell-cycle arrest and attenuation of cancer cells proliferation, reduction in the number of cancer stem cells, induction of apoptosis, modulation of oncogene signaling pathways, inhibition of matrix metalloproteinases, prevention of metastasis, anti-angiogenesis, anti-inflammatory effects accompanied by the modulation of the tumor microenvironment (by modifying macrophage activation and polarization), epigenetic regulation, antiviral and bactericidal activities, modulation of gut microbiota, and attenuation of chemotherapy-induced deleterious side effects. Ingredients from propolis also "sensitize" cancer cells to chemotherapeutic agents, likely by blocking the activation of the transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). In this review, we summarize the current knowledge related to the the effects of flavonoids and other polyphenolic compounds from propolis on tumor growth and metastasizing ability, and discuss possible molecular and cellular mechanisms involved in the modulation of inflammatory pathways and cellular processes that affect survival, proliferation, invasion, angiogenesis, and metastasis of the tumor.
Collapse
|
4
|
Chiawpanit C, Panwong S, Sawasdee N, Yenchitsomanus PT, Panya A. Genistein Sensitizes Human Cholangiocarcinoma Cell Lines to Be Susceptible to Natural Killer Cells. BIOLOGY 2022; 11:biology11081098. [PMID: 35892954 PMCID: PMC9330512 DOI: 10.3390/biology11081098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022]
Abstract
Cholangiocarcinoma (CCA) is a lethal bile duct cancer, which has poor treatment outcomes due to its high resistance to chemotherapy and cancer recurrence. Activation of aberrant anti-apoptotic signaling pathway has been reported to be a mechanism of chemoresistance and immune escape of CCA. Therefore, reversal of anti-apoptotic signaling pathway represents a feasible approach to potentiate effective treatments, especially for CCA with high chemoresistance. In this study, we demonstrated the effects of genistein on reactivation of apoptosis cascade and increase the susceptibility of CCA cells to natural killer (NK-92) cells. Genistein at 50 and 100 µM significantly activated extrinsic apoptotic pathway in CCA cells (KKU055, KKU100, and KKU213A), which was evident by reduction of procaspase-8 and -3 expression. Pretreatment of CCA cells with genistein at 50 µM, but not NK-92 cells, significantly increased NK-92 cell killing ability over the untreated control, suggesting the ability of genistein to sensitize CCA cells. Interestingly, genistein treatment could greatly lower the expression of cFLIP, an anti-apoptotic protein involved in the immune escape pathway, in addition to upregulation of death receptors, Fas- and TRAIL-receptors, in CCA cells, which might be the underlying molecular mechanism of genistein to sensitize CCA to be susceptible to NK-92 cells. Taken together, this finding revealed the benefit of genistein as a sensitizer to enhance the efficiency of NK cell immunotherapy for CCA.
Collapse
Affiliation(s)
- Chutipa Chiawpanit
- Doctoral Program in Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand;
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Suthida Panwong
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand;
- Doctoral Program in Applied Microbiology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nunghathai Sawasdee
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (N.S.); (P.-t.Y.)
- Division of Molecular Medicine, Research Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Pa-thai Yenchitsomanus
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (N.S.); (P.-t.Y.)
- Division of Molecular Medicine, Research Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Aussara Panya
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand;
- Research Center in Bioresources for Agriculture, Industry and Medicine, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence: ; Tel.: +66-53-943346
| |
Collapse
|
5
|
Lu C, Miao J, Li M, Zheng Q, Xu F, Pan Y, Wang Y, Yang Z, Xia X, Zhu H, Chen J, Bao S. Characterization of the Estrogen Response Helps to Predict Prognosis and Identify Potential Therapeutic Targets in Cholangiocarcinoma. Front Oncol 2022; 12:870840. [PMID: 35664769 PMCID: PMC9162778 DOI: 10.3389/fonc.2022.870840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/19/2022] [Indexed: 11/21/2022] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive malignancy originating from the epithelium of the bile duct. The prognosis of patients is poor regardless of radical resection and chemoradiotherapy. The current classification and prognostic model of CCA are unable to satisfy the requirements for predicting the clinical outcome and exploring therapeutic targets. Estrogen signaling is involved in diverse cancer types, and it has long been established that CCA could be regulated by estrogen. In our study, estrogen response was identified to be significantly and stably correlated with poor prognosis in CCA. Employing several algorithms, CCA was classified into ES cluster A and B. ES cluster B was mainly composed of patients with fluke infection and overlapped with CCA cluster 1/2, and ES cluster A was mainly composed of patients without fluke infection and overlapped with CCA cluster 3/4. COMT and HSD17B1 were identified to be responsible for the differential estrogen response between ES clusters A and B, and the estrogen response may be correlated with the differentiation and cancer stemness of CCA at the single-cell level. Complement activation and the expression of C3 and C5, which are mainly expressed by CCA cells, were significantly downregulated in ES cluster B. An estrogen response risk score (ESRS) model was constructed to predict the prognosis of CCA, followed by a nomogram integrating ESRS and clinical features. Finally, altered pathways, applicable drugs and sensitivity to chemical drugs were analyzed specific to the estrogen response. In summary, our results provide insights into the role of the estrogen response in CCA progression as well as applicable drugs and potential therapeutic targets in estrogen metabolism, the complement system and ESRS-related pathways.
Collapse
Affiliation(s)
- Chenglin Lu
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ji Miao
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Minhuan Li
- Department of Andrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qisi Zheng
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Feng Xu
- Department of General Surgery, Changshu NO.1 People’s Hospital, The Affiliated Hospital of Soochow University, Changshu, China
| | - Yiming Pan
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yizhou Wang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhi Yang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xuefeng Xia
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Hao Zhu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- *Correspondence: Shanhua Bao, ; Jie Chen, ; Hao Zhu,
| | - Jie Chen
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- *Correspondence: Shanhua Bao, ; Jie Chen, ; Hao Zhu,
| | - Shanhua Bao
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- *Correspondence: Shanhua Bao, ; Jie Chen, ; Hao Zhu,
| |
Collapse
|
6
|
Wang J, Li D, Zhao B, Kim J, Sui G, Shi J. Small Molecule Compounds of Natural Origin Target Cellular Receptors to Inhibit Cancer Development and Progression. Int J Mol Sci 2022; 23:ijms23052672. [PMID: 35269825 PMCID: PMC8911024 DOI: 10.3390/ijms23052672] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/16/2022] [Accepted: 02/25/2022] [Indexed: 01/03/2023] Open
Abstract
Receptors are macromolecules that transmit information regulating cell proliferation, differentiation, migration and apoptosis, play key roles in oncogenic processes and correlate with the prognoses of cancer patients. Thus, targeting receptors to constrain cancer development and progression has gained widespread interest. Small molecule compounds of natural origin have been widely used as drugs or adjuvant chemotherapeutic agents in cancer therapies due to their activities of selectively killing cancer cells, alleviating drug resistance and mitigating side effects. Meanwhile, many natural compounds, including those targeting receptors, are still under laboratory investigation for their anti-cancer activities and mechanisms. In this review, we classify the receptors by their structures and functions, illustrate the natural compounds targeting these receptors and discuss the mechanisms of their anti-cancer activities. We aim to provide primary knowledge of mechanistic regulation and clinical applications of cancer therapies through targeting deregulated receptors.
Collapse
Affiliation(s)
| | | | | | | | - Guangchao Sui
- Correspondence: (G.S.); (J.S.); Tel.: +86-451-82191081 (G.S. & J.S.)
| | - Jinming Shi
- Correspondence: (G.S.); (J.S.); Tel.: +86-451-82191081 (G.S. & J.S.)
| |
Collapse
|
7
|
Kazybay B, Ahmad A, Mu C, Mengdesh D, Xie Y. Omicron N501Y mutation among SARS-CoV-2 lineages: Insilico analysis of potent binding to tyrosine kinase and hypothetical repurposed medicine. Travel Med Infect Dis 2022; 45:102242. [PMID: 34929375 PMCID: PMC8677628 DOI: 10.1016/j.tmaid.2021.102242] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022]
Abstract
Variants of SARS-CoV-2 lineages including the most recently circulated Omicron, and previous pandemic B.1.351, B.1.1.7, which have been public concerns, contain a N501Y mutation located in the spike receptor binding domain. However, the potential interactions with host cells linking N501Y mutation to pathogenic relevance remain elusive. Recently, we and others report that kinases such as PI3K/AKT signaling are essential in SARS-CoV-2 entry. Here we analyzed the predicted potential kinases interacting with the mutation. Bioinformatics tools including structure-prediction based molecular docking analysis were applied. We found kinases such as EGFR might potentially act as new factors involving the N501Y mutation binding through possible phosphorylation at Y501 and enhanced affinity in certain variants. To our surprise, the Omicron receptor binding domain harboring N501Y mutation did not enhance binding to EGFR which might be due to the mutations of charged polar to uncharged polar side chains located on the interaction interfaces. Similarly, potent gains of phosphorylation in B.1.351 and B.1.1.7 by mutations were predicted and interaction networks were analyzed with enrichment of pathways. Given kinases might be elevated in cancer patients, the N501Y mutation containing lineages may be possibly much more infectious and additional care for cancer management might be taken into consideration by precision prevention, therapy or recovery.
Collapse
Affiliation(s)
- Bexultan Kazybay
- Biology Department, Nazarbayev University, Nur-Sultan, 010000, Kazakhstan
| | - Ashfaq Ahmad
- Department of Bioinformatics, Hazara University, Mansehra, 21300, Pakistan
| | - Chenglin Mu
- Zhejiang University, Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Diana Mengdesh
- Biology Department, Nazarbayev University, Nur-Sultan, 010000, Kazakhstan
| | - Yingqiu Xie
- Biology Department, Nazarbayev University, Nur-Sultan, 010000, Kazakhstan.
| |
Collapse
|
8
|
Li T, Kuang T, Yang Z, Zhang Q, Zhang W, Fan Y. Co-treatment With Everolimus, an mTOR-Specific Antagonist, or Downregulation of ELK1 Enhances the Sensitivity of Pancreatic Cancer Cells to Genistein. Front Cell Dev Biol 2021; 9:633035. [PMID: 34540820 PMCID: PMC8448347 DOI: 10.3389/fcell.2021.633035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 08/06/2021] [Indexed: 12/24/2022] Open
Abstract
Genistein is a natural isoflavone with pharmacological or potentially anti-tumor properties. However, the resistance of cancer cells to genistein remains a major obstacle. This study focused on the mechanism implicated in the resistance of pancreatic cancer (PC) cells to genistein and the mechanism of action. First, key molecules and signaling pathways related to genistein resistance in PC cells were explored using bioinformatics tools. DEP domain containing MTOR interacting protein (DEPTOR), a typical inhibitor of the mammalian target of rapamycin (mTOR) signaling, was predicted to be poorly expressed in the genistein-resistant PC cells. Thereafter, genistein-resistant PC cells (Panc-1 and PaCa) were constructed. Altered expression of DEPTOR was introduced in cells, and everolimus (ELM), an mTOR-specific antagonist, was administrated in cells as well to examine their roles in genistein resistance. The cell apoptosis was examined in vitro and in vivo in mouse xenograft tumors. The upstream regulator of DEPTOR was predicted via bioinformatic tools. The bioinformatic analyses showed that the PI3K/AKT/mTOR signaling pathway was activated in the setting of DEPTOR downregulation in genistein-resistant PC cells. DEPTOR overexpression reduced the 50% inhibiting concentration (IC50) of genistein in PC cells and suppressed mTOR phosphorylation, and it increased caspase-3 activity, LDH release and apoptosis in PC cells. ELM treatment enhanced the sensitivity of PC cells to genistein in vitro and it strengthened the tumor-eliminating role of genistein in mice. ETS transcription factor ELK1 (ELK1), a transcription factor that negatively regulated DEPTOR transcription, was suppressed by genistein. Upregulation of ELK1 suppressed DEPTOR transcription and reduced the genistein sensitivity of cells, and it also blocked the genistein-sensitizing roles of ELM in PC cells. In conclusion, this study demonstrated that ELK1 reduces DEPTOR transcription, leading to mTOR phosphorylation and the drug resistance of PC cells.
Collapse
Affiliation(s)
- Tianyu Li
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tiantao Kuang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhaoshuo Yang
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiqi Zhang
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wen Zhang
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yue Fan
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Tsugami Y, Wakasa H, Nishimura T, Kobayashi K. Genistein Directly Represses the Phosphorylation of STAT5 in Lactating Mammary Epithelial Cells. ACS OMEGA 2021; 6:22765-22772. [PMID: 34514247 PMCID: PMC8427774 DOI: 10.1021/acsomega.1c03107] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/16/2021] [Indexed: 06/13/2023]
Abstract
Genistein is a soy isoflavone and shows various physiological activities, such as affinities for estrogen receptors (ERs) and inhibitory effects on the epidermal growth factor receptor (EGFR) pathway. A previous study reported that genistein downregulates milk production ability in mammary epithelial cells (MECs) while decreasing the phosphorylation of STAT5. The ER and EGFR pathways indirectly regulate STAT5. In this study, the repressing mechanism of genistein against the phosphorylation of STAT5 was investigated using a culture model of mouse MECs with milk production ability. The results revealed that genistein did not influence the behavior of ERα and ERβ, whereas genistein immediately repressed the phosphorylation of ERK1/2. However, the decrease in phosphorylated STAT5 occurred independent of the phosphorylation of EGFR. Genistein repressed new phosphorylation of STAT5 by prolactin without influencing the phosphorylation of JAK2. In conclusion, this study indicates that genistein directly inhibits the phosphorylation of STAT5 in lactating MECs.
Collapse
Affiliation(s)
- Yusaku Tsugami
- Laboratory
of Animal Histophysiology, Graduate School of Integrated Sciences
for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima 739-8528 Hiroshima, Japan
| | - Haruka Wakasa
- Laboratory
of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9 060-8589 Sapporo, Japan
| | - Takanori Nishimura
- Laboratory
of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9 060-8589 Sapporo, Japan
| | - Ken Kobayashi
- Laboratory
of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9 060-8589 Sapporo, Japan
| |
Collapse
|
10
|
Ponte LGS, Pavan ICB, Mancini MCS, da Silva LGS, Morelli AP, Severino MB, Bezerra RMN, Simabuco FM. The Hallmarks of Flavonoids in Cancer. Molecules 2021; 26:2029. [PMID: 33918290 PMCID: PMC8038160 DOI: 10.3390/molecules26072029] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
Flavonoids represent an important group of bioactive compounds derived from plant-based foods and beverages with known biological activity in cells. From the modulation of inflammation to the inhibition of cell proliferation, flavonoids have been described as important therapeutic adjuvants against several diseases, including diabetes, arteriosclerosis, neurological disorders, and cancer. Cancer is a complex and multifactor disease that has been studied for years however, its prevention is still one of the best known and efficient factors impacting the epidemiology of the disease. In the molecular and cellular context, some of the mechanisms underlying the oncogenesis and the progression of the disease are understood, known as the hallmarks of cancer. In this text, we review important molecular signaling pathways, including inflammation, immunity, redox metabolism, cell growth, autophagy, apoptosis, and cell cycle, and analyze the known mechanisms of action of flavonoids in cancer. The current literature provides enough evidence supporting that flavonoids may be important adjuvants in cancer therapy, highlighting the importance of healthy and balanced diets to prevent the onset and progression of the disease.
Collapse
Affiliation(s)
- Luis Gustavo Saboia Ponte
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Isadora Carolina Betim Pavan
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
- Laboratory of Signal Mechanisms (LMS), School of Pharmaceutical Sciences (FCF), University of Campinas (UNICAMP), Campinas, São Paulo 13083-871, Brazil
| | - Mariana Camargo Silva Mancini
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Luiz Guilherme Salvino da Silva
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Ana Paula Morelli
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Matheus Brandemarte Severino
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Rosangela Maria Neves Bezerra
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Fernando Moreira Simabuco
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| |
Collapse
|
11
|
Yang W, Sun Y. Promising Molecular Targets for the Targeted Therapy of Biliary Tract Cancers: An Overview. Onco Targets Ther 2021; 14:1341-1366. [PMID: 33658799 PMCID: PMC7920611 DOI: 10.2147/ott.s297643] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Biliary tract cancer (BTC) is a leading cause of cancer-related death, due to the limited benefits of current systematic therapies and the heterogeneity of the tumor itself. High heterogeneity means that the clinical and molecular features vary between different subtypes of BTC, while the underlying molecular mechanisms remain unclear. Targeted therapy, where inhibitors are developed to selectively combine with targeted molecules in order to block abnormal signaling pathways in BTC, has shown promise as an emerging form of treatment for various types of cancer. In this article, a comprehensive review is conducted to examine potential molecular targets for BTC targeted therapy and their mechanisms. Furthermore, preliminary data published from clinical trials is utilized to analyze the main drugs used to combat BTC. The collective information presented in this article has provided useful insights into the current understanding of BTC.
Collapse
Affiliation(s)
- Wenwei Yang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Yongkun Sun
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| |
Collapse
|
12
|
Gill MSA, Saleem H, Ahemad N. Plant Extracts and their Secondary Metabolites as Modulators of Kinases. Curr Top Med Chem 2021; 20:1093-1104. [PMID: 32091334 DOI: 10.2174/1568026620666200224100219] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/20/2020] [Accepted: 01/24/2020] [Indexed: 12/20/2022]
Abstract
Natural Products (NP), specifically from medicinal plants or herbs, have been extensively utilized to analyze the fundamental mechanisms of ultimate natural sciences as well as therapeutics. Isolation of secondary metabolites from these sources and their respective biological properties, along with their lower toxicities and cost-effectiveness, make them a significant research focus for drug discovery. In recent times, there has been a considerable focus on isolating new chemical entities from natural flora to meet the immense demand for kinase modulators, and also to overcome major unmet medical challenges in relation to signal transduction pathways. The signal transduction systems are amongst the foremost pathways involved in the maintenance of life and protein kinases play an imperative part in these signaling pathways. It is important to find a kinase inhibitor, as it can be used not only to study cell biology but can also be used as a drug candidate for cancer and metabolic disorders. A number of plant extracts and their isolated secondary metabolites such as flavonoids, phenolics, terpenoids, and alkaloids have exhibited activities against various kinases. In the current review, we have presented a brief overview of some important classes of plant secondary metabolites as kinase modulators. Moreover, a number of phytocompounds with kinase inhibition potential, isolated from different plant species, are also discussed.
Collapse
Affiliation(s)
- Muhammad Shoaib Ali Gill
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Petaling Jaya 47500, Selangor Darul Ehsan, Malaysia.,Institute of Pharmaceutical Sciences (IPS), University of Veterinary and Animal Sciences (UVAS), Lahore 54000, Pakistan
| | - Hammad Saleem
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Petaling Jaya 47500, Selangor Darul Ehsan, Malaysia.,Institute of Pharmaceutical Sciences (IPS), University of Veterinary and Animal Sciences (UVAS), Lahore 54000, Pakistan
| | - Nafees Ahemad
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Petaling Jaya 47500, Selangor Darul Ehsan, Malaysia.,Tropical Medicine and Biology Platform, Monash University Malaysia, Jalan Lagoon Selatan, Petaling Jaya 47500, Selangor Darul Ehsan, Malaysia.,Global Asia in 21st Century Platform, Monash University Malaysia, Jalan Lagoon Selatan, Petaling Jaya 47500, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
13
|
Chen Z, Lin T, Liao X, Li Z, Lin R, Qi X, Chen G, Sun L, Lin L. Network pharmacology based research into the effect and mechanism of Yinchenhao Decoction against Cholangiocarcinoma. Chin Med 2021; 16:13. [PMID: 33478536 PMCID: PMC7818939 DOI: 10.1186/s13020-021-00423-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/01/2021] [Accepted: 01/04/2021] [Indexed: 12/14/2022] Open
Abstract
Background Cholangiocarcinoma refers to an epithelial cell malignancy with poor prognosis. Yinchenhao decoction (YCHD) showed positive effects on cancers, and associations between YCHD and cholangiocarcinoma remain unclear. This study aimed to screen out the effective active components of Yinchenhao decoction (YCHD) using network pharmacology, estimate their potential targets, screen out the pathways, as well as delve into the potential mechanisms on treating cholangiocarcinoma. Methods By the traditional Chinese medicine system pharmacology database and analysis platform (TCMSP) as well as literature review, the major active components and their corresponding targets were estimated and screened out. Using the software Cytoscape 3.6.0, a visual network was established using the active components of YCHD and the targets of cholangiocarcinoma. Based on STRING online database, the protein interaction network of vital targets was built and analyzed. With the Database for Annotation, Visualization, and Integrated Discovery (DAVID) server, the gene ontology (GO) biological processes and the Kyoto encyclopedia of genes and genomes (KEGG) signaling pathways of the targets enrichment were performed. The AutoDock Vina was used to perform molecular docking and calculate the binding affinity. The PyMOL software was utilized to visualize the docking results of active compounds and protein targets. In vivo experiment, the IC50 values and apoptosis rate in PI-A cells were detected using CCK-8 kit and Cell Cycle Detection Kit. The predicted targets were verified by the real-time PCR and western blot methods. Results 32 effective active components with anti-tumor effects of YCHD were sifted in total, covering 209 targets, 96 of which were associated with cancer. Quercetin, kaempferol, beta-sitosterol, isorhamnetin, and stigmasterol were identified as the vital active compounds, and AKT1, IL6, MAPK1, TP53 as well as VEGFA were considered as the major targets. The molecular docking revealed that these active compounds and targets showed good binding interactions. These 96 putative targets exerted therapeutic effects on cancer by regulating signaling pathways (e.g., hepatitis B, the MAPK signaling pathway, the PI3K-Akt signaling pathway, and MicroRNAs in cancer). Our in vivo experimental results confirmed that YCHD showed therapeutic effects on cholangiocarcinoma by decreasing IC50 values, down-regulating apoptosis rate of cholangiocarcinoma cells, and lowering protein expressions. Conclusions As predicted by network pharmacology strategy and validated by the experimental results, YCHD exerts anti-tumor effectsthrough multiple components, targets, and pathways, thereby providing novel ideas and clues for the development of preparations and the treatment of cholangiocarcinoma.
Collapse
Affiliation(s)
- Zhiqiang Chen
- The First School of Clinical Medical Sciences, Guangzhou University of Chinese Medicine, 510405, Guangzhou, China
| | - Tong Lin
- The First School of Clinical Medical Sciences, Guangzhou University of Chinese Medicine, 510405, Guangzhou, China
| | - Xiaozhong Liao
- The First School of Clinical Medical Sciences, Guangzhou University of Chinese Medicine, 510405, Guangzhou, China
| | - Zeyun Li
- The First School of Clinical Medical Sciences, Guangzhou University of Chinese Medicine, 510405, Guangzhou, China
| | - Ruiting Lin
- The First School of Clinical Medical Sciences, Guangzhou University of Chinese Medicine, 510405, Guangzhou, China
| | - Xiangjun Qi
- The First School of Clinical Medical Sciences, Guangzhou University of Chinese Medicine, 510405, Guangzhou, China
| | - Guoming Chen
- The First School of Clinical Medical Sciences, Guangzhou University of Chinese Medicine, 510405, Guangzhou, China
| | - Lingling Sun
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 16, Jichang Road, Baiyun District, 510405, Guangzhou, China
| | - Lizhu Lin
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 16, Jichang Road, Baiyun District, 510405, Guangzhou, China.
| |
Collapse
|
14
|
Chen L, Yang ZS, Zhou YZ, Deng Y, Jiang P, Tan SL. Dihydromyricetin inhibits cell proliferation, migration, invasion and promotes apoptosis via regulating miR-21 in Human Cholangiocarcinoma Cells. J Cancer 2020; 11:5689-5699. [PMID: 32913463 PMCID: PMC7477438 DOI: 10.7150/jca.45970] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022] Open
Abstract
Dihydromyricetin, the most abundant natural flavonoid isolated from Ampelopsis grossedentata, exhibits broad anti-tumor effects. However, the effects of dihydromyricetin on cholangiocarcinoma remain unclear. This study examined the anti-tumor effects of dihydromyricetin in two human cholangiocarcinoma cell lines HCCC9810 and TFK-1, and the underlying mechanism was also investigated. Our study was the first to show that dihydromyricetin significantly inhibited cell proliferation, migration, invasion and promoted apoptosis in cholangiocarcinoma cells. By analyzing the TCGA dataset, we found that expression of miR-21, an oncogene and a potential target of anticancer drugs for cholangiocarcinoma, was upregulated in cholangiocarcinoma tissues compared to paired control tissues. Moreover, dihydromyricetin significantly reduced the expression of miR-21 in a dose-dependent manner. Overexpression of miR-21 remarkably abolished the inhibitory effects of dihydromyricetin on cell proliferation, migration, invasion and abrogated its effect of promoting cell apoptosis in both HCCC9810 and TFK-1 cells. Dihydromyricetin remarkably increased the expression of PTEN and decreased the expression of phosphorylated Akt, while overexpression of miR-21 abrogated the modulation of PTEN/ Akt pathway by dihydromyricetin. Taken together, our study demonstrates that dihydromyricetin inhibits cell proliferation, migration, invasion and promotes apoptosis in cholangiocarcinoma cells via regulating miR-21.
Collapse
Affiliation(s)
- Lei Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China, 410011.,Institute of Clinical Pharmacy, Central South University, Changsha, China, 410011
| | - Zhou-Sheng Yang
- Department of Pharmacy, The People's Hopital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China, 530021
| | - Yang-Zhao Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China, 410011
| | - Yang Deng
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, China, 410015
| | - Pei Jiang
- Department of Clinical Pharmacy and Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China, 272000
| | - Sheng-Lan Tan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China, 410011.,Institute of Clinical Pharmacy, Central South University, Changsha, China, 410011
| |
Collapse
|
15
|
James CD, Morgan IM, Bristol ML. The Relationship between Estrogen-Related Signaling and Human Papillomavirus Positive Cancers. Pathogens 2020; 9:E403. [PMID: 32455952 PMCID: PMC7281727 DOI: 10.3390/pathogens9050403] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/15/2022] Open
Abstract
High risk-human papillomaviruses (HPVs) are known carcinogens. Numerous reports have linked the steroid hormone estrogen, and the expression of estrogen receptors (ERs), to HPV-related cancers, although the exact nature of the interactions remains to be fully elucidated. Here we will focus on estrogen signaling and describe both pro and potentially anti-cancer effects of this hormone in HPV-positive cancers. This review will summarize: (1) cell culture-related evidence, (2) animal model evidence, and (3) clinical evidence demonstrating an interaction between estrogen and HPV-positive cancers. This comprehensive review provides insights into the potential relationship between estrogen and HPV. We suggest that estrogen may provide a potential therapeutic for HPV-related cancers, however additional studies are necessary.
Collapse
Affiliation(s)
- Claire D. James
- School of Dentistry, Philips Institute for Oral Health Research, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA;
| | - Iain M. Morgan
- School of Dentistry, Philips Institute for Oral Health Research, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA;
- VCU Massey Cancer Center, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA
| | - Molly L. Bristol
- School of Dentistry, Philips Institute for Oral Health Research, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA;
| |
Collapse
|
16
|
Network Pharmacology-Based Strategy for Predicting Therapy Targets of Traditional Chinese Medicine Xihuang Pill on Liver Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:6076572. [PMID: 32256653 PMCID: PMC7102465 DOI: 10.1155/2020/6076572] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 01/16/2020] [Accepted: 01/22/2020] [Indexed: 02/07/2023]
Abstract
Objective To investigate the potential therapy targets and pharmacological mechanism of traditional Chinese medicine (TCM) Xihuang pill in liver cancer based on network pharmacology. Methods Drug ingredients-target network was constructed based on the target sets of Xihuang pill and liver cancer. The overlapping genes between Xihuang pill targets and liver cancer-related molecular targets were investigated using comparative analysis. Moreover, the PPI network and module was constructed based on overlapping genes and hub nodes, respectively, followed by the pathway enrichment analysis. Results A drug ingredients-target network was established with 1184 nodes and 11035 interactions. Moreover, a total of 106 overlapping genes were revealed between drug targets and liver cancer molecular targets. Furthermore, a PPI network and 4 modules were further investigated based on overlapping genes, respectively. These hub nodes such as VEGFA and EGFR were mainly enriched in GO functions including positive regulation of MAP kinase activity, activation of protein kinase activity, regulation of MAP kinase activity, and pathways like proteoglycans in cancer, bladder cancer, and estrogen signaling. Conclusion VEGFA and EGFR might be potential therapy targets of Xihuang pill in liver cancer. Furthermore, the effect of Xihuang pill on liver cancer might be realized by targeting VEGFA and EGFR in pathways like proteoglycans in cancer and estrogen signaling.
Collapse
|
17
|
Sato K, Francis H, Zhou T, Meng F, Kennedy L, Ekser B, Baiocchi L, Onori P, Mancinelli R, Gaudio E, Franchitto A, Glaser S, Alpini G. Neuroendocrine Changes in Cholangiocarcinoma Growth. Cells 2020; 9:E436. [PMID: 32069926 PMCID: PMC7072848 DOI: 10.3390/cells9020436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/09/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
Cholangiocarcinoma (CCA) is a highly aggressive malignancy that emerges from the biliary tree. There are three major classes of CCA-intrahepatic, hilar (perihilar), or distal (extrahepatic)-according to the location of tumor development. Although CCA tumors are mainly derived from biliary epithelia (i.e., cholangiocytes), CCA can be originated from other cells, such as hepatic progenitor cells and hepatocytes. This heterogeneity of CCA may be responsible for poor survival rates of patients, limited effects of chemotherapy and radiotherapy, and the lack of treatment options and novel therapies. Previous studies have identified a number of neuroendocrine mediators, such as hormones, neuropeptides, and neurotransmitters, as well as corresponding receptors. The mediator/receptor signaling pathways play a vital role in cholangiocyte proliferation, as well as CCA progression and metastases. Agonists or antagonists for candidate pathways may lead to the development of novel therapies for CCA patients. However, effects of mediators may differ between healthy or cancerous cholangiocytes, or between different subtypes of receptors. This review summarizes current understandings of neuroendocrine mediators and their functional roles in CCA.
Collapse
Affiliation(s)
- Keisaku Sato
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Heather Francis
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - Tianhao Zhou
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX 77807, USA
| | - Fanyin Meng
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - Lindsey Kennedy
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Burcin Ekser
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Leonardo Baiocchi
- Liver Unit, Department of Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Paolo Onori
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Romina Mancinelli
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | | | - Shannon Glaser
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX 77807, USA
| | - Gianfranco Alpini
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
18
|
Zhou M, Zhu Y, Hou R, Mou X, Tan J. Identification of candidate genes for the diagnosis and treatment of cholangiocarcinoma using a bioinformatics approach. Oncol Lett 2019; 18:5459-5467. [PMID: 31612054 PMCID: PMC6781666 DOI: 10.3892/ol.2019.10904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 08/20/2019] [Indexed: 12/13/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a biliary epithelial tumor with poor prognosis. As the key genes and signaling pathways underlying the disease have not been fully elucidated, the aim of the present study was to improve the understanding of the molecular mechanisms associated with CCA. The microarray datasets GSE26566 and GSE89749 were downloaded from the Gene Expression Omnibus and differentially expressed genes (DEGs) between CCA and normal bile duct samples were identified. Gene and pathway enrichment analyses were performed, and a protein-protein interaction network was constructed and analyzed. A total of 159 DEGs and 10 hub genes were identified. The functions and pathways of the DEGs were mainly enriched in ‘heparin binding’, ‘serine-type endopeptidase activity’, ‘calcium ion binding’, ‘pancreatic secretion’, ‘fat digestion and absorption’ and ‘protein digestion and absorption’. Survival analysis revealed that the upregulated expression of carboxypeptidase B1 and Kruppel like factor 4 was significantly associated with lower overall survival rate. In summary, the present study identified DEGs and hub genes associated with CCA, which may serve as potential diagnostic and therapeutic targets for the disease.
Collapse
Affiliation(s)
- Mi Zhou
- Department of Cell Biology, The Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Yabin Zhu
- Department of Cell Biology, The Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Ruixia Hou
- Department of Cell Biology, The Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xianbo Mou
- Department of Cell Biology, The Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jun Tan
- Department of Hepatology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
19
|
Tong Y, Wang M, Huang H, Zhang J, Huang Y, Chen Y, Pan H. Inhibitory effects of genistein in combination with gefitinib on the hepatocellular carcinoma Hep3B cell line. Exp Ther Med 2019; 18:3793-3800. [PMID: 31611933 PMCID: PMC6781792 DOI: 10.3892/etm.2019.8027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 07/08/2019] [Indexed: 01/27/2023] Open
Abstract
Combination therapy is an important method for treating advanced hepatocellular carcinoma (HCC). Gefitinib is an epidermal growth factor receptor (EGFR) inhibitor, which has profound effects on HCC. The purpose of the present study was to investigate the effects of genistein in combination with gefitinib on the proliferation and apoptosis of HCC cells and the associated mechanism. Cell counting kit-8 assay was performed to calculate the IC50 values and cytotoxicity, whilst flow cytometry was used to assess cell apoptosis. Protein expression was detected using western blot analysis. The IC50 of genistein and gefitinib on Hep3B cells were calculated to be 128.078 and 13.657 µM, respectively. Genistein in combination with gefitinib significantly inhibited cell viability, promoted apoptosis and reduced EGFR, vascular endothelial growth factor receptor and platelet-derived growth factor receptor phosphorylation. Genistein in combination with gefitinib promoted the expression of cleaved caspase-3 and cleaved poly ADP-ribose polymerase. In addition, combined treatment of genistein and gefitinib strongly inhibited the activation of the Akt/Erk/mTOR signaling pathway. In conclusion, findings from the present study suggest that genistein in combination with gefitinib inhibit HCC cell proliferation and promote apoptosis by inhibiting the Akt/Erk/mTOR pathway.
Collapse
Affiliation(s)
- Yongxi Tong
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Mingshan Wang
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Haijun Huang
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Jiajie Zhang
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Yicheng Huang
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Yingjun Chen
- Department of Infectious Diseases, Tiantai People's Hospital of Zhejiang Province, Taizhou, Zhejiang 317200, P.R. China
| | - Hongying Pan
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
20
|
Guerra B, Issinger OG. Natural Compounds and Derivatives as Ser/Thr Protein Kinase Modulators and Inhibitors. Pharmaceuticals (Basel) 2019; 12:E4. [PMID: 30609679 PMCID: PMC6469162 DOI: 10.3390/ph12010004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022] Open
Abstract
The need for new drugs is compelling, irrespective of the disease. Focusing on medical problems in the Western countries, heart disease and cancer are at the moment predominant illnesses. Owing to the fact that ~90% of all 21,000 cellular proteins in humans are regulated by phosphorylation/dephosphorylation it is not surprising that the enzymes catalysing these reactions (i.e., protein kinases and phosphatases, respectively) have attracted considerable attention in the recent past. Protein kinases are major team players in cell signalling. In tumours, these enzymes are found to be mutated disturbing the proper function of signalling pathways and leading to uncontrolled cellular growth and sustained malignant behaviour. Hence, the search for small-molecule inhibitors targeting the altered protein kinase molecules in tumour cells has become a major research focus in the academia and pharmaceutical companies.
Collapse
Affiliation(s)
- Barbara Guerra
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark.
| | - Olaf-Georg Issinger
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark.
| |
Collapse
|