1
|
Abbenante D, Scotti B, Baraldi C, LA Placa M, Bardazzi F. Multiple disseminated clear-cell acanthomas associated with colon cancer: more than a mere coincidence? Ital J Dermatol Venerol 2023; 158:166-167. [PMID: 37153955 DOI: 10.23736/s2784-8671.23.07493-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Affiliation(s)
- Diego Abbenante
- Unit of Dermatology, IRCCS AOU Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Biagio Scotti
- Unit of Dermatology, IRCCS AOU Bologna, Bologna, Italy -
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Carlotta Baraldi
- Unit of Dermatology, IRCCS AOU Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Michelangelo LA Placa
- Unit of Dermatology, IRCCS AOU Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | | |
Collapse
|
2
|
Lin G, Yang J, Liu J, Shangguan J, Pan H, Zhang Y, Ran K, Li D, Yu F, Xu H. In situ polyphenol-adhesive hydrogel enhanced the noncarcinogenic repairing of KGF on the gut epithelial barrier on TNBS-induced colitis rats. Int J Biol Macromol 2023; 231:123323. [PMID: 36669631 DOI: 10.1016/j.ijbiomac.2023.123323] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/09/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023]
Abstract
Ulcerative colitis (UC) is a chronic recurrent disease affecting the gastrointestinal tract especially colorectum. Keratinocyte growth factor (KGF) plays the vital roles in maintaining the colonic mucosal barrier. The poor stability and off-target of KGF were two hindering factors for its clinical application. Herein, in situ hydrogel (PE) with mucoadhesive ability was constructed by using temperature-sensitive poloxamer and EGCG as hydrogel-forming material and adhesive enhancer, respectively. Incorporation of EGCG led to the slight decrease of the gelled temperature and shortened the gelled time of PE hydrogel. When the concentration of EGCG is 0.1 %, PE hydrogel exhibits the suitable viscosity of 280 ± 20 Pa·s and the strong adhesive force of 725 ± 25 mN. KGF was soluble in cold PE solution to obtain KGF-loaded PE hydrogel (KGF@PE). PE hydrogel could improve the stability of KGF in vitro. KGF@PE not only could recover greatly the body weight of TNBS-induced rats but also repair their colonic morphology and goblet cell function. Moreover, the potential of repairing the epithelial barrier was indicated by upregulating tight junction proteins. Importantly, the safety of KGF@PE hydrogel for colitis was also confirmed on AOM/DSS-induced mice models. Conclusively, KGF@PE may be a promising therapeutic platform without obvious side effect for ulcerative colitis.
Collapse
Affiliation(s)
- Gaolong Lin
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Jiaojiao Yang
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Jiayi Liu
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Jianxun Shangguan
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Hanxiao Pan
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Yingying Zhang
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Kunjie Ran
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Dingwei Li
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Fengnan Yu
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Helin Xu
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China.
| |
Collapse
|
3
|
Shantha Kumara HMC, Shah A, Miyagaki H, Yan X, Cekic V, Hedjar Y, Whelan RL. Plasma Levels of Keratinocyte Growth Factor Are Significantly Elevated for 5 Weeks After Minimally Invasive Colorectal Resection Which May Promote Cancer Recurrence and Metastasis. Front Surg 2021; 8:745875. [PMID: 34820416 PMCID: PMC8606552 DOI: 10.3389/fsurg.2021.745875] [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: 07/22/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Human Keratinocyte Growth Factor (KGF) is an FGF family protein produced by mesenchymal cells. KGF promotes epithelial cell proliferation, plays a role in wound healing and may also support tumor growth. It is expressed by some colorectal cancers (CRC). Surgery's impact on KGF levels is unknown. This study's purpose was to assess plasma KGF levels before and after minimally invasive colorectal resection (MICR) for CRC. Aim: To determine plasma KGF levels before and after minimally invasive colorectal resection surgery for cancer pathology. Method: CRC MICR patients (pts) in an IRB approved data/plasma bank were studied. Pre-operative (pre-op) and post-operative (post-op) plasma samples were taken/stored. Late samples were bundled into 7 day blocks and considered as single time points. KGF levels (pg/ml) were measured via ELISA (mean ± SD). The Wilcoxon paired t-test was used for statistical analysis. Results: Eighty MICR CRC patients (colon 61%; rectal 39%; mean age 65.8 ± 13.3) were studied. The mean incision length was 8.37 ± 3.9 and mean LOS 6.5 ± 2.6 days. The cancer stage breakdown was; I (23), II (26), III (27), and IV (4). The median pre-op KGF level was 17.1 (95 %CI: 14.6-19.4; n = 80); significantly elevated (p < 0.05) median levels (pg/ml) were noted on post-op day (POD) 1 (23.4 pg/ml; 95% CI: 21.4-25.9; n = 80), POD 3 (22.5 pg/ml; 95% CI: 20.7-25.9; n = 76), POD 7-13 (21.8 pg/ml; 95% CI: 17.7-25.4; n = 50), POD 14-20 (20.1 pg/ml; 95% CI: 17.1-23.9; n = 33), POD 21-27 (19.6 pg/ml; 95% CI: 15.2-24.9; n = 15) and on POD 28-34 (16.7 pg/ml; 95% CI: 14.0-25.8; n = 12). Conclusion: Plasma KGF levels were significantly elevated for 5 weeks after MICR for CRC. The etiology of these changes is unclear, surgical trauma related acute inflammatory response and wound healing process may play a role. These changes, may stimulate angiogenesis in residual tumor deposits after surgery.
Collapse
Affiliation(s)
- H M C Shantha Kumara
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, United States
| | - Abhinit Shah
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, United States
| | | | - Xiaohong Yan
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, United States
| | - Vesna Cekic
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, United States
| | - Yanni Hedjar
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, United States
| | - Richard L Whelan
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| |
Collapse
|
4
|
Pashirzad M, Khorasanian R, Fard MM, Arjmand MH, Langari H, Khazaei M, Soleimanpour S, Rezayi M, Ferns GA, Hassanian SM, Avan A. The Therapeutic Potential of MAPK/ERK Inhibitors in the Treatment of Colorectal Cancer. Curr Cancer Drug Targets 2021; 21:932-943. [PMID: 34732116 DOI: 10.2174/1568009621666211103113339] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/16/2021] [Accepted: 08/24/2021] [Indexed: 11/22/2022]
Abstract
The MAPK/ERK signaling pathway regulates cancer cell proliferation, apoptosis, inflammation, angiogenesis, metastasis and drug resistance. Mutations and up-regulation of components of the MAPK/ERK signaling pathway, as well as over-activation of this critical signaling pathway, are frequently observed in colorectal carcinomas. Targeting the MAPK/ERK signaling pathway, using specific pharmacological inhibitors, elicits potent anti-tumor effects, supporting the therapeutic potential of these inhibitors in the treatment of CRC. Several drugs have recently been developed for the inhibition of the MEK/ERK pathway in preclinical and clinical settings, such as MEK162 and MK-2206. MEK1/2 inhibitors demonstrate promising efficacy and anticancer activity for the treatment of this malignancy. This review summarizes the current knowledge on the role of the MAPK/ERK signaling pathway in the pathogenesis of CRC and the potential clinical value of synthetic inhibitors of this pathway in preventing CRC progression for a better understanding, and hence, better management of colorectal cancer.
Collapse
Affiliation(s)
- Mehran Pashirzad
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad. Iran
| | - Reihaneh Khorasanian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad. Iran
| | - Maryam Mahmoudi Fard
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad. Iran
| | - Mohammad-Hassan Arjmand
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord. Iran
| | - Hadis Langari
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord. Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad. Iran
| | - Saman Soleimanpour
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad. Iran
| | - Majid Rezayi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord. Iran
| | - Gordon A Ferns
- Division of Pulmonary and Critical Care Medicine, Washington University, School of Medicine, Saint Louis, MO. United States
| | - Seyed Mahdi Hassanian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad. Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad. Iran
| |
Collapse
|
5
|
Pulido T, Velarde MC, Alimirah F. The senescence-associated secretory phenotype: Fueling a wound that never heals. Mech Ageing Dev 2021; 199:111561. [PMID: 34411604 DOI: 10.1016/j.mad.2021.111561] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 07/29/2021] [Accepted: 08/12/2021] [Indexed: 12/15/2022]
Abstract
Wound healing is impaired with advanced age and certain chronic conditions, such as diabetes and obesity. Moreover, common cancer treatments, including chemotherapy and radiation, can cause unintended tissue damage and impair wound healing. Available wound care treatments are not always effective, as some wounds fail to heal or recur after treatment. Hence, a more thorough understanding of the pathophysiology of chronic, nonhealing wounds may offer new ideas for the development of effective wound care treatments. Cancers are sometimes referred to as wounds that never heal, sharing mechanisms similar to wound healing. We describe in this review how cellular senescence and the senescence-associated secretory phenotype (SASP) contribute to chronic wounds versus cancer.
Collapse
Affiliation(s)
- Tanya Pulido
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Michael C Velarde
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, 1101, Philippines.
| | | |
Collapse
|
6
|
Farajihaye Qazvini F, Samadi N, Saffari M, Emami-Razavi AN, Shirkoohi R. Fibroblast growth factor-10 and epithelial-mesenchymal transition in colorectal cancer. EXCLI JOURNAL 2019; 18:530-539. [PMID: 31611737 PMCID: PMC6785779 DOI: 10.17179/excli2018-1784] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 07/10/2019] [Indexed: 12/12/2022]
Abstract
As an inducer of epithelial-mesenchymal transition (EMT), fibroblast growth factor-10 (FGF-10) has a role in cell proliferation and differentiation in the embryo in addition to invasion and metastasis during carcinogenesis. In this study, we aimed to investigate the FGF-10 gene expression in tumor tissues based on the pathological feature of tumor related to EMT and metastasis. 62 tumors were obtained from 62 colorectal cancer patients during surgery. The pathological characteristics of the patients were carefully collected and classified by Iran National Tumor Bank. To quantify FGF-10 gene expression, RNA extraction, reverse transcription-PCR and real-time PCR were respectively performed. In addition, three colorectal cancer cell lines including LS174T, SW-948 and SW-480 were collected and cultured for further molecular analysis. Consequently, FGF-10 gene expression showed increased expression level in LS174T and SW-948 while it displayed decreased level in SW-480. Considering the tumor samples, we found an upregulation of FGF-10 gene expression in 52.1 % of all tumors in stage III and only in 9.09 % of all tumors in stage I. Also, there were an upregulation of FGF-10 gene expression in 50 % of all positive lymph invasion patients. Besides, FGF-10 gene upregulation was observed in 50 % of all tumors with a size larger than 5 cm (P value < 0.05) and 69 % of all tumors located in the colon (P value < 0.05). To our knowledge, this is the first time that FGF-10 expression is reported based on pathological features of colorectal cancer.
Collapse
Affiliation(s)
- Fatemeh Farajihaye Qazvini
- Department of Clinical Biochemistry and Laboratory Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Group of Genetics, Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Nasser Samadi
- Department of Clinical Biochemistry and Laboratory Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mojtaba Saffari
- Group of Genetics, Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Amir Nader Emami-Razavi
- Iran National Tumor Bank, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Shirkoohi
- Group of Genetics, Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Patel A, Tripathi G, McTernan P, Gopalakrishnan K, Ali O, Spector E, Williams N, Arasaradnam RP. Fibroblast growth factor 7 signalling is disrupted in colorectal cancer and is a potential marker of field cancerisation. J Gastrointest Oncol 2019; 10:429-436. [PMID: 31183192 DOI: 10.21037/jgo.2019.02.11] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Field cancerisation proposes that there are pre-malignant genetic mutations in the macroscopically normal mucosal tissue around colorectal cancer. This study aims to evaluate fibroblast growth factor 7 (FGF7) tissue expression in the mucosal field around colorectal cancer. Methods Gene and protein expression of FGF7, its receptor, FGFR2 and its downstream targets; FRS2α, Erk 1/2 and Akt was measured from mucosal samples in 34 control subjects and 17 cancer patients. Serial samples from tumour, adjacent to tumour and at the resection margin were utilised. Results FGF7 gene expression was significantly higher in tumour (2.3-fold), adjacent mucosa (3.2-fold) and resection margin (2.8-fold) of cancer patients compared with control subjects (P<0.01 respectively). However, FGFR2 was down regulated (3.5-fold) in the tumour tissue (P<0.001). Protein expression of FRS2α and Akt was significantly lower in tumour tissue compared with the resection margin in cancer patients (P<0.05 respectively). No differences in protein expression of Erk 1/2 were detected. Conclusions FGF7 was elevated in the mucosal field of cancer patients supporting its potential as a biomarker of field cancerisation. Changes in FRS2α, Akt and Erk 1/2 expression in the tumour tissue indicate that with malignant transformation, FGF7 loses its ability to regulate cellular differentiation.
Collapse
Affiliation(s)
- Abhilasha Patel
- Department of Colorectal Surgery, University Hospitals of Coventry and Warwickshire NHS Trust, Coventry, UK.,Warwick Medical School, University of Warwick, Coventry, UK
| | | | | | - Kishore Gopalakrishnan
- Department of Pathology, University Hospitals of Coventry and Warwickshire NHS Trust, Coventry, UK
| | - Omar Ali
- Warwick Medical School, University of Warwick, Coventry, UK
| | - Emma Spector
- Warwick Medical School, University of Warwick, Coventry, UK
| | - Nigel Williams
- Department of Colorectal Surgery, University Hospitals of Coventry and Warwickshire NHS Trust, Coventry, UK
| | - Ramesh P Arasaradnam
- Warwick Medical School, University of Warwick, Coventry, UK.,Department of Gastroenterology, University Hospitals of Coventry and Warwickshire NHS Trust, Coventry, UK.,University of Coventry, Coventry, UK.,University of Leicester, Leicester, UK
| |
Collapse
|
8
|
Jafari B, Hamzeh-Mivehroud M, Moosavi-Movahedi AA, Dastmalchi S. Identification of Novel Single-Domain Antibodies against FGF7 Using Phage Display Technology. SLAS DISCOVERY 2017; 23:193-201. [DOI: 10.1177/2472555217728520] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Fibroblast growth factor 7 (FGF7) is a member of the fibroblast growth factor (FGF) family of proteins. FGF7 is of stromal origin and produces a paracrine effect on epithelial cells. In the current investigation, we aimed to identify new single-domain antibodies (sdAbs) against FGF7 using phage display technology. The vector harboring the codon-optimized DNA sequence for FGF7 protein was transformed into Escherichia coli BL21 (DE3) pLysS, and then the protein was expressed at the optimized condition. Enzyme-linked immunosorbent assay, circular dichroism spectropolarimetry, and in vitro scratch assay experiments were used to confirm the proper folding and functionality of the purified FGF7 protein. The purity of the produced FGF7 was 92%, with production yield of 3.5 mg/L of culture. Panning against the purified FGF7 was performed, and the identified single-domain antibodies showed significant affinity. Further investigation on one of the selected sdAb displaying phage clones showed concentration-dependent binding to FGF7. The selected sdAb can be used for developing novel tumor-suppressing agents where inhibition of FGF7 is required.
Collapse
Affiliation(s)
- Behzad Jafari
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hamzeh-Mivehroud
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
9
|
Abstract
Colorectal cancers develop through at least 3 major pathways, including chromosomal instability, mismatch repair, and methylator phenotype. These pathways can coexist in a single individual and occur in both sporadic and inherited colorectal cancers. In spite of the unique molecular and genetic signatures of colorectal cancers, nonspecific chemotherapy based on the antineoplastic effects of 5-fluorouracil is the cornerstone of therapy for stage III and some stage II disease. Techniques to recognize colorectal cancer at the molecular level have facilitated development of new signature drugs designed to inhibit the unique pathways of colorectal cancer growth and immunity.
Collapse
|
10
|
Keratinocyte Growth Factor Regulation of Aryl Hydrocarbon Receptor Activation in Colorectal Cancer Cells. Dig Dis Sci 2016; 61:444-52. [PMID: 26514676 DOI: 10.1007/s10620-015-3908-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 09/28/2015] [Indexed: 12/09/2022]
Abstract
BACKGROUND Keratinocyte growth factor (KGF) stimulates normal growth, development and intestinal epithelial cell proliferation. Cyclin D1 promotes the cell cycle by inhibiting retinoblastoma protein (RB1). The activated aryl hydrocarbon receptor (AhR) has an important influence on the development of tumors through its interactions with the cell cycle. AIM The aim of the present study was to explore a new role for AhR in KGF-induced colon cancer cell growth. MATERIALS AND METHODS Real-time PCR, western blot or immunofluorescence analysis were used to detect the expression of KGF, AhR, cyclin D1 and CYP1A1. Immunohistochemistry was used to observe the localization of AhR. MTT assay and flow cytometric analyses were performed to measure cell viability and the cell cycle. RESULTS Real-time PCR analysis revealed that KGF, AhR, and CYP1A1 mRNAs were overexpressed in colorectal cancer tissues. Meanwhile, overexpression of AhR was primarily observed in epithelial cells. In in vitro assay, KGF promoted colon cancer cell growth, as well as up-regulated and activated AhR. At the same time, AhR-knockdown colon cancer cells were less responsive to KGF. Western blot analysis, real-time PCR, or immunofluorescence data indicated that cyclin D1 expression was up-regulated by KGF but this up-regulation was compromised when AhR was silenced, and the cell cycle was arrested in the G0/G1 stage in these cells. CONCLUSIONS Our study suggests that KGF, AhR, and CYP1A1 are overexpressed in colorectal cancer tissues. Moreover, we reveal a new mechanism by which KGF promotes cell proliferation through the AhR-cyclin D1 pathway in colon cancer cells.
Collapse
|
11
|
Clinicopathological correlation of keratinocyte growth factor and matrix metalloproteinase-9 expression in human gastric cancer. TUMORI JOURNAL 2015; 101:566-71. [PMID: 26350198 DOI: 10.5301/tj.5000367] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2015] [Indexed: 02/01/2023]
Abstract
AIMS AND BACKGROUND Keratinocyte growth factor (KGF) is reported to be implicated in the growth of some cancer cells. Matrix metalloproteinase 9 (MMP-9) is thought to enhance the tumor invasion and metastasis ability. This study was aimed at analyzing the relationship between KGF and MMP-9 expression and patients' clinicopathological characteristics to clarify the clinical significance of the expression of KGF and MMP-9 in gastric cancer. METHODS Tissue samples from 161 patients with primary gastric cancer were investigated using immunohistochemistry. The relationship between KGF and/or MMP-9 expression and clinicopathological characteristics was analyzed. RESULTS KGF expression and MMP-9 expression in gastric cancer tissue were observed in 62 cases (38.5%) and 97 cases (60.2%), respectively. MMP-9 was significantly associated with depth of invasion, lymph node metastasis and TNM stage. The prognosis of MMP-9-positive patients was significantly poorer than that of MMP-9-negative patients (p = 0.009). KGF expression was positively correlated with MMP-9 expression in gastric cancer, and the prognosis of patients with both KGF- and MMP-9-positive tumors was significantly worse than that of patients with negative tumors for either factor (p = 0.045). Expression of MMP-9 was revealed to be an independent prognostic factor (p = 0.026). CONCLUSIONS Coexpression of KGF and MMP-9 in gastric cancer could be a useful prognostic factor, and MMP-9 might also serve as a novel target for both prognostic prediction and therapeutics.
Collapse
|
12
|
Hong L, Han Y, Liu J, Fan D. Keratinocyte growth factor receptor: a therapeutic target in solid cancer. Expert Opin Ther Targets 2015. [PMID: 26200212 DOI: 10.1517/14728222.2015.1062474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The treatment effects of advanced solid cancer are unsatisfactory, and novel therapeutic approaches are much needed. Keratinocyte growth factor receptor (KGFR) is a receptor tyrosine kinase that is primarily localized on epithelial cells. KGFR may play important roles in epithelial cell proliferation and differentiation, epithelial wound repair, embryonic development, immunity, tumor formation and development. AREAS COVERED This review summarizes the expression, function and mechanism of KGFR in solid cancer, and analyzes its value for the cancer therapy. Furthermore, this study discusses the limitations of KGFR-based therapy, and envisages future developments in the clinical applications of KGFR. EXPERT OPINION KGFR may function as an ideal therapeutic target for solid cancer. Continued basic investigation of KGFR-mediated pathways will push insight into the novel strategies of target therapy. More in vivo studies and clinical trials should be performed to promote the translational bridging of the latest research into clinical application.
Collapse
Affiliation(s)
- Liu Hong
- a 1 Fourth Military Medical University, Xijing Hospital of Digestive Diseases , Xi'an 710032, Shaanxi Province, China +86 29 84771531 ; +86 29 82539041 ;
| | - Yu Han
- b 2 Fourth Military Medical University, Xijing Hospital, Department of Otolaryngology , Xi'an 710032, Shaanxi Province, China
| | - Jinqiang Liu
- a 1 Fourth Military Medical University, Xijing Hospital of Digestive Diseases , Xi'an 710032, Shaanxi Province, China +86 29 84771531 ; +86 29 82539041 ;
| | - Daiming Fan
- a 1 Fourth Military Medical University, Xijing Hospital of Digestive Diseases , Xi'an 710032, Shaanxi Province, China +86 29 84771531 ; +86 29 82539041 ;
| |
Collapse
|
13
|
GLP-1R agonists promote normal and neoplastic intestinal growth through mechanisms requiring Fgf7. Cell Metab 2015; 21:379-91. [PMID: 25738454 DOI: 10.1016/j.cmet.2015.02.005] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 12/09/2014] [Accepted: 02/06/2015] [Indexed: 12/28/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) secreted from enteroendocrine L cells promotes nutrient disposal via the incretin effect. However, the majority of L cells are localized to the distal gut, suggesting additional biological roles for GLP-1. Here, we demonstrate that GLP-1 receptor (GLP-1R) signaling controls mucosal expansion of the small bowel (SB) and colon. These actions did not require the epidermal growth factor (EGF) or intestinal epithelial insulin-like growth factor (IGF1) receptors but were absent in Glp1r(-/-) mice. Polyp number and size were increased in SB of exendin-4-treated Apc(Min/+) mice, whereas polyp number was reduced in SB and colon of Glp1r(-/-):Apc(Min/+) mice. Exendin-4 increased fibroblast growth factor 7 (Fgf7) expression in colonic polyps of Apc(Min/+) mice and failed to increase intestinal growth in mice lacking Fgf7. Exogenous exendin-4 and Fgf7 regulated an overlapping set of genes important for intestinal growth. Thus, gain and loss of GLP-1R signaling regulates gut growth and intestinal tumorigenesis.
Collapse
|
14
|
Fan EW, Li CC, Wu WJ, Huang CN, Li WM, Ke HL, Yeh HC, Wu TF, Liang PI, Ma LJ, Li CF. FGF7 Over Expression is an Independent Prognosticator in Patients with Urothelial Carcinoma of the Upper Urinary Tract and Bladder. J Urol 2015; 194:223-9. [PMID: 25623741 DOI: 10.1016/j.juro.2015.01.073] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2015] [Indexed: 12/14/2022]
Abstract
PURPOSE Urothelial carcinoma of the bladder and upper tract is the most common tumor type in the urinary tract but its molecular pathogenesis and survival determinants remain obscure. By data mining a published transcriptomic database of bladder urothelial carcinoma (GSE31684) we identified FGF7 as the most significant gene up-regulated during urothelial carcinoma progression. We then used our well characterized urothelial carcinoma cohort to analyze FGF7 transcript and protein expression, and its clinicopathological significance. MATERIALS AND METHODS We performed real-time reverse transcriptase-polymerase chain reaction assay to determine the FGF7 transcript level in 30 fresh samples each of upper tract and bladder urothelial carcinoma. Immunohistochemistry evaluated by H-score was used to determine FGF7 protein expression in 340 upper tract and 295 bladder urothelial carcinomas. Transcript and protein expression were correlated with clinicopathological features. We further evaluated the prognostic significance of FGF7 protein expression for disease specific and metastasis-free survival. RESULTS An increased FGF7 transcript level was associated with higher pT stage in upper tract and bladder urothelial carcinoma (p = 0.003 and <0.001, respectively). In the upper tract and bladder carcinoma groups FGF7 protein over expression was also significantly associated with advanced pT status (each p <0.001), lymph node metastasis (p = 0.002 and <0.001), high histological grade (p = 0.019 and <0.001), vascular invasion (each p <0.001), perineural invasion (p = 0.002 and 0.021) and frequent mitoses (p = 0.002 and 0.042, respectively). FGF7 over expression predicted dismal disease specific and metastasis-free survival on univariate and multivariate analysis. CONCLUSIONS Our study shows that FGF7 over expression is associated with advanced clinical features in patients with upper tract and bladder urothelial carcinoma, justifying its potential prognostic value for urothelial carcinoma.
Collapse
Affiliation(s)
- Eric W Fan
- Division of Urology, Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan, Republic of China
| | - Ching-Chia Li
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China; Department of Urology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China; Department of Urology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan, Republic of China
| | - Wen-Jeng Wu
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China; Department of Urology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China; Department of Urology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
| | - Chun-Nung Huang
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China; Department of Urology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
| | - Wei-Ming Li
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China; Department of Urology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China; Pingtung Hospital, Ministry of Health and Welfare, Executive Yuan, Pingtung, Taiwan, Republic of China
| | - Hung-Lung Ke
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China; Department of Urology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
| | - Hsin-Chih Yeh
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China; Department of Urology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China; Department of Urology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan, Republic of China
| | - Ting-Feng Wu
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan, Republic of China
| | - Peir-In Liang
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
| | - Li-Jung Ma
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan, Republic of China.
| | - Chien-Feng Li
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan, Republic of China; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan, Republic of China; Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China.
| |
Collapse
|
15
|
Cheng M, Ho S, Yoo JH, Tran DHY, Bakirtzi K, Su B, Tran DHN, Kubota Y, Ichikawa R, Koon HW. Cathelicidin suppresses colon cancer development by inhibition of cancer associated fibroblasts. Clin Exp Gastroenterol 2014; 8:13-29. [PMID: 25565877 PMCID: PMC4274046 DOI: 10.2147/ceg.s70906] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Cathelicidin (LL-37 in humans and mCRAMP in mice) represents a family of endogenous antimicrobial and anti-inflammatory peptides. Cancer-associated fibroblasts can promote the proliferation of colon cancer cells and growth of colon cancer tumors. METHODS We examined the role of cathelicidin in the development of colon cancer, using subcutaneous human HT-29 colon-cancer-cell-derived tumor model in nude mice and azoxymethane- and dextran sulfate-mediated colon cancer model in C57BL/6 mice. We also determined the indirect antitumoral mechanism of cathelicidin via the inhibition of epithelial-mesenchymal transition (EMT) of colon cancer cells and fibroblast-supported colon cancer cell proliferation. RESULTS Intravenous administration of cathelicidin expressing adeno-associated virus significantly reduced the size of tumors, tumor-derived collagen expression, and tumor-derived fibroblast expression in HT-29-derived subcutaneous tumors in nude mice. Enema administration of the mouse cathelicidin peptide significantly reduced the size and number of colonic tumors in azoxymethane- and dextran sulfate-treated mice without inducing apoptosis in tumors and the adjacent normal colonic tissues. Cathelicidin inhibited the collagen expression and vimentin-positive fibroblast expression in colonic tumors. Cathelicidin did not directly affect HT-29 cell viability, but did significantly reduce tumor growth factor-β1-induced EMT of colon cancer cells. Media conditioned by the human colonic CCD-18Co fibroblasts promoted human colon cancer HT-29 cell proliferation. Cathelicidin pretreatment inhibited colon cancer cell proliferation mediated by media conditioned by human colonic CCD-18Co fibroblasts. Cathelicidin disrupted tubulin distribution in colonic fibroblasts. Disruption of tubulin in fibroblasts reduced fibroblast-supported colon cancer cell proliferation. CONCLUSION Cathelicidin effectively inhibits colon cancer development by interfering with EMT and fibroblast-supported colon cancer cell proliferation.
Collapse
Affiliation(s)
- Michelle Cheng
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Samantha Ho
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jun Hwan Yoo
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA ; Digestive Disease Center, CHA University Bundang Medical Center, Seongnam, Republic of Korea
| | - Deanna Hoang-Yen Tran
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Kyriaki Bakirtzi
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Bowei Su
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Diana Hoang-Ngoc Tran
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Yuzu Kubota
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ryan Ichikawa
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Hon Wai Koon
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
16
|
What could Nintedanib (BIBF 1120), a triple inhibitor of VEGFR, PDGFR, and FGFR, add to the current treatment options for patients with metastatic colorectal cancer? Crit Rev Oncol Hematol 2014; 92:83-106. [PMID: 24924525 DOI: 10.1016/j.critrevonc.2014.05.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 05/02/2014] [Indexed: 12/29/2022] Open
Abstract
Increasing knowledge of the pro-angiogenic processes involved in the progression of metastatic colorectal cancer (mCRC) has resulted in the clinical development of several anti-angiogenic agents, with bevacizumab currently being the only approved agent for mCRC. Nintedanib (BIBF 1120) has been shown to block the vascular endothelial growth factor receptor (VEGFR), the platelet-derived growth factor receptor (PDGFR), and the fibroblast growth factor receptor (FGFR). By targeting FGFR signaling, nintedanib may overcome resistance to previous anti-VEGF treatments, and may represent a better approach in patients with high basal levels of circulating FGFs. In this article, the angiogenic mechanisms implicated in mCRC are reviewed (focusing on the signaling pathways activated by VEGFR, PDGFR, and FGFR), along with the clinical data for nintedanib in the context of other anti-angiogenic tyrosine kinase inhibitors under clinical development for mCRC. Biomarkers that could predict response to nintedanib are also discussed.
Collapse
|
17
|
Fibroblast growth factor receptor 2: expression, roles, and potential as a novel molecular target for colorectal cancer. PATHOLOGY RESEARCH INTERNATIONAL 2012; 2012:574768. [PMID: 22701813 PMCID: PMC3373204 DOI: 10.1155/2012/574768] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 03/28/2012] [Indexed: 12/19/2022]
Abstract
The fibroblast growth factor receptor (FGFR) family consists of four members, named FGFR1, 2, 3, and 4. All 4 FGFRs and their ligands, fibroblast growth factors (FGFs), are expressed in colorectal cancer (CRC). Recent studies have shown that FGFR2 plays important roles in cancer progression; therefore, it is of great interest as a novel target for cancers. Expression of FGFR2 regulates migration, invasion, and growth in CRC. Expression of the FGFR2 isoform FGFR2 IIIb was associated with well-differentiated histological types, and its specific ligand, FGF7, enhanced angiogenesis and adhesion to type-IV collagen via FGFR2 IIIb in CRC. FGFR2 IIIc is detected in CRC, but its roles have not been well elucidated. Interactions between FGFR2 IIIb and IIIc and FGFs may play important roles in CRC via autocrine and/or paracrine signaling. Several kinds of molecular-targeting agents against FGFR2 have been developed; however, it is not clear how a cancer treatment can most effectively inhibit FGFR2 IIIb or FGFR2 IIIc, or both isoforms. The aim of this paper is to summarize the roles of FGFR2 and its isoforms in CRC and clarify whether they are potent therapeutic targets for CRC.
Collapse
|
18
|
Inactivation of Rb in stromal fibroblasts promotes epithelial cell invasion. EMBO J 2012; 31:3092-103. [PMID: 22643222 PMCID: PMC3400012 DOI: 10.1038/emboj.2012.153] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Accepted: 04/27/2012] [Indexed: 12/18/2022] Open
Abstract
Stromal-derived growth factors are required for normal epithelial growth but are also implicated in tumour progression. We have observed inactivation of the retinoblastoma protein (Rb), through phosphorylation, in cancer-associated fibroblasts in oro-pharyngeal cancer specimens. Rb is well known for its cell-autonomous effects on cancer initiation and progression; however, cell non-autonomous functions of Rb are not well described. We have identified a cell non-autonomous role of Rb, using three-dimensional cultures, where depletion of Rb in stromal fibroblasts enhances invasive potential of transformed epithelia. In part, this is mediated by upregulation of keratinocyte growth factor (KGF), which is produced by the depleted fibroblasts. KGF drives invasion of epithelial cells through induction of MMP1 expression in an AKT- and Ets2-dependent manner. Our data identify that stromal fibroblasts can alter the invasive behaviour of the epithelium, and we show that altered expression of KGF can mediate these functions.
Collapse
|
19
|
Ceccarelli S, Romano F, Angeloni A, Marchese C. Potential dual role of KGF/KGFR as a target option in novel therapeutic strategies for the treatment of cancers and mucosal damages. Expert Opin Ther Targets 2012; 16:377-93. [PMID: 22443411 DOI: 10.1517/14728222.2012.671813] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Keratinocyte growth factor (KGF) and its receptor KGFR play a pivotal role in regulating cell proliferation, migration, differentiation and survival, in response to injury and tissue repair. Altered expression of this pathway in cancer opened the way to the development of targeted therapy to achieve KGFR inhibition. Nevertheless, KGF administration has been demonstrated to ameliorate oral mucositis resulting from chemoradiotherapy, besides protecting epithelial cells against radiation-induced damage. AREAS COVERED This review focuses on the potential therapeutic interest of KGF/KGFR in two different areas: selective inhibition of KGFR signaling for the treatment of cancers characterized by upregulation of this pathway and administration of KGF to protect epithelial cells from induced damage. The review presents an overview of therapeutic strategies in both directions. EXPERT OPINION KGF/KGFR signaling can contribute to enhancing the malignant potential of epithelial cells and to promoting tumorigenesis. On the other hand, the therapeutic use of KGF in cancer patients provides epithelial protection, reducing chemotherapy side effects. FGFRs have become attractive antitumor targets and various inhibitors have been used to contrast tumor cell growth. The identification of KGFR-specific molecules might represent a promising therapeutic strategy that could increase the window of available agents and treatment methods.
Collapse
Affiliation(s)
- Simona Ceccarelli
- Sapienza University of Rome, Department of Experimental Medicine, Roma, Italy
| | | | | | | |
Collapse
|
20
|
Cho K, Matsuda Y, Ueda J, Uchida E, Naito Z, Ishiwata T. Keratinocyte growth factor induces matrix metalloproteinase-9 expression and correlates with venous invasion in pancreatic cancer. Int J Oncol 2011; 40:1040-8. [PMID: 22159401 PMCID: PMC3584520 DOI: 10.3892/ijo.2011.1280] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 10/24/2011] [Indexed: 12/15/2022] Open
Abstract
Keratinocyte growth factor (KGF), also known as fibroblast growth factor-7, and KGF receptor (KGFR) play important roles in the growth of epithelial cells and are overexpressed in a variety of malignant epithelial tumors, including pancreatic ductal adenocarcinoma (PDAC). We previously reported that co-expression of KGF and KGFR in PDAC is associated with venous invasion, enhanced vascular endothelial growth factor A expression and poor prognosis. Matrix metalloproteinase-9 (MMP-9) is known to participate in the degradation of type IV collagen, which is a primary component of extracellular matrices in the vascular basement membrane. In the present study, we examined the expression and roles of KGF, KGFR and MMP-9 in human PDAC cell lines and tissues. Quantitative real-time polymerase chain reaction analysis demonstrated the expression of MMP-9 mRNA in all eight PDAC cell lines. KGF, KGFR and MMP-9 were, respectively, expressed in 27 (43%), 23 (37%) and 35 (56%) of 63 patients. Each expression of KGF, KGFR or MMP-9 correlated positively with venous invasion. Furthermore, expression of KGF or MMP-9 correlated positively with liver metastasis. KGF-positive cases exhibited shorter survival than KGF-negative cases, while KGFR and MMP-9 expression were unrelated to prognosis. Administration of recombinant human KGF increased MMP-9 expression in PDAC cells, while transient transfection with short hairpin RNAs targeting KGF transcripts reduced MMP-9 expression in PDAC cells. Moreover, recombinant human KGF significantly enhanced migration and invasion of PDAC cells. These findings suggest that KGF and KGFR promote venous invasion via MMP-9 in PDAC, and closely correlate with liver metastasis. The KGF/KGFR pathway may be a critical therapeutic target for PDAC metastasis.
Collapse
Affiliation(s)
- Kazumitsu Cho
- Departments of Pathology and Integrative Oncological Pathology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | | | | | | | | | | |
Collapse
|
21
|
Matsuda Y, Ishiwata T, Yamahatsu K, Kawahara K, Hagio M, Peng WX, Yamamoto T, Nakazawa N, Seya T, Ohaki Y, Naito Z. Overexpressed fibroblast growth factor receptor 2 in the invasive front of colorectal cancer: a potential therapeutic target in colorectal cancer. Cancer Lett 2011; 309:209-19. [PMID: 21745712 DOI: 10.1016/j.canlet.2011.06.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 06/06/2011] [Accepted: 06/12/2011] [Indexed: 12/31/2022]
Abstract
Fibroblast growth factor receptor 2 (FGFR2) is considered a novel therapeutic target for various cancer. We used a silencing strategy to clarify the effect of reduced FGFR2 expression in human colorectal cancer (CRC) cells. The invasive front of cancer cells exhibited stronger FGFR2 expression than the surface area of the cancers. FGFR2 shRNA-transfected LoVo cells inhibited cell migration, invasion and tumor growth in vitro and in vivo. Thus, FGFR2 plays important roles in CRC progression in association with tumor cell migration, invasion and growth, and FGFR2 might be a novel therapeutic target for CRC.
Collapse
Affiliation(s)
- Yoko Matsuda
- Departments of Pathology and Integrative Oncological Pathology, Nippon Medical School, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Hille A, Grüger S, Christiansen H, Wolff HA, Volkmer B, Lehmann J, Dörr W, Rave-Fränk M. Effect of tumour-cell-derived or recombinant keratinocyte growth factor (KGF) on proliferation and radioresponse of human epithelial tumour cells (HNSCC) and normal keratinocytes in vitro. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2010; 49:261-270. [PMID: 20213138 PMCID: PMC2855434 DOI: 10.1007/s00411-010-0271-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Accepted: 02/05/2010] [Indexed: 05/28/2023]
Abstract
Purpose of this work was to test the effect of tumour-cell-derived keratinocyte growth factor (KGF) or recombinant KGF (palifermin) on cell proliferation and radiation response of human HNSCC cells and normal keratinocytes in vitro. Four tumour cell cultures derived from head and neck squamous cell carcinomas, primary keratinocytes, and immortalized keratinocytes were analysed. Fibroblasts, the natural source of KGF protein, served as controls. KGF expression was observed in primary and immortalized keratinocytes, fibroblasts, and in tumour cells, while significant KGF receptor expression was only found in keratinocytes. Recombinant KGF as well as tumour-cell-derived KGF caused a significant growth stimulation and radioprotection in keratinocytes, which was abolished by a neutralizing anti-KGF antibody. This indicates that tumour-cell-derived KGF is biologically active. In the tumour cell lines, no significant growth stimulation was induced by recombinant KGF, and the neutralizing antibody did not influence tumour cell growth or radiation response. Our results indicate that the normal, paracrine KGF regulatory mechanisms, which are based on KGF receptor expression, are lost in malignant cells, with the consequence of irresponsiveness of the tumour cells to exogenous KGF. In face of the amelioration of the radiation response of normal epithelia, demonstrated in various clinical and various preclinical animal studies, recombinant KGF represents a candidate for the selective protection of normal epithelia during radio(chemo) therapy of squamous cell carcinoma.
Collapse
Affiliation(s)
- Andrea Hille
- Department of Radiotherapy and Radiooncology, University Medicine Göttingen, Göttingen, Germany
| | - Susanne Grüger
- Department of Radiotherapy and Radiooncology, University Medicine Göttingen, Göttingen, Germany
| | - Hans Christiansen
- Department of Radiotherapy and Radiooncology, University Medicine Göttingen, Göttingen, Germany
| | - Hendrik A. Wolff
- Department of Radiotherapy and Radiooncology, University Medicine Göttingen, Göttingen, Germany
| | - Beate Volkmer
- Dermatology Centre Elbeklinikum Buxtehude, Buxtehude, Germany
| | - Jörg Lehmann
- Department of Radiation Oncology, University of California Davis School of Medicine, Sacramento, CA USA
| | - Wolfgang Dörr
- Department of Radiotherapy and Radiation Oncology, Medical Faculty Carl Gustav Carus, University of Technology, Dresden, Germany
| | - Margret Rave-Fränk
- Department of Radiotherapy and Radiooncology, University Medicine Göttingen, Göttingen, Germany
| |
Collapse
|
23
|
Drozdowski L, Thomson ABR. Intestinal hormones and growth factors: effects on the small intestine. World J Gastroenterol 2009; 15:385-406. [PMID: 19152442 PMCID: PMC2653359 DOI: 10.3748/wjg.15.385] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
There are various hormones and growth factors which may modify the intestinal absorption of nutrients, and which might thereby be useful in a therapeutic setting, such as in persons with short bowel syndrome. In part I, we focus first on insulin-like growth factors, epidermal and transferring growth factors, thyroid hormones and glucocorticosteroids. Part II will detail the effects of glucagon-like peptide (GLP)-2 on intestinal absorption and adaptation, and the potential for an additive effect of GLP2 plus steroids.
Collapse
|
24
|
Abstract
Prostanoid pathway in hair follicle gained closer attention since trichogenic side-effects on hair growth has been observed concomitantly with prostaglandin F(2alpha) receptor (FP) agonist treatment of intraocular pressure. We thus investigated prostanoid receptor distribution in anagen hair follicle and different cell types from hair and skin. Using RT-PCR, Western blot and immunohistochemistry (IHC), we found that all receptors were present in hair follicle. This data shed new light on an underestimated complex network involved in hair growth control. Indeed most of these receptors showed a wide spectrum of expression in cultured cells and the whole hair follicle. Using IHC, we observed that expression of prostaglandin E(2) receptors (EP(2), EP(3), EP(4)), prostaglandin D(2) receptor (DP(2)), prostanoid thromboxane A(2) receptor (TP) and to a lesser extent EP(1) involved several hair follicle compartments. On the opposite, Prostaglandin I(2) receptor (IP) and DP(1) were more specifically expressed in hair cuticle layer and outer root sheath (ORS) basal layer, respectively. FP expression was essentially restricted to ORS companion layer and dermal papilla (DP). Although extracting a clear functional significance from this intricate network remains open challenge, FP labelling, i.e. could explain the biological effect of PGF(2alpha) on hair regrowth, by directly modulating DP function.
Collapse
|
25
|
Kudo M, Ishiwata T, Nakazawa N, Kawahara K, Fujii T, Teduka K, Naito Z. Keratinocyte growth factor-transfection-stimulated adhesion of colorectal cancer cells to extracellular matrices. Exp Mol Pathol 2007; 83:443-52. [PMID: 17706640 DOI: 10.1016/j.yexmp.2007.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Revised: 07/06/2007] [Accepted: 07/06/2007] [Indexed: 11/18/2022]
Abstract
The keratinocyte growth factor (KGF) regulates cell growth and behavior in an autocrine or paracrine manner. In colorectal cancer tissues, KGF is expressed in tumor cells and adjacent stromal fibroblasts. We have constructed a KGF-gene-transfected cell line (HCT15-KGF) from a colorectal cancer cell line, HCT-15, that expresses the KGF receptor, and studied the effects of KGF on cell behavior, particularly growth and adhesion to extracellular matrices (ECMs). The amount of KGF secreted from HCT15-KGF was significantly higher than that from a mock-transfected cell line (HCT15-MOCK). The modes of growth of these cell lines were similar. The degree of adhesion of HCT15-KGF to ECMs, including type-IV collagen and fibronectin was higher than that of HCT15-MOCK. The expressions of integrins in both cell lines were not significantly different. However, extracellular-regulated kinase-1 and -2 (ERK1/2) phosphorylation and focal adhesion kinase (FAK) expression that regulate the adhesive functions of integrin families were enhanced in HCT15-KGF. U0126, an inhibitor of the ERK upstream regulator MEK, attenuated the adhesion and spreading of HCT15-KGF cells to type-IV collagen. These results indicate that KGF enhances the adhesion of colorectal cancer cells to type-IV collagen through ERK and FAK signaling pathways.
Collapse
Affiliation(s)
- Mitsuhiro Kudo
- Department of Integrative Oncological Pathology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan.
| | | | | | | | | | | | | |
Collapse
|
26
|
Thomas RS, Pluta L, Yang L, Halsey TA. Application of genomic biomarkers to predict increased lung tumor incidence in 2-year rodent cancer bioassays. Toxicol Sci 2007; 97:55-64. [PMID: 17311802 DOI: 10.1093/toxsci/kfm023] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Rodent cancer bioassays are part of a legacy of safety testing that has not changed significantly over the past 30 years. The bioassays are expensive, time consuming, and use hundreds of animals. Fewer than 1500 chemicals have been tested in a rodent cancer bioassay compared to the thousands of environmental and industrial chemicals that remain untested for carcinogenic activity. In this study, we used existing data generated by the National Toxicology Program (NTP) to identify gene expression biomarkers that can predict results from a rodent cancer bioassay. A set of 13 diverse chemicals was selected from those tested by the NTP. Seven chemicals were positive for increased lung tumor incidence in female B6C3F1 mice and six were negative. Female mice were exposed subchronically to each of the 13 chemicals, and microarray analysis was performed on the lung. Statistical classification analysis using the gene expression profiles identified a set of eight probe sets corresponding to six genes whose expression correctly predicted the increase in lung tumor incidence with 93.9% accuracy. The sensitivity and specificity were 95.2 and 91.8%, respectively. Among the six genes in the predictive signature, most were enzymes involved in endogenous and xenobiotic metabolism, and one gene was a growth factor receptor involved in lung development. The results demonstrate that increases in chemically induced lung tumor incidence in female mice can be predicted using gene biomarkers from a subchronic exposure and may form the basis of a more efficient and economical approach for evaluating the carcinogenic activity of chemicals.
Collapse
Affiliation(s)
- Russell S Thomas
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709-2137, USA.
| | | | | | | |
Collapse
|
27
|
Shaoul R, Eliahu L, Sher I, Hamlet Y, Miselevich I, Goldshmidt O, Ron D. Elevated expression of FGF7 protein is common in human gastric diseases. Biochem Biophys Res Commun 2006; 350:825-33. [PMID: 17049492 DOI: 10.1016/j.bbrc.2006.08.198] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Accepted: 08/30/2006] [Indexed: 10/24/2022]
Abstract
Growth alterations within the gastric mucosa during chronic gastric inflammation are key steps in gastric cancer development. FGF7, a specific mitogen for epithelial cells, is implicated in epithelial tissue repair and cancer. We investigated FGF7 expression in normal human stomach, and in 35 cases from various gastric pathologies including 23 gastritis and 8 adenocarcinoma cases. Modest FGF7 protein levels were detected in the normal mucosal gland epithelium and in stromal fibroblasts. FGF7 protein levels, however, were markedly increased in the mucosal epithelium of all gastric inflammation cases. A similar elevated expression was also observed in gastric adenocarcinoma. Upregulation of FGF7 protein was associated with a modest increase in FGF7 mRNA expression. Interestingly, high levels of FGF7 anti-sense (AS) RNA were observed in the gastric pathologies, at the same sites where FGF7 protein was upregulated. Altogether, these findings suggest a role for FGF7 in maintaining gastric mucosa integrity, and that FGF7 protein levels are regulated mainly by posttranscriptional mechanisms. The elevated FGF7 protein levels in gastric inflammation and gastric cancer, together with the known oncogenic potential of FGF7, implicate excessive FGF7 signaling in gastric tumorigenesis, and point to FGF7 as an attractive target for gastric cancer prevention and treatment.
Collapse
Affiliation(s)
- Ron Shaoul
- Department of Pediatrics, Bnai Zion Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | | | | | | | | | | | | |
Collapse
|
28
|
Finch PW, Rubin JS. Keratinocyte growth factor expression and activity in cancer: implications for use in patients with solid tumors. J Natl Cancer Inst 2006; 98:812-24. [PMID: 16788155 DOI: 10.1093/jnci/djj228] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Keratinocyte growth factor (KGF) is a locally acting epithelial mitogen that is produced by cells of mesenchymal origin and has an important role in protecting and repairing epithelial tissues. Use of recombinant human KGF (palifermin) in patients with hematologic malignancies reduces the incidence and duration of severe oral mucositis experienced after intensive chemoradiotherapy. These results suggest that KGF may be useful in the treatment of patients with other kinds of tumors, including those of epithelial origin. However, its application in this context raises issues that were not pertinent to its use in hematologic cancer because epithelial tumor cells, unlike blood cells, often express the KGF receptor (FGFR2b). Thus, it is important to examine whether KGF could promote the growth of epithelial tumors or protect such tumor cells from the effects of chemotherapy agents. Analyses of KGF and FGFR2b expression in tumor specimens and of KGF activity on transformed cells in vitro and in vivo do not indicate a definitive role for KGF in tumorigenesis. On the contrary, restoring FGFR2b expression to certain malignant cells can induce cell differentiation or apoptosis. However, other observations suggest that, in specific situations, KGF may contribute to epithelial tumorigenesis. Thus, further studies are warranted to examine the nature and extent of KGF involvement in these settings. In addition, clinical trials in patients with solid tumors are underway to assess the potential benefits of using KGF to protect normal tissue from the adverse effects of chemoradiotherapy and its possible impact on clinical outcome.
Collapse
Affiliation(s)
- Paul W Finch
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, Building 37, Room 2042, 37 Convent Drive, MSC 4256, Bethesda, MD 20892-4256, USA
| | | |
Collapse
|
29
|
Abstract
There are three major subfamilies of mitogen-activated protein kinases (MAPK): the extracellular-signal-regulated kinases (ERK MAPK); the c-jun N-terminal kinase or stress-activated protein kinases (JNK or SAPK); and MAPK14. The ERK MAPK pathway is one of the most important for cell proliferation. The MAPK pathways are located downstream of many growth-factor receptors, including that for epidermal growth factor. Overexpression and activation of this receptor are commonly detected in colorectal cancer, and several lines of evidence indicate that overexpression and activation of ERK MAPK play an important part in progression of this cancer. ERK MAPK could be a molecular target for treatment of the disorder. This review focuses on the ERK MAPK signal-transduction pathway, the consequences of its dysregulation in colorectal cancer, and its potential as an approach to cancer treatment. Future challenges for the assessment of these targeted agents in the clinic are also presented.
Collapse
|
30
|
Finch PW, Rubin JS. Keratinocyte growth factor/fibroblast growth factor 7, a homeostatic factor with therapeutic potential for epithelial protection and repair. Adv Cancer Res 2004; 91:69-136. [PMID: 15327889 DOI: 10.1016/s0065-230x(04)91003-2] [Citation(s) in RCA: 170] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Keratinocyte growth factor (KGF) is a paracrine-acting, epithelial mitogen produced by cells of mesenchymal origin. It is a member of the fibroblast growth factor (FGF) family, and acts exclusively through a subset of FGF receptor isoforms (FGFR2b) expressed predominantly by epithelial cells. The upregulation of KGF after epithelial injury suggested it had an important role in tissue repair. This hypothesis was reinforced by evidence that intestinal damage was worse and healing impaired in KGF null mice. Preclinical data from several animal models demonstrated that recombinant human KGF could enhance the regenerative capacity of epithelial tissues and protect them from a variety of toxic exposures. These beneficial effects are attributed to multiple mechanisms that collectively act to strengthen the integrity of the epithelial barrier, and include the stimulation of cell proliferation, migration, differentiation, survival, DNA repair, and induction of enzymes involved in the detoxification of reactive oxygen species. KGF is currently being evaluated in clinical trials to test its ability to ameliorate severe oral mucositis (OM) that results from cancer chemoradiotherapy. In a phase 3 trial involving patients who were treated with myeloablative chemoradiotherapy before autologous peripheral blood progenitor cell transplantation for hematologic malignancies, KGF significantly reduced both the incidence and duration of severe OM. Similar investigations are underway in patients being treated for solid tumors. On the basis of its success in ameliorating chemoradiotherapy-induced OM in humans and tissue damage in a variety of animal models, additional clinical applications of KGF are worthy of investigation.
Collapse
Affiliation(s)
- Paul W Finch
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
31
|
Yeh BK, Igarashi M, Eliseenkova AV, Plotnikov AN, Sher I, Ron D, Aaronson SA, Mohammadi M. Structural basis by which alternative splicing confers specificity in fibroblast growth factor receptors. Proc Natl Acad Sci U S A 2003; 100:2266-71. [PMID: 12591959 PMCID: PMC151329 DOI: 10.1073/pnas.0436500100] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2002] [Indexed: 12/28/2022] Open
Abstract
Binding specificity between fibroblast growth factors (FGFs) and their receptors (FGFRs) is essential for mammalian development and is regulated primarily by two alternatively spliced exons, IIIb ("b") and IIIc ("c"), that encode the second half of Ig-like domain 3 (D3) of FGFRs. FGF7 and FGF10 activate only the b isoform of FGFR2 (FGFR2b). Here, we report the crystal structure of the ligand-binding portion of FGFR2b bound to FGF10. Unique contacts between divergent regions in FGF10 and two b-specific loops in D3 reveal the structural basis by which alternative splicing provides FGF10-FGFR2b specificity. Structure-based mutagenesis of FGF10 confirms the importance of the observed contacts for FGF10 biological activity. Interestingly, FGF10 binding induces a previously unobserved rotation of receptor Ig domain 2 (D2) to introduce specific contacts with FGF10. Hence, both D2 and D3 of FGFR2b contribute to the exceptional specificity between FGF10 and FGFR2b. We propose that ligand-induced conformational change in FGFRs may also play an important role in determining specificity for other FGF-FGFR complexes.
Collapse
MESH Headings
- Alternative Splicing
- Amino Acid Sequence
- Crystallography, X-Ray
- DNA/metabolism
- Exons
- Fibroblast Growth Factor 10
- Fibroblast Growth Factors/metabolism
- Humans
- Hydrogen Bonding
- Ligands
- Models, Molecular
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Protein Binding
- Protein Conformation
- Protein Isoforms
- Protein Structure, Secondary
- Protein Structure, Tertiary
- Receptor, Fibroblast Growth Factor, Type 2
- Receptors, Fibroblast Growth Factor/chemistry
- Receptors, Fibroblast Growth Factor/metabolism
- Selenomethionine/chemistry
- Sequence Homology, Amino Acid
- Structure-Activity Relationship
- Tissue Distribution
Collapse
Affiliation(s)
- Brian K Yeh
- Department of Pharmacology, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Ishiwata T, Naito Z, Lu YP, Kawahara K, Fujii T, Kawamoto Y, Teduka K, Sugisaki Y. Differential distribution of fibroblast growth factor (FGF)-7 and FGF-10 in L-arginine-induced acute pancreatitis. Exp Mol Pathol 2002; 73:181-90. [PMID: 12565793 DOI: 10.1006/exmp.2002.2472] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The regenerative process of the pancreas after acute pancreatitis (AP) is characterized by acinar and ductal cell proliferation with synthesis and transient deposition of extracellular matrices. Various growth factors were reported to be highly expressed in AP, but their regulation has not yet been clarified. Fibroblast growth factor (FGF)-7, also known as keratinocyte growth factor (KGF), and FGF-10 are members of the FGF family and show high structural homology and similar biological characteristics. Both are mainly synthesized by mesenchymal cells and stimulate epithelial cells via KGF receptor (KGFR) which is a splice variant of FGFR-2. In the present study, we attempted to immunohistochemically determine the localization of FGF-7 and FGF-10 in pancreatic tissues of an L-arginine-induced rat pancreatitis model. Furthermore, highly specific KGFR antibodies were prepared and used for Western blot analysis and immunohistochemistry. In the normal pancreas, FGF-7 was localized in alpha cells of islets, but FGF-10 was not detected. KGFR was also localized in islet cells, ductal cells, and centroacinar cells in the normal pancreas. In the pancreatic tissues of rats with L-arginine-induced pancreatitis, FGF-7 was localized in alpha cells, whereas FGF-10 was expressed in vascular smooth muscle cells (VSMCs). KGFR was not expressed in centroacinar cells and its level decreased after L-arginine treatment. However, KGFR was detected instead in some acinar cells and VSMCs in addition to islet cells. These findings suggest that FGF-7 and FGF-10 contribute to the regeneration and differentiation of acinar cells and angiogenesis in AP through KGFR.
Collapse
|
33
|
Lu YP, Ishiwata T, Kawahara K, Watanabe M, Naito Z, Moriyama Y, Sugisaki Y, Asano G. Expression of lumican in human colorectal cancer cells. Pathol Int 2002; 52:519-26. [PMID: 12366811 DOI: 10.1046/j.1440-1827.2002.01384.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Lumican is a member of a small leucine-rich proteoglycan family and its overexpression in human breast cancer tissues is reported to influence the growth of cancer cells. In the present study, we aimed to clarify the expression of lumican mRNA and its protein in human colorectal cancer cell lines and their localization in normal and cancerous colorectal tissues. Reverse transcription-polymerase chain reaction and western blot analysis revealed lumican mRNA and its protein expression in COLO 205, DLD-1, HCT-15, SW 480 and WiDr colorectal cancer cell lines. The lumican in colorectal cancer cells had non-sulfated or poorly sulfated polylactosamine side chains. Based on its immunoreactivity, the lumican protein was found to be localized in fibroblasts and stromal tissues of normal colorectal tissues, but not in colorectal epithelial cells. In colorectal cancer tissues, the lumican was strongly localized in cancer cells in eight of 12 cancer cases. The lumican protein was also localized in epithelial cells with mild reactive dysplasia and fibroblasts adjacent to cancer cells. Lumican mRNA was expressed in cancer cells and adjacent fibroblasts, and epithelial cells. These findings may indicate that the lumican protein synthesized by cancer cells, fibroblasts and epithelial cells with mild reactive dysplasia found adjacent to cancer cells may affect the growth of human colorectal cancer cells.
Collapse
Affiliation(s)
- Yue Ping Lu
- Department of Pathology, Nippon Medical School, Sendagi, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Mishima K, Inoue K, Hayashi Y. Overexpression of extracellular-signal regulated kinases on oral squamous cell carcinoma. Oral Oncol 2002; 38:468-74. [PMID: 12110341 DOI: 10.1016/s1368-8375(01)00104-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Mitogen-activated protein kinases (MAPKs) consist of major three subfamilies, extracellular-signal regulated kinases (ERK MAPKs), the c-Jun N-terminal kinases/stress activated protein kinases (JNK MAPKs/SAP MAPKs), and p38 MAPKs. ERK MAPKs pathway is one of the most important pathways for cell proliferation. ERK MAPKs are located at downstream of a lot of growth factors (epidermal growth factor (EGF), nerve growth factor (NGF), platelet-derived growth factor (PDGF), etc.), the overexpressions and activation of which are frequently detected on a number of cancers including oral squamous cell carcinoma (OSCC). These data indicate that overexpression and activation of ERK MAPKs play an important role in cancer progression. On the contrary, JNK MAPKs are possible regulators of cell death induced by chemotherapeutic agents. p38 MAPKs are activated by pro-inflammatory cytokines and inflammatory drugs (non-steroidal anti-inflammatory drug), which are known to suppress cancer growth. These findings imply that each MAPKs can be molecular targets for cancer therapy in OSCC and its investigation is very important things in OSCC.
Collapse
Affiliation(s)
- Kenji Mishima
- Department of Pathology, Tokushima University of Dentistry, 3-18-15, Kuramoto-cho, Japan.
| | | | | |
Collapse
|
35
|
Yeakley JM, Fan JB, Doucet D, Luo L, Wickham E, Ye Z, Chee MS, Fu XD. Profiling alternative splicing on fiber-optic arrays. Nat Biotechnol 2002; 20:353-8. [PMID: 11923840 DOI: 10.1038/nbt0402-353] [Citation(s) in RCA: 159] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The human transcriptome is marked by extensive alternative mRNA splicing and the expression of many closely related genes, which may be difficult to distinguish using standard microarray techniques. Here we describe a sensitive and specific assay for parallel analysis of mRNA isoforms on a fiber-optic microarray platform. The method permits analysis of mRNA transcripts without prior RNA purification or cDNA synthesis. Using an endogenously expressed viral transcript as a model, we demonstrated that the assay readily detects mRNA isoforms from as little as 10-100 pg of total cellular RNA or directly from a few cells. Multiplexed analysis of human cancer cell lines revealed differences in mRNA splicing and suggested a potential autocrine mechanism in the development of choriocarcinomas. Our approach may be useful in the large-scale analysis of the role of alternative splicing in development and disease.
Collapse
Affiliation(s)
- Joanne M Yeakley
- Department of Cellular and Molecular Medicine, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Onda M, Ishiwata T, Kawahara K, Wang R, Naito Z, Sugisaki Y. Expression of lumican in thickened intima and smooth muscle cells in human coronary atherosclerosis. Exp Mol Pathol 2002; 72:142-9. [PMID: 11890723 DOI: 10.1006/exmp.2002.2425] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Lumican is a member of a small leucine-rich proteoglycan family. Members of this family play an important role in cell migration and proliferation during embryonic development, tissue repair, and tumor growth. Lumican is reported to be overexpressed during the wound-healing process in the cornea and ischemic and reperfused heart. Recently, we found that lumican mRNA and its protein are expressed in cultured vascular smooth muscle cells (VSMCs) from the rat aorta. However, the expression and role of lumican in human atherosclerotic tissues are not clearly elucidated. In the present study, we aimed to clarify whether lumican is expressed in VSMCs and its localization in human coronary atherosclerotic tissues. The lumican protein and its mRNA were expressed in a small number of VSMCs in the media of normal coronary artery, but the lumican protein was not localized in the medial stroma. In contrast, the lumican protein and its mRNA were expressed in most of VSMCs that migrated into the thickened intima, but not in infiltrating foamy macrophages. The lumican protein was prominently localized in the thickened intimal stroma. The lumican protein and its mRNA were also expressed in VSMCs in the inner layer of the media and its protein was localized in medial stromal tissues. These findings indicate that the lumican protein is mainly synthesized by intimal and medial VSMCs in coronary atherosclerosis and that lumican contributes to collagen fibrillogenesis of coronary atherosclerosis.
Collapse
Affiliation(s)
- Munehiko Onda
- Department of Pathology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan.
| | | | | | | | | | | |
Collapse
|
37
|
Ping Lu Y, Ishiwata T, Asano G. Lumican expression in alpha cells of islets in pancreas and pancreatic cancer cells. J Pathol 2002; 196:324-30. [PMID: 11857496 DOI: 10.1002/path.1037] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Lumican is a member of a small leucine-rich proteoglycan family, members of which play an important role in cell migration and proliferation during embryonic development, tissue repair, and tumour growth. Lumican is reported to be overexpressed during the wound healing process in the cornea and in human breast cancer tissues, but its expression and localization in normal pancreas and pancreatic cancer tissues are not known. The present study aimed to clarify the expression of lumican protein and its mRNA in human pancreatic cancer cell lines and their localization in normal and cancerous human pancreatic tissues. Reverse transcription-polymerase chain reaction (RT-PCR) and western blot analysis revealed lumican mRNA and its protein expression in PK-8 and MIA-PaCa-2 human pancreatic cancer cells. The tumour lumican had non- or poorly sulphated polylactosamine side-chains rather than highly sulphated keratan sulphate chains. Immunoreactivity of the lumican protein was localized in alpha cells of islets and stromal tissues of a normal pancreas. In pancreatic cancer tissues, the lumican protein was strongly localized in cancer cells, and in acinar and islet cells in chronic pancreatitis-like lesions adjacent to the cancer cells. It was also localized in fibroblasts and collagen fibres close to cancer cells. Lumican mRNA was expressed in cancer cells, in acinar and islet cells in chronic pancreatitis-like lesions, and in stromal fibroblasts in the pancreatic cancer tissues. This is the first report that lumican is synthesized in endocrine and cancer cells. Lumican protein may play a role in the maintenance of islet cell function in normal pancreas and the lumican protein synthesized by cancer cells, acinar and islet cells, and stromal fibroblasts may play a role in the growth of human pancreatic cancer cells.
Collapse
Affiliation(s)
- Yue Ping Lu
- Department of Pathology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | | | | |
Collapse
|
38
|
Wirth K, Mertelsmann R. Cytoprotective function of keratinocyte growth factor in tumour therapy-induced tissue damage. Br J Haematol 2002; 116:505-10. [PMID: 11849205 DOI: 10.1046/j.0007-1048.2001.03290.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
39
|
Qin H, Ishiwata T, Asano G. Effects of the extracellular matrix on lumican expression in rat aortic smooth muscle cells in vitro. J Pathol 2001; 195:604-8. [PMID: 11745697 DOI: 10.1002/path.994] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Lumican is a small leucine-rich proteoglycan (SLRP), which contributes to cell migration, proliferation, tissue hydration, and collagen fibrillogenesis. Whether lumican is localized in rat aortic smooth muscle cells (SMCs) and what its relationships might be to other extracellular matrix components have not yet been elucidated. In this study, using reverse transcription-polymerase chain reaction (RT-PCR), competitive RT-PCR, and western blot, lumican messenger ribonucleic acid (mRNA) was expressed in cultured rat aortic SMCs. SMCs cultured in serum-free medium showed four bands at 68, 62, 50, and 37 kD. The 68 and 62 kD bands corresponded to proteoglycan, the 50 kD band to glycoprotein, and the 37 kD band to the core protein form of lumican. The relationships of lumican to fibronectin and laminin were also investigated. The lumican mRNA level in SMCs cultured on fibronectin was highest at day 1, but it increased at day 3 in SMCs cultured on laminin. On the fibronectin or laminin-coated plates, SMCs expressed only the 68 and 62 kD bands, corresponding to proteoglycan. Pretreatment with anti-beta1 integrin receptor antibody revealed a decrease in the proteoglycan forms of lumican protein and an additional two bands at 50 and 37 kD, indicating glycoprotein and the core protein of lumican. These results show that lumican was synthesized in cultured rat aortic SMCs as proteoglycan, glycoprotein, and core protein. The extracellular matrix (ECM) affected lumican protein production and restricted the lumican protein form to proteoglycan via the beta1 integrin receptor in SMCs.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/cytology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Blotting, Western
- Cell Culture Techniques
- Chondroitin Sulfate Proteoglycans/biosynthesis
- Chondroitin Sulfate Proteoglycans/genetics
- Extracellular Matrix Proteins/pharmacology
- Fibronectins/pharmacology
- Gene Expression Regulation/drug effects
- Integrin beta1/physiology
- Keratan Sulfate/biosynthesis
- Keratan Sulfate/genetics
- Laminin/pharmacology
- Lumican
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- RNA, Messenger/genetics
- Rats
Collapse
Affiliation(s)
- H Qin
- Department of Pathology, Nippon Medical School, Tokyo 113-8602, Japan
| | | | | |
Collapse
|
40
|
Matsuike A, Ishiwata T, Watanabe M, Asano G. Expression of fibroblast growth factor (FGF)-10 in human colorectal adenocarcinoma cells. J NIPPON MED SCH 2001; 68:397-404. [PMID: 11598623 DOI: 10.1272/jnms.68.397] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Fibroblast growth factor (FGF) -10 (keratinocyte growth factor 2, KGF 2) is a new member of the FGF family that is mainly synthesized by mesenchymal cells and acts predominantly on epithelial cells in a paracrine manner. Its actions are dependent on its binding to the iiib isoform of the cell-surface FGF receptor 2 (FGFR2 iiib). FGF-10 is known to play an important role in fetal limb and lung development, skin wound healing and prostatic epithelial cell growth. In the present study, the expression of FGF-10 and FGFR2 iiib in five cultured human colorectal adenocarcinoma cell lines (COLO 205, DLD-1, HCT-15, SW 480 and WiDr) and the localization of FGF-10 messenger RNA (mRNA) and its protein in human colorectal cancer tissues from 10 patients were determined. All five colorectal cancer cell lines expressed FGF-10 mRNA and its protein. FGFR2 iiib mRNAs were expressed in these cells and the recombinant FGF-10 (1 ng/ml) increased the growth rate of COLO 205 cells. To determine the localization of FGF-10 protein and its mRNA in normal and cancerous human colorectal tissues, immunohistochemistry and in situ hybridization were performed. In normal colorectal tissues, FGF-10 and its mRNA were not detected. In contrast, moderate immunoreactivity was present in cancer cells in 5 of 10 colorectal cancer cases and mild immunoreactivity was recognized in adjacent fibroblasts. By using in situ hybridization, FGF-10 mRNA was observed in colorectal cancer cells and fibroblasts adjacent to cancer cells. These findings indicate that FGF-10 and its receptor, FGFR2 iiib expression in colorectal adenocarcinoma cells and FGF-10 may contribute to the growth of cells of this type.
Collapse
Affiliation(s)
- A Matsuike
- Department of Pathology, Nippon Medical School, Tokyo, Japan
| | | | | | | |
Collapse
|
41
|
Kurban G, Ishiwata T, Lu YP, Fujii T, Kawahara K, Naito Z, Yamada N, Asano G. [Expression and intracytoplasmic signal transduction pathway of fibroblast growth factor (FGF)-10 in human cervical cancer cell lines]. J NIPPON MED SCH 2001; 68:253-8. [PMID: 11404772 DOI: 10.1272/jnms.68.253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Fibroblast growth factor (FGF) -10 is a new member of the FGF family initially reported in Japan. It is mainly synthesized by mesenchymal cells and acts on epithelial cells in a paracrine manner. FGF-10 actions are dependent on their binding to the iiib form of FGF receptor 2 (FGFR2) iiib, also known as keratinocyte growth factor receptor (KGFR). FGF-10 has high amino acid homology to keratinocyte growth factor (KGF) and plays an important role in fetal limb and lung development and skin wound healing. In the present study, the expression of FGF-10 and FGFR2 iiib messenger RNA (mRNA) in two different human uterine cervical cancer cell lines (CaSki and ME-180) were examined. Both CaSki and ME-180 cells expressed FGFR2 iiib mRNA, while only CaSki cells expressed FGF-10 mRNA and protein. Recombinant FGF-10 (1 ng/ml) increased the growth rate of ME-180 cells and also enhanced mitogen-activated protein kinase (MAPK) phosphorylation of the cells. These data indicate that FGF-10 may directly promote the growth of squamous cell cancer in the uterine cervix via the MAPK pathway.
Collapse
Affiliation(s)
- G Kurban
- Department of Pathology, Nippon Medical School, Japan
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Baba H, Ishiwata T, Takashi E, Xu G, Asano G. Expression and Localization of Lumican in the Ischemic and Reperfused Rat Heart. ACTA ACUST UNITED AC 2001; 65:445-50. [PMID: 11348051 DOI: 10.1253/jcj.65.445] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Lumican belongs to the small leucine-rich proteoglycan family and has an important role in the regulation of corneal and dermal collagen fiber assembly. Recently, lumican mRNA was found in the heart and its high expression was reported during wound healing of the cornea. In the present study, the expression and role of lumican in fibrosis of an ischemic and reperfused rat heart were examined. The expression level of lumican mRNA increased in the ischemic and reperfused rat heart and was highest on the fourth week. Lumican protein existed in the forms of core protein and proteoglycan in the control rat heart. The amount of lumican in the form of proteoglycan increased, and that as the form glycoprotein was newly detected in the ischemic and reperfused rat heart on the fourth week. In the control heart, lumican was weakly expressed in the collagen fibers of the perivascular area, but it was expressed strongly in many capillary endothelial cells in the ischemic lesion on day 1. After 3 days, lumican was localized in collagen fibers and in the fibroblasts of fibrotic lesions. A few myocardial cells close to the ischemic lesion expressed lumican mRNA. Lumican is considered to play an important role in the fibrillogenesis of the ischemic and reperfused rat heart.
Collapse
Affiliation(s)
- H Baba
- Department of Pathology, Nippon Medical School, Tokyo, Japan
| | | | | | | | | |
Collapse
|