1
|
Yu S, Kalinin AA, Paraskevopoulou MD, Maruggi M, Cheng J, Tang J, Icke I, Luo Y, Wei Q, Scheibe D, Hunter J, Singh S, Nguyen D, Carpenter AE, Horman SR. Integrating inflammatory biomarker analysis and artificial-intelligence-enabled image-based profiling to identify drug targets for intestinal fibrosis. Cell Chem Biol 2023; 30:1169-1182.e8. [PMID: 37437569 PMCID: PMC10529501 DOI: 10.1016/j.chembiol.2023.06.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 03/11/2023] [Accepted: 06/13/2023] [Indexed: 07/14/2023]
Abstract
Intestinal fibrosis, often caused by inflammatory bowel disease, can lead to intestinal stenosis and obstruction, but there are no approved treatments. Drug discovery has been hindered by the lack of screenable cellular phenotypes. To address this, we used a scalable image-based morphology assay called Cell Painting, augmented with machine learning algorithms, to identify small molecules that could reverse the activated fibrotic phenotype of intestinal myofibroblasts. We then conducted a high-throughput small molecule chemogenomics screen of approximately 5,000 compounds with known targets or mechanisms, which have achieved clinical stage or approval by the FDA. By integrating morphological analyses and AI using pathologically relevant cells and disease-relevant stimuli, we identified several compounds and target classes that are potentially able to treat intestinal fibrosis. This phenotypic screening platform offers significant improvements over conventional methods for identifying a wide range of drug targets.
Collapse
Affiliation(s)
- Shan Yu
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA.
| | | | | | - Marco Maruggi
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Jie Cheng
- Takeda Development Center Americas, Inc., Cambridge, MA 02142, USA
| | - Jie Tang
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Ilknur Icke
- Takeda Development Center Americas, Inc., Cambridge, MA 02142, USA
| | - Yi Luo
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Qun Wei
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Dan Scheibe
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Joel Hunter
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Shantanu Singh
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Deborah Nguyen
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | | | - Shane R Horman
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA.
| |
Collapse
|
2
|
Kim SW, Choi JW, Kim JM, Yoon HY, Bae K, Yoon KA, Kim JH. Case report: Toceranib as adjuvant chemotherapy in a dog with incompletely resected combined hepatocellular-cholangiocarcinoma. Front Vet Sci 2023; 9:963390. [PMID: 36686162 PMCID: PMC9845258 DOI: 10.3389/fvets.2022.963390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/17/2022] [Indexed: 01/05/2023] Open
Abstract
An 11-year-old intact female mixed breed dog was presented with abdominal distention and elevated hepatic enzyme levels. Computed tomography revealed a multicystic hepatic mass at the left medial lobe adjacent to the diaphragm and caudal vena cava. The mass was surgically removed with partial hepatectomy, but it could not be removed completely because of adhesion to the diaphragm. The tissue was submitted for histopathologic evaluation, and the patient was diagnosed with stage IIIA combined hepatocellular-cholangiocarcinoma (cHCC-CC). Considering the residual tumor tissue from incomplete surgical excision, adjuvant chemotherapy was recommended. Tumor tissue obtained from the patient was assessed using an anticancer drug response prediction test, and the results showed that toceranib phosphate was the most effective chemotherapeutic agent for this patient. Toceranib was initiated (3.1 mg/kg, PO, q48 h), and routine adverse effect assessment, including systemic blood pressure measurement, complete blood count, serum biochemical evaluations, and urinalysis were performed at two-week intervals for the first 2 months and every 2 months thereafter. Radiography and ultrasonography were conducted at one-month intervals for the first two months and then every 2 months subsequently. Concurrent hyperadrenocorticism was managed with trilostane (1 to 5 mg/kg, PO, q12h). The patient showed no critical adverse effects of chemotherapy, obvious recurrence, or metastasis. The response to toceranib was assessed as a partial response, and the patient is still alive over 23 months after tumor excision. This is the first case report describing chemotherapy for a dog with cHCC-CC.
Collapse
Affiliation(s)
- Sang-Won Kim
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul, South Korea,Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Ju-Won Choi
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul, South Korea
| | - Jeon-Mo Kim
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Hun-Young Yoon
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul, South Korea,Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Kieun Bae
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul, South Korea,Department of Veterinary Biochemistry, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Kyong-Ah Yoon
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul, South Korea,Department of Veterinary Biochemistry, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Jung-Hyun Kim
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul, South Korea,Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, South Korea,*Correspondence: Jung-Hyun Kim
| |
Collapse
|
3
|
Malik IA, Rajput M, Werner R, Fey D, Salehzadeh N, von Arnim CAF, Wilting J. Differential in vitro effects of targeted therapeutics in primary human liver cancer: importance for combined liver cancer. BMC Cancer 2022; 22:1193. [PMCID: PMC9675209 DOI: 10.1186/s12885-022-10247-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/29/2022] [Indexed: 11/21/2022] Open
Abstract
The incidence of primary liver tumors, hepatocellular carcinoma (HCC), intrahepatic cholangiocellular carcinoma (ICC), and combined HCC/ICC (cHCC/CC) is increasing. For ICC, targeted therapy exists only for a small subpopulation of patients, while for HCC, Sorafenib and Lenvatinib are in use. Diagnosis of cHCC/CC is a great challenge and its incidence is underestimated, bearing the risk of unintended non-treatment of ICC. Here, we investigated effects of targeted inhibitors on human ICC cell lines (HUH28, RBE, SSP25), in comparison to extrahepatic (E)CC lines (EGI1, CCC5, TFK1), and HCC/hepatoblastoma cell lines (HEP3B, HUH7, HEPG2). Cells were challenged with: AKT inhibitor MK-2206; multikinase inhibitors Sorafenib, Lenvatinib and Dasatinib; PI3-kinase inhibitors BKM-120, Wortmannin, LY294002, and CAL-101; and mTOR inhibitor Rapamycin. Dosage of the substances was based on the large number of published data of recent years. Proliferation was analyzed daily for four days. All cell lines were highly responsive to MK-2206. Thereby, MK-2206 reduced expression of phospho(p)-AKT in all ICC, ECC, and HCC lines, which mostly corresponded to reduction of p-mTOR, whereas p-ERK1/2 was upregulated in many cases. Lenvatinib showed inhibitory effects on the two HCC cell lines, but not on HEPG2, ICCs and ECCs. Sorafenib inhibited proliferation of all cells, except the ECC line CCC5. However, at reduced dosage, we observed increased cell numbers in some ICC experiments. Dasatinib was highly effective especially in ICC cell lines. Inhibitory effects were observed with all four PI3-kinase inhibitors. However, cell type-specific differences were also evident here. Rapamycin was most effective in the two HCC cell lines. Our studies show that the nine inhibitors differentially target ICC, ECC, and HCC/hepatoblastoma lines. Caution should be taken with Lenvatinib and Sorafenib administration in patients with cHCC/CC as the drugs may have no effects on, or might even stimulate, ICC.
Collapse
Affiliation(s)
- Ihtzaz Ahmed Malik
- grid.411984.10000 0001 0482 5331Department of Geriatrics, University Medical Center Goettingen, Waldweg 33, D-37073 Goettingen, Germany
| | - Mansi Rajput
- grid.411984.10000 0001 0482 5331Department of Geriatrics, University Medical Center Goettingen, Waldweg 33, D-37073 Goettingen, Germany
| | - Rieke Werner
- grid.411984.10000 0001 0482 5331Department of Anatomy and Cell Biology, University Medical Center Goettingen, Kreuzbergring 36, 37075 Goettingen, Germany
| | - Dorothea Fey
- grid.411984.10000 0001 0482 5331Department of Geriatrics, University Medical Center Goettingen, Waldweg 33, D-37073 Goettingen, Germany ,grid.411984.10000 0001 0482 5331Department of Anatomy and Cell Biology, University Medical Center Goettingen, Kreuzbergring 36, 37075 Goettingen, Germany
| | - Niloofar Salehzadeh
- grid.411984.10000 0001 0482 5331Department of Geriatrics, University Medical Center Goettingen, Waldweg 33, D-37073 Goettingen, Germany ,grid.411984.10000 0001 0482 5331Department of Anatomy and Cell Biology, University Medical Center Goettingen, Kreuzbergring 36, 37075 Goettingen, Germany
| | - Christine A. F. von Arnim
- grid.411984.10000 0001 0482 5331Department of Geriatrics, University Medical Center Goettingen, Waldweg 33, D-37073 Goettingen, Germany
| | - Jörg Wilting
- grid.411984.10000 0001 0482 5331Department of Anatomy and Cell Biology, University Medical Center Goettingen, Kreuzbergring 36, 37075 Goettingen, Germany
| |
Collapse
|
4
|
Association between Immunologic Markers and Cirrhosis in Individuals from a Prospective Chronic Hepatitis C Cohort. Cancers (Basel) 2022; 14:cancers14215280. [PMID: 36358697 PMCID: PMC9657502 DOI: 10.3390/cancers14215280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/19/2022] [Accepted: 10/23/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Chronic hepatitis C virus (HCV) infection can affect immune response and inflammatory pathways, leading to severe liver diseases such as cirrhosis and hepatocellular carcinoma (HCC). Methods: In a prospective cohort of chronically HCV-infected individuals, we sampled 68 individuals who developed cirrhosis, 91 controls who did not develop cirrhosis, and 94 individuals who developed HCC. Unconditional odds ratios (ORs) from polytomous logistic regression models and canonical discriminant analyses (CDAs) were used to compare categorical (C) baseline plasma levels for 102 markers in individuals who developed cirrhosis vs. controls and those who developed HCC vs. cirrhosis. Leave-one-out cross validation was used to produce receiver operating characteristic curves to assess predictive ability of markers. Lastly, biological pathways were assessed in association with cirrhotic development compared to controls. Results: After multivariable adjustment, DEFA-1 (OR: C2v.C1 = 7.73; p < 0.0001), ITGAM (OR: C2v.C1 = 4.03; p = 0.0002), SCF (OR: C4v.C1 = 0.19; p-trend = 0.0001), and CCL11 (OR: C4v.C1 = 0.31; p-trend= 0.002) were all associated with development of cirrhosis compared to controls; these markers, together with clinical/demographics variables, improved prediction of cirrhosis from 55.7% (in clinical/demographic-only model) to 74.9% accuracy. A twelve-marker model based on CDA results further increased prediction of cirrhosis to 88.0%. While six biological pathways were found to be associated with cirrhosis, cell adhesion was the only pathway associated with cirrhosis after Bonferroni correction. In contrast to cirrhosis, DEFA-1 and ITGAM levels were inversely associated with HCC risk. Conclusions: Pending validation, these findings highlight the important role of immunological markers in predicting HCV-related cirrhosis even 11 years post-enrollment.
Collapse
|
5
|
Tacconi C, Plein A, Colletto C, Villa E, Denti L, Barone C, Javanmardi Y, Moeendarbary E, Azzoni E, Fantin A, Ruhrberg C. KIT is dispensable for physiological organ vascularisation in the embryo. Angiogenesis 2022; 25:343-353. [PMID: 35416527 PMCID: PMC9249691 DOI: 10.1007/s10456-022-09837-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 03/25/2022] [Indexed: 11/08/2022]
Abstract
Blood vessels form vast networks in all vertebrate organs to sustain tissue growth, repair and homeostatic metabolism, but they also contribute to a range of diseases with neovascularisation. It is, therefore, important to define the molecular mechanisms that underpin blood vessel growth. The receptor tyrosine kinase KIT is required for the normal expansion of hematopoietic progenitors that arise during embryogenesis from hemogenic endothelium in the yolk sac and dorsal aorta. Additionally, KIT has been reported to be expressed in endothelial cells during embryonic brain vascularisation and has been implicated in pathological angiogenesis. However, it is neither known whether KIT expression is widespread in normal organ endothelium nor whether it promotes blood vessel growth in developing organs. Here, we have used single-cell analyses to show that KIT is expressed in endothelial cell subsets of several organs, both in the adult and in the developing embryo. Knockout mouse analyses revealed that KIT is dispensable for vascularisation of growing organs in the midgestation embryo, including the lung, liver and brain. By contrast, vascular changes emerged during late-stage embryogenesis in these organs from KIT-deficient embryos, concurrent with severe erythrocyte deficiency and growth retardation. These findings suggest that KIT is not required for developmental tissue vascularisation in physiological conditions, but that KIT deficiency causes foetal anaemia at late gestation and thereby pathological vascular remodelling.
Collapse
Affiliation(s)
- Carlotta Tacconi
- Department of Biosciences, University of Milan, Via G. Celoria 26, 20133, Milan, Italy
| | - Alice Plein
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Chiara Colletto
- Department of Biosciences, University of Milan, Via G. Celoria 26, 20133, Milan, Italy
| | - Emanuela Villa
- Department of Biosciences, University of Milan, Via G. Celoria 26, 20133, Milan, Italy
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Laura Denti
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Cristiana Barone
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Yousef Javanmardi
- UCL Department of Mechanical Engineering, University College London, London, UK
| | - Emad Moeendarbary
- UCL Department of Mechanical Engineering, University College London, London, UK
| | - Emanuele Azzoni
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Alessandro Fantin
- Department of Biosciences, University of Milan, Via G. Celoria 26, 20133, Milan, Italy.
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| |
Collapse
|
6
|
Maeda A, Goto S, Iwasaki R, Yamada K, Murakami M, Sakai H, Mori T. Outcome of Localized Bile Duct Carcinoma in Six Dogs Treated with Liver Lobectomy. J Am Anim Hosp Assoc 2022; 58:189-193. [PMID: 35793487 DOI: 10.5326/jaaha-ms-7199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2021] [Indexed: 11/11/2022]
Abstract
The prognosis for bile duct carcinoma in dogs is generally believed to be poor. However, only a few studies have evaluated the postoperative outcomes in such cases. The objective of this case series was to describe the postoperative outcomes of localized intrahepatic bile duct carcinoma in dogs. The electronic medical records of 16 dogs with bile duct carcinoma were reviewed, and 6 dogs were included in the study. All cases were diagnosed as bile duct carcinoma using postoperative pathology, and five of them had already been diagnosed using preoperative core biopsy. The tumors in all of the dogs were confirmed as completely resected on histopathological examination. Two dogs received toceranib following the surgery. The median follow-up time was 693 days (range, 420-1386 days), with a median survival time of 894 days (range, 420-1386 days). Local recurrence or distant metastases were detected in two of the six dogs (33%) on 354 and 398 days following surgery, respectively. The median progression-free survival was 492 days (range, 354-1386 days). In conclusion, dogs with localized intrahepatic bile duct carcinoma had a good prognosis following complete surgical resection.
Collapse
Affiliation(s)
- Atsushi Maeda
- From the Laboratory of Veterinary Clinical Oncology, Department of Veterinary Medicine (A.M., M.M., T.M.), Gifu University, Gifu, Japan
| | - Sho Goto
- From the Animal Medical Center (S.G., R.I.), Gifu University, Gifu, Japan
| | - Ryota Iwasaki
- From the Animal Medical Center (S.G., R.I.), Gifu University, Gifu, Japan
| | - Koji Yamada
- From the Awano Animal Hospital, Gifu, Japan (K.Y.)
| | - Mami Murakami
- From the Laboratory of Veterinary Clinical Oncology, Department of Veterinary Medicine (A.M., M.M., T.M.), Gifu University, Gifu, Japan
| | - Hiroki Sakai
- From the Laboratory of Veterinary Pathology, Department of Veterinary Medicine (H.S.), Gifu University, Gifu, Japan.,From the Center for Highly Advanced Integration of Nano and Life Sciences (H.S., T.M.), Gifu University, Gifu, Japan
| | - Takashi Mori
- From the Laboratory of Veterinary Clinical Oncology, Department of Veterinary Medicine (A.M., M.M., T.M.), Gifu University, Gifu, Japan.,From the Center for Highly Advanced Integration of Nano and Life Sciences (H.S., T.M.), Gifu University, Gifu, Japan
| |
Collapse
|
7
|
Jaffar FHF, Osman K, Hui CK, Zulkefli AF, Ibrahim SF. Long-Term Wi-Fi Exposure From Pre-Pubertal to Adult Age on the Spermatogonia Proliferation and Protective Effects of Edible Bird’s Nest Supplementation. Front Physiol 2022; 13:828578. [PMID: 35360230 PMCID: PMC8963498 DOI: 10.3389/fphys.2022.828578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Children are vulnerable to the radiofrequency radiation (RFR) emitted by Wi-Fi devices. Nevertheless, the severity of the Wi-Fi effect on their reproductive development has been sparsely available. Therefore, this study was conducted to evaluate the Wi-Fi exposure on spermatogonia proliferation in the testis. This study also incorporated an approach to attenuate the effect of Wi-Fi by giving concurrent edible bird’s nest (EBN) supplementation. It was predicted that Wi-Fi exposure reduces spermatogonia proliferation while EBN supplementation protects against it. A total of 30 (N = 30) 3-week-old Sprague Dawley weanlings were divided equally into five groups; Control, Control EBN, Wi-Fi, Sham Wi-Fi, and Wi-Fi + EBN. 2.45 GHz Wi-Fi exposure and 250 mg/kg EBN supplementation were conducted for 14 weeks. Findings showed that the Wi-Fi group had decreased in spermatogonia mitosis status. However, the mRNA and protein expression of c-Kit-SCF showed no significant decrease. Instead, the reproductive hormone showed a reduction in FSH and LH serum levels. Of these, LH serum level was decreased significantly in the Wi-Fi group. Otherwise, supplementing the Wi-Fi + EBN group with 250 mg/kg EBN resulted in a significant increase in spermatogonia mitotic status. Even though EBN supplementation improved c-Kit-SCF mRNA and protein expression, the effects were insignificant. The improvement of spermatogonia mitosis appeared to be associated with a significant increase in blood FSH levels following EBN supplementation. In conclusion, the long-term Wi-Fi exposure from pre-pubertal to adult age reduces spermatogonia proliferation in the testis. On the other hand, EBN supplementation protects spermatogonia proliferation against Wi-Fi exposure.
Collapse
Affiliation(s)
| | - Khairul Osman
- Faculty of Health Sciences, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Chua Kien Hui
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Aini Farzana Zulkefli
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Siti Fatimah Ibrahim
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
- *Correspondence: Siti Fatimah Ibrahim,
| |
Collapse
|
8
|
Hämäläinen S, Kareinen L, Sukura A, Kareinen I. Carboxypeptidase A3 expression in canine mast cell tumors and tissue-resident mast cells. Vet Pathol 2021; 59:236-243. [PMID: 34894899 PMCID: PMC8928232 DOI: 10.1177/03009858211062636] [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] [Indexed: 11/16/2022]
Abstract
Mast cell tumors (MCTs) are one of the most common cutaneous malignancies in dogs. Previous studies have reported expression of mast cell–specific proteases chymase and tryptase in canine cutaneous MCTs and in connective tissue and mucosal mast cells. In humans and rodents, mast cells express an additional specific protease, carboxypeptidase A3 (CPA3). In this article, we describe CPA3 immunoreactivity in connective tissue, visceral, mucosal, and neoplastic mast cells in dogs. Positive immunolabeling for CPA3 was observed in nonneoplastic mast cells in 20/20 formalin-fixed paraffin-embedded normal tissues (skin, liver, spleen, intestine), and in 63/63 MCTs irrespective of their histological grade. CPA3 protein expression was comparable to that of c-kit in both the nonneoplastic and neoplastic mast cells. Three distinct labeling patterns (membranous, diffuse, and focal cytoplasmic) were observed for CPA3 in MCTs. The focal cytoplasmic labeling pattern was associated with high-grade MCTs staged with the Kiupel 2-tier grading criteria. We propose CPA3 as a novel immunohistochemical marker for canine mast cells in health and disease.
Collapse
|
9
|
Aimaitijiang A, Tabu K, Wang W, Nobuhisa I, Taga T. Glioma cells remotely promote erythropoiesis as a self-expanding strategy of cancer stem cells. Genes Cells 2021; 27:25-42. [PMID: 34837452 DOI: 10.1111/gtc.12908] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 11/18/2021] [Indexed: 11/27/2022]
Abstract
Cancer stem cells are a promising target for cancer eradication due to their responsibility for therapy-resistance and cancer recurrence. Previously, we have demonstrated that glioma stem cells (GSCs) recruit and induce the differentiation of bone marrow (BM) monocytes into tumor-infiltrating macrophages, which phagocytose hemorrhaged erythrocytes and store GSC-beneficial iron in mouse xenografts, suggesting a self-expanding strategy of GSCs that exploits host hematopoiesis of myeloid cells. However, it remains unclear whether a self-advantageous effect of GSCs also occurs on erythroid cells during glioma development. Here, we found that, in the primary cultures of mouse fetal liver proerythroblasts (proEs), conditioned media prepared from glioma cells including patient-derived glioblastoma (GBM) cells significantly facilitated the differentiation of proEs into erythroblasts. Importantly, in-vivo erythroid analysis in intracranially GSC-transplanted mice showed an enhanced erythropoiesis in the BM. In addition, the sphere forming ability of patient-derived GBM cells was significantly suppressed by hypoxia treatment and iron chelation, suggesting higher demands of GSCs for oxygen and iron, which may be supplied by GSCs- and their progeny-induced erythrocyte production. Our findings provide a new insight into survival and expanding strategies of GSCs that systemically exploit host erythropoiesis.
Collapse
Affiliation(s)
- Alapati Aimaitijiang
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kouichi Tabu
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Wenqian Wang
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Ikuo Nobuhisa
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tetsuya Taga
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
10
|
Wang W, Shui L, Liu Y, Zheng M. C-Kit, a Double-Edged Sword in Liver Regeneration and Diseases. Front Genet 2021; 12:598855. [PMID: 33603771 PMCID: PMC7884772 DOI: 10.3389/fgene.2021.598855] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/08/2021] [Indexed: 12/24/2022] Open
Abstract
Previous studies have reported an important role of c-kit in embryogenesis and adulthood. Activation of the SCF/KIT signal transduction pathway is customarily linked to cell proliferation, migration and survival thus influence hematopoiesis, pigmentation, and spermatogenesis. The role of c-kit in the liver is controversial, it is however argued that it is a double-edged sword in liver regeneration and diseases. First, liver c-kit+ cells, including oval cells, bile epithelial cells, and part of hepatocytes, participate in liver tissue repair by regenerating target cells according to the type of liver injury. At the same time, c-kit+ mast cells, act as immature progenitors in circulation, playing a critical role in liver fibrosis. Furthermore, c-kit is also a proto-oncogene. Notably, c-kit overexpression regulates gastrointestinal stromal tumors. Various studies have explored on c-kit and hepatocellular carcinoma, nevertheless, the intricate roles of c-kit in the liver are largely understudied. Herein, we extensively summarize previous studies geared toward providing hints for future clinical and basic research.
Collapse
Affiliation(s)
- Weina Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Liyan Shui
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yanning Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Min Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Lee PC, Hsieh YC, Huo TI, Yang UC, Lin CH, Li CP, Huang YH, Hou MC, Lin HC, Lee KC. Active Vitamin D 3 Treatment Attenuated Bacterial Translocation via Improving Intestinal Barriers in Cirrhotic Rats. Mol Nutr Food Res 2021; 65:e2000937. [PMID: 33258263 DOI: 10.1002/mnfr.202000937] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/23/2020] [Indexed: 12/19/2022]
Abstract
SCOPE Pathological bacterial translocation from the disrupted intestinal barrier leads to substantial complications and mortality in liver cirrhosis. Vitamin D is reported as beneficial to gut barriers in some animal models. However, its effect on cirrhotic bacterial translocation is unknown. The authors aim to investigate the effects of calcitriol on bacterial translocation in cirrhotic rats. METHODS AND RESULTS Cirrhotic rats are administrated with a 2-week course of active vitamin D3 (calcitriol, 0.1 μg kg-1 per day) or vehicle by oral gavage after thioacetamide (TAA) injection for 16 weeks. Bacterial translocation, gut permeability, gut microbiota, and associated mechanisms are investigated. Calcitriol treatment significantly attenuates bacterial translocation and reduces intestinal permeability in TAA-induced cirrhotic rats. It upregulates the expressions of occludin in the small intestine and claudin-1 in the colon of cirrhotic rats directly independent of intrahepatic status. Even when a short period of calcitriol treatment do not reduce intestinal bacterial overgrowth, it induces a remarkable change of bacterial diversities and enrichment of Muribaculaceae, Bacteroidales, Allobaculum, Anaerovorax, and Ruminococcaceae. CONCLUSION Calcitriol treatment attenuates intestinal permeability, reduces bacterial translocation, and enriches potentially beneficial gut microbiota in cirrhotic rats that may enable it as a potential therapeutic agent to prevent cirrhotic complications.
Collapse
Affiliation(s)
- Pei-Chang Lee
- Institute of Pharmacology, National Yang Ming University, 155, Sec. 2, Li-Nong Street, Taipei, 11217, Taiwan
- Faculty of Medicine, National Yang Ming University, Taipei 11221, Taiwan., 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, 201 Shih-Pai Road, Sec. 2, Taipei, 11217, Taiwan
| | - Yun-Chen Hsieh
- Institute of Pharmacology, National Yang Ming University, 155, Sec. 2, Li-Nong Street, Taipei, 11217, Taiwan
- Faculty of Medicine, National Yang Ming University, Taipei 11221, Taiwan., 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, 201 Shih-Pai Road, Sec. 2, Taipei, 11217, Taiwan
| | - Teh-Ia Huo
- Institute of Pharmacology, National Yang Ming University, 155, Sec. 2, Li-Nong Street, Taipei, 11217, Taiwan
- Faculty of Medicine, National Yang Ming University, Taipei 11221, Taiwan., 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, 201 Shih-Pai Road, Sec. 2, Taipei, 11217, Taiwan
| | - Ueng-Cheng Yang
- Institute of Biomedical Informatics, National Yang Ming University, 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan
| | - Chao-Hsiung Lin
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming University, 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan
- Proteomics Research Center, National Yang Ming University, 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan
| | - Chung-Pin Li
- Faculty of Medicine, National Yang Ming University, Taipei 11221, Taiwan., 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, 201 Shih-Pai Road, Sec. 2, Taipei, 11217, Taiwan
| | - Yi-Hsiang Huang
- Faculty of Medicine, National Yang Ming University, Taipei 11221, Taiwan., 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, 201 Shih-Pai Road, Sec. 2, Taipei, 11217, Taiwan
- Institute of Clinical Medicine, National Yang Ming University, 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan
| | - Ming-Chih Hou
- Faculty of Medicine, National Yang Ming University, Taipei 11221, Taiwan., 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, 201 Shih-Pai Road, Sec. 2, Taipei, 11217, Taiwan
| | - Han-Chieh Lin
- Faculty of Medicine, National Yang Ming University, Taipei 11221, Taiwan., 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, 201 Shih-Pai Road, Sec. 2, Taipei, 11217, Taiwan
| | - Kuei-Chuan Lee
- Faculty of Medicine, National Yang Ming University, Taipei 11221, Taiwan., 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, 201 Shih-Pai Road, Sec. 2, Taipei, 11217, Taiwan
| |
Collapse
|
12
|
Brandi G, Tavolari S. In Vitro and In Vivo Model Systems of Cholangiocarcinoma. DIAGNOSIS AND MANAGEMENT OF CHOLANGIOCARCINOMA 2021:471-494. [DOI: 10.1007/978-3-030-70936-5_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
13
|
Fantin A, Tacconi C, Villa E, Ceccacci E, Denti L, Ruhrberg C. KIT Is Required for Fetal Liver Hematopoiesis. Front Cell Dev Biol 2021; 9:648630. [PMID: 34395414 PMCID: PMC8358609 DOI: 10.3389/fcell.2021.648630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 06/23/2021] [Indexed: 01/22/2023] Open
Abstract
In the mouse embryo, endothelial cell (EC) progenitors almost concomitantly give rise to the first blood vessels in the yolk sac and the large vessels of the embryo proper. Although the first blood cells form in the yolk sac before blood vessels have assembled, consecutive waves of hematopoietic progenitors subsequently bud from hemogenic endothelium located within the wall of yolk sac and large intraembryonic vessels in a process termed endothelial-to-hematopoietic transition (endoHT). The receptor tyrosine kinase KIT is required for late embryonic erythropoiesis, but KIT is also expressed in hematopoietic progenitors that arise via endoHT from yolk sac hemogenic endothelium to generate early, transient hematopoietic waves. However, it remains unclear whether KIT has essential roles in early hematopoiesis. Here, we have combined single-cell expression studies with the analysis of knockout mice to show that KIT is dispensable for yolk sac endoHT but required for transient definitive hematopoiesis in the fetal liver.
Collapse
Affiliation(s)
- Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
- Department of Biosciences, University of Milan, Milan, Italy
- *Correspondence: Alessandro Fantin,
| | | | - Emanuela Villa
- Department of Biosciences, University of Milan, Milan, Italy
| | - Elena Ceccacci
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Laura Denti
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
- Christiana Ruhrberg,
| |
Collapse
|
14
|
Fiume D, Lenci I, Milana M, Manzia TM, Massoud R, Tariciotti L, Russo C, Toti L, Baiocchi L. Serum Levels of Granulocyte-Macrophage-colony-stimulating Factor and Stem-cell Factor During Liver Regeneration after Partial Hepatectomy in Humans. Rev Recent Clin Trials 2020; 15:131-136. [PMID: 31971114 DOI: 10.2174/1574887115666200123113623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/01/2020] [Accepted: 01/03/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Multiple biological functions have been recognized regarding Granulocyte Macrophage-Colony Stimulating Factor (GM-CSF) and Stem Cell Factor (SCF). AIM To evaluate the serum changes of GM-CSF and SCF in patients undergoing surgical resection for liver tumor, in the regenerative phase after surgery in order to identify the possible relationship with the patient, tumor or surgical variables. METHODS Thirty-two consecutive patients (50% male, median age 66), undergoing hepatic resection of liver neoplasm, were evaluated. The liver tumor was Hepatocellular Carcinoma (HCC) in 44% of cases. Other tumors were cholangiocarcinoma and metastasis. Serum levels of GM-CSF and SCF were assessed at baseline and 2 days, 7 days and 4 weeks after surgery. Personal and clinical patient data were also recorded. The statistical analysis was carried out using t-test for unpaired data or ANOVA (repeated measure) for continuous variables and Fisher test for discrete variables. RESULTS GM-CSF levels remained constant after surgery and were compared to baseline values. SCF levels, on the other hand, increased during the time, after surgery. The evaluation of SCF levels (fold increase) according to surgical, patient and tumor variables evidenced some differences. At day 7 and week 4, SCF levels were statistically increased: i) in patients undergoing a large resection in comparison with others (p<0.05); ii) in patients non-cirrhotic in comparison with cirrhotic ones (p=0.02) and finally; iii) in patients with non-HCC tumor in comparison with HCC ones (p=0.02). CONCLUSION During liver regeneration in humans, SCF serum levels are increased allowing to hypothesize a possible role of this chemokine during tissue growth and remodeling.
Collapse
Affiliation(s)
- Diego Fiume
- Department of Experimental Medicine Tor Vergata Policlinic, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy
| | - Ilaria Lenci
- Hepatology Unit, Tor Vergata Policlinic, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy
| | - Martina Milana
- Hepatology Unit, Tor Vergata Policlinic, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy
| | - Tommaso M Manzia
- Unit of Transplant Surgery, Tor Vergata Policlinic, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy
| | - Renato Massoud
- Department of Experimental Medicine Tor Vergata Policlinic, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy
| | - Laura Tariciotti
- Unit of Transplant Surgery, Tor Vergata Policlinic, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy
| | - Carmelo Russo
- Department of Experimental Medicine Tor Vergata Policlinic, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy
| | - Luca Toti
- Hepatology Unit, Tor Vergata Policlinic, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy
| | - Leonardo Baiocchi
- Hepatology Unit, Tor Vergata Policlinic, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy
| |
Collapse
|
15
|
Meadows V, Kennedy L, Hargrove L, Demieville J, Meng F, Virani S, Reinhart E, Kyritsi K, Invernizzi P, Yang Z, Wu N, Liangpunsakul S, Alpini G, Francis H. Downregulation of hepatic stem cell factor by Vivo-Morpholino treatment inhibits mast cell migration and decreases biliary damage/senescence and liver fibrosis in Mdr2 -/- mice. Biochim Biophys Acta Mol Basis Dis 2019; 1865:165557. [PMID: 31521820 PMCID: PMC6878979 DOI: 10.1016/j.bbadis.2019.165557] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/06/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022]
Abstract
Primary sclerosing cholangitis (PSC) is characterized by increased mast cell (MC) infiltration, biliary damage and hepatic fibrosis. Cholangiocytes secrete stem cell factor (SCF), which is a chemoattractant for c-kit expressed on MCs. We aimed to determine if blocking SCF inhibits MC migration, biliary damage and hepatic fibrosis. METHODS FVB/NJ and Mdr2-/- mice were treated with Mismatch or SCF Vivo-Morpholinos. We measured (i) SCF expression and secretion; (ii) hepatic damage; (iii) MC migration/activation and histamine signaling; (iv) ductular reaction and biliary senescence; and (v) hepatic fibrosis. In human PSC patients, SCF expression and secretion were measured. In vitro, cholangiocytes were evaluated for SCF expression and secretion. Biliary proliferation/senescence was measured in cholangiocytes pretreated with 0.1% BSA or the SCF inhibitor, ISK03. Cultured HSCs were stimulated with cholangiocyte supernatant and activation measured. MC migration was determined with cholangiocytes pretreated with BSA or ISK03 loaded into the bottom of Boyden chambers and MCs into top chamber. RESULTS Biliary SCF expression and SCF serum levels increase in human PSC. Cholangiocytes, but not hepatocytes, from SCF Mismatch Mdr2-/- mice have increased SCF expression and secretion. Inhibition of SCF in Mdr2-/- mice reduced (i) hepatic damage; (ii) MC migration; (iii) histamine and SCF serum levels; and (iv) ductular reaction/biliary senescence/hepatic fibrosis. In vitro, cholangiocytes express and secrete SCF. Blocking biliary SCF decreased MC migration, biliary proliferation/senescence, and HSC activation. CONCLUSION Cholangiocytes secrete increased levels of SCF inducing MC migration, contributing to biliary damage/hepatic fibrosis. Targeting MC infiltration may be an option to ameliorate PSC progression.
Collapse
Affiliation(s)
- Vik Meadows
- Research, Central Texas Veterans Health Care System, United States of America; Department of Medical Physiology, Texas A&M University College of Medicine, United States of America
| | - Lindsey Kennedy
- Department of Medical Physiology, Texas A&M University College of Medicine, United States of America
| | - Laura Hargrove
- Department of Medical Physiology, Texas A&M University College of Medicine, United States of America
| | - Jennifer Demieville
- Research, Central Texas Veterans Health Care System, United States of America
| | - Fanyin Meng
- Research, Central Texas Veterans Health Care System, United States of America
| | - Shohaib Virani
- Department of Medical Physiology, Texas A&M University College of Medicine, United States of America
| | - Evan Reinhart
- Department of Medical Physiology, Texas A&M University College of Medicine, United States of America
| | - Konstantina Kyritsi
- Department of Medical Physiology, Texas A&M University College of Medicine, United States of America
| | | | - Zhihong Yang
- Richard L. Roudebush VA Medical Center, Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, United States of America
| | - Nan Wu
- Richard L. Roudebush VA Medical Center, Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, United States of America
| | - Suthat Liangpunsakul
- Richard L. Roudebush VA Medical Center, Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, United States of America
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, United States of America; Department of Medical Physiology, Texas A&M University College of Medicine, United States of America
| | - Heather Francis
- Research, Central Texas Veterans Health Care System, United States of America; Department of Medical Physiology, Texas A&M University College of Medicine, United States of America.
| |
Collapse
|
16
|
Models for Understanding Resistance to Chemotherapy in Liver Cancer. Cancers (Basel) 2019; 11:cancers11111677. [PMID: 31671735 PMCID: PMC6896032 DOI: 10.3390/cancers11111677] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 12/19/2022] Open
Abstract
The lack of response to pharmacological treatment constitutes a substantial limitation in the handling of patients with primary liver cancers (PLCs). The existence of active mechanisms of chemoresistance (MOCs) in hepatocellular carcinoma, cholangiocarcinoma, and hepatoblastoma hampers the usefulness of chemotherapy. A better understanding of MOCs is needed to develop strategies able to overcome drug refractoriness in PLCs. With this aim, several experimental models are commonly used. These include in vitro cell-free assays using subcellular systems; studies with primary cell cultures; cancer cell lines or heterologous expression systems; multicellular models, such as spheroids and organoids; and a variety of in vivo models in rodents, such as subcutaneous and orthotopic tumor xenografts or chemically or genetically induced liver carcinogenesis. Novel methods to perform programmed genomic edition and more efficient techniques to isolate circulating microvesicles offer new opportunities for establishing useful experimental tools for understanding the resistance to chemotherapy in PLCs. In the present review, using three criteria for information organization: (1) level of research; (2) type of MOC; and (3) type of PLC, we have summarized the advantages and limitations of the armamentarium available in the field of pharmacological investigation of PLC chemoresistance.
Collapse
|
17
|
Development of A New Mouse Model for Intrahepatic Cholangiocellular Carcinoma: Accelerating Functions of Pecam-1. Cancers (Basel) 2019; 11:cancers11081045. [PMID: 31344919 PMCID: PMC6721446 DOI: 10.3390/cancers11081045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 01/09/2023] Open
Abstract
Due to the lack of suitable in-vivo models, the etiology of intrahepatic cholangiocellular carcinoma (ICC) is poorly understood. We previously showed the involvement of platelet endothelial cell adhesion molecule-1 (Pecam-1/CD31) in acute liver damage. Here, we developed a model of ICC using thioacetamide (TAA) in drinking water of wild-type (WT)-mice and Pecam-1-knock-out (KO)-mice. Gross inspection and microscopy revealed liver-cirrhosis and ICC in both groups after 22 weeks of TAA. The severity of cirrhosis and ICC (Ck-19-positive) was reduced in Pecam-1 KO mice (stage-4 cirrhosis in WT vs. stage-3 in KO mice). Tumor networks (accompanied by neutrophils) were predominantly located in portal areas, with signs of epithelial-to-mesenchymal transition (EMT). In serum, TAA induced an increase in hepatic damage markers, with lower levels in Pecam-1 null mice. With qPCR of liver, elevated expression of Pecam-1 mRNA was noted in WT mice, in addition to Icam-1, EpCam, cytokines, cMyc, and Mmp2. Thereby, levels of EpCAM, cytokines, cMyc, and Mmp2 were significantly lower in Pecam-1 null mice. Lipocalin-2 and Ccl5 were elevated significantly in both WT and Pecam-1 null mice after TAA administration. Also, EMT marker Wnt5a (not Twist-1) was increased in both groups after TAA. We present a highly reproducible mouse model for ICC and show protective effects of Pecam-1 deficiency.
Collapse
|
18
|
Loeuillard E, Fischbach SR, Gores GJ, Ilyas SI. Animal models of cholangiocarcinoma. Biochim Biophys Acta Mol Basis Dis 2019; 1865:982-992. [PMID: 29627364 PMCID: PMC6177316 DOI: 10.1016/j.bbadis.2018.03.026] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/23/2018] [Accepted: 03/29/2018] [Indexed: 12/18/2022]
Abstract
Cholangiocarcinoma (CCA) is an aggressive biliary tract malignancy with a poor overall prognosis. There is a critical need to develop effective targeted therapies for the treatment of this lethal disease. In an effort to address this challenge, preclinical in vivo studies have become paramount in understanding CCA carcinogenesis, progression, and therapy. Various CCA animal models exist including carcinogen-based models in which animals develop CCA after exposure to a carcinogen, genetically engineered mouse models in which genetic changes are induced in mice leading to CCA, murine syngeneic orthotopic models, as well as xenograft tumors derived from xenotransplantation of CCA cells, organoids, and patient-derived tissue. Each type has distinct advantages as well as shortcomings. In the ideal animal model of CCA, the tumor arises from the biliary tract in an immunocompetent host with a species-matched tumor microenvironment. Such a model would also be time-efficient, recapitulate the genetic and histopathological features of human CCA, and predict therapeutic response in humans. Recently developed biliary tract transduction and orthotopic syngeneic transplant mouse models encompass several of these elements. Herein, we review the different animal models of CCA, their advantages and deficiencies, as well as features which mimic human CCA.
Collapse
Affiliation(s)
- Emilien Loeuillard
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Samantha R Fischbach
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Sumera I Ilyas
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
19
|
The stem cell factor (SCF)/c-KIT signalling in testis and prostate cancer. J Cell Commun Signal 2017; 11:297-307. [PMID: 28656507 DOI: 10.1007/s12079-017-0399-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 06/15/2017] [Indexed: 01/17/2023] Open
Abstract
The stem cell factor (SCF) is a cytokine that specifically binds the tyrosine kinase receptor c-KIT. The SCF/c-KIT interaction leads to receptor dimerization, activation of kinase activity and initiation of several signal transduction pathways that control cell proliferation, apoptosis, differentiation and migration in several tissues. The activity of SCF/c-KIT system is linked with the phosphatidylinositol 3-kinase (PI3-K), the Src, the Janus kinase/signal transducers and activators of transcription (JAK/STAT), the phospholipase-C (PLC-γ) and the mitogen-activated protein kinase (MAPK) pathways. Moreover, it has been reported that cancer cases display an overactivation of c-KIT due to the presence of gain-of-function mutations or receptor overexpression, which renders c-KIT a tempting target for cancer treatment. In the case of male cancers the most documented activated pathways are the PI3-K and Src, both enhancing abnormal cell proliferation. It is also known that the Src activity in prostate cancer cases depends on the presence of tr-KIT, the cytoplasmic truncated variant of c-KIT that is specifically expressed in tumour tissues and, thus, a very interesting target for drug development. The present review provides an overview of the signalling pathways activated by SCF/c-KIT and discusses the potential application of c-KIT inhibitors for treatment of testicular and prostatic cancers.
Collapse
|
20
|
Matkowskyj KA, Rao MS, Yang GY. Pathologic Features of Primary and Metastatic Hepatic Malignancies. Cancer Treat Res 2016; 168:257-293. [PMID: 29206377 DOI: 10.1007/978-3-319-34244-3_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In the mammalian liver, 60 % of the cellular components are hepatocytes while the remainder (35 %) includes biliary epithelium, Kupffer cells, endothelial cells, fat storing cells and connective tissue cells. Although neoplasms of hepatocytes are the most common, a significant number of both benign and malignant primary liver neoplasms arising from other cell types can develop, such as tumors of bile duct epithelium (Table 1). In addition, the liver is one of the most susceptible sites for metastatic tumors arising from other organs of the body. Not too long ago, liver tumors were left untreated because the liver was considered a complex and mysterious organ inaccessible to surgery. Advances in imaging procedures and surgical techniques over the past 40 years have revolutionized the approaches to the treatment of benign and malignant liver tumors. Subsegmentectomy, segmentectomy, lobectomy, and transplantation are routinely performed for the treatment of primary and metastatic liver tumors with minimal morbidity and mortality. Since accurate diagnosis remains the key to clinical and surgical management, the emphasis of this chapter is on classification, morphological features and differential diagnosis of malignant neoplasms of the liver.
Collapse
|
21
|
Romano M, De Francesco F, Gringeri E, Giordano A, Ferraro GA, Di Domenico M, Cillo U. Tumor Microenvironment Versus Cancer Stem Cells in Cholangiocarcinoma: Synergistic Effects? J Cell Physiol 2015; 231:768-76. [PMID: 26357947 DOI: 10.1002/jcp.25190] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 09/09/2015] [Indexed: 12/19/2022]
Abstract
Cholangiocarcinoma (CCAs) may be defined as tumors that derived from the biliary tree with the differentiation in the biliary epithelial cells. This tumor is malignant, extremely aggressive with a poor prognosis. It can be treated surgically and its pathogenesis is poorly understood. The tumor microenvironment (TME) is a very important factor in the regulation of tumor angiogenesis, invasion, and metastasis. Besides cancer stem cells (CSCs) can modulate tumor growth, stroma formation, and migratory capability. The initial stage of tumorigenesis is characterized by genetic mutations and epigenetic alterations due to intrinsic factors which lead to the generation of oncogenes thus inducing tumorigenesis. CSCs may result from precancerous stem cells, cell de-differentiation, normal stem cells, or an epithelial-mesenchymal transition (EMT). CSCs have been found in the cancer niche, and EMT may occur early within the tumor microenvironment. Previous studies have demonstrated evidence of cholangiocarcinoma stem cells (CD133, CD24, EpCAM, CD44, and others) and the presence of these markers has been associated with malignant potential. The interaction between TME and cholangiocarcinoma stem cells via signaling mediators may create an environment that accommodates tumor growth, yielding resistance to cytotoxic insults (chemotherarapeutic). While progress has been made in the understanding of the mechanisms, the interactions in the tumorigenic process still remain a major challenge. Our review, addresses recent concepts of TME-CSCs interaction and will emphasize the importance of early detection with the use of novel diagnostic mechanisms such as CCA-CSC biomarkers and the importance of tumor stroma to define new treatments. J. Cell. Physiol. 231: 768-776, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Maurizio Romano
- Department of Surgery, Oncology and Gastroenterology, Hepatobiliary Surgery and Liver Transplantation, Padua University Hospital, Padua, Italy
| | - Francesco De Francesco
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples, Naples, Italy
| | - Enrico Gringeri
- Department of Surgery, Oncology and Gastroenterology, Hepatobiliary Surgery and Liver Transplantation, Padua University Hospital, Padua, Italy
| | - Antonio Giordano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, Pennsylvania
| | - Giuseppe A Ferraro
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples, Naples, Italy
| | - Marina Di Domenico
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Umberto Cillo
- Department of Surgery, Oncology and Gastroenterology, Hepatobiliary Surgery and Liver Transplantation, Padua University Hospital, Padua, Italy
| |
Collapse
|
22
|
Saadi T, Nayshool O, Carmel J, Ariche A, Bramnik Z, Mironi-Harpaz I, Seliktar D, Baruch Y. Cellularized biosynthetic microhydrogel polymers for intravascular liver tissue regeneration therapy. Tissue Eng Part A 2014; 20:2850-9. [PMID: 24797901 PMCID: PMC4229865 DOI: 10.1089/ten.tea.2013.0494] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Accepted: 04/16/2014] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION The liver is the natural microenvironment for hepatocytes transplantation but unfortunately engraftment efficiency is low. Cell-laden microhydrogels made of fibrinogen attached to poly(ethylene glycol) (PEG)-diacrylate side chains, were used as a cell carrier, for intravascular transplantation. This approach may reduce shear stress and immediate immunological pressure after intravascular transplantation and provide biomatrix for environmental support. AIMS In vitro assessment of HuH-7 viability and function after polymerization within PEGylated fibrinogen-hydrogel. In vivo assessment of intraportal transplantation of cell-laden microhydrogels with rat adult parenchymal cells. METHODS (1) In vitro assessment of HuH-7 cell viability and function, after cell-laden hydrogel (hydrogel volume 30 μL) fabrication, by propidium iodide (PI)/fluorescein diacetate (FDA), and MTT assays, albumin concentration and CYP1A activity. (2) Fabrication of cell-laden microhydrogels and their intraportal transplantion. Engraftment efficiency in vivo was evaluated by real-time qPCR of Y chromosome (SRY gene) and histology. RESULTS The viability of cells in hydrogels in culture was comparable to viability of not embedded cells during the first 48 h. However, the viability of cells in hydrogels was reduced after 72 h compared with not embedded cells. Activity of CYP1A in hydrogel was comparable to that of not embedded cells (4.33±1 pmole/μg DNA/4 h vs. 5.13±1 pmole/μg DNA/4 h, respectively). Albumin concentration increased at day 3 in hydrogels to 1.4±0.6 μg/10(4)/24 h and was greater to that of free cells, 0.3±0.1 μg/10(4)/24 h. Cell-laden microhydrogels at a size of 150-150-600 μm (6×10(6) cells/rat) showed better engraftment efficiency at 21 days post-transplantation, compared with isolated cell transplantation (54.6%±5% vs. 1.8%±1.2%, p<0.001). CONCLUSIONS The in vitro HuH-7 viability and function after polymerization in PEGylated fibrinogen hydrogel was comparable to cells without the hydrogel. Long-term survival and engraftment efficiency of intravascular transplanted adult hepatocytes is much better in within cell-laden microhydrogels compared with isolated cells. The overall efficiency of the procedure needs to be improved.
Collapse
Affiliation(s)
- Tarek Saadi
- Liver Unit, Rambam—Health Care Campus, Haifa, Israel
| | - Omri Nayshool
- Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| | - Julie Carmel
- Liver Unit, Rambam—Health Care Campus, Haifa, Israel
| | - Arie Ariche
- Department of Surgery B, Rambam—Health Care Campus, Haifa, Israel
| | - Zakhar Bramnik
- Department of Surgery B, Rambam—Health Care Campus, Haifa, Israel
| | - Iris Mironi-Harpaz
- Department of Biomedical Engineering, Technion- Israel Institute of Technology, Haifa, Israel
| | - Dror Seliktar
- Department of Biomedical Engineering, Technion- Israel Institute of Technology, Haifa, Israel
| | - Yaacov Baruch
- Liver Unit, Rambam—Health Care Campus, Haifa, Israel
- Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
23
|
Cardoso HJ, Figueira MI, Correia S, Vaz CV, Socorro S. The SCF/c-KIT system in the male: Survival strategies in fertility and cancer. Mol Reprod Dev 2014; 81:1064-79. [PMID: 25359157 DOI: 10.1002/mrd.22430] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 09/25/2014] [Indexed: 12/18/2022]
Abstract
Maintaining the delicate balance between cell survival and death is of the utmost importance for the proper development of germ cells and subsequent fertility. On the other hand, the fine regulation of tissue homeostasis by mechanisms that control cell fate is a factor that can prevent carcinogenesis. c-KIT is a type III receptor tyrosine kinase activated by its ligand, stem cell factor (SCF). c-KIT signaling plays a crucial role in cell fate decisions, specifically controlling cell proliferation, differentiation, survival, and apoptosis. Indeed, deregulating the SCF/c-KIT system by attenuation or overactivation of its signaling strength is linked to male infertility and cancer, and rebalancing its activity via c-KIT inhibitors has proven beneficial in treating human tumors that contain gain-of-function mutations or overexpress c-KIT. This review addresses the roles of SCF and c-KIT in the male reproductive tract, and discusses the potential application of c-KIT target therapies in disorders of the reproductive system.
Collapse
Affiliation(s)
- Henrique J Cardoso
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | | | | | | | | |
Collapse
|
24
|
TGF-β signaling alters the pattern of liver tumorigenesis induced by Pten inactivation. Oncogene 2014; 34:3273-82. [PMID: 25132272 PMCID: PMC4333137 DOI: 10.1038/onc.2014.258] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 06/04/2014] [Accepted: 06/15/2014] [Indexed: 12/14/2022]
Abstract
Hepatocarcinogenesis results from the accumulation of genetic and epigenetic changes in liver cells. A common mechanism through which these alterations induce liver cancer is by deregulating signaling pathways. A number of signaling pathways, including the PI3K/PTEN/AKT and transforming growth factor β (TGF-β) pathways have been implicated in normal liver development as well as in cancer formation. In this study, we assessed the effect of the TGF-β signaling pathway on liver tumors induced by phosphatase and tensin homolog (Pten) loss. Inactivation of only the TGF-β receptor type II, Tgfbr2, in the mouse liver (Tgfbr2(LKO)) had no overt phenotype, while inactivation of Pten alone (Pten(LKO)), resulted in the formation of both hepatocellular carcinomas and cholangiocarcinomas (CC). Interestingly, deletion of both Pten and Tgfbr2 (Pten(LKO);Tgfbr2(LKO)) in the mouse liver resulted in a dramatic shift in tumor type to predominantly CC. Assessment of the PI3K/PTEN/AKT pathway revealed increased phosphorylation of AKT and glycogen synthase kinase 3 beta (GSK-3β) in both the Pten(LKO) and Pten(LKO);Tgfbr2(LKO) mice, suggesting that this pathway is constitutively active regardless of the status of the TGF-β signaling pathway. However, phosphorylation of p70 S6 kinase was observed in the liver of all three phenotypes (Tgfbr2(LKO), Pten(LKO), Pten(LKO);Tgfbr2(LKO)) indicating that the loss of Tgfbr2 and/or Pten leads to an increase in this signaling pathway. Analysis of markers of liver progenitor/stem cells revealed that the loss of TGF-β signaling resulted in increased expression of c-Kit and CD133. Furthermore, in addition to increased c-Kit and CD133, Scf and EpCam expression were also increased in the double knock-out mice. These results suggest that the alteration in tumor types between the Pten(LKO) mice and Pten(LKO);Tgfbr2(LKO) mice is secondary to the altered regulation of stem-cell features induced by the loss of TGF-β signaling.
Collapse
|
25
|
Wang X, Ren H, Zhao T, Chen J, Sun W, Sun Y, Ma W, Wang J, Gao C, Gao S, Lang M, Jia L, Hao J. Stem cell factor is a novel independent prognostic biomarker for hepatocellular carcinoma after curative resection. Carcinogenesis 2014; 35:2283-90. [PMID: 25086759 DOI: 10.1093/carcin/bgu162] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Stem cell factor (SCF), a ligand of c-kit, is a hematopoietic growth factor. Uncontrolled activity of SCF/c-kit signaling pathway contributes to the formation of a variety of human malignancies. In this study, we determined whether SCF expression could risk-stratify patients with hepatocellular carcinoma (HCC) after curative resection. HCC tissues from 160 patients were collected during curative resection and stained with SCF and CD34, a marker for microvessel density (MVD), using immunohistochemistry. Two statistical analyses were performed: an independent continuous and a multivariate categorical analysis, with test/validation set-defined cut points, and Kaplan-Meier estimated outcome measures of overall survival (OS) and relapse-free survival (RFS). We found that higher levels of SCF confer worse OS (continuous P = 0.014; and categorical P = 0.009), and RFS (continuous P = 0.002; categorical P = 0.003) of patients with HCC. SCF varies independently from MVD-CD34, tumor node metastasis, histologic grade, age and gender, and retains prognostic significance when analysed as a categorical variable in a multivariate analysis . We confirmed that MVD-CD34 is also an independent prognostic marker for patients with HCC. The levels of SCF and CD34 showed a positive and significant correlation (P < 0.0001) and double low expression confers superior OS (median = 48 months) and RFS (median = 24 months), whereas double high expression confers shortest RFS (median = 10.5 months) compared with single measurements. The prognostic values of SCF and CD34 were independently determined in this study and we propose that both of them are independent prognostic markers for HCC.
Collapse
Affiliation(s)
- Xiuchao Wang
- Department of Abdominal Oncology and Department of Pathology, National Clinical Research Center for Cancer, Key Lab of Cancer Treatment and Prevention, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China and Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - He Ren
- Department of Abdominal Oncology and Department of Pathology, National Clinical Research Center for Cancer, Key Lab of Cancer Treatment and Prevention, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China and Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Tiansuo Zhao
- Department of Abdominal Oncology and Department of Pathology, National Clinical Research Center for Cancer, Key Lab of Cancer Treatment and Prevention, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China and Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Jing Chen
- Department of Abdominal Oncology and Department of Pathology, National Clinical Research Center for Cancer, Key Lab of Cancer Treatment and Prevention, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China and Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Wei Sun
- Department of Abdominal Oncology and Department of Pathology, National Clinical Research Center for Cancer, Key Lab of Cancer Treatment and Prevention, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China and Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Yan Sun
- Department of Pathology, National Clinical Research Center for Cancer, Key Lab of Cancer Treatment and Prevention, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China and
| | - Weidong Ma
- Department of Abdominal Oncology and Department of Pathology, National Clinical Research Center for Cancer, Key Lab of Cancer Treatment and Prevention, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China and Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Jian Wang
- Department of Abdominal Oncology and Department of Pathology, National Clinical Research Center for Cancer, Key Lab of Cancer Treatment and Prevention, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China and Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Chuntao Gao
- Department of Abdominal Oncology and Department of Pathology, National Clinical Research Center for Cancer, Key Lab of Cancer Treatment and Prevention, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China and Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Song Gao
- Department of Abdominal Oncology and Department of Pathology, National Clinical Research Center for Cancer, Key Lab of Cancer Treatment and Prevention, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China and Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Mingxiao Lang
- Department of Abdominal Oncology and Department of Pathology, National Clinical Research Center for Cancer, Key Lab of Cancer Treatment and Prevention, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China and Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Li Jia
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Jihui Hao
- Department of Abdominal Oncology and Department of Pathology, National Clinical Research Center for Cancer, Key Lab of Cancer Treatment and Prevention, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China and Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| |
Collapse
|
26
|
Jo HJ, Kim N, Nam RH, Kang JM, Kim JH, Choe G, Lee HS, Park JH, Chang H, Kim H, Lee MY, Kim YS, Kim JS, Jung HC. Fat deposition in the tunica muscularis and decrease of interstitial cells of Cajal and nNOS-positive neuronal cells in the aged rat colon. Am J Physiol Gastrointest Liver Physiol 2014; 306:G659-69. [PMID: 24525022 DOI: 10.1152/ajpgi.00304.2012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Little is known about the time course of aging on interstitial cells of Cajal (ICC) of colon. The aim of this study was to investigate the change of morphology, ICC, and neuronal nitric oxide synthase (nNOS)-immunoreactive cells in the aged rat. The proximal colon of 344 Fischer rats at four different ages (6, 31, 74 wk, and 2 yr) were studied. The immunoreactivity of c-Kit, nNOS, anti-protein gene product 9.5, and synaptophysin were counted after immunohistochemistry. The c-kit, stem cell factor (ligand of Kit), and nNOS mRNA were measured by real-time PCR. c-Kit and nNOS protein were assessed by Western blot. Isovolumetric contractile force measurement and electrical field stimulation (EFS) were conducted. The area of intramuscular fat deposition significantly increased with age after 31 wk. c-Kit-immunoreactive ICC and nNOS-immunoreactive neurons and nerve fibers significantly declined with age. mRNA and protein expression of c-kit and nNOS decreased with aging. The functional study showed that the spontaneous contractility was decreased in aged rat, whereas EFS responses in the presence of atropine and L-NG-Nitroarginine methyl ester were increased in aged rat. In conclusion, the decrease of proportion of proper smooth muscle, the density of ICC and nNOS-immunoreactive neuronal fibers, and the number of nNOS-immunoreactive neurons during the aging process may explain the aging-associated colonic dysmotility.
Collapse
|
27
|
Induction of chemokines and cytokines before neutrophils and macrophage recruitment in different regions of rat liver after TAA administration. J Transl Med 2014; 94:235-47. [PMID: 24276236 DOI: 10.1038/labinvest.2013.134] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 09/20/2013] [Accepted: 10/16/2013] [Indexed: 12/13/2022] Open
Abstract
Single-dose thioacetamide (TAA) administration induces inflammation and acute liver damage. The mechanism of inflammatory cell recruitment in the liver is still unclear. The aim of this study was to examine the sequence and recruitment of inflammatory cells in different liver regions in relation to CXC- and CC-chemokine and cytokine expression during acute liver injury. Single-dose TAA was administered to rats intraperitoneally, and animals were killed at different time points thereafter. Serum and liver tissue were taken and frozen immediately. Tissue was used for immunostaining cryostat sections, RNA, and protein extraction. RT-PCR and western blotting were performed for RNA and protein analysis, respectively. An early increase (3 h) in CXCL8/IL-8 levels was measured followed by a marked release in MCP1/CCL2 (24 h) serum levels after TAA administration compared with controls. Similarly, an early increase in specific RNA of hepatic chemokines CXCL1/KC and CXCL8/IL-8 was found at 3 h, followed by an upregulation of CXCL5/LIX (6 h), CXCL2/MIP-2 (12 h), and MCP1/CCL2 gene expression at 24-48 h. Further, an induction of pro-inflammatory cytokines IFN-γ and IL-1β followed by IL-6 and TNF-α was observed with a maximum at 12 h. The magnitude of increase in gene expression of TNF-α and MCP1/CCL2 was the highest among all cytokines and chemokines, respectively. By means of immunohistochemistry, an early (12-24 h) increase in the number of only neutrophil granulocytes (NGs) attached to and around portal vessel walls was observed, followed by increased numbers of mononuclear phagocytes (24-48 h) along the sinusoids. Treatment of the human monocytic cell line U-937 with TNF-α increased the gene expression of CXCL1/KC, CXCL8/IL-8, and MCP1/CCL2. Conversely, adding of infliximab (IFX) to the culture medium inhibited this upregulation significantly. In conclusion, single-dose TAA administration induces a sequence of events with a defined upregulation of gene expression of inflammatory chemokines and cytokines and a transient accumulation of NGs within the portal area and macrophages along the sinusoids throughout the liver. Periportal inflammation seems to precede hepatocellular damage.
Collapse
|
28
|
Vu NB, Nguyen TT, Tran LCD, Do CD, Nguyen BH, Phan NK, Pham PV. Doxorubicin and 5-fluorouracil resistant hepatic cancer cells demonstrate stem-like properties. Cytotechnology 2013; 65:491-503. [PMID: 23104270 PMCID: PMC3720974 DOI: 10.1007/s10616-012-9511-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 10/16/2012] [Indexed: 12/11/2022] Open
Abstract
The efficacy of hepatocellular carcinoma (HCC) treatment is very low because of the high percentage of recurrence and resistance to anticancer agents. Hepatic cancer stem cells (HCSCs) are considered the origin of such recurrence and resistance. Our aim was to evaluate the stemness of doxorubicin and 5-fluorouracil resistant hepatic cancer cells and establish the new method to isolate the HCSCs from primary cultured HCC tumors. HCC biopsies were used to establish primary cultures. Then, primary cells were selected for HCSCs by culture in medium supplemented with doxorubicin (0, 0.1, 0.25, 0.5 or 1 μg/mL), 5-fluorouracil (0, 0.1, 0.25, 0.5 or 1 μg/mL) or their combination. Selection was confirmed by detection of HCSC markers such as CD133, CD13, CD90, and the side population was identified by rhodamine 123 efflux. The cell population with the strongest expression of these markers was used to evaluate the cell cycle, gene expression profile, tumor sphere formation, marker protein expression, and in vivo tumorigenesis. Selective culture of primary cells in medium supplemented with 0.5 μg/mL doxorubicin and 1 μg/mL 5-fluorouracil selected cancer cells with the highest stemness properties. Selected cells strongly expressed CD13, CD133, CD90, and CD326, efflux rhodamine 123 and formed tumor spheres in suspension. Moreover, selected cells were induced to differentiate into cells with high expression of CD19 and AFP (alpha-fetoprotein), and importantly, could form tumors in NOD/SCID mice upon injection of 1 × 10(5) cells/mouse. Selective culture with doxorubicin and 5-fluorouracil will enrich HCSCs, is an easy method to obtain HCSCs that can be used to develop better therapeutic strategies for patients with HCC, and particularly HCSC-targeting therapy.
Collapse
Affiliation(s)
- Ngoc Bich Vu
- />Laboratory of Stem Cell Research and Application, University of Science, VNU-HCM, HCM City, Vietnam
| | - Tam Thanh Nguyen
- />Laboratory of Stem Cell Research and Application, University of Science, VNU-HCM, HCM City, Vietnam
| | - Long Cong-Duy Tran
- />University of Medical Center, Ho Chi Minh University of Medicine and Pharmacy, HCM City, Vietnam
| | - Cong Dinh Do
- />University of Medical Center, Ho Chi Minh University of Medicine and Pharmacy, HCM City, Vietnam
| | - Bac Hoang Nguyen
- />University of Medical Center, Ho Chi Minh University of Medicine and Pharmacy, HCM City, Vietnam
| | - Ngoc Kim Phan
- />Laboratory of Stem Cell Research and Application, University of Science, VNU-HCM, HCM City, Vietnam
| | - Phuc Van Pham
- />Laboratory of Stem Cell Research and Application, University of Science, VNU-HCM, HCM City, Vietnam
| |
Collapse
|
29
|
Minicis SD, Kisseleva T, Francis H, Baroni GS, Benedetti A, Brenner D, Alvaro D, Alpini G, Marzioni M. Liver carcinogenesis: rodent models of hepatocarcinoma and cholangiocarcinoma. Dig Liver Dis 2013; 45. [PMID: 23177172 PMCID: PMC3716909 DOI: 10.1016/j.dld.2012.10.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Hepatocellular carcinoma and cholangiocarcinoma are primary liver cancers, both represent a growing challenge for clinicians due to their increasing morbidity and mortality. In the last few years a number of in vivo models of hepatocellular carcinoma and cholangiocarcinoma have been developed. The study of these models is providing a significant contribution in unveiling the pathophysiology of primary liver malignancies. They are also fundamental tools to evaluate newly designed molecules to be tested as new potential therapeutic agents in a pre-clinical set. Technical aspects of each model are critical steps, and they should always be considered in order to appropriately interpret the findings of a study or its planning. The purpose of this review is to describe the technical and experimental features of the most significant rodent models, highlighting similarities or differences between the corresponding human diseases. The first part is dedicated to the discussion of models of hepatocellular carcinoma, developed using toxic agents, or through dietary or genetic manipulations. In the second we will address models of cholangiocarcinoma developed in rats or mice by toxin administration, genetic manipulation and/or bile duct incannulation or surgery. Xenograft or syngenic models are also proposed.
Collapse
Affiliation(s)
- Samuele De Minicis
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy
| | - Tatiana Kisseleva
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, CA, United States
| | - Heather Francis
- Division Research, Central Texas Veterans Health Care System, Scott & White Digestive Disease Research Center, Department of Medicine, Division Gastroenterology, Scott & White Hospital and Texas A&M Health Science Center, College of Medicine, Temple, TX, United States
| | | | - Antonio Benedetti
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy
| | - David Brenner
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, CA, United States
| | - Domenico Alvaro
- Division of Gastroenterology, Polo Pontino, Università degli Studi “La Sapienza”, Rome, Italy
| | - Gianfranco Alpini
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, CA, United States,Co-corresponding author. Tel.: +1 254 743 1041/1044; fax: +1 254 743 0378/0555. (M. Marzioni)
| | - Marco Marzioni
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy,Corresponding author at: Department of Gastroenterology, Università Politecnica delle Marche, Nuovo Polo Didattico, III Piano, Via Tronto 10, 60020 Ancona, Italy. Tel.: +39 0712206043; fax: +39 0712206044
| |
Collapse
|
30
|
Characteristics of hepatic stem/progenitor cells in the fetal and adult liver. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2013; 19:587-93. [PMID: 23010995 DOI: 10.1007/s00534-012-0544-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND The liver is an essential organ that maintains vital activity through its numerous important functions. It has a unique capability of fully regenerating after injury. Regulating a balance between self-renewal and differentiation of hepatic stem cells that are resources for functional mature liver cells is required for maintenance of tissue homeostasis. METHODS This review describes the characteristics of hepatic stem/progenitor cells and the regulatory mechanism of their self-renewal and differentiation capacity. RESULTS In liver organogenesis, undifferentiated hepatic stem/progenitor cells expand their pool by repeated self-renewal in the early stage of liver development and then differentiate into two different types of cell lineage, namely hepatocytes and cholangiocytes. Liver development is regulated by expression of stem cell transcription factors in a complex multistep process. Recent studies suggest that stem cells are maintained by integrative regulation of gene expression patterns related to self-renewal and differentiation by epigenetic mechanisms such as histone modification and DNA methylation. CONCLUSIONS Analysis of the proper regulatory mechanism of hepatic stem/progenitor cells is important for regenerative medicine that utilizes hepatic stem cells and for preventing liver cancer through clarification of the carcinogenetic mechanism involved in stem cell system failure.
Collapse
|
31
|
Recent advances in cancer stem cell research for cholangiocarcinoma. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2013; 19:606-13. [PMID: 22907641 DOI: 10.1007/s00534-012-0542-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer stem cells have been identified as cells with the capacity to self-renew and differentiate into multiple lineages of human malignancies. Cholangiocarcinoma is one of the most difficult intra-abdominal malignancies that can be treated using a surgical approach. Chemotherapy in addition to surgery is necessary to improve patient survival. However, its clinical benefit is limited, and, to date, no other effective anticancer drug is available for this disease. Several reports have shown the existence of cholangiocarcinoma stem cells. Cell surface antigens such as CD133, CD24, EpCAM, CD44, and others have been used to isolate cholangiocarcinoma stem cells. In general, enhanced expression of these markers in resected specimens of cholangiocarcinoma was associated with malignant potential. Distinct and specific pathways are expected to be present in cancer stem cells compared to other cancer cells that have no stem cell properties. To date, reports showing possible signaling pathways in cholangiocarcinoma stem cells are limited. More research is anticipated. Targeting therapies for surface molecular markers or specific signaling pathways of cholangiocarcinoma stem cells may be important in order to change the clinical outcome of patients with this disease. However, no clinical trial has been performed so far. This review will focus on the markers and signaling pathways used to define cholangiocarcinoma stem cells. A novel therapeutic approach of targeting cholangiocarcinoma stem cells will also be discussed.
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Even though recent accumulated data can help to understand fundamental molecular mechanisms of progression of cholangiocarcinoma (CCA), its incidence and mortality still keep increasing worldwide with poor prognosis. As appropriate animal disease models are critical to fill the gap between the findings from in vitro and the applications to human diseases, lack of effective and patient-like CCA animal models may contribute to limits of controlling progression of CCA. This review is focusing to provide the information about recently developed CCA animal models. RECENT FINDINGS Recent advancements in cell and molecular biology make it possible to mimic the pathogenicity of human CCA using various animal models. In this review, several up-to-date techniques and the examples to induce CCA in animal models (xenograft and orthotopic models, carcinogen-induced CCA model, genetically engineered mouse model for CCA) with resemblance of human CCA are discussed. SUMMARY Not only establishing animal models relevant to CCA is beneficial for its early diagnosis and therapy but also well suited experimental CCA models will guide the development of applicable treatment strategy for the hard-to-cure CCA.
Collapse
|
33
|
Fan H, Yuan Y, Wang J, Zhou F, Zhang M, Giercksky KE, Nesland JM, Suo Z. CD117 expression in operable oesophageal squamous cell carcinomas predicts worse clinical outcome. Histopathology 2013; 62:1028-37. [PMID: 23570416 PMCID: PMC3712472 DOI: 10.1111/his.12111] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 02/06/2013] [Indexed: 12/11/2022]
Abstract
Aims To investigate the aberrant expression of CD117 in oesophageal squamous cell carcinoma (SCC) and its prognostic significance. Methods and results Immunohistochemical staining for CD117 was performed on tissue microarray and routine tissue sections from 157 oesophageal SCC patients and 10 normal oesophageal epithelia adjacent to tumour. The positive rate of CD117 expression was 29.9% in oesophageal SCC tissues, whereas no CD117 expression was detected in the 10 normal oesophageal epithelia. CD117 expression was significantly associated with T stage (P < 0.001), distant metastasis (P = 0.015), lymph node metastasis (P = 0.019), and clinical stage (P = 0.021). Progression-free survival in the patients with CD117-positive tumours was shorter than that in the patients with CD117-negative tumours (P = 0.010). In univariate analyses, CD117 expression was the most significant factor for overall survival of oesophageal SCC patients (P < 0.001), followed by lymph node metastasis (P = 0.001), T stage (P = 0.002), clinical stage (P = 0.006), distant metastasis (P = 0.020), and histological grade (P = 0.027). Multivariate analyses verified that CD117 expression was an independent prognostic marker for oesophageal SCC patients (P = 0.002). In addition, CD117 expression predicted poorer survival in patients without distant metastases. Conclusions CD117 expression in operable oesophageal SCC may be a valuable prognostic marker, and detection of its expression in clinical samples may be useful in defining a subclass of oesophageal SCCs with extremely poor clinical outcome, which may require a specially targeted treatment modality.
Collapse
Affiliation(s)
- Huijie Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Sekiya S, Suzuki A. Intrahepatic cholangiocarcinoma can arise from Notch-mediated conversion of hepatocytes. J Clin Invest 2013; 122:3914-8. [PMID: 23023701 DOI: 10.1172/jci63065] [Citation(s) in RCA: 259] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 08/02/2012] [Indexed: 12/15/2022] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is the second most common primary malignancy in the liver. ICC has been classified as a malignant tumor arising from cholangiocytes; however, the co-occurrence of ICC and viral hepatitis suggests that ICC originates in hepatocytes. In order to determine the cellular origin of ICC, we used a mouse model of ICC in which hepatocytes and cholangiocytes were labeled with heritable, cell type–specific reporters. Our studies reveal that ICC is generated by biliary lineage cells derived from hepatocytes, rather than cholangiocytes. Additionally, we found that Notch activation is critical for hepatocyte conversion into biliary lineage cells during the onset of ICC and its subsequent malignancy and progression. These findings will help to elucidate the pathogenic mechanism of ICC and to develop therapeutic strategies for this refractory disease.
Collapse
Affiliation(s)
- Sayaka Sekiya
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
35
|
Mrzljak A, Košuta I, Škrtićl A, Kardum-Skelin I, Vrhovac R. Metachronous gastrointestinal stromal tumor and acute leukemia after liver transplantation for cholangiocellular carcinoma: is there a link? Case Rep Oncol 2013; 6:163-8. [PMID: 23626555 PMCID: PMC3636964 DOI: 10.1159/000348817] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The synchronous or metachronous coexistence of gastrointestinal stromal tumors (GISTs) with solid and hematologic neoplasms has been addressed in a non-transplant population. However, the association with primary hepatic neoplasms and leukemias is uncommon. Scarce data exist considering association of GISTs and other neoplasms in a transplant population where long-term immunosuppression carries the additional burden of de novo malignancy. We present a case of posttransplant metachronous GIST and acute biphenotypic leukemia in a patient transplanted for intrahepatic cholangiocellular carcinoma, emphasizing the possible link between mechanisms of carcinogenesis and influence of other factors upon their development.
Collapse
Affiliation(s)
- Anna Mrzljak
- Department of Medicine, University Hospital Merkur, School of Medicine, University of Zagreb, Zagreb, Croatia
- *Anna Mrzljak, Department of Medicine, University Hospital Merkur, School of Medicine, University of Zagreb, Zajševa 19, HR-10000 Zagreb (Croatia), E-Mail
| | - Iva Košuta
- Department of Medicine, University Hospital Merkur, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Anita Škrtićl
- Department of Pathology and Cytology, University Hospital Merkur, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ika Kardum-Skelin
- Department of Pathology and Cytology, University Hospital Merkur, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Radovan Vrhovac
- Department of Medicine, University Hospital Merkur, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
36
|
Peripheral intrahepatic cholangiocarcinoma occurring in patients without cirrhosis or chronic bile duct diseases: epidemiology and histopathology of distant nontumoral liver in 57 White patients. Eur J Gastroenterol Hepatol 2013; 25:94-8. [PMID: 23085575 DOI: 10.1097/meg.0b013e328357cdd7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND/AIM Peripheral intrahepatic cholangiocarcinoma (ICC) occurring mainly in the absence of cirrhosis represents an increasing subgroup of primary liver tumors in Western countries. Histopathologic changes in the non-neoplastic liver in this context are not well characterized. PATIENTS AND METHODS We assessed the clinical characteristics and histopathologic changes in the distant nontumoral liver of 57 consecutive White patients (34 men, mean age 59 years) referred to one medical and one surgical liver institution over a 16-year period who developed a peripheral ICC in the absence of cirrhosis or bile duct disease. RESULTS High alcohol consumption was observed in 11 patients (20%), 38 patients (66%) had a BMI of 25 kg/m or more, 22 patients (40%) had diabetes, two patients had hepatitis B virus infection, two others had hepatitis C virus infection, three patients had genetic hemochromatosis, and two patients had cutaneous porphyria tarda. The distant nontumoral liver was normal in 10 patients (18%). The two main histopathologic changes observed were macrovesicular steatosis (>10% of hepatocytes) in 38 patients (66%), including 11 patients (19%) with steatohepatitis, and moderate or intense hepatocyte iron overload in 22 patients (38%). CONCLUSION This study shows a high prevalence of macrovesicular steatosis associated or not with steatohepatitis and iron overload in patients who develop peripheral ICC in the absence of cirrhosis or bile duct disease.
Collapse
|
37
|
Duda DG. Molecular Biomarkers of Response to Antiangiogenic Therapy for Cancer. ACTA ACUST UNITED AC 2012; 2012. [PMID: 24340224 DOI: 10.5402/2012/587259] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Antiangiogenic therapy for cancer has gone from an intriguing hypothesis in the 1970s to an accepted treatment approach for many cancer types. It has also become a standard of care for certain eye diseases. Yet, despite the use of molecularly targeted drugs with well defined targets, to date there are no biomarkers to guide the use of antiangiogenic therapy in patients. The mechanisms of action of these drugs are also being debated. This paper discusses some of the emerging biomarker candidates for this type of cancer therapy, which have provided mechanistic insight and might be useful in the future for optimizing cancer treatment.
Collapse
Affiliation(s)
- Dan G Duda
- Steele Laboratory for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
38
|
Moriconi F, Ramadori P, Schultze FC, Blaschke M, Amanzada A, Khan S, Ramadori G. Characterization of the erythropoietin/erythropoietin receptor axis in a rat model of liver damage and cholangiocarcinoma development. Histochem Cell Biol 2012; 139:473-85. [PMID: 23052842 PMCID: PMC3573187 DOI: 10.1007/s00418-012-1037-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2012] [Indexed: 01/28/2023]
Abstract
It has been recently shown that the biological effects of erythropoietin (EPO) are not limited to the hematopoietic compartment but, as pleiotropic glycoprotein, this hormone can exert pro-angiogenic and tissue-protective functions also in a wide range of non-hematopoietic organs. The role of EPO and the effective functionality of its receptor in solid tumors are still a controversial point of debate. In the present work we analyzed the gene expression of EPO and its cognate receptor (EpoR) in a rat model of thioacetamide-induced damage and tumor. An analysis of the EPO/EpoR axis was also performed on human cholangiocarcinoma (CC) cell lines. A progressive increase of EPO and EpoR mRNA can already be observed during the fibrotic–cirrhotic development with a peak of expression rising at tumor formation (24.7 ± 9.9-fold increase and 15.5 ± 1.1-fold increase, respectively, for the two genes). Co-localization studies by immunofluorescence revealed hepatocytes in the regenerative cirrhotic nodules (Hep Par-1+) and in the dysplastic bile duct cells (CK19+) as the major EPO producers in this specific condition. The same cell populations, together with endothelial cells, exhibited an increased expression of EpoR, although all the non-parenchymal cell populations in the liver exhibited modest basal mRNA levels. Challenging human CC cells, Mz-Cha-2, with a combination of EPO and SCF resulted in a synergistic effect on the gene expression of EPO, CyclinD1 and PCNA. This study suggests that the autocrine and paracrine release of endogenous EPO in the microenvironment may contribute to the development and maintenance of the CC possibly in cooperation with other signaling pathways.
Collapse
Affiliation(s)
- Federico Moriconi
- Department of Gastroenterology and Endocrinology, Center of Internal Medicine, University of Göttingen, Robert Koch Strasse 40, 37075 Göttingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
39
|
Hata M, Iida H, Yamanegi K, Yamada N, Ohyama H, Hirano H, Nakasho K, Terada N. Phenotypic characteristics and proliferative activity of hyperplastic ductule cells in cholangiofibrosis induced by thioacetamide in rats. ACTA ACUST UNITED AC 2011; 65:351-6. [PMID: 22206608 DOI: 10.1016/j.etp.2011.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 11/14/2011] [Accepted: 11/29/2011] [Indexed: 01/16/2023]
Abstract
The oral administration of thioacetamide to rats induces cholangiofibrosis characterized by hyperplasia of ductules surrounded by fibrous tissue. In the present study, we examined the expression of markers of cholangiocyte and hepatocyte phenotypes in these hyperplastic ductule cells and their proliferative activity immunohistochemically. The oral administration of thioacetamide to 21-day-old male Fisher 344 rats for 12 weeks induced multiple areas of various sizes with hyperplastic ductules. The ductules consisted of two types of ductules; ductules composed of cholangiocyte-like cuboidal cells with transparent nuclei and cytoplasm, and of intestinal epithelium-like (IE-like) cells of basophilic nuclei and cytoplasm, and the transition of these two types of cells in the same ductule was sometimes observed. The cholangiocyte-like cells expressed cytokeratin (CK)-7, CK-19 and OV-6 (cholangiocyte phenotype markers) but not Hep Par-1 antigen or HNF4α (hepatocyte phenotype markers). In contrast, the IE-like cells expressed Hep Par-1 antigen and HNF4α but not CK-7, CK-19 or OV-6. The examination of Ki-67 expression showed a much higher proliferative activity for the IE-like cells compared to the cholangiocyte-like cells. The present results show that the hyperplastic ductules induced by thioacetamide are composed of IE-like cells with a high proliferative activity expressing the hepatocyte phenotype markers and of cholangiocyte-like cells with a low proliferative activity expressing the cholangiocyte phenotype markers.
Collapse
Affiliation(s)
- Masaki Hata
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501 Japan.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Immunodetection of cyclooxygenase-2 (COX-2) is restricted to tissue macrophages in normal rat liver and to recruited mononuclear phagocytes in liver injury and cholangiocarcinoma. Histochem Cell Biol 2011; 137:217-33. [PMID: 22131058 PMCID: PMC3262142 DOI: 10.1007/s00418-011-0889-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2011] [Indexed: 12/11/2022]
Abstract
It has been suggested that cyclooxygenase-2 (COX-2)-mediated prostaglandin synthesis is associated with liver inflammation and carcinogenesis. The aim of this study is to identify the cellular source of COX-2 expression in different stages, from acute liver injury through liver fibrosis to cholangiocarcinoma (CC). We induced in rats acute and "chronic" liver injury (thioacetamide (TAA) or carbon tetrachloride (CCl(4))) and CC development (TAA) and assessed COX-2 gene expression in normal and damaged liver tissue by RT-PCR of total RNA. The cellular localization of COX-2 protein in liver tissue was analyzed by immunohistochemistry as well as in isolated rat liver cells by Western blotting. The findings were compared with those obtained in human cirrhotic liver tissue. The specificity of the antibodies was tested by 2-DE Western blot and mass spectrometric identification of the positive protein spots. RT-PCR analysis of total RNA revealed an increase of hepatic COX-2 gene expression in acutely as well as "chronically" damaged liver. COX-2-protein was detected in those ED1(+)/ED2(+) cells located in the non-damaged tissue (resident tissue macrophages). In addition COX-2 positivity in inflammatory mononuclear phagocytes (ED1(+)/ED2(-)), which were also present within the tumoral tissue was detected. COX-2 protein was clearly detectable in isolated Kupffer cells as well as (at lower level) in isolated "inflammatory" macrophages. Similar results were obtained in human cirrhotic liver. COX-2 protein is constitutively detectable in liver tissue macrophages. Inflammatory mononuclear phagocytes contribute to the increase of COX-2 gene expression in acute and chronic liver damage induced by different toxins and in the CC microenvironment.
Collapse
|
41
|
Abstract
Hepatocellular carcinoma (HCC), the most common primary liver tumor, is notoriously resistant to systemic therapies, and often recurs even after aggressive local therapies. HCCs rely on the formation of new blood vessels for growth, and VEGF is critical in this process. A hallmark of new vessel formation in tumors is their structural and functional abnormality. This leads to an abnormal tumor microenvironment characterized by low oxygen tension. The liver is perfused by both arterial and venous blood and the resulting abnormal microenvironment selects for more-aggressive malignancies. Anti-VEGF therapy with sorafenib was the first systemic therapy to demonstrate improved survival in patients with advanced-stage HCC. This important development in the treatment of HCC raises hope as well as critical questions on the future development of targeted agents including other antiangiogenic agents, which hold promise to further increase survival in this aggressive disease.
Collapse
|
42
|
Sirica AE, Dumur CI, Campbell DJW, Almenara JA, Ogunwobi OO, Dewitt JL. Intrahepatic cholangiocarcinoma progression: prognostic factors and basic mechanisms. Clin Gastroenterol Hepatol 2009; 7:S68-78. [PMID: 19896103 PMCID: PMC3795391 DOI: 10.1016/j.cgh.2009.08.023] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 08/13/2009] [Accepted: 08/14/2009] [Indexed: 02/07/2023]
Abstract
In this review, we will examine various molecular biomarkers for their potential to serve as independent prognostic factors for predicting survival outcome in postoperative patients with progressive intrahepatic cholangiocarcinoma. Specific rodent models of intrahepatic cholangiocarcinoma that mimic relevant cellular, molecular, and clinical features of the human disease are also described, not only in terms of their usefulness in identifying molecular pathways and mechanisms linked to cholangiocarcinoma development and progression, but also for their potential value as preclinical platforms for suggesting and testing novel molecular strategies for cholangiocarcinoma therapy. Last, recent studies aimed at addressing the role of desmoplastic stroma in promoting intrahepatic cholangiocarcinoma progression are highlighted in an effort to underline the potential value of targeting tumor stromal components together with that of cholangiocarcinoma cells as a novel therapeutic option for this devastating cancer.
Collapse
Affiliation(s)
- Alphonse E Sirica
- Division of Cellular and Molecular Pathogenesis, Department of Pathology, Virginia Commonwealth University School of Medicine, Medical College of Virginia Campus, Richmond, Virginia 23298-0297, USA.
| | | | | | | | | | | |
Collapse
|