1
|
Pozzi S, Scomparin A, Israeli Dangoor S, Rodriguez Ajamil D, Ofek P, Neufeld L, Krivitsky A, Vaskovich-Koubi D, Kleiner R, Dey P, Koshrovski-Michael S, Reisman N, Satchi-Fainaro R. Meet me halfway: Are in vitro 3D cancer models on the way to replace in vivo models for nanomedicine development? Adv Drug Deliv Rev 2021; 175:113760. [PMID: 33838208 DOI: 10.1016/j.addr.2021.04.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/24/2021] [Accepted: 04/01/2021] [Indexed: 12/12/2022]
Abstract
The complexity and diversity of the biochemical processes that occur during tumorigenesis and metastasis are frequently over-simplified in the traditional in vitro cell cultures. Two-dimensional cultures limit researchers' experimental observations and frequently give rise to misleading and contradictory results. Therefore, in order to overcome the limitations of in vitro studies and bridge the translational gap to in vivo applications, 3D models of cancer were developed in the last decades. The three dimensions of the tumor, including its cellular and extracellular microenvironment, are recreated by combining co-cultures of cancer and stromal cells in 3D hydrogel-based growth factors-inclusive scaffolds. More complex 3D cultures, containing functional blood vasculature, can integrate in the system external stimuli (e.g. oxygen and nutrient deprivation, cytokines, growth factors) along with drugs, or other therapeutic compounds. In this scenario, cell signaling pathways, metastatic cascade steps, cell differentiation and self-renewal, tumor-microenvironment interactions, and precision and personalized medicine, are among the wide range of biological applications that can be studied. Here, we discuss a broad variety of strategies exploited by scientists to create in vitro 3D cancer models that resemble as much as possible the biology and patho-physiology of in vivo tumors and predict faithfully the treatment outcome.
Collapse
Affiliation(s)
- Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anna Scomparin
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Turin, Italy
| | - Sahar Israeli Dangoor
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Daniel Rodriguez Ajamil
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Paula Ofek
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Lena Neufeld
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Adva Krivitsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Daniella Vaskovich-Koubi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ron Kleiner
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Pradip Dey
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shani Koshrovski-Michael
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Noa Reisman
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
2
|
Novel patient-derived preclinical models of liver cancer. J Hepatol 2020; 72:239-249. [PMID: 31954489 DOI: 10.1016/j.jhep.2019.09.028] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/24/2019] [Accepted: 09/28/2019] [Indexed: 12/25/2022]
Abstract
Preclinical models of cancer based on the use of human cancer cell lines and mouse models have enabled discoveries that have been successfully translated into patients. And yet the majority of clinical trials fail, emphasising the urgent need to improve preclinical research to better interrogate the potential efficacy of each therapy and the patient population most likely to benefit. This is particularly important for liver malignancies, which lack highly efficient treatments and account for hundreds of thousands of deaths around the globe. Given the intricate network of genetic and environmental factors that contribute to liver cancer development and progression, the identification of new druggable targets will mainly depend on establishing preclinical models that mirror the complexity of features observed in patients. The development of new 3D cell culture systems, originating from cells/tissues isolated from patients, might create new opportunities for the generation of more specific and personalised therapies. However, these systems are unable to recapitulate the tumour microenvironment and interactions with the immune system, both proven to be critical influences on therapeutic outcomes. Patient-derived xenografts, in particular with humanised mouse models, more faithfully mimic the physiology of human liver cancer but are costly and time-consuming, which can be prohibitive for personalising therapies in the setting of an aggressive malignancy. In this review, we discuss the latest advances in the development of more accurate preclinical models to better understand liver cancer biology and identify paradigm-changing therapies, stressing the importance of a bi-directional communicative flow between clinicians and researchers to establish reliable model systems and determine how best to apply them to expanding our current knowledge.
Collapse
|
3
|
Roelants C, Pillet C, Franquet Q, Sarrazin C, Peilleron N, Giacosa S, Guyon L, Fontanell A, Fiard G, Long JA, Descotes JL, Cochet C, Filhol O. Ex-Vivo Treatment of Tumor Tissue Slices as a Predictive Preclinical Method to Evaluate Targeted Therapies for Patients with Renal Carcinoma. Cancers (Basel) 2020; 12:cancers12010232. [PMID: 31963500 PMCID: PMC7016787 DOI: 10.3390/cancers12010232] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/08/2020] [Accepted: 01/15/2020] [Indexed: 12/14/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the third type of urologic cancer. At time of diagnosis, 30% of cases are metastatic with no effect of chemotherapy or radiotherapy. Current targeted therapies lead to a high rate of relapse and resistance after a short-term response. Thus, a major hurdle in the development and use of new treatments for ccRCC is the lack of good pre-clinical models that can accurately predict the efficacy of new drugs and allow the stratification of patients into the correct treatment regime. Here, we describe different 3D cultures models of ccRCC, emphasizing the feasibility and the advantage of ex-vivo treatment of fresh, surgically resected human tumor slice cultures of ccRCC as a robust preclinical model for identifying patient response to specific therapeutics. Moreover, this model based on precision-cut tissue slices enables histopathology measurements as tumor architecture is retained, including the spatial relationship between the tumor and tumor-infiltrating lymphocytes and the stromal components. Our data suggest that acute treatment of tumor tissue slices could represent a benchmark of further exploration as a companion diagnostic tool in ccRCC treatment and a model to develop new therapeutic drugs.
Collapse
Affiliation(s)
- Caroline Roelants
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
- Inovarion, 75005 Paris, France
| | - Catherine Pillet
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biologie à Grande Echelle, UMR 1038, F-38000 Grenoble, France;
| | - Quentin Franquet
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043 Grenoble CEDEX 9, France; (A.F.); (G.F.); (J.-A.L.); (J.-L.D.)
| | - Clément Sarrazin
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043 Grenoble CEDEX 9, France; (A.F.); (G.F.); (J.-A.L.); (J.-L.D.)
| | - Nicolas Peilleron
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043 Grenoble CEDEX 9, France; (A.F.); (G.F.); (J.-A.L.); (J.-L.D.)
| | - Sofia Giacosa
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
| | - Laurent Guyon
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
| | - Amina Fontanell
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043 Grenoble CEDEX 9, France; (A.F.); (G.F.); (J.-A.L.); (J.-L.D.)
| | - Gaëlle Fiard
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043 Grenoble CEDEX 9, France; (A.F.); (G.F.); (J.-A.L.); (J.-L.D.)
| | - Jean-Alexandre Long
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043 Grenoble CEDEX 9, France; (A.F.); (G.F.); (J.-A.L.); (J.-L.D.)
| | - Jean-Luc Descotes
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043 Grenoble CEDEX 9, France; (A.F.); (G.F.); (J.-A.L.); (J.-L.D.)
| | - Claude Cochet
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
| | - Odile Filhol
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
- Correspondence: ; Tel.: +33-(0)4-38785645; Fax: +33-(0)4-38785058
| |
Collapse
|
4
|
Simeone K, Guay-Lord R, Lateef MA, Péant B, Kendall-Dupont J, Orimoto AM, Carmona E, Provencher D, Saad F, Gervais T, Mes-Masson AM. Paraffin-embedding lithography and micro-dissected tissue micro-arrays: tools for biological and pharmacological analysis of ex vivo solid tumors. LAB ON A CHIP 2019; 19:693-705. [PMID: 30671574 DOI: 10.1039/c8lc00982a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
There is an urgent need and strong clinical and pharmaceutical interest in developing assays that allow for the direct testing of therapeutic agents on primary tissues. Current technologies fail to provide the required sample longevity, throughput, and integration with standard clinically proven assays to make the approach viable. Here we report a microfluidic micro-histological platform that enables ex vivo culture of a large array of prostate and ovarian cancer micro-dissected tissue (MDT) followed by direct on-chip fixation and paraffination, a process we term paraffin-embedding lithography (PEL). The result is a high density MDT-Micro Array (MDTMA) compatible with standard clinical histopathology that can be used to analyse ex vivo tumor response or resistance to therapeutic agents. The cellular morphology and tissue architecture are preserved in MDTs throughout the 15 day culture period. We also demonstrate how this methodology can be used to study molecular pathways involved in cancer by performing in-depth characterization of biological and pharmacological mechanisms such as p65 nuclear translocation via TNF stimuli, and to predict the treatment outcome in the clinic via MDT response to taxane-based therapies.
Collapse
Affiliation(s)
- Kayla Simeone
- Centre de recherche du CHUM (CRCHUM)/Institut du Cancer de Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Huang X, Cai H, Ammar R, Zhang Y, Wang Y, Ravi K, Thompson J, Jarai G. Molecular characterization of a precision-cut rat liver slice model for the evaluation of antifibrotic compounds. Am J Physiol Gastrointest Liver Physiol 2019; 316:G15-G24. [PMID: 30406699 PMCID: PMC6383382 DOI: 10.1152/ajpgi.00281.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Precision-cut liver tissue slice (PCLS) contains all major cell types of the liver parenchyma and preserves the original cell-cell and cell-matrix contacts. It represents a promising ex vivo model to study liver fibrosis and test the antifibrotic effect of experimental compounds in a physiological environment. In this study using RNA sequencing, we demonstrated that various pathways functionally related to fibrotic mechanisms were dysregulated in PCLSs derived from rats subjected to bile duct ligation. The activin receptor-like kinase-5 (Alk5) inhibitor SB525334, nintedanib, and sorafenib each reversed a subset of genes dysregulated in fibrotic PCLSs, and of those genes we identified 608 genes whose expression was reversed by all three compounds. These genes define a molecular signature characterizing many aspects of liver fibrosis pathology and its attenuation in the model. A panel of 12 genes and 4 secreted biomarkers including procollagen I, hyaluronic acid (HA), insulin-like growth factor binding protein 5 (IGFBP5), and WNT1-inducible signaling pathway protein 1 (WISP1) were further validated as efficacy end points for the evaluation of antifibrotic activity of experimental compounds. Finally, we showed that blockade of αV-integrins with a small molecule inhibitor attenuated the fibrotic phenotype in the model. Overall, our results suggest that the rat fibrotic PCLS model may represent a valuable system for target validation and determining the efficacy of experimental compounds. NEW & NOTEWORTHY We investigated the antifibrotic activity of three compounds, the activin receptor-like kinase-5 (Alk5) inhibitor SB525334, nintedanib, and sorafenib, in a rat fibrotic precision-cut liver tissue slice model using RNA sequencing analysis. A panel of 12 genes and 4 secreted biomarkers including procollagen I, hyaluronic acid (HA), insulin-like growth factor binding protein 5 (IGFBP5), and WNT1-inducible signaling pathway protein 1 (WISP1) were then established as efficacy end points to validate the antifibrotic activity of the αV-integrin inhibitor CWHM12. This study demonstrated the value of the rat fibrotic PCLS model for the evaluation of antifibrotic drugs.
Collapse
Affiliation(s)
| | - Hong Cai
- Bristol-Myers Squibb, Pennington, New Jersey
| | - Ron Ammar
- Bristol-Myers Squibb, Pennington, New Jersey
| | - Yan Zhang
- Bristol-Myers Squibb, Pennington, New Jersey
| | - Yihe Wang
- Bristol-Myers Squibb, Pennington, New Jersey
| | | | | | - Gabor Jarai
- Bristol-Myers Squibb, Pennington, New Jersey
| |
Collapse
|
6
|
Starokozhko V, Abza GB, Maessen HC, Merema MT, Kuper F, Groothuis GMM. Viability, function and morphological integrity of precision-cut liver slices during prolonged incubation: Effects of culture medium. Toxicol In Vitro 2015; 30:288-99. [PMID: 26514934 DOI: 10.1016/j.tiv.2015.10.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 10/20/2015] [Accepted: 10/24/2015] [Indexed: 02/07/2023]
Abstract
Precision-cut liver slices (PCLS) are an ex vivo model for metabolism and toxicity studies. However, data on the maintenance of the morphological integrity of the various cell types in the slices during prolonged incubation are lacking. Therefore, our aims were to characterize morphological and functional changes in rat PCLS during five days of incubation in a rich medium, RegeneMed®, and a standard medium, Williams' Medium E. Although cells of all types in the slices remain viable, profound changes in morphology were observed, which were more prominent in RegeneMed®. Slices underwent notable fibrosis, bile duct proliferation and fat deposition. Slice thickness increased, resulting in necrotic areas, while slice diameter decreased, possibly indicating cell migration. An increased proliferation of parenchymal and non-parenchymal cells (NPCs) was observed. Glycogen, albumin and Cyp3a1 were maintained albeit to a different level in two media. In conclusion, both hepatocytes and NPCs remain viable and functional, enabling five-day toxicity studies. Tissue remodeling and formation of a new capsule-like cell lining around the slices are evident after 3–4 days. The differences in effects between media emphasize the importance of media selection and of the recognition of morphological changes in PCLS, when interpreting results from toxicological or pharmacological studies.
Collapse
Affiliation(s)
- Viktoriia Starokozhko
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Getahun B Abza
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Hedy C Maessen
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Marjolijn T Merema
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Frieke Kuper
- TNO, Utrechtseweg 48, 3704 HE Zeist, The Netherlands
| | - Geny M M Groothuis
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
7
|
Hauff P, Gottwald U, Ocker M. Early to Phase II drugs currently under investigation for the treatment of liver fibrosis. Expert Opin Investig Drugs 2015; 24:309-327. [PMID: 25547844 DOI: 10.1517/13543784.2015.997874] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Chronic liver diseases represent a high unmet medical need and are characterized by persistent inflammation, parenchymal damage and fibrotic remodeling, leading eventually to cirrhosis and hepatic failure. Besides the persisting high prevalence of chronic viral hepatitis B and C, the dramatic increase in nonalcoholic steatohepatitis is now considered to be a major pathophysiologic driver for fibrosis development and subsequently cirrhosis. Increasing evidence suggests that also liver cirrhosis can regress when treated adequately. AREAS COVERED Herein, the authors review the underlying pathophysiologic mechanisms leading to fibrotic remodeling in the liver. They also highlight the options for novel treatment strategies by using molecular targeted agents. EXPERT OPINION New in vitro and preclinical animal models, and the careful selection of patients with high disease dynamics for clinical studies, provide a sound basis for the clinical development of antifibrotic agents in humans. Surrogate parameters of liver function, inflammation, tissue remodeling and damage, as well as noninvasive imaging techniques, can be applied in clinical trials to provide fast readouts and novel and reliable endpoints for trial design, and provide an attractive regulatory environment for this emerging disease area.
Collapse
|
8
|
Freudlsperger C, Horn D, Weißfuß S, Weichert W, Weber KJ, Saure D, Sharma S, Dyckhoff G, Grabe N, Plinkert P, Hoffmann J, Freier K, Hess J. Phosphorylation of AKT(Ser473) serves as an independent prognostic marker for radiosensitivity in advanced head and neck squamous cell carcinoma. Int J Cancer 2014; 136:2775-85. [PMID: 25388642 DOI: 10.1002/ijc.29328] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 10/22/2014] [Indexed: 12/30/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is frequently characterized by high resistance to radiotherapy, which critically depends on both altered signaling pathways within tumor cells and their dynamic interaction with the tumor microenvironment. This study evaluated the prognostic value of the phosphorylation status of AKT on Ser473 and Thr308 for the clinical outcome of patients with advanced HNSCC on radiotherapy. Furthermore, we investigated the impact of AKT(Ser473) phosphorylation [p-AKT(Ser473)] in the context of radioresistance using ex vivo tissue cultures that resemble the complex tissue architecture and paracrine interaction with the tumor microenvironment. In a cohort of 120 patients with advanced HNSCC, who were treated with primary or adjuvant radiotherapy, a significant association was found between relative p-AKT(Ser473) levels and overall survival (p = 0.006) as well as progression-free survival (p = 0.021), while no significant correlation was revealed for relative p-AKT(Thr308) levels. In ex vivo tissue cultures p-AKT(Ser473) levels were increased upon irradiation and treatment with the PI3K inhibitor LY294002 inhibited both basal and irradiation induced AKT(Ser473) phosphorylation. Strikingly, pretreatment with LY294002 sensitized tissue cultures derived from primary and recurrent tumors to radiotherapy as determined by impaired tumor cell proliferation and enhanced DNA damage. In conclusion, phosphorylation status of AKT(Ser473) in tumor specimens serves as a novel biomarker to identify patients with advanced HNSCC at high risk for treatment failure following radiotherapy, and our data from ex vivo tissue cultures support the assumption that pharmacological inhibition of AKT(Ser473) phosphorylation might circumvent radioresistance to improve efficiency and reduce toxicity of current treatment modalities.
Collapse
|
9
|
Wells RG. The portal fibroblast: not just a poor man's stellate cell. Gastroenterology 2014; 147:41-7. [PMID: 24814904 PMCID: PMC4090086 DOI: 10.1053/j.gastro.2014.05.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Revised: 05/02/2014] [Accepted: 05/06/2014] [Indexed: 12/12/2022]
Abstract
Portal fibroblasts, the resident fibroblasts of the portal tract, are found in the mesenchyme surrounding the bile ducts. Their roles in liver homeostasis and response to injury are undefined and controversial. Although portal fibroblasts almost certainly give rise to myofibroblasts during the development of biliary fibrosis, recent lineage tracing studies suggest that their contribution to fibrogenesis is limited compared with that of hepatic stellate cells. Other functions of portal fibroblasts include participation in the peribiliary stem cell niche, regulation of cholangiocyte proliferation, and deposition of specific matrix proteins. Portal fibroblasts synthesize elastin and other components of microfibrils; these may serve structural roles, providing stability to ducts and the vasculature under conditions of increased ductal pressure, or could regulate the bioavailability of the fibrogenic transforming growth factor β in response to injury. Viewing portal fibroblasts in the context of fibroblast populations throughout the body and studying their niche-specific roles in matrix deposition and epithelial regulation could yield new insights into their contributions in the normal and injured liver. Understanding the functions of portal fibroblasts will require us to view them as more than just an alternative to hepatic stellate cells in fibrosis.
Collapse
Affiliation(s)
- Rebecca G Wells
- Departments of Medicine (GI) and Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
10
|
Szalowska E, Stoopen G, Rijk JCW, Wang S, Hendriksen PJM, Groot MJ, Ossenkoppele J, Peijnenburg AACM. Effect of oxygen concentration and selected protocol factors on viability and gene expression of mouse liver slices. Toxicol In Vitro 2013; 27:1513-24. [PMID: 23531554 DOI: 10.1016/j.tiv.2013.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 03/10/2013] [Accepted: 03/15/2013] [Indexed: 01/14/2023]
Abstract
Precision cut liver slices (PCLSs) are widely used as a model to study hepatotoxicity. For culturing of PCLS diverse protocols are used which could affect slices viability and results. We aimed to identify the most optimal culture protocol for mouse PCLS. Slices were cultured for 24h under different concentrations of serum, glucose, insulin, and oxygen. Thereafter, slices viability was assessed by biochemical methods. Transcriptome analysis was performed to identify changes introduced by culture at different oxygen concentrations (20%, 40%, 60%, and 80% of oxygen). Medium composition did not affect the slices viability. Although metabolic competence was unaffected by oxygen concentrations, culturing at 80% of oxygen yielded slices with the best biochemical characteristics. The comparison of uncultured vs. cultured slices revealed 2524 genes to be differentially expressed. Genes involved in drug metabolism, peroxisomal and mitochondrial functions were down-regulated while several adaptive/stress response processes were up-regulated. Moreover, 80% of oxygen was the most favorable condition with respect to maintenance of expression of genes involved in drug and energy metabolism. The outcome of this study indicates that mouse PCLS are a valuable tool in research on hepatic functions and toxicity, particularly if they are cultured under a controlled oxygen concentration of 80%.
Collapse
Affiliation(s)
- Ewa Szalowska
- RIKILT - Institute of Food Safety, Wageningen UR, PO Box 230, 6700 AE Wageningen, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Karim S, Liaskou E, Hadley S, Youster J, Faint J, Adams DH, Lalor PF. An in vitro model of human acute ethanol exposure that incorporates CXCR3- and CXCR4-dependent recruitment of immune cells. Toxicol Sci 2013; 132:131-41. [PMID: 23300006 DOI: 10.1093/toxsci/kfs337] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Alcoholic liver disease (ALD) is one of the commonest causes of cirrhosis and liver failure in the developed world. Hepatic inflammation is the critical stage in progression of both ALD and non-ALD, but it remains difficult to study the underlying mechanisms in a human system, and current animal models do not fully recapitulate human liver disease. We developed a human tissue-based system to study lymphocyte recruitment in response to ethanol challenge. Precision-cut liver slices (PCLS) from human livers were incubated in culture, and hepatic function was determined by albumin production, 3-(4,5-dimethylthiazol)-2,5-diphenyl tetrazolium bromide assay, glucose uptake responses, and morphometric assessment. Responses of tissue and lymphocytes to ethanol exposure were determined by PCR, flow cytometry, histology, and lymphocyte infiltration assays. Human PCLS demonstrated appropriate upregulation of CYP2E1, ADH1α, and ADH3 in response to ethanol treatment. Ethanol also induced expression of endothelial VCAM-1 and ICAM-1, production of sICAM-1 and CXCL8, and the chemokine receptors CXCR3 and CXCR4 on CD4 and CD8 lymphocytes. CXCR3- and CXCR4-dependent migration of lymphocytes into the tissue increased significantly in response to treatment with ethanol. We have demonstrated that ethanol increases chemokine receptor expression and lymphocyte recruitment into human liver tissue, suggesting that it may operate directly to promote hepatitis in ALD. The physiological and pathophysiological responses of the PCLS to ethanol in vitro highlight the potential of this assay for dissecting the molecular mechanisms underlying human liver inflammation and as a screening tool for novel therapeutics.
Collapse
Affiliation(s)
- Sumera Karim
- Institute of Biomedical Research, University of Birmingham, Birmingham, UK
| | | | | | | | | | | | | |
Collapse
|
12
|
Westra IM, Pham BT, Groothuis GMM, Olinga P. Evaluation of fibrosis in precision-cut tissue slices. Xenobiotica 2012; 43:98-112. [DOI: 10.3109/00498254.2012.723151] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
13
|
Hsu CY, Lee FY, Huo TI, Chan CY, Huang HC, Lin HC, Chang CC, Teng TH, Wang SS, Lee SD. Lack of therapeutic effects of gabexate mesilate on the hepatic encephalopathy in rats with acute and chronic hepatic failure. J Gastroenterol Hepatol 2010; 25:1321-8. [PMID: 20594263 DOI: 10.1111/j.1440-1746.2010.06235.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIM Inflammation plays a pivotal role in liver injury. Gabexate mesilate (GM, a protease inhibitor) inhibits inflammation by blocking various serine proteases. This study examined the effects of GM on hepatic encephalopathy in rats with acute and chronic liver failure. METHODS Acute and chronic liver failure (cirrhosis) were induced by intraperitoneal TAA administration (350 mg/kg/day for 3 days) and common bile duct ligation, respectively, in male Sprague-Dawley rats. Rats were randomized to receive either GM (50 mg/10 mL/kg) or saline intraperitoneally for 5 days. Severity of encephalopathy was assessed by the Opto-Varimex animal activity meter and hemodynamic parameters, mean arterial pressure and portal pressure, were measured (only in chronic liver failure rats). Plasma levels of liver biochemistry, ammonia, nitrate/nitrite, interleukins (IL) and tumor necrosis factor (TNF)-alpha were determined. RESULTS In rats with acute liver failure, GM treatment significantly decreased the plasma levels of alanine aminotransferase (P = 0.02), but no significant difference of motor activity, plasma levels of ammonia, IL-1beta, IL-6, IL-10 and TNF-alpha or survival was found. In chronic liver failure rats, GM significantly lowered the plasma TNF-alpha levels (P = 0.04). However, there was no significant difference of motor activity, other biochemical tests or survival found. GM-treated chronic liver failure rats had higher portal pressure (P = 0.04) but similar mean arterial pressure in comparison with saline-treated rats. CONCLUSIONS Chronic GM treatment does not have a major effect on hepatic encephalopathy in rats with TAA-induced acute liver failure and rats with chronic liver failure induced by common bile duct ligation.
Collapse
Affiliation(s)
- Chia-Yang Hsu
- Divisions of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Preclinical model of organotypic culture for pharmacodynamic profiling of human tumors. Proc Natl Acad Sci U S A 2010; 107:8352-6. [PMID: 20404174 DOI: 10.1073/pnas.0907676107] [Citation(s) in RCA: 203] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Predicting drug response in cancer patients remains a major challenge in the clinic. We have perfected an ex vivo, reproducible, rapid and personalized culture method to investigate antitumoral pharmacological properties that preserves the original cancer microenvironment. Response to signal transduction inhibitors in cancer is determined not only by properties of the drug target but also by mutations in other signaling molecules and the tumor microenvironment. As a proof of concept, we, therefore, focused on the PI3K/Akt signaling pathway, because it plays a prominent role in cancer and its activity is affected by epithelial-stromal interactions. Our results show that this culture model preserves tissue 3D architecture, cell viability, pathway activity, and global gene-expression profiles up to 5 days ex vivo. In addition, we show pathway modulation in tumor cells resulting from pharmacologic intervention in ex vivo culture. This technology may have a significant impact on patient selection for clinical trials and in predicting response to small-molecule inhibitor therapy.
Collapse
|
15
|
Khimji AK, Shao R, Rockey DC. Divergent transforming growth factor-beta signaling in hepatic stellate cells after liver injury: functional effects on ECE-1 regulation. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:716-27. [PMID: 18753413 DOI: 10.2353/ajpath.2008.071121] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
In liver wound healing, transforming growth factor-beta (TGF-beta) plays a critical role in stellate cell activation as well as signaling cascades in the fibrogenic response to injury. We postulate that the TGF-beta-dependent downstream signaling pathway may vary according to the mechanism of stellate cell activation; this study was undertaken to ascertain whether the downstream signaling pathways mediated by TGF-beta vary in different liver injury models. We measured Smad3 and MAP kinase activation after isolating stellate cells from rat livers injured by either bile duct ligation (BDL) or repeated carbon tetrachloride (CCl(4)) administration. Phospho-Smad3 was dramatically up-regulated in stellate cells after CCl(4) injury, but not after BDL-induced injury. TGF-beta signaling in stellate cells activated after BDL was mediated prominently through ERK activation, whereas activation induced by CCl(4) injury or culture led to a cross-signaling mechanism involving both Smad3 and p38. The divergent Smad signaling pathways observed appeared to be attributable to the differential regulation of the early growth response gene-1 (Egr-1), an apparent negative transcriptional factor for Smad3 in our system. In addition, inhibition of ERK activation in stellate cells from BDL-injured liver led to a decrease in expression of endothelin-converting enzyme-1, a critical regulator of endothelin-1. We speculate that TGF-beta signaling proceeds through differential signaling pathways depending on the mechanism of liver injury that leads to stellate cell activation.
Collapse
Affiliation(s)
- Al-Karim Khimji
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | | | | |
Collapse
|
16
|
Ohayon O, Mawasi N, Pevzner A, Tryvitz A, Gildor T, Pines M, Rojkind M, Paizi M, Spira G. Halofuginone upregulates the expression of heparanase in thioacetamide-induced liver fibrosis in rats. J Transl Med 2008; 88:627-33. [PMID: 18458672 DOI: 10.1038/labinvest.2008.30] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Advanced hepatic fibrosis is characterized by excessive extracellular matrix deposition, where collagen and proteoglycans are the main constituents of scar tissue. In previous studies, we showed that heparanase, a heparan sulfate-degrading enzyme, and vascular endothelial growth factor (VEGF) play an important role during liver development and remodeling. In this communication, we investigated the relationship between heparanase and VEGF in thioacetamide-induced liver fibrosis in rats. Our study shows that heparanase mRNA expression levels correlate with those of VEGF during the induction and recovery stages of liver fibrosis. We further demonstrated that treating fibrotic rat livers with halofuginone (HF), a multipotent antifibrogenic drug, and subsequently subjecting them to hydrodynamics-based transfection with human VEGF-165 resulted in elevated expression of heparanase mRNA. Moreover, these rats demonstrated an improved capacity to regenerate following 70% partial hepatectomy. In vitro, HF stimulated heparanase and VEGF mRNA expression in hepatic stellate cells. Taken together, our results suggest that in addition to the known multiple functions of HF, it also enhances heparanase and VEGF expression and promotes liver regeneration. Accordingly, HF seems to possess ideal properties required to become an excellent antifibrogenic agent in humans.
Collapse
Affiliation(s)
- Olga Ohayon
- Department of Anatomy and Cell Biology, The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Strnad P, Tao GZ, Zhou Q, Harada M, Toivola DM, Brunt EM, Omary MB. Keratin mutation predisposes to mouse liver fibrosis and unmasks differential effects of the carbon tetrachloride and thioacetamide models. Gastroenterology 2008; 134:1169-79. [PMID: 18395095 PMCID: PMC2692280 DOI: 10.1053/j.gastro.2008.01.035] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2007] [Accepted: 01/04/2008] [Indexed: 01/07/2023]
Abstract
BACKGROUND & AIMS Keratins 8 and 18 (K8/K18) are important hepatoprotective proteins. Animals expressing K8/K18 mutants show a marked susceptibility to acute/subacute liver injury. K8/K18 variants predispose to human end-stage liver disease and associate with fibrosis progression during chronic hepatitis C infection. We sought direct evidence for a keratin mutation-related predisposition to liver fibrosis using transgenic mouse models because the relationship between keratin mutations and cirrhosis is based primarily on human association studies. METHODS Mouse hepatofibrosis was induced by carbon tetrachloride (CCl(4)) or thioacetamide. Nontransgenic mice, or mice that over express either human Arg89-to-Cys (R89C mice) or wild-type K18 (WT mice) were used. The extent of fibrosis was evaluated by quantitative real-time reverse-transcription polymerase chain reaction of fibrosis-related genes, liver hydroxyproline measurement, and Picro-Sirius red staining and collagen immunofluorescence staining. RESULTS Compared with control animals, CCl(4) led to similar liver fibrosis but increased injury in K18 R89C mice. In contrast, thioacetamide caused more severe liver injury and fibrosis in K18 R89C as compared with WT and nontransgenic mice and resulted in increased messenger RNA levels of collagen, tissue inhibitor of metalloproteinase 1, matrix metalloproteinase 2, and matrix metalloproteinase 13. Analysis in nontransgenic mice showed that thioacetamide and CCl(4) have dramatically different molecular expression responses involving cytoskeletal and chaperone proteins. CONCLUSIONS Over expression of K18 R89C predisposes transgenic mice to thioacetamide- but not CCl(4)-induced liver fibrosis. Differences in the keratin mutation-associated fibrosis response among the 2 models raise the hypothesis that keratin variants may preferentially predispose to fibrosis in unique human liver diseases. Findings herein highlight distinct differences in the 2 widely used fibrosis models.
Collapse
Affiliation(s)
| | | | | | - Masaru Harada
- Department of Medicine, Kurume University School of Medicine, Kurume 830-0011, Japan
| | | | - Elizabeth M. Brunt
- Department of Pathology and Immunology, Washington University School of Medicine, 660 S. Euclid Ave, Campus Box 8118, St. Louis, MO 63110
| | - M. Bishr Omary
- Corresponding Author Address: Bishr Omary, Palo Alto VA Medical Center, 3801 Miranda Avenue, Mail code 154J, Palo Alto, CA 94304, Tel: (650) 493-5000, x63140; Fax: (650) 852-3259, E-Mail:
| |
Collapse
|