1
|
Wang C, Felli E, Fallowfield JA, Dietrich CF, Rockey D, Hennig J, Teng GJ, Gracia-Sancho J, Qi X. Vasomics of the liver. Gut 2025:gutjnl-2024-334133. [PMID: 40044498 DOI: 10.1136/gutjnl-2024-334133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/07/2025] [Indexed: 03/09/2025]
Abstract
Chronic liver disease is a cluster of disorders associated with complex haemodynamic alterations, which is characterised by structural and functional disruptions of the intrahepatic and extrahepatic vasculature. 'Vasomics' is an emerging omics discipline that comprehensively analyses and models the vascular system by integrating pathophysiology of disease, biomechanics, medical imaging, computational science and artificial intelligence. Vasomics is further typified by its multidimensional, multiscale and high-throughput nature, which depends on the rapid and robust extraction of well-defined vascular phenotypes with clear clinical and/or biological interpretability. By leveraging multimodality medical imaging techniques, vascular functional assessments, pathological image evaluation, and related computational methods, integrated vasomics provides a deeper understanding of the associations between the vascular system and disease. This in turn reveals the crucial role of the vascular system in disease occurrence, progression and treatment responses, thereby supporting precision medicine approaches. Pathological vascular features have already demonstrated their key role in different clinical scenarios. Despite this, vasomics is yet to be widely recognised. Therefore, we furnished a comprehensive definition of vasomics providing a classification of existing hepatic vascular phenotypes into the following categories: anatomical, biomechanical, biochemical, pathophysiological and composite.
Collapse
Affiliation(s)
- Chengyan Wang
- State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Southeast University, Nanjing, China
- Shanghai Pudong Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Eric Felli
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Bern, Switzerland
| | | | | | - Don Rockey
- Digestive Disease Research Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jürgen Hennig
- Department of Radiology, Medical Center, University of Freiburg, Freiburg im Breisgau, Baden-Württemberg, Germany
| | - Gao-Jun Teng
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Southeast University, Nanjing, China
- Liver Disease Center of Integrated Traditional Chinese and Western Medicine, Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Nanjing, China
| | - Jordi Gracia-Sancho
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Liver Vascular Biology Lab, Liver Unit IDIBAPS, Hospital Clínic Barcelona-CIBEREHD, Barcelona, Spain
| | - Xiaolong Qi
- State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Southeast University, Nanjing, China
- Liver Disease Center of Integrated Traditional Chinese and Western Medicine, Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Nanjing, China
| |
Collapse
|
2
|
Fan H, Shang J, Li J, Yang B, Zhou D, Jiang S, Fan Y, Zhou Y, Wang Y, Liu P, Li C, Chen Z, Chen P. High-Throughput Formation of Pre-Vascularized hiPSC-Derived Hepatobiliary Organoids on a Chip via Nonparenchymal Cell Grafting. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407945. [PMID: 39755926 PMCID: PMC11848576 DOI: 10.1002/advs.202407945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/17/2024] [Indexed: 01/06/2025]
Abstract
Liver organoids have been increasingly adopted as a critical in vitro model to study liver development and diseases. However, the pre-vascularization of liver organoids without affecting liver parenchymal specification remains a long-lasting challenge, which is essential for their application in regenerative medicine. Here, the large-scale formation of pre-vascularized human hepatobiliary organoids (vhHBOs) is presented without affecting liver epithelial specification via a novel strategy, namely nonparenchymal cell grafting (NCG). Endothelial and mesenchymal cells are grafted to human hepatobiliary organoids (hHBOs) at the different liver epithelial differentiation stages without supplementing with nonparenchymal culture medium and growth factors. Endothelial grafting at the stage of hepatic maturation offers an optimal integration efficiency compared to the stage of hepatic specification. Additionally, grafting with mesenchymal proves crucial in endothelial invading and sprouting into the liver epithelial cells during the establishment of vhHBOs. Ectopic liver implants into mice further displayed integration of vhHBOs into mice vascular networks. Notably, transplanted vhHBOs self-organized into native liver tissue like hepatic zone and bile ducts, indicating their potential to regenerate damaged hepatic and bile duct tissues. It is believed that nonparenchymal cell grafting will offer a novel technical route to form a high-fidelity complex in vitro model for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Han Fan
- Tissue Engineering and Organ Manufacturing (TEOM) LabDepartment of Biomedical EngineeringWuhan University TaiKang Medical School (School of Basic Medical Sciences)Wuhan430071China
| | - Jia Shang
- Department of Biological RepositoriesZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Junbo Li
- Key Laboratory of Organ TransplantationInstitute of Organ TransplantationTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Bo Yang
- Key Laboratory of Organ TransplantationInstitute of Organ TransplantationTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Ding Zhou
- Tissue Engineering and Organ Manufacturing (TEOM) LabDepartment of Biomedical EngineeringWuhan University TaiKang Medical School (School of Basic Medical Sciences)Wuhan430071China
| | - Shanqing Jiang
- Tissue Engineering and Organ Manufacturing (TEOM) LabDepartment of Biomedical EngineeringWuhan University TaiKang Medical School (School of Basic Medical Sciences)Wuhan430071China
| | - Yuhang Fan
- Tissue Engineering and Organ Manufacturing (TEOM) LabDepartment of Biomedical EngineeringWuhan University TaiKang Medical School (School of Basic Medical Sciences)Wuhan430071China
| | - Ying Zhou
- Research Center for Medicine and Structural Biology of Wuhan UniversityWuhan UniversityWuhanHubei430071China
| | - Yuwen Wang
- Tissue Engineering and Organ Manufacturing (TEOM) LabDepartment of Biomedical EngineeringWuhan University TaiKang Medical School (School of Basic Medical Sciences)Wuhan430071China
| | - Peidi Liu
- Tissue Engineering and Organ Manufacturing (TEOM) LabDepartment of Biomedical EngineeringWuhan University TaiKang Medical School (School of Basic Medical Sciences)Wuhan430071China
| | - Changyong Li
- Department of PhysiologyWuhan University TaiKang Medical School (School of Basic Medical Sciences)WuhanHubei430071China
| | - Zhishui Chen
- Key Laboratory of Organ TransplantationInstitute of Organ TransplantationTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Pu Chen
- Tissue Engineering and Organ Manufacturing (TEOM) LabDepartment of Biomedical EngineeringWuhan University TaiKang Medical School (School of Basic Medical Sciences)Wuhan430071China
- TaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430071China
| |
Collapse
|
3
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2024:1-95. [DOI: 10.1016/b978-0-7020-8228-3.00001-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Lotto J, Stephan TL, Hoodless PA. Fetal liver development and implications for liver disease pathogenesis. Nat Rev Gastroenterol Hepatol 2023; 20:561-581. [PMID: 37208503 DOI: 10.1038/s41575-023-00775-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 05/21/2023]
Abstract
The metabolic, digestive and homeostatic roles of the liver are dependent on proper crosstalk and organization of hepatic cell lineages. These hepatic cell lineages are derived from their respective progenitors early in organogenesis in a spatiotemporally controlled manner, contributing to the liver's specialized and diverse microarchitecture. Advances in genomics, lineage tracing and microscopy have led to seminal discoveries in the past decade that have elucidated liver cell lineage hierarchies. In particular, single-cell genomics has enabled researchers to explore diversity within the liver, especially early in development when the application of bulk genomics was previously constrained due to the organ's small scale, resulting in low cell numbers. These discoveries have substantially advanced our understanding of cell differentiation trajectories, cell fate decisions, cell lineage plasticity and the signalling microenvironment underlying the formation of the liver. In addition, they have provided insights into the pathogenesis of liver disease and cancer, in which developmental processes participate in disease emergence and regeneration. Future work will focus on the translation of this knowledge to optimize in vitro models of liver development and fine-tune regenerative medicine strategies to treat liver disease. In this Review, we discuss the emergence of hepatic parenchymal and non-parenchymal cells, advances that have been made in in vitro modelling of liver development and draw parallels between developmental and pathological processes.
Collapse
Affiliation(s)
- Jeremy Lotto
- Terry Fox Laboratory, BC Cancer, Vancouver, BC, Canada
- Cell and Developmental Biology Program, University of British Columbia, Vancouver, BC, Canada
| | - Tabea L Stephan
- Terry Fox Laboratory, BC Cancer, Vancouver, BC, Canada
- Cell and Developmental Biology Program, University of British Columbia, Vancouver, BC, Canada
| | - Pamela A Hoodless
- Terry Fox Laboratory, BC Cancer, Vancouver, BC, Canada.
- Cell and Developmental Biology Program, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
5
|
Romanzi A, Villa E. Angiogenesis: the Yin and Yang in intrahepatic cholangiocarcinoma. HEPATOMA RESEARCH 2023. [DOI: 10.20517/2394-5079.2023.53] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
The tumor microenvironment (TME) constitutes a complex structure comprising different cell types and soluble factors that surround the tumor and promote its progression. Primarily for its pivotal role in malignant growth, TME has become a potential therapeutic objective for developing new targeted therapy and a marker for assessing therapeutic response. In intrahepatic cholangiocarcinoma (iCCA), the second most common primary liver malignancy, TME has also gained a central role in understanding the mechanisms underlying tumor progression. In this review, we focused on the role of angiogenic factors and their pathway in iCCA and analyzed possible therapeutic and prognostic implications.
Collapse
|
6
|
Gannoun L, De Schrevel C, Belle M, Dauguet N, Achouri Y, Loriot A, Vanderaa C, Cordi S, Dili A, Heremans Y, Rooman I, Leclercq IA, Jacquemin P, Gatto L, Lemaigre FP. Axon guidance genes control hepatic artery development. Development 2023; 150:dev201642. [PMID: 37497580 DOI: 10.1242/dev.201642] [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: 02/03/2023] [Accepted: 07/19/2023] [Indexed: 07/28/2023]
Abstract
Earlier data on liver development demonstrated that morphogenesis of the bile duct, portal mesenchyme and hepatic artery is interdependent, yet how this interdependency is orchestrated remains unknown. Here, using 2D and 3D imaging, we first describe how portal mesenchymal cells become organised to form hepatic arteries. Next, we examined intercellular signalling active during portal area development and found that axon guidance genes are dynamically expressed in developing bile ducts and portal mesenchyme. Using tissue-specific gene inactivation in mice, we show that the repulsive guidance molecule BMP co-receptor A (RGMA)/neogenin (NEO1) receptor/ligand pair is dispensable for portal area development, but that deficient roundabout 2 (ROBO2)/SLIT2 signalling in the portal mesenchyme causes reduced maturation of the vascular smooth muscle cells that form the tunica media of the hepatic artery. This arterial anomaly does not impact liver function in homeostatic conditions, but is associated with significant tissular damage following partial hepatectomy. In conclusion, our work identifies new players in development of the liver vasculature in health and liver regeneration.
Collapse
Affiliation(s)
- Lila Gannoun
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| | - Catalina De Schrevel
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| | - Morgane Belle
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Department of Development, Rue Moreau 17, Paris 75012, France
| | - Nicolas Dauguet
- Flow cytometry CYTF platform, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Younes Achouri
- Transgene Technology Platform TRSG, Université Catholique de Louvain, Brussels, Avenue Hippocrate 75, Belgium 1200
| | - Axelle Loriot
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| | - Christophe Vanderaa
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| | - Sabine Cordi
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| | - Alexandra Dili
- HPB Surgery Unit, Centre Hospitalier Universitaire UCL Namur, Site Mont-Godinne, Avenue du Dr. Thérasse 1, Yvoir 5530, Belgium
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Avenue Mounier 53, Brussels 1200, Belgium
| | - Yves Heremans
- Visual & Spatial Tissue Analysis (VSTA) core facility, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels 1090, Belgium
| | - Ilse Rooman
- Laboratory of Medical and Molecular Oncology, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels 1090, Belgium
| | - Isabelle A Leclercq
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Avenue Mounier 53, Brussels 1200, Belgium
| | - Patrick Jacquemin
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| | - Laurent Gatto
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| | - Frédéric P Lemaigre
- de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Brussels 1200, Belgium
| |
Collapse
|
7
|
Koike N, Tadokoro T, Ueno Y, Okamoto S, Kobayashi T, Murata S, Taniguchi H. Development of the nervous system in mouse liver. World J Hepatol 2022; 14:386-399. [PMID: 35317173 PMCID: PMC8891673 DOI: 10.4254/wjh.v14.i2.386] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/12/2021] [Accepted: 01/20/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The role of the hepatic nervous system in liver development remains unclear. We previously created functional human micro-hepatic tissue in mice by co-culturing human hepatic endodermal cells with endothelial and mesenchymal cells. However, they lacked Glisson’s sheath [the portal tract (PT)]. The PT consists of branches of the hepatic artery (HA), portal vein, and intrahepatic bile duct (IHBD), collectively called the portal triad, together with autonomic nerves.
AIM To evaluate the development of the mouse hepatic nervous network in the PT using immunohistochemistry.
METHODS Liver samples from C57BL/6J mice were harvested at different developmental time periods, from embryonic day (E) 10.5 to postnatal day (P) 56. Thin sections of the surface cut through the hepatic hilus were examined using protein gene product 9.5 (PGP9.5) and cytokeratin 19 (CK19) antibodies, markers of nerve fibers (NFs), and biliary epithelial cells (BECs), respectively. The numbers of NFs and IHBDs were separately counted in a PT around the hepatic hilus (center) and the peripheral area (periphery) of the liver, comparing the average values between the center and the periphery at each developmental stage. NF-IHBD and NF-HA contacts in a PT were counted, and their relationship was quantified. SRY-related high mobility group-box gene 9 (SOX9), another BEC marker; hepatocyte nuclear factor 4α (HNF4α), a marker of hepatocytes; and Jagged-1, a Notch ligand, were also immunostained to observe the PT development.
RESULTS HNF4α was expressed in the nucleus, and Jagged-1 was diffusely positive in the primitive liver at E10.5; however, the PGP9.5 and CK19 were negative in the fetal liver. SOX9-positive cells were scattered in the periportal area in the liver at E12.5. The Jagged-1 was mainly expressed in the periportal tissue, and the number of SOX9-positive cells increased at E16.5. SOX9-positive cells constructed the ductal plate and primitive IHBDs mainly at the center, and SOX-9-positive IHBDs partly acquired CK19 positivity at the same period. PGP9.5-positive bodies were first found at E16.5 and HAs were first found at P0 in the periportal tissue of the center. Therefore, primitive PT structures were first constructed at P0 in the center. Along with remodeling of the periportal tissue, the number of CK19-positive IHBDs and PGP9.5-positive NFs gradually increased, and PTs were also formed in the periphery until P5. The numbers of NFs and IHBDs were significantly higher in the center than in the periphery from E16.5 to P5. The numbers of NFs and IHBDs reached the adult level at P28, with decreased differences between the center and periphery. NFs associated more frequently with HAs than IHBDs in PTs at the early phase after birth, after which the number of NF-IHBD contacts gradually increased.
CONCLUSION Mouse hepatic NFs first emerge at the center just before birth and extend toward the periphery. The interaction between NFs and IHBDs or HAs plays important roles in the morphogenesis of PT structure.
Collapse
Affiliation(s)
- Naoto Koike
- Department of Surgery, Seirei Sakura Citizen Hospital, Sakura 285-8765, Chiba, Japan
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Kanagawa, Japan
| | - Tomomi Tadokoro
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Kanagawa, Japan
| | - Yasuharu Ueno
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Kanagawa, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Satoshi Okamoto
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Kanagawa, Japan
| | - Tatsuya Kobayashi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Kanagawa, Japan
| | - Soichiro Murata
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Kanagawa, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Kanagawa, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
8
|
Abstract
Yes-associated protein 1 (YAP1) is a transcriptional coactivator that activates transcriptional enhanced associate domain transcription factors upon inactivation of the Hippo signaling pathway, to regulate biological processes like proliferation, survival, and differentiation. YAP1 is most prominently expressed in biliary epithelial cells (BECs) in normal adult livers and during development. In the current review, we will discuss the multiple roles of YAP1 in the development and morphogenesis of bile ducts inside and outside the liver, as well as in orchestrating the cholangiocyte repair response to biliary injury. We will review how biliary repair can occur through the process of hepatocyte-to-BEC transdifferentiation and how YAP1 is pertinent to this process. We will also discuss the liver's capacity for metabolic reprogramming as an adaptive mechanism in extreme cholestasis, such as when intrahepatic bile ducts are absent due to YAP1 loss from hepatic progenitors. Finally, we will discuss the roles of YAP1 in the context of pediatric pathologies afflicting bile ducts, such as Alagille syndrome and biliary atresia. In conclusion, we will comprehensively discuss the spatiotemporal roles of YAP1 in biliary development and repair after biliary injury while describing key interactions with other well-known developmental pathways.
Collapse
Affiliation(s)
- Laura Molina
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine
| | - Kari Nejak-Bowen
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine,Pittsburgh Liver Research Center, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Satdarshan P. Monga
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine,Pittsburgh Liver Research Center, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania,Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh and UPMC, Pittsburgh, Pennsylvania
| |
Collapse
|
9
|
Wu LN, Zhu ZJ, Sun LY. Genetic Factors and Their Role in the Pathogenesis of Biliary Atresia. Front Pediatr 2022; 10:912154. [PMID: 35844731 PMCID: PMC9277099 DOI: 10.3389/fped.2022.912154] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/08/2022] [Indexed: 12/12/2022] Open
Abstract
Biliary Atresia, a common basis for neonatal cholestasis and primary indication for Liver Transplantation, accounts for 60% of pediatric Liver Transplantations. While the pathogenesis of Biliary Atresia remains obscure, abnormalities within bile ducts and the liver, inflammation, fibrosis and cilia defects are thought to comprise the pathological basis for this condition. The findings of genetic variants in Biliary Atresia, such as Copy Number Variations and Single Nucleotide Polymorphism, are considered as essential factors in the development of this condition. In this review, we summarize and analyze these Biliary Atresia variants from a perspective of their pathological characteristics. In conclusion, such analyses may offer novel insights into the pathogenesis of Biliary Atresia and provide a foundation for future studies directed toward a better understanding and treatment of Biliary Atresia.
Collapse
Affiliation(s)
- Li-Na Wu
- Department of Critical Liver Diseases, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
| | - Zhi-Jun Zhu
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
| | - Li-Ying Sun
- Department of Critical Liver Diseases, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Baiocchi L, Glaser S, Francis H, Kennedy L, Felli E, Alpini G, Gracia‐Sancho J. Impact of Aging on Liver Cells and Liver Disease: Focus on the Biliary and Vascular Compartments. Hepatol Commun 2021; 5:1125-1137. [PMID: 34278165 PMCID: PMC8279468 DOI: 10.1002/hep4.1725] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/26/2021] [Accepted: 03/16/2021] [Indexed: 12/11/2022] Open
Abstract
The aging process is represented by the time-dependent decay in physiologic functions of living beings. Major interest has been focused in recent years on the determinants of this progressive condition due to its correlative relationship with the onset of diseases. Several hallmark features have been observed in aging, such as genetic alterations, mitochondrial impairment, and telomere shortening. At the cellular level, a senescent phenotype has been identified in response to aging that is characterized by a flat appearance, proliferative arrest, and production of specific molecules. The net effect of these cells in the course of diseases is an argument of debate. In fact, while the onset of a senescent phenotype may prevent tumor spreading, these cells appear to support pathological processes in some conditions. Several studies are now focused on clarifying the specific molecular pathways of aging/senescence in different cells, tissues, or organs. Biliary and vascular components, within the liver, have emerged as important determinants of some form of liver disease. In this review we summarize the most recent achievements on aging/senescence, focusing on the biliary and vascular liver system. Conclusion: Several findings, in both preclinical animal models and on human liver specimens, converge in supporting the presence of specific aging hallmarks in the diseases involving these hepatic compartments.
Collapse
Affiliation(s)
- Leonardo Baiocchi
- Hepatology UnitDepartment of MedicineUniversity of Tor VergataRomeItaly
| | - Shannon Glaser
- Medical PhysiologyTexas A&M College of MedicineBryanTXUSA
| | - Heather Francis
- Hepatology and MedicineIndiana UniversityIndianapolisINUSA
- Richard L. Roudebush VA Medical CenterIndianapolisINUSA
| | - Lindsey Kennedy
- Hepatology and MedicineIndiana UniversityIndianapolisINUSA
- Richard L. Roudebush VA Medical CenterIndianapolisINUSA
| | - Eric Felli
- HepatologyDepartment of Biomedical ResearchInselspitalBernSwitzerland
| | - Gianfranco Alpini
- Hepatology and MedicineIndiana UniversityIndianapolisINUSA
- Richard L. Roudebush VA Medical CenterIndianapolisINUSA
| | - Jordi Gracia‐Sancho
- Liver Vascular BiologyIDIBAPS Biomedical Research Institute and CIBEREHDBarcelonaSpain
- HepatologyDepartment of Biomedical ResearchInselspitalBernSwitzerland
| |
Collapse
|
11
|
Engineering the Vasculature of Stem-Cell-Derived Liver Organoids. Biomolecules 2021; 11:biom11070966. [PMID: 34208902 PMCID: PMC8301828 DOI: 10.3390/biom11070966] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 12/28/2022] Open
Abstract
The vasculature of stem-cell-derived liver organoids can be engineered using methods that recapitulate embryonic liver development. Hepatic organoids with a vascular network offer great application prospects for drug screening, disease modeling, and therapeutics. However, the application of stem cell-derived organoids is hindered by insufficient vascularization and maturation. Here, we review different theories about the origin of hepatic cells and the morphogenesis of hepatic vessels to provide potential approaches for organoid generation. We also review the main protocols for generating vascularized liver organoids from stem cells and consider their potential and limitations in the generation of vascularized liver organoids.
Collapse
|
12
|
Mariotti V, Fiorotto R, Cadamuro M, Fabris L, Strazzabosco M. New insights on the role of vascular endothelial growth factor in biliary pathophysiology. JHEP Rep 2021; 3:100251. [PMID: 34151244 PMCID: PMC8189933 DOI: 10.1016/j.jhepr.2021.100251] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
The family of vascular endothelial growth factors (VEGFs) includes 5 members (VEGF-A to -D, and placenta growth factor), which regulate several critical biological processes. VEGF-A exerts a variety of biological effects through high-affinity binding to tyrosine kinase receptors (VEGFR-1, -2 and -3), co-receptors and accessory proteins. In addition to its fundamental function in angiogenesis and endothelial cell biology, VEGF/VEGFR signalling also plays a role in other cell types including epithelial cells. This review provides an overview of VEGF signalling in biliary epithelial cell biology in both normal and pathologic conditions. VEGF/VEGFR-2 signalling stimulates bile duct proliferation in an autocrine and paracrine fashion. VEGF/VEGFR-1/VEGFR-2 and angiopoietins are involved at different stages of biliary development. In certain conditions, cholangiocytes maintain the ability to secrete VEGF-A, and to express a functional VEGFR-2 receptor. For example, in polycystic liver disease, VEGF secreted by cystic cells stimulates cyst growth and vascular remodelling through a PKA/RAS/ERK/HIF1α-dependent mechanism, unveiling a new level of complexity in VEFG/VEGFR-2 regulation in epithelial cells. VEGF/VEGFR-2 signalling is also reactivated during the liver repair process. In this context, pro-angiogenic factors mediate the interactions between epithelial, mesenchymal and inflammatory cells. This process takes place during the wound healing response, however, in chronic biliary diseases, it may lead to pathological neo-angiogenesis, a condition strictly linked with fibrosis progression, the development of cirrhosis and related complications, and cholangiocarcinoma. Novel observations indicate that in cholangiocarcinoma, VEGF is a determinant of lymphangiogenesis and of the immune response to the tumour. Better insights into the role of VEGF signalling in biliary pathophysiology might help in the search for effective therapeutic strategies.
Collapse
Key Words
- ADPKD, adult dominant polycystic kidney disease
- Anti-Angiogenic therapy
- BA, biliary atresia
- BDL, bile duct ligation
- CCA, cholangiocarcinoma
- CCl4, carbon tetrachloride
- CLDs, chronic liver diseases
- Cholangiocytes
- Cholangiopathies
- DP, ductal plate
- DPM, ductal plate malformation
- DRCs, ductular reactive cells
- Development
- HIF-1α, hypoxia-inducible factor type 1α
- HSCs, hepatic stellate cells
- IHBD, intrahepatic bile ducts
- IL-, interleukin-
- LECs, lymphatic endothelial cells
- LSECs, liver sinusoidal endothelial cells
- Liver repair
- MMPs, matrix metalloproteinases
- PBP, peribiliary plexus
- PC, polycystin
- PDGF, platelet-derived growth factor
- PIGF, placental growth factor
- PLD, polycystic liver diseases
- Polycystic liver diseases
- SASP, senescence-associated secretory phenotype
- TGF, transforming growth factor
- VEGF, vascular endothelial growth factors
- VEGF-A
- VEGF/VEGFR-2 signalling
- VEGFR-1/2, vascular endothelial growth factor receptor 1/2
- mTOR, mammalian target of rapamycin
Collapse
Affiliation(s)
- Valeria Mariotti
- Section of Digestive Diseases, Liver Center, Yale University, New Haven, CT, USA
| | - Romina Fiorotto
- Section of Digestive Diseases, Liver Center, Yale University, New Haven, CT, USA
| | - Massimiliano Cadamuro
- Department of Molecular Medicine, University of Padua, School of Medicine, Padua, Italy
| | - Luca Fabris
- Section of Digestive Diseases, Liver Center, Yale University, New Haven, CT, USA.,Department of Molecular Medicine, University of Padua, School of Medicine, Padua, Italy
| | - Mario Strazzabosco
- Section of Digestive Diseases, Liver Center, Yale University, New Haven, CT, USA
| |
Collapse
|
13
|
Tassinari R, Narciso L, Tait S, Busani L, Martinelli A, Di Virgilio A, Carli F, Deodati A, La Rocca C, Maranghi F. Juvenile Toxicity Rodent Model to Study Toxicological Effects of Bisphenol A (BPA) at Dose Levels Derived From Italian Children Biomonitoring Study. Toxicol Sci 2020; 173:387-401. [PMID: 31697385 DOI: 10.1093/toxsci/kfz226] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Bisphenol A (BPA) is a plasticizer with endocrine disrupting properties particularly relevant for children health. Recently BPA has been associated with metabolic dysfunctions but no data are yet available in specific, long-term studies. This study aimed to evaluate BPA modes of action and hazards during animal juvenile life-stage, corresponding to childhood. Immature Sprague-Dawley rats of both sexes were orally treated with 0 (vehicle only-olive oil), 2, 6, and 18 mg/kg bw per day of BPA for 28 days, from weaning to sexual maturity. Dose levels were obtained from the PERSUADED biomonitoring study in Italian children. Both no-observed-adverse-effect-level (NOAEL)/low-observed-adverse-effect-level (LOAEL) and estimated benchmark dose (BMD) approaches were applied. General toxicity, parameters of sexual development, endocrine/reproductive/functional liver and kidney biomarkers, histopathology of target tissues, and gene expression in hypothalamic-pituitary area and liver were studied. No mortality or general toxicity occurred. Sex-specific alterations were observed in liver, thyroid, spleen, leptin/adiponectin serum levels, and hypothalamic-pituitary gene expression. Thyroid homeostasis and liver were the most sensitive targets of BPA exposure in the peripubertal phase. The proposed LOAEL was 2 mg/kg bw, considering as critical effect the liver endpoints, kidney weight in male and adrenal histomorphometrical alterations and osteopontin upregulation in female rats. The BMD lower bounds were 0.05 and 1.33 mg/kg bw in males and females, considering liver and thyroid biomarkers, respectively. Overall, BPA evaluation at dose levels derived from children biomonitoring study allowed to identify sex-specific, targeted toxicological effects that may have significant impact on risk assessment for children.
Collapse
Affiliation(s)
| | | | | | | | - Andrea Martinelli
- Experimental Animal Welfare Sector, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Antonio Di Virgilio
- Experimental Animal Welfare Sector, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Fabrizia Carli
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Annalisa Deodati
- Dipartimento Pediatrico Universitario Ospedaliero "Bambino Gesù".,Children's Hospital-Tor Vergata University, Rome, Italy
| | | | | | | |
Collapse
|
14
|
Fabris L, Fiorotto R, Spirli C, Cadamuro M, Mariotti V, Perugorria MJ, Banales JM, Strazzabosco M. Pathobiology of inherited biliary diseases: a roadmap to understand acquired liver diseases. Nat Rev Gastroenterol Hepatol 2019; 16:497-511. [PMID: 31165788 PMCID: PMC6661007 DOI: 10.1038/s41575-019-0156-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bile duct epithelial cells, also known as cholangiocytes, regulate the composition of bile and its flow. Acquired, congenital and genetic dysfunctions in these cells give rise to a set of diverse and complex diseases, often of unknown aetiology, called cholangiopathies. New knowledge has been steadily acquired about genetic and congenital cholangiopathies, and this has led to a better understanding of the mechanisms of acquired cholangiopathies. This Review focuses on findings from studies on Alagille syndrome, polycystic liver diseases, fibropolycystic liver diseases (Caroli disease and congenital hepatic fibrosis) and cystic fibrosis-related liver disease. In particular, knowledge on the role of Notch signalling in biliary repair and tubulogenesis has been advanced by work on Alagille syndrome, and investigations in polycystic liver diseases have highlighted the role of primary cilia in biliary pathophysiology and the concept of biliary angiogenic signalling and its role in cyst growth and biliary repair. In fibropolycystic liver disease, research has shown that loss of fibrocystin generates a signalling cascade that increases β-catenin signalling, activates the NOD-, LRR- and pyrin domain-containing 3 inflammasome, and promotes production of IL-1β and other chemokines that attract macrophages and orchestrate the process of pericystic and portal fibrosis, which are the main mechanisms of progression in cholangiopathies. In cystic fibrosis-related liver disease, lack of cystic fibrosis transmembrane conductance regulator increases the sensitivity of epithelial Toll-like receptor 4 that sustains the secretion of nuclear factor-κB-dependent cytokines and peribiliary inflammation in response to gut-derived products, providing a model for primary sclerosing cholangitis. These signalling mechanisms may be targeted therapeutically and they offer a possibility for the development of novel treatments for acquired cholangiopathies.
Collapse
Affiliation(s)
- Luca Fabris
- Liver Center, Department of Medicine, Yale University, New Haven, CT, USA
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Romina Fiorotto
- Liver Center, Department of Medicine, Yale University, New Haven, CT, USA
| | - Carlo Spirli
- Liver Center, Department of Medicine, Yale University, New Haven, CT, USA
| | | | - Valeria Mariotti
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Maria J Perugorria
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III), Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III), Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Mario Strazzabosco
- Liver Center, Department of Medicine, Yale University, New Haven, CT, USA.
- Department of Molecular Medicine, University of Padova, Padova, Italy.
| |
Collapse
|
15
|
Franchitto A, Overi D, Mancinelli R, Mitterhofer AP, Muiesan P, Tinti F, Umbro I, Hubscher SG, Onori P, Gaudio E, Carpino G. Peribiliary gland damage due to liver transplantation involves peribiliary vascular plexus and vascular endothelial growth factor. Eur J Histochem 2019; 63:3022. [PMID: 31113191 PMCID: PMC6517787 DOI: 10.4081/ejh.2019.3022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
Extrahepatic bile ducts are characterized by the presence of peribiliary glands (PBGs), which represent stem cell niches implicated in biliary regeneration. Orthotopic liver transplantation may be complicated by non-anastomotic strictures (NAS) of the bile ducts, which have been associated with ischemic injury of PBGs and occur more frequently in livers obtained from donors after circulatory death than in those from brain-dead donors. The aims of the present study were to investigate the PBG phenotype in bile ducts after transplantation, the integrity of the peribiliary vascular plexus (PVP) around PBGs, and the expression of vascular endothelial growth factor-A (VEGF-A) by PBGs. Transplanted ducts obtained from patients who underwent liver transplantation were studied (N=62). Controls included explanted bile duct samples not used for transplantation (N=10) with normal histology. Samples were processed for histology, immunohistochemistry and immunofluorescence. Surface epithelium is severely injured in transplanted ducts; PBGs are diffusely damaged, particularly in ducts obtained from circulatory-dead compared to brain-dead donors. PVP is reduced in transplanted compared to controls. PBGs in transplanted ducts contain more numerous progenitor and proliferating cells compared to controls, show higher positivity for VEGF-A compared to controls, and express VEGF receptor-2. In conclusion, PBGs and associated PVP are damaged in transplanted extrahepatic bile ducts; however, an activation of the PBG niche takes place and is characterized by proliferation and VEGF-A expression. This response could have a relevant role in reconstituting biliary epithelium and vascular plexus and could be implicated in the genesis of non-anastomotic strictures.
Collapse
Affiliation(s)
- Antonio Franchitto
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Radhakrishnan H, Walther W, Zincke F, Kobelt D, Imbastari F, Erdem M, Kortüm B, Dahlmann M, Stein U. MACC1-the first decade of a key metastasis molecule from gene discovery to clinical translation. Cancer Metastasis Rev 2019; 37:805-820. [PMID: 30607625 DOI: 10.1007/s10555-018-9771-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Deciphering the paths to metastasis and identifying key molecules driving this process is one important issue for understanding and treatment of cancer. Such a key driver molecule is Metastasis Associated in Colon Cancer 1 (MACC1). A decade long research on this evolutionarily conserved molecule with features of a transcription factor as well as an adapter protein for versatile protein-protein interactions has shown that it has manifold properties driving tumors to their metastatic stage. MACC1 transcriptionally regulates genes involved in epithelial-mesenchymal transition (EMT), including those which are able to directly induce metastasis like c-MET, impacts tumor cell migration and invasion, and induces metastasis in solid cancers. MACC1 has proven as a valuable biomarker for prognosis of metastasis formation linked to patient survival and gives promise to also act as a predictive marker for individualized therapies in a broad variety of cancers. This review discusses the many features of MACC1 in the context of the hallmarks of cancer and the potential of this molecule as biomarker and novel therapeutic target for restriction and prevention of metastasis.
Collapse
Affiliation(s)
- Harikrishnan Radhakrishnan
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Wolfgang Walther
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Fabian Zincke
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Dennis Kobelt
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Francesca Imbastari
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Müge Erdem
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Benedikt Kortüm
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Mathias Dahlmann
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany. .,German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
17
|
Blau BJ, Miki T. The role of cellular interactions in the induction of hepatocyte polarity and functional maturation in stem cell-derived hepatic cells. Differentiation 2019; 106:42-48. [PMID: 30878880 DOI: 10.1016/j.diff.2019.02.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 02/08/2023]
Abstract
The unique microenvironment found within the liver in vivo plays a key role in the induction of functional maturation in the developing hepatocyte. During organogenesis, hepatocytes acquire a polar phenotype that allows them to perform their functions of bile production and transport, protein synthesis, metabolism, and detoxification simultaneously, independently, and efficiently. It is thought that the induction of polarity and functional maturation in hepatocytes is dependent on the complex interplay of cell-cell and cell-extracellular matrix (ECM) interactions. While this process is highly efficient in the human liver, it has been shown that hepatocytes rapidly lose their functions when placed in cell culture. This poses a challenge for the development of a bioartificial liver (BAL) support system, which utilizes a live cellular source to perform hepatic functions in the event of acute liver failure or primary nonfunction. However, once the molecular mechanisms underlying the induction of hepatocyte polarity are fully identified, it will be possible to develop highly functional hepatic cells from human pluripotent stem cells (hPSCs). This new cell line would be an ideal cellular source for a BAL system, as it would have both the functionality and longevity to support a patient through the entire clinical course of treatment. In this review, we explore the literature that has examined the potential mechanisms that induce polarity in the developing hepatocyte and discuss the future implications of this knowledge in a clinical setting from a bioengineering perspective.
Collapse
Affiliation(s)
- Brandon J Blau
- Department of Surgery, Keck School of Medicine, University of Southern California, USA
| | - Toshio Miki
- Department of Surgery, Keck School of Medicine, University of Southern California, USA.
| |
Collapse
|
18
|
Fibroinflammatory Liver Injuries as Preneoplastic Condition in Cholangiopathies. Int J Mol Sci 2018; 19:ijms19123875. [PMID: 30518128 PMCID: PMC6321547 DOI: 10.3390/ijms19123875] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 11/29/2018] [Accepted: 12/01/2018] [Indexed: 02/08/2023] Open
Abstract
The cholangipathies are a class of liver diseases that specifically affects the biliary tree. These pathologies may have different etiologies (genetic, autoimmune, viral, or toxic) but all of them are characterized by a stark inflammatory infiltrate, increasing overtime, accompanied by an excess of periportal fibrosis. The cellular types that mount the regenerative/reparative hepatic response to the damage belong to different lineages, including cholagiocytes, mesenchymal and inflammatory cells, which dynamically interact with each other, exchanging different signals acting in autocrine and paracrine fashion. Those messengers may be proinflammatory cytokines and profibrotic chemokines (IL-1, and 6; CXCL1, 10 and 12, or MCP-1), morphogens (Notch, Hedgehog, and WNT/β-catenin signal pathways) and finally growth factors (VEGF, PDGF, and TGFβ, among others). In this review we will focus on the main molecular mechanisms mediating the establishment of a fibroinflammatory liver response that, if perpetuated, can lead not only to organ dysfunction but also to neoplastic transformation. Primary Sclerosing Cholangitis and Congenital Hepatic Fibrosis/Caroli’s disease, two chronic cholangiopathies, known to be prodrome of cholangiocarcinoma, for which several murine models are also available, were also used to further dissect the mechanisms of fibroinflammation leading to tumor development.
Collapse
|
19
|
Fiorotto R, Amenduni M, Mariotti V, Fabris L, Spirli C, Strazzabosco M. Liver diseases in the dish: iPSC and organoids as a new approach to modeling liver diseases. Biochim Biophys Acta Mol Basis Dis 2018; 1865:920-928. [PMID: 30264693 DOI: 10.1016/j.bbadis.2018.08.038] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/10/2018] [Accepted: 08/28/2018] [Indexed: 02/07/2023]
Abstract
Liver diseases negatively impact the quality of life and survival of patients, and often require liver transplantation in cases that progress to organ failure. Understanding the cellular and molecular mechanisms of liver development and pathogenesis has been a challenging task, in part for the lack of adequate cellular models directly relevant to the human diseases. Recent technological advances in the stem cell field have shown the potentiality of induced pluripotent stem cells (iPSC) and liver organoids as the next generation tool to model in vitro liver diseases. Hepatocyte-like cells and cholangiocyte are currently being generated from skin fibroblasts and mononuclear blood cells reprogrammed into iPSC and have been successfully used for disease modeling, drug testing and gene editing, with the hope to be able to find application also in regenerative medicine. Protocols to generate other liver cell types are still under development, but the field is advancing rapidly. On the other end, liver cells can now be isolated from liver specimens (liver explants or liver biopsies) and cultured in specific conditions to form polarized 3D organoids. The purpose of this review is to summarize all these recent technological advances and their potential applications but also to analyze the current issues to be addressed before the technology can reach its full potential.
Collapse
Affiliation(s)
- Romina Fiorotto
- Digestive Disease Section, Yale Liver Center, Yale University School of Medicine, New Haven, CT, (USA)
| | - Mariangela Amenduni
- Digestive Disease Section, Yale Liver Center, Yale University School of Medicine, New Haven, CT, (USA)
| | - Valeria Mariotti
- Department of Molecular Medicine, University of Padova School of Medicine, Padova, Italy
| | - Luca Fabris
- Department of Molecular Medicine, University of Padova School of Medicine, Padova, Italy
| | - Carlo Spirli
- Digestive Disease Section, Yale Liver Center, Yale University School of Medicine, New Haven, CT, (USA)
| | - Mario Strazzabosco
- Digestive Disease Section, Yale Liver Center, Yale University School of Medicine, New Haven, CT, (USA).
| |
Collapse
|
20
|
Tam PKH, Yiu RS, Lendahl U, Andersson ER. Cholangiopathies - Towards a molecular understanding. EBioMedicine 2018; 35:381-393. [PMID: 30236451 PMCID: PMC6161480 DOI: 10.1016/j.ebiom.2018.08.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/06/2018] [Accepted: 08/09/2018] [Indexed: 12/14/2022] Open
Abstract
Liver diseases constitute an important medical problem, and a number of these diseases, termed cholangiopathies, affect the biliary system of the liver. In this review, we describe the current understanding of the causes of cholangiopathies, which can be genetic, viral or environmental, and the few treatment options that are currently available beyond liver transplantation. We then discuss recent rapid progress in a number of areas relevant for decoding the disease mechanisms for cholangiopathies. This includes novel data from analysis of transgenic mouse models and organoid systems, and we outline how this information can be used for disease modeling and potential development of novel therapy concepts. We also describe recent advances in genomic and transcriptomic analyses and the importance of such studies for improving diagnosis and determining whether certain cholangiopathies should be viewed as distinct or overlapping disease entities.
Collapse
Affiliation(s)
- Paul K H Tam
- Department of Surgery, Li Ka Shing Faculty of Medicine, and Dr. Li Dak-Sum Research Centre, The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, and The University of Hong Kong, Hong Kong.
| | - Rachel S Yiu
- Department of Surgery, Li Ka Shing Faculty of Medicine, and Dr. Li Dak-Sum Research Centre, The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, and The University of Hong Kong, Hong Kong
| | - Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Emma R Andersson
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden.
| |
Collapse
|
21
|
Ober EA, Lemaigre FP. Development of the liver: Insights into organ and tissue morphogenesis. J Hepatol 2018; 68:1049-1062. [PMID: 29339113 DOI: 10.1016/j.jhep.2018.01.005] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 12/29/2017] [Accepted: 01/06/2018] [Indexed: 02/08/2023]
Abstract
Recent development of improved tools and methods to analyse tissues at the three-dimensional level has expanded our capacity to investigate morphogenesis of foetal liver. Here, we review the key morphogenetic steps during liver development, from the prehepatic endoderm stage to the postnatal period, and consider several model organisms while focussing on the mammalian liver. We first discuss how the liver buds out of the endoderm and gives rise to an asymmetric liver. We next outline the mechanisms driving liver and lobe growth, and review morphogenesis of the intra- and extrahepatic bile ducts; morphogenetic responses of the biliary tract to liver injury are discussed. Finally, we describe the mechanisms driving formation of the vasculature, namely venous and arterial vessels, as well as sinusoids.
Collapse
Affiliation(s)
- Elke A Ober
- Novo Nordisk Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
22
|
Vyas D, Baptista PM, Brovold M, Moran E, Brovold M, Gaston B, Booth C, Samuel M, Atala A, Soker S. Self-assembled liver organoids recapitulate hepatobiliary organogenesis in vitro. Hepatology 2018; 67:750-761. [PMID: 28834615 PMCID: PMC5825235 DOI: 10.1002/hep.29483] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 08/01/2017] [Accepted: 08/16/2017] [Indexed: 12/17/2022]
Abstract
Several three-dimensional cell culture systems are currently available to create liver organoids. In gneral, these systems display better physiologic and metabolic aspects of intact liver tissue compared with two-dimensional culture systems. However, none reliably mimic human liver development, including parallel formation of hepatocyte and cholangiocyte anatomical structures. Here, we show that human fetal liver progenitor cells self-assembled inside acellular liver extracellular matrix scaffolds to form three-dimensional liver organoids that recapitulated several aspects of hepatobiliary organogenesis and resulted in concomitant formation of progressively more differentiated hepatocytes and bile duct structures. The duct morphogenesis process was interrupted by inhibiting Notch signaling, in an attempt to create a liver developmental disease model with a similar phenotype to Alagille syndrome. Conclusion: In the current study, we created an in vitro model of human liver development and disease, physiology, and metabolism, supported by liver extracellular matrix substrata; we envision that it will be used in the future to study mechanisms of hepatic and biliary development and for disease modeling and drug screening. (Hepatology 2018;67:750-761).
Collapse
Affiliation(s)
- Dipen Vyas
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Pedro M. Baptista
- Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain,CIBERehd, Spain,Corresponding Authors: Shay Soker, Wake Forest Institute for Regenerative Medicine, 391 Technology Way, Winston Salem, NC 27101. Phone: 336-713-7295; Fax: 336-713-7290. ; Pedro M. Baptista, Aragon’s Health Research Institute (IIS Aragon), Zaragoza, Spain.
| | - Matthew Brovold
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Emma Moran
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Matthew Brovold
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Brandon Gaston
- Department of Surgery, University of California, San Francisco, CA, USA
| | - Chris Booth
- John Hopkins Medical Institute, Baltimore, MD, USA
| | - Michael Samuel
- Mass Spectrometry Core Facility, Lipid Sciences Department, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Health, Winston-Salem, NC, USA,Corresponding Authors: Shay Soker, Wake Forest Institute for Regenerative Medicine, 391 Technology Way, Winston Salem, NC 27101. Phone: 336-713-7295; Fax: 336-713-7290. ; Pedro M. Baptista, Aragon’s Health Research Institute (IIS Aragon), Zaragoza, Spain.
| |
Collapse
|
23
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function, and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2018:1-87. [DOI: 10.1016/b978-0-7020-6697-9.00001-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
24
|
Animal models of biliary injury and altered bile acid metabolism. Biochim Biophys Acta Mol Basis Dis 2017; 1864:1254-1261. [PMID: 28709963 DOI: 10.1016/j.bbadis.2017.06.027] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/28/2017] [Accepted: 06/30/2017] [Indexed: 12/14/2022]
Abstract
In the last 25years, a number of animal models, mainly rodents, have been generated with the goal to mimic cholestatic liver injuries and, thus, to provide in vivo tools to investigate the mechanisms of biliary repair and, eventually, to test the efficacy of innovative treatments. Despite fundamental limitations applying to these models, such as the distinct immune system and the different metabolism regulating liver homeostasis in rodents when compared to humans, multiple approaches, such as surgery (bile duct ligation), chemical-induced (3,5-diethoxycarbonyl-1,4-dihydrocollidine, DDC, α-naphthylisothiocyanate, ANIT), viral infections (Rhesus rotavirustype A, RRV-A), and genetic manipulation (Mdr2, Cftr, Pkd1, Pkd2, Prkcsh, Sec63, Pkhd1) have been developed. Overall, they have led to a range of liver phenotypes recapitulating the main features of biliary injury and altered bile acid metabolisms, such as ductular reaction, peribiliary inflammation and fibrosis, obstructive cholestasis and biliary dysgenesis. Although with a limited translability to the human setting, these mouse models have provided us with the ability to probe over time the fundamental mechanisms promoting cholestatic disease progression. Moreover, recent studies from genetically engineered mice have unveiled 'core' pathways that make the cholangiocyte a pivotal player in liver repair. In this review, we will highlight the main phenotypic features, the more interesting peculiarities and the different drawbacks of these mouse models. This article is part of a Special Issue entitled: Cholangiocytes in Health and Disease edited by Jesus Banales, Marco Marzioni, Nicholas LaRusso and Peter Jansen.
Collapse
|
25
|
Hall C, Sato K, Wu N, Zhou T, Kyritsi K, Meng F, Glaser S, Alpini G. Regulators of Cholangiocyte Proliferation. Gene Expr 2017; 17:155-171. [PMID: 27412505 PMCID: PMC5494439 DOI: 10.3727/105221616x692568] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cholangiocytes, a small population of cells within the normal liver, have been the focus of a significant amount of research over the past two decades because of their involvement in cholangiopathies such as primary sclerosing cholangitis and primary biliary cholangitis. This article summarizes landmark studies in the field of cholangiocyte physiology and aims to provide an updated review of biliary pathogenesis. The historical approach of rodent extrahepatic bile duct ligation and the relatively recent utilization of transgenic mice have led to significant discoveries in cholangiocyte pathophysiology. Cholangiocyte physiology is a complex system based on heterogeneity within the biliary tree and a number of signaling pathways that serve to regulate bile composition. Studies have expanded the list of neuropeptides, neurotransmitters, and hormones that have been shown to be key regulators of proliferation and biliary damage. The peptide histamine and hormones, such as melatonin and angiotensin, angiotensin, as well as numerous sex hormones, have been implicated in cholangiocyte proliferation during cholestasis. Numerous pathways promote cholangiocyte proliferation during cholestasis, and there is growing evidence to suggest that cholangiocyte proliferation may promote hepatic fibrosis. These pathways may represent significant therapeutic potential for a subset of cholestatic liver diseases that currently lack effective therapies.
Collapse
Affiliation(s)
- Chad Hall
- *Research, Central Texas Veterans Health Care System, Temple, TX, USA
- †Baylor Scott & White Digestive Disease Research Center, Temple, TX, USA
- ‡Department of Surgery, Baylor Scott & White and Texas A&M Health Science Center, Temple, TX, USA
| | - Keisaku Sato
- §Operational Funds, Baylor Scott & White, Temple, TX, USA
| | - Nan Wu
- §Operational Funds, Baylor Scott & White, Temple, TX, USA
| | - Tianhao Zhou
- §Operational Funds, Baylor Scott & White, Temple, TX, USA
| | | | - Fanyin Meng
- *Research, Central Texas Veterans Health Care System, Temple, TX, USA
- §Operational Funds, Baylor Scott & White, Temple, TX, USA
- ¶Department of Medicine, Baylor Scott & White and Texas A&M Health Science Center, Temple, TX, USA
| | - Shannon Glaser
- *Research, Central Texas Veterans Health Care System, Temple, TX, USA
- §Operational Funds, Baylor Scott & White, Temple, TX, USA
- ¶Department of Medicine, Baylor Scott & White and Texas A&M Health Science Center, Temple, TX, USA
| | - Gianfranco Alpini
- ‡Department of Surgery, Baylor Scott & White and Texas A&M Health Science Center, Temple, TX, USA
- §Operational Funds, Baylor Scott & White, Temple, TX, USA
- ¶Department of Medicine, Baylor Scott & White and Texas A&M Health Science Center, Temple, TX, USA
| |
Collapse
|
26
|
Spirli C, Villani A, Mariotti V, Fabris L, Fiorotto R, Strazzabosco M. Posttranslational regulation of polycystin-2 protein expression as a novel mechanism of cholangiocyte reaction and repair from biliary damage. Hepatology 2015; 62:1828-39. [PMID: 26313562 PMCID: PMC4681612 DOI: 10.1002/hep.28138] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 08/12/2015] [Indexed: 12/12/2022]
Abstract
UNLABELLED Polycystin-2 (PC2 or TRPPC2), a member of the transient receptor potential channel family, is a nonselective calcium channel. Mutations in PC2 are associated with polycystic liver diseases. PC2-defective cholangiocytes show increased production of cyclic adenosine monophosphate, protein kinase A-dependent activation of the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway, hypoxia-inducible factor 1α (HIF-1α)-mediated vascular endothelial growth factor (VEGF) production, and stimulation of cyst growth and progression. Activation of the ERK/HIF-1α/VEGF pathway in cholangiocytes plays a key role during repair from biliary damage. We hypothesized that PC2 levels are modulated during biliary damage/repair, resulting in activation of the ERK/HIF-1α/VEGF pathway. PC2 protein expression, but not its gene expression, was significantly reduced in mouse livers with biliary damage (Mdr2(-/-) knockout, bile duct ligation, 3,5-diethoxycarbonyl-1,4-dihydrocollidine treatment). Treatment of cholangiocytes with proinflammatory cytokines, nitric oxide donors, and endoplasmic reticulum stressors increased ERK1/2 phosphorylation, HIF-1α transcriptional activity, secretion of VEGF, and VEGF receptor type 2 phosphorylation and down-regulated PC2 protein expression without affecting PC2 gene expression. Expression of homocysteine-responsive endoplasmic reticulum-resident ubiquitin-like domain member 1 protein and NEK, ubiquitin-like proteins that promote proteosomal PC2 degradation, was increased. Pretreatment with the proteasome inhibitor MG-132 restored the expression of PC2 in cells treated with cytokines but not in cells treated with nitric oxide donors or with endoplasmic reticulum stressors. In these conditions, PC2 degradation was instead inhibited by interfering with the autophagy pathway. Treatment of 3,5-diethoxycarbonyl-1,4-dihydrocollidine mice and of Mdr2(-/-) mice with the proteasome inhibitor bortezomib restored PC2 expression and significantly reduced the ductular reaction, fibrosis, and phosphorylated ERK1/2. CONCLUSION In response to biliary damage, PC2 expression is modulated posttranslationally by the proteasome or the autophagy pathway, and PC2 down-regulation is associated with activation of ERK1/2 and an increase of HIF-1α-mediated VEGF secretion; treatments able to restore PC2 expression and to reduce ductular reaction and fibrosis may represent a new therapeutic approach in biliary diseases.
Collapse
Affiliation(s)
- Carlo Spirli
- Section of Digestive Diseases, Yale University, New Haven, Connecticut, USA
| | - Ambra Villani
- Section of Digestive Diseases, Yale University, New Haven, Connecticut, USA
| | - Valeria Mariotti
- Section of Digestive Diseases, Yale University, New Haven, Connecticut, USA,Department of Surgery and Interdisciplinary Medicine, University of Milan-Bicocca, Milan, Italy
| | - Luca Fabris
- Section of Digestive Diseases, Yale University, New Haven, Connecticut, USA,Department of Molecular Medicine, University of Padova, Italy
| | - Romina Fiorotto
- Section of Digestive Diseases, Yale University, New Haven, Connecticut, USA
| | - Mario Strazzabosco
- Section of Digestive Diseases, Yale University, New Haven, Connecticut, USA,Department of Surgery and Interdisciplinary Medicine, University of Milan-Bicocca, Milan, Italy
| |
Collapse
|
27
|
Abstract
The liver is a central regulator of metabolism, and liver failure thus constitutes a major health burden. Understanding how this complex organ develops during embryogenesis will yield insights into how liver regeneration can be promoted and how functional liver replacement tissue can be engineered. Recent studies of animal models have identified key signaling pathways and complex tissue interactions that progressively generate liver progenitor cells, differentiated lineages and functional tissues. In addition, progress in understanding how these cells interact, and how transcriptional and signaling programs precisely coordinate liver development, has begun to elucidate the molecular mechanisms underlying this complexity. Here, we review the lineage relationships, signaling pathways and transcriptional programs that orchestrate hepatogenesis.
Collapse
Affiliation(s)
- Miriam Gordillo
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Valerie Gouon-Evans
- Department of Developmental and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
28
|
Abstract
Cholangiocytes are the epithelial cells that line the bile ducts. Along the biliary tree, two different kinds of cholangiocytes exist; small and large cholangiocytes. Each type has important differences in their biological role in physiological and pathological conditions. In response to injury, cholangiocytes become reactive and acquire a neuroendocrine-like phenotype with the secretion of a number of peptides. These molecules act in an autocrine/paracrine fashion to modulate cholangiocyte biology and determine the evolution of biliary damage. The failure of such mechanisms is believed to influence the progression of cholangiopathies, a group of diseases that selectively target biliary cells. Therefore, the understanding of mechanisms regulating cholangiocyte response to injury is expected to foster the development of new therapeutic options to treat biliary diseases. In the present review, we will discuss the most recent findings in the mechanisms driving cholangiocyte adaptation to damage, with particular emphasis on molecular pathways that are susceptible of therapeutic intervention. Morphogenic pathways (Hippo, Notch, Hedgehog), which have been recently shown to regulate biliary ontogenesis and response to injury, will also be reviewed. In addition, the results of ongoing clinical trials evaluating new drugs for the treatment of cholangiopathies will be discussed.
Collapse
|
29
|
Cast AE, Walter TJ, Huppert SS. Vascular patterning sets the stage for macro and micro hepatic architecture. Dev Dyn 2014; 244:497-506. [PMID: 25370311 DOI: 10.1002/dvdy.24222] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/29/2014] [Accepted: 10/30/2014] [Indexed: 01/20/2023] Open
Abstract
Background The liver is a complex organ with a variety of tissue components that require a precise architecture for optimal function of metabolic and detoxification processes. As a result of the delicate orchestration required between the various hepatic tissues, it is not surprising that impairment of hepatic function can be caused by a variety of factors leading to chronic liver disease. Results Despite the growing rate of chronic liver disease, there are currently few effective treatment options besides orthotopic liver transplantation. Better therapeutic options reside in the potential for genetic and cellular therapies that promote progenitor cell activation aiding de novo epithelial and vascular regeneration, cell replacement, or population of bioartificial hepatic devices. In order to explore this area of new therapeutic potential, it is crucial to understand the factors that promote hepatic function through regulating cell identities and tissue architecture. Conclusions In this commentary, we review the signals regulating liver cell fates during development and regeneration and highlight the importance of patterning the hepatic vascular systems to set the groundwork for the macro and micro hepatic architecture of the epithelium.
Collapse
Affiliation(s)
- Ashley E Cast
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | | | |
Collapse
|
30
|
Walter TJ, Cast AE, Huppert KA, Huppert SS. Epithelial VEGF signaling is required in the mouse liver for proper sinusoid endothelial cell identity and hepatocyte zonation in vivo. Am J Physiol Gastrointest Liver Physiol 2014; 306:G849-62. [PMID: 24650547 PMCID: PMC4024728 DOI: 10.1152/ajpgi.00426.2013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Vascular endothelial growth factor (VEGF) is crucial for vascular development in several organs. However, the specific contribution of epithelial-VEGF signaling in the liver has not been tested. We used a mouse model to specifically delete Vegf from the liver epithelial lineages during midgestational development and assessed the cell identities and architectures of epithelial and endothelial tissues. We find that without epithelial-derived VEGF, the zonal endothelial and hepatocyte cell identities are altered. We also find decreased portal vein and hepatic artery branching coincident with an increase in hepatic hypoxia postnatally. Together, these data indicate that VEGF secreted from the hepatic epithelium is required for normal differentiation of cells and establishment of three-dimensional vascular branching and zonal architectures in both epithelial and endothelial hepatic tissues.
Collapse
Affiliation(s)
- Teagan J. Walter
- 1Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee; and ,2Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ashley E. Cast
- 2Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kari A. Huppert
- 2Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stacey S. Huppert
- 2Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
31
|
Renzi A, Mancinelli R, Onori P, Franchitto A, Alpini G, Glaser S, Gaudio E. Inhibition of the liver expression of arylalkylamine N-acetyltransferase increases the expression of angiogenic factors in cholangiocytes. Hepatobiliary Surg Nutr 2014; 3:4-10. [PMID: 24696833 DOI: 10.3978/j.issn.2304-3881.2014.01.02] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 01/24/2014] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND AIMS Reduction of biliary serotonin N-acetyltransferase (AANAT) expression and melatonin administration/secretion in cholangiocytes increases biliary proliferation and the expression of SR, CFTR and Cl(-)/HCO3 (-) AE2. The balance between biliary proliferation/damage is regulated by several autocrine neuroendocrine factors including vascular endothelial growth factor-A/C (VEGF-A/C). VEGFs are secreted by several epithelia, where they modulate cell growth by autocrine and paracrine mechanisms. No data exists regarding the effect of AANAT modulation on the expressions of VEGFs by cholangiocytes. METHODS In this study, we evaluated the effect of local modulation of biliary AANAT expression on the cholangiocytes synthesis of VEGF-A/C. RESULTS The decrease in AANAT expression and subsequent lower melatonin secretion by cholangiocytes was associated with increased expression of VEGF-A/C. Overexpression of AANAT in cholangiocyte lines decreased the expression of VEGF-A/C. CONCLUSIONS Modulation of melatonin synthesis may affect the expression of VEGF-A/C by cholangiocytes and may modulate the hepatic microvascularization through the regulation of VEGF-A/C expression regulating biliary functions.
Collapse
Affiliation(s)
- Anastasia Renzi
- 1 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza, Rome, Italy ; 2 Eleonora Lorillard Spencer Cenci Foundation, Rome, Italy ; 3 Research, Central Texas Veterans Health Care System, 4 Scott & White Digestive Disease Research Center, Scott & White, Academic Operations, Scott & White, 5 Department of Medicine, Division Gastroenterology, Texas A&M Health Science Center, College of Medicine, Temple, TX 76504, USA
| | - Romina Mancinelli
- 1 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza, Rome, Italy ; 2 Eleonora Lorillard Spencer Cenci Foundation, Rome, Italy ; 3 Research, Central Texas Veterans Health Care System, 4 Scott & White Digestive Disease Research Center, Scott & White, Academic Operations, Scott & White, 5 Department of Medicine, Division Gastroenterology, Texas A&M Health Science Center, College of Medicine, Temple, TX 76504, USA
| | - Paolo Onori
- 1 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza, Rome, Italy ; 2 Eleonora Lorillard Spencer Cenci Foundation, Rome, Italy ; 3 Research, Central Texas Veterans Health Care System, 4 Scott & White Digestive Disease Research Center, Scott & White, Academic Operations, Scott & White, 5 Department of Medicine, Division Gastroenterology, Texas A&M Health Science Center, College of Medicine, Temple, TX 76504, USA
| | - Antonio Franchitto
- 1 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza, Rome, Italy ; 2 Eleonora Lorillard Spencer Cenci Foundation, Rome, Italy ; 3 Research, Central Texas Veterans Health Care System, 4 Scott & White Digestive Disease Research Center, Scott & White, Academic Operations, Scott & White, 5 Department of Medicine, Division Gastroenterology, Texas A&M Health Science Center, College of Medicine, Temple, TX 76504, USA
| | - Gianfranco Alpini
- 1 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza, Rome, Italy ; 2 Eleonora Lorillard Spencer Cenci Foundation, Rome, Italy ; 3 Research, Central Texas Veterans Health Care System, 4 Scott & White Digestive Disease Research Center, Scott & White, Academic Operations, Scott & White, 5 Department of Medicine, Division Gastroenterology, Texas A&M Health Science Center, College of Medicine, Temple, TX 76504, USA
| | - Shannon Glaser
- 1 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza, Rome, Italy ; 2 Eleonora Lorillard Spencer Cenci Foundation, Rome, Italy ; 3 Research, Central Texas Veterans Health Care System, 4 Scott & White Digestive Disease Research Center, Scott & White, Academic Operations, Scott & White, 5 Department of Medicine, Division Gastroenterology, Texas A&M Health Science Center, College of Medicine, Temple, TX 76504, USA
| | - Eugenio Gaudio
- 1 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza, Rome, Italy ; 2 Eleonora Lorillard Spencer Cenci Foundation, Rome, Italy ; 3 Research, Central Texas Veterans Health Care System, 4 Scott & White Digestive Disease Research Center, Scott & White, Academic Operations, Scott & White, 5 Department of Medicine, Division Gastroenterology, Texas A&M Health Science Center, College of Medicine, Temple, TX 76504, USA
| |
Collapse
|
32
|
Maroni L, Pierantonelli I, Benedetti A, Marzioni M. Angiogenic factors in chronic liver diseases: the effects on hepatic progenitor cells. Hepatobiliary Surg Nutr 2014; 2:61-4. [PMID: 24570917 DOI: 10.3978/j.issn.2304-3881.2012.12.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 12/12/2012] [Indexed: 12/21/2022]
Affiliation(s)
- Luca Maroni
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy; ; Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Irene Pierantonelli
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy; ; Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Antonio Benedetti
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy
| | - Marco Marzioni
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
33
|
Spirli C, Strazzabosco M. Vascular endothelial growth factors in progenitor cells mediated liver repair. Hepatobiliary Surg Nutr 2014; 2:65-7. [PMID: 24570918 DOI: 10.3978/j.issn.2304-3881.2012.12.05] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 12/20/2012] [Indexed: 01/14/2023]
Affiliation(s)
- Carlo Spirli
- Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, CT, USA
| | - Mario Strazzabosco
- Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, CT, USA; ; Department of Clinical Medicine, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
34
|
Han Y, Glaser S, Meng F, Francis H, Marzioni M, McDaniel K, Alvaro D, Venter J, Carpino G, Onori P, Gaudio E, Alpini G, Franchitto A. Recent advances in the morphological and functional heterogeneity of the biliary epithelium. Exp Biol Med (Maywood) 2013; 238:549-65. [PMID: 23856906 DOI: 10.1177/1535370213489926] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review focuses on the recent advances related to the heterogeneity of different-sized bile ducts with regard to the morphological and phenotypical characteristics, and the differential secretory, apoptotic and proliferative responses of small and large cholangiocytes to gastrointestinal hormones/peptides, neuropeptides and toxins. We describe several in vivo and in vitro models used for evaluating biliary heterogeneity. Subsequently, we discuss the heterogeneous proliferative and apoptotic responses of small and large cholangiocytes to liver injury and the mechanisms regulating the differentiation of small into large (more differentiated) cholangiocytes. Following a discussion on the heterogeneity of stem/progenitor cells in the biliary epithelium, we outline the heterogeneity of bile ducts in human cholangiopathies. After a summary section, we discuss the future perspectives that will further advance the field of the functional heterogeneity of the biliary epithelium.
Collapse
Affiliation(s)
- Yuyan Han
- Department of Medicine, Division Gastroenterology, Texas A&M Health Science Center, College of Medicine, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Morell CM, Fabris L, Strazzabosco M. Vascular biology of the biliary epithelium. J Gastroenterol Hepatol 2013; 28 Suppl 1:26-32. [PMID: 23855292 PMCID: PMC3721432 DOI: 10.1111/jgh.12022] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/01/2012] [Indexed: 01/13/2023]
Abstract
Cholangiocytes are involved in a variety of processes essential for liver pathophysiology. To meet their demanding metabolic and functional needs, bile ducts are nourished by their own arterial supply, the peribiliary plexus. This capillary network originates from the hepatic artery and is strictly arranged around the intrahepatic bile ducts. Biliary and vascular structures are linked by a close anatomic and functional association necessary for liver development, normal organ physiology, and liver repair. This strong association is finely regulated by a range of angiogenic signals, enabling the cross talk between cholangiocytes and the different vascular cell types. This review will briefly illustrate the "vascular" properties of cholangiocytes, their underlying molecular mechanisms and the relevant pathophysiological settings.
Collapse
Affiliation(s)
- Carola M. Morell
- Department of Surgery and Interdisciplinary Medicine, University of Milano-Bicocca, Milan, Italy
| | - Luca Fabris
- Department of Surgery and Interdisciplinary Medicine, University of Milano-Bicocca, Milan, Italy,Department of Surgery, Oncology and Gastroenterology, Università di Padova, Padova, Italy
| | - Mario Strazzabosco
- Department of Surgery and Interdisciplinary Medicine, University of Milano-Bicocca, Milan, Italy,Liver Center & Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
| |
Collapse
|
36
|
Masyuk TV, Radtke BN, Stroope AJ, Banales JM, Gradilone SA, Huang B, Masyuk AI, Hogan MC, Torres VE, LaRusso NF. Pasireotide is more effective than octreotide in reducing hepatorenal cystogenesis in rodents with polycystic kidney and liver diseases. Hepatology 2013; 58:409-21. [PMID: 23172758 PMCID: PMC3616157 DOI: 10.1002/hep.26140] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 11/06/2012] [Indexed: 12/21/2022]
Abstract
UNLABELLED In polycystic liver (PLD) and kidney (PKD) diseases, increased cyclic adenosine monophosphate (cAMP) levels trigger hepatorenal cystogenesis. A reduction of the elevated cAMP by targeting somatostatin receptors (SSTRs) with octreotide (OCT; a somatostatin analog that preferentially binds to SSTR2) inhibits cyst growth. Here we compare the effects of OCT to pasireotide (PAS; a more potent somatostatin analog with broader receptor specificity) on: (1) cAMP levels, cell cycle, proliferation, and cyst expansion in vitro using cholangiocytes derived from control and PCK rats (a model of autosomal recessive PKD [ARPKD]), healthy human beings, and patients with autosomal dominant PKD (ADPKD); and (2) hepatorenal cystogenesis in vivo in PCK rats and Pkd2(WS25/-) mice (a model of ADPKD). Expression of SSTRs was assessed in control and cystic cholangiocytes of rodents and human beings. Concentrations of insulin-like growth factor 1 (IGF1) and vascular endothelial growth factor (VEGF) (both involved in indirect action of somatostatin analogs), and expression and localization of SSTRs after treatment were evaluated. We found that PAS was more potent (by 30%-45%) than OCT in reducing cAMP and cell proliferation, affecting cell cycle distribution, decreasing growth of cultured cysts in vitro, and inhibiting hepatorenal cystogenesis in vivo in PCK rats and Pkd2(WS25/-) mice. The levels of IGF1 (but not VEGF) were reduced only in response to PAS. Expression of SSTR1 and SSTR2 (but not SSTR3 and SSTR5) was decreased in cystic cholangiocytes compared to control. Although both OCT and PAS increased the immunoreactivity of SSTR2, only PAS up-regulated SSTR1; neither drug affected cellular localization of SSTRs. CONCLUSION PAS is more effective than OCT in reducing hepatorenal cystogenesis in rodent models; therefore, it might be more beneficial for the treatment of PKD and PLD.
Collapse
Affiliation(s)
- Tatyana V Masyuk
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN USA
| | - Brynn N Radtke
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN USA
| | - Angela J Stroope
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN USA
| | - Jesús M Banales
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN USA,IKERBASQUE, Basque Foundation of Science, Division of Hepatology, Biodonostia Institute, Donostia Hospital, CIBERehd, University of Basque Country, San Sebastián, Spain
| | - Sergio A Gradilone
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN USA
| | - Bing Huang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN USA
| | - Anatoliy I Masyuk
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN USA
| | - Marie C Hogan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN USA
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN USA
| | - Nicholas F LaRusso
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
37
|
Iguchi H, Oda M, Yamazaki H, Yokomori H. Participation of aquaporin-1 in vascular changes and remodeling in cirrhotic liver. Med Mol Morphol 2013; 46:123-32. [PMID: 23549977 DOI: 10.1007/s00795-013-0039-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 02/13/2013] [Indexed: 12/22/2022]
Abstract
The pathophysiology of arterial capillary proliferation accompanying fibrosis in human cirrhosis remains unclear. However, evidence regarding the molecules participating in the pathophysiological process has been accumulating. Water channel proteins known as aquaporins (AQP)s, notably AQP-1, appear to be involved in the arterial capillary proliferation in the cirrhotic liver.
Collapse
Affiliation(s)
- Hiroyoshi Iguchi
- Department of Radiology, Kitasato University Medical Center, Saitama, 364-8501, Japan
| | | | | | | |
Collapse
|
38
|
Strazzabosco M, Fabris L. Development of the bile ducts: essentials for the clinical hepatologist. J Hepatol 2012; 56:1159-1170. [PMID: 22245898 PMCID: PMC3328609 DOI: 10.1016/j.jhep.2011.09.022] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 09/08/2011] [Accepted: 09/13/2011] [Indexed: 02/07/2023]
Abstract
Several cholangiopathies result from a perturbation of developmental processes. Most of these cholangiopathies are characterised by the persistence of biliary structures with foetal configuration. Developmental processes are also relevant in acquired liver diseases, as liver repair mechanisms exploit a range of autocrine and paracrine signals transiently expressed in embryonic life. We briefly review the ontogenesis of the intra- and extrahepatic biliary tree, highlighting the morphogens, growth factors, and transcription factors that regulate biliary development, and the relationships between developing bile ducts and other branching biliary structures. Then, we discuss the ontogenetic mechanisms involved in liver repair, and how these mechanisms are recapitulated in ductular reaction, a common reparative response to many forms of biliary and hepatocellular damage. Finally, we discuss the pathogenic aspects of the most important primary cholangiopathies related to altered biliary development, i.e. polycystic and fibropolycystic liver diseases, Alagille syndrome.
Collapse
Affiliation(s)
- Mario Strazzabosco
- Section of Digestive Diseases, Yale University, New Haven, CT, USA; Department of Clinical Medicine, University of Milan-Bicocca, Milan, Italy.
| | - Luca Fabris
- Department of Clinical Medicine, University of Milan-Bicocca, Milan, Italy,Department of Surgical and Gastroenterological Sciences, University of Padova, Italy
| |
Collapse
|
39
|
Vestentoft PS, Jelnes P, Hopkinson BM, Vainer B, Møllgård K, Quistorff B, Bisgaard HC. Three-dimensional reconstructions of intrahepatic bile duct tubulogenesis in human liver. BMC DEVELOPMENTAL BIOLOGY 2011; 11:56. [PMID: 21943389 PMCID: PMC3192761 DOI: 10.1186/1471-213x-11-56] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 09/26/2011] [Indexed: 01/05/2023]
Abstract
Background During liver development, intrahepatic bile ducts are thought to arise by a unique asymmetric mode of cholangiocyte tubulogenesis characterized by a series of remodeling stages. Moreover, in liver diseases, cells lining the Canals of Hering can proliferate and generate new hepatic tissue. The aim of this study was to develop protocols for three-dimensional visualization of protein expression, hepatic portal structures and human hepatic cholangiocyte tubulogenesis. Results Protocols were developed to digitally visualize portal vessel branching and protein expression of hepatic cell lineage and extracellular matrix deposition markers in three dimensions. Samples from human prenatal livers ranging from 7 weeks + 2 days to 15½ weeks post conception as well as adult normal and acetaminophen intoxicated liver were used. The markers included cytokeratins (CK) 7 and 19, the epithelial cell adhesion molecule (EpCAM), hepatocyte paraffin 1 (HepPar1), sex determining region Y (SRY)-box 9 (SOX9), laminin, nestin, and aquaporin 1 (AQP1). Digital three-dimensional reconstructions using CK19 as a single marker protein disclosed a fine network of CK19 positive cells in the biliary tree in normal liver and in the extensive ductular reactions originating from intrahepatic bile ducts and branching into the parenchyma of the acetaminophen intoxicated liver. In the developing human liver, three-dimensional reconstructions using multiple marker proteins confirmed that the human intrahepatic biliary tree forms through several developmental stages involving an initial transition of primitive hepatocytes into cholangiocytes shaping the ductal plate followed by a process of maturation and remodeling where the intrahepatic biliary tree develops through an asymmetrical form of cholangiocyte tubulogenesis. Conclusions The developed protocols provide a novel and sophisticated three-dimensional visualization of vessels and protein expression in human liver during development and disease.
Collapse
Affiliation(s)
- Peter S Vestentoft
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen N, Denmark
| | | | | | | | | | | | | |
Collapse
|
40
|
Truesdale AE, Caldwell SH, Shah NL, Argo CK, Al-Osaimi AMS, Schmitt TM, Northup PG. Sorafenib therapy for hepatocellular carcinoma prior to liver transplant is associated with increased complications after transplant. Transpl Int 2011; 24:991-8. [PMID: 21777298 DOI: 10.1111/j.1432-2277.2011.01299.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This study compared post-transplant outcomes of patients with hepatocellular carcinoma (HCC) who took sorafenib prior to orthotopic liver transplantation (OLT) with those patients who were not treated with sorafenib. Thirty-three patients with HCC who were listed for liver transplantation were studied: 10 patients were treated with sorafenib prior to transplantation in an attempt to prevent progression of HCC while awaiting transplant. The remaining 23 patients were considered controls. The mean duration of sorafenib use was 19.2 (SD 25.2) weeks. Overall death rates were similar between the sorafenib group and control group (20% vs. 8.7%, respectively, P = 0.56). However, the patients in the sorafenib group had a higher incidence of acute cellular rejection following transplantation (67% vs. 22%, OR = 7.2, 95% CI 1.3-39.6, P = 0.04). The sorafenib group also had a higher rate of early biliary complications (67% vs. 17%, OR = 9.5, 1.6-55.0, P = 0.01). The use of sorafenib was found to be an independent predictor of post-transplant biliary complications (OR 12.6, 1.4-116.2, P = 0.03). Sorafenib administration prior to OLT appears to be associated with an increase in biliary complications and possibly in acute rejection following liver transplantation. Caution should be taken in this setting until larger studies are completed.
Collapse
Affiliation(s)
- Aimee E Truesdale
- Division of Gastroenterology and Hepatology, University of Virginia, Charlottesville, VA 22908-0708, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Colombo F, Baldan F, Mazzucchelli S, Martin-Padura I, Marighetti P, Cattaneo A, Foglieni B, Spreafico M, Guerneri S, Baccarin M, Bertolini F, Rossi G, Mazzaferro V, Cadamuro M, Maggioni M, Agnelli L, Rebulla P, Prati D, Porretti L. Evidence of distinct tumour-propagating cell populations with different properties in primary human hepatocellular carcinoma. PLoS One 2011; 6:e21369. [PMID: 21731718 PMCID: PMC3121782 DOI: 10.1371/journal.pone.0021369] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Accepted: 05/27/2011] [Indexed: 12/14/2022] Open
Abstract
Background and Aims Increasing evidence that a number of malignancies are characterised by tumour cell heterogeneity has recently been published, but there is still a lack of data concerning liver cancers. The aim of this study was to investigate and characterise tumour-propagating cell (TPC) compartments within human hepatocellular carcinoma (HCC). Methods After long-term culture, we identified three morphologically different tumour cell populations in a single HCC specimen, and extensively characterised them by means of flow cytometry, fluorescence microscopy, karyotyping and microarray analyses, single cell cloning, and xenotransplantation in NOD/SCID/IL2Rγ−/− mice. Results The primary cell populations (hcc-1, -2 and -3) and two clones generated by means of limiting dilutions from hcc-1 (clone-1/7 and -1/8) differently expressed a number of tumour-associated stem cell markers, including EpCAM, CD49f, CD44, CD133, CD56, Thy-1, ALDH and CK19, and also showed different doubling times, drug resistance and tumorigenic potential. Moreover, we found that ALDH expression, in combination with CD44 or Thy-1 negativity or CD56 positivity identified subpopulations with a higher clonogenic potential within hcc-1, hcc-2 and hcc-3 primary cell populations, respectively. Karyotyping revealed the clonal evolution of the cell populations and clones within the primary tumour. Importantly, the primary tumour cell population with the greatest tumorigenic potential and drug resistance showed more chromosomal alterations than the others and contained clones with epithelial and mesenchymal features. Conclusions Individual HCCs can harbor different self-renewing tumorigenic cell types expressing a variety of morphological and phenotypical markers, karyotypic evolution and different gene expression profiles. This suggests that the models of hepatic carcinogenesis should take into account TPC heterogeneity due to intratumour clonal evolution.
Collapse
Affiliation(s)
- Federico Colombo
- Experimental Hepatology Laboratory, Centre of Transfusion Medicine, Cellular Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Polycystic liver diseases (PLD) are inherited disorders of the biliary epithelium, caused by genetic defects in proteins associated with intracellular organelles, mainly the endoplasmic reticulum and the cilium. PLD are characterized by the formation and progressive enlargement of multiple cysts scattered throughout the liver parenchyma, and include different entities, classified based on their pathology, inheritance pattern, involvement of the kidney and clinical features. PLD should be considered as congenital diseases of cholangiocyte signaling. Here, we will review the changes in signaling pathways involved in liver cyst formation and progression, and their impact on cholangiocyte physiology. Each pathway represents a potential target for therapies aimed at reducing disease progression.
Collapse
Affiliation(s)
- Mario Strazzabosco
- Liver Center and Section of Digestive Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut 06520, USA.
| | | |
Collapse
|
43
|
Desmet VJ. Ductal plates in hepatic ductular reactions. Hypothesis and implications. II. Ontogenic liver growth in childhood. Virchows Arch 2011; 458:261-70. [PMID: 21298286 DOI: 10.1007/s00428-011-1049-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2010] [Revised: 01/18/2011] [Accepted: 01/18/2011] [Indexed: 01/09/2023]
Abstract
This article discusses the processes of bile duct growth and new lobule formation in the liver during childhood in the light of the ductal plate (DP) hypothesis. Unlike in other organs in which tubular elongation and branching ends with the creation of the organ-specific terminal differentiation products, in the liver a steadily enlarging parenchymal mass needs to establish continuity of its canalicular network with the existing bile duct system. The hypothesis suggests that this occurs by DP formation, like in the embryonic liver, and further assumes that pathological ductular reactions (DRs) induced by cholestasis or hypoxia are amplified equivalents of similar mechanisms operating at low level during liver growth. The concept is confronted with data on porcine liver growth, since swine and non-swine liver growth is thought to be comparable. Relative bile acid load may be the driving force for establishment of new canaliculo-ductular connections, supported in zones of relative hypoxia by hypoxia-inducible factor 1 alpha secreted by hepatocytes. The latter mechanism is at the base for induction of appropriate vascular changes in selected sinusoids, resulting in the development of portal inlet venules and additional draining central veins. The process gives rise to the formation of new single lobules by formation of new portal tracts or to the transformation of single lobules in compound lobules by development of new vascular septa. The concept of postnatal DP formation is important in the elucidation of several unexplained findings in adult liver diseases.
Collapse
Affiliation(s)
- Valeer J Desmet
- Department of Pathology, University Hospital K.U.Leuven, Rafael, Leuven, Belgium.
| |
Collapse
|
44
|
Abstract
In most cholangiopathies, liver diseases of different etiologies in which the biliary epithelium is the primary target in the pathogenic sequence, the central mechanism involves inflammation. Inflammation, characterized by pleomorphic peribiliary infiltrate containing fibroblasts, macrophages, lymphocytes, as well as endothelial cells and pericytes, is associated to the emergence of "reactive cholangiocytes." These biliary cells do not possess bile secretory functions, are in contiguity with terminal cholangioles, and are of a less-differentiated phenotype. They have acquired several mesenchymal properties, including motility and ability to secrete a vast number of proinflammatory chemo/cytokines and growth factors along with de novo expression of a rich receptor machinery. These functional properties enable reactive cholangiocytes to establish intimate contacts and to mutually exchange a variety of paracrine signals with the different mesenchymal cell types populating the portal infiltrate. The extensive crosstalk between the epithelial and mesenchymal compartments is the driver of liver repair mechanisms in cholangiopathies, ultimately evolving toward portal fibrosis. Herein, the authors first review the properties of the different cell types involved in their interaction, and then analyze the underlying molecular mechanisms as they relate to liver repair in cholangiopathies.
Collapse
Affiliation(s)
- Luca Fabris
- Department of Surgical and Gastroenterological Sciences, University of Padua, Padova, Italy
- Center for Liver Research (CeLiveR), Bergamo, Italy
| | - Mario Strazzabosco
- Center for Liver Research (CeLiveR), Bergamo, Italy
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut
- Department of Clinical Medicine, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
45
|
Bateman AC, Hübscher SG. Cytokeratin expression as an aid to diagnosis in medical liver biopsies. Histopathology 2011; 56:415-25. [PMID: 20459548 DOI: 10.1111/j.1365-2559.2009.03391.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The study of cytokeratin expression has provided a valuable insight into the biliary microanatomy of the liver in health and disease. The canals of Hering are a putative site of origin for progenitor cells, which may repopulate the liver after cellular damage and loss. Normal bile ducts and the bile ductular reaction that occurs in many chronic liver diseases - especially chronic biliary tract disease - express cytokeratin (CK) 7 and CK19. Therefore, both ductopenia and the process of bile ductular reaction can be highlighted with immunohistochemistry for these cytokeratins. Furthermore, CK7 is usually expressed in an increasingly widespread manner by hepatocytes as chronic cholestatic liver disease progresses. For these reasons, CK immunohistochemistry is a very useful adjunct to morphological assessment and histochemical stains for copper retention when a diagnosis of chronic biliary disease is being considered. This review describes the anatomical theory behind the use of CK immunohistochemistry for the assessment of bile duct number and distribution in the liver and provides practical advice for the application of this technique in the diagnostic setting of common medical liver diseases.
Collapse
Affiliation(s)
- Adrian C Bateman
- Department of Cellular Pathology, Southampton General Hospital, Southampton, UK.
| | | |
Collapse
|
46
|
Gouw ASH, Zeng W, Buiskool M, Platteel I, van den Heuvel MC, Poppema S, de Jong KP, Molema G. Molecular characterization of the vascular features of focal nodular hyperplasia and hepatocellular adenoma: a role for angiopoietin-1. Hepatology 2010; 52:540-9. [PMID: 20683953 DOI: 10.1002/hep.23700] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
UNLABELLED Focal nodular hyperplasia (FNH) and hepatocellular adenoma (HCA) are two hepatic nodular lesions of different etiologies. FNH, a polyclonal lesion, is assumed to be a regenerative reaction following a vascular injury, whereas HCA is a monoclonal, benign neoplastic lesion. In addition to features that are predominantly found in either FNH or HCA (e.g., dystrophic vessels in FNH and single arteries in HCA), FNH and HCA share morphological vascular abnormalities such as dilated sinusoids. We hypothesized that these anomalous vascular features are associated with altered expression of growth factors involved in vascular remodeling. This was based on reports of morphologically abnormal hepatic vasculature and nodular lesions in transgenic models of hepatocytic overexpression of angiopoietin-1 (Ang-1), a member of the angiopoietin family, which is crucially involved in vascular morphogenesis and homeostasis. We investigated gene and protein expression of members of the angiopoietin system and vascular endothelial growth factor A (VEGF-A) and its receptors in 9 FNH samples, 13 HCA samples, and 9 histologically normal livers. In comparison with normal samples, a significant increase in Ang-1 was found in FNH (P < 0.01) and HCA (P < 0.05), whereas no significant changes in Ang-2, receptor tyrosine kinase with immunoglobulin-like and EGF-like domains 2, VEGF-A, or vascular endothelial growth factor receptor 2 (VEGFR-2) were observed. CONCLUSION Because of the different etiological contexts of a preceding vascular injury in FNH and a neoplastic growth in HCA, Ang-1 might exert different effects on the vasculature in these lesions. In FNH, it could predominantly stimulate recruitment of myofibroblasts and result in dystrophic vessels, whereas in HCA, it may drive vascular remodeling that produces enlarged vessels and arterial sprouting that generates single arteries.
Collapse
Affiliation(s)
- Annette S H Gouw
- Pathology Section, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Huebert RC, Vasdev MM, Shergill U, Das A, Huang BQ, Charlton MR MR, LaRusso NF, Shah VH. Aquaporin-1 facilitates angiogenic invasion in the pathological neovasculature that accompanies cirrhosis. Hepatology 2010; 52:238-48. [PMID: 20578142 PMCID: PMC2928054 DOI: 10.1002/hep.23628] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
UNLABELLED Increasing evidence suggests that hepatic fibrosis and pathological angiogenesis are interdependent processes that occur in parallel. Endothelial cell invasion is requisite for angiogenesis, and thus studies of the mechanisms governing liver endothelial cell (LEC) invasion during cirrhosis are of great importance. Emerging research implicates amoeboid-type motility and membrane blebbing as features that may facilitate invasion through matrix-rich microenvironments. Aquaporins (AQPs) are integral membrane water channels, recognized for their importance in epithelial secretion and absorption. However, recent studies also suggest links between water transport and cell motility or invasion. Therefore, the purpose of this study was to test the hypothesis that AQP-1 is involved in amoeboid motility and angiogenic invasion during cirrhosis. AQP-1 expression and localization was examined in normal and cirrhotic liver tissues derived from human and mouse. AQP-1 levels were modulated in LEC using retroviral overexpression or small interfering RNA (siRNA) knockdown and functional effects on invasion, membrane blebbing dynamics, and osmotic water permeability were assayed. Results demonstrate that AQP-1 is up-regulated in the small, angiogenic, neovasculature within the fibrotic septa of cirrhotic liver. AQP-1 overexpression promotes fibroblast growth factor (FGF)-induced dynamic membrane blebbing in LEC, which is sufficient to augment invasion through extracellular matrix. Additionally, AQP-1 localizes to plasma membrane blebs, where it increases osmotic water permeability and locally facilitates the rapid, trans-membrane flux of water. CONCLUSION AQP-1 enhances osmotic water permeability and FGF-induced dynamic membrane blebbing in LEC and thereby drives invasion and pathological angiogenesis during cirrhosis.
Collapse
Affiliation(s)
- Robert C. Huebert
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN 55905
- Department of Internal Medicine, Mayo Clinic and Foundation, Rochester, MN 55905
| | - Meher M. Vasdev
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN 55905
| | - Uday Shergill
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN 55905
| | - Amitava Das
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN 55905
| | - Bing Q. Huang
- Center for Basic Research in Digestive Diseases, Mayo Clinic and Foundation, Rochester, MN 55905
| | - Michael R. Charlton MR
- Center for Basic Research in Digestive Diseases, Mayo Clinic and Foundation, Rochester, MN 55905
- Department of Internal Medicine, Mayo Clinic and Foundation, Rochester, MN 55905
| | - Nicholas F. LaRusso
- Center for Basic Research in Digestive Diseases, Mayo Clinic and Foundation, Rochester, MN 55905
- Department of Internal Medicine, Mayo Clinic and Foundation, Rochester, MN 55905
- Center for Cell Signaling, Mayo Clinic and Foundation, Rochester, MN 55905
| | - Vijay H. Shah
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN 55905
- Department of Internal Medicine, Mayo Clinic and Foundation, Rochester, MN 55905
- Center for Cell Signaling, Mayo Clinic and Foundation, Rochester, MN 55905
| |
Collapse
|
48
|
Arterial stiffness in hypertensives in relation to expression of angiopoietin-1 and 2 genes in peripheral monocytes. J Hum Hypertens 2010; 24:306-11. [PMID: 20072144 DOI: 10.1038/jhh.2009.95] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Angiopoietins (Angs) are important angiogenic and endothelial cell growth factors with many functions, including influence on the vascular wall. Pulse-wave velocity (pwv) is an independent marker of cardiovascular adverse outcome in hypertensives, although all the pathophysiological mechanisms that affect it have not yet been determined. We investigated the relationship between arterial stiffness and Ang-1 and Ang-2 gene expression in the peripheral blood monocytes of hypertensive patients. We studied 53 patients who had untreated grade-1 or grade-2 essential hypertension and no indications of other organic heart disease. Carotid-femoral (c-f) and carotid-radial (c-r) artery waveforms were measured and pwv was determined. The monocytes were isolated using anti-CD14(+) antibodies and mRNAs were estimated by real-time quantitative reverse transcription-PCR. Ang-1 gene expression was strongly correlated with both c-f-pwv (r=0.952, P<0.001) and c-r-pwv (r=0.898, P<0.001). Similarly, Ang-2 gene expression was significantly correlated with both c-f-pwv (r=0.471, P=0.002) and c-r-pwv (r=0.437, P=0.003). Our data provide important evidence that Ang-1 and Ang-2 gene expression levels in peripheral monocytes are closely related with pwv in patients with essential hypertension. This positive correlation may suggest a link between angiogenesis and arterial stiffness in those patients.
Collapse
|
49
|
Spirli C, Okolicsanyi S, Fiorotto R, Fabris L, Cadamuro M, Lecchi S, Tian X, Somlo S, Strazzabosco M. ERK1/2-dependent vascular endothelial growth factor signaling sustains cyst growth in polycystin-2 defective mice. Gastroenterology 2010; 138:360-371.e7. [PMID: 19766642 PMCID: PMC3000794 DOI: 10.1053/j.gastro.2009.09.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2008] [Revised: 09/04/2009] [Accepted: 09/09/2009] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Severe polycystic liver disease can complicate adult dominant polycystic kidney disease, a genetic disease caused by defects in polycystin-1 (Pkd1) or polycystin-2 (Pkd2). Liver cyst epithelial cells (LCECs) express vascular endothelial growth factor (VEGF) and its receptor, VEGFR-2. We investigated the effects of VEGF on liver cyst growth and autocrine VEGF signaling in mice with Pkd1 and Pkd2 conditional knockouts. METHODS We studied mice in which Pkd1 or Pkd2 were conditionally inactivated following exposure to tamoxifen; these mice were called Pkd1(flox/-):pCxCreER (Pkd1KO) and Pkd2(flox/-):pCxCreER (Pkd2KO). RESULTS Pkd1KO and Pkd2KO mice developed liver defects; their LCECs expressed VEGF, VEGFR-2, hypoxia-inducible factor (HIF)-1alpha, phosphorylated extracellular signal-regulated kinase 1/2 (pERK1/2), and proliferating cell nuclear antigen (PCNA). In Pkd2KO but not Pkd1KO mice, exposure to the VEGFR-2 inhibitor SU5416 significantly reduced liver cyst development, liver/body weight ratio, and expression of pERK and PCNA. VEGF secretion and phosphorylation of ERK1/2 and VEGFR-2 were significantly increased in cultured LCECs from Pkd2KO compared with Pkd1KO mice. Inhibition of protein kinase A (PKA) reduced VEGF secretion and pERK1/2 expression. Addition of VEGF to LCECs from Pkd2KO mice increased phosphorylated VEGFR-2 and phosphorylated mitogen signal-regulated kinase (MEK) expression and induced phosphorylation of ERK1/2; this was inhibited by SU5416. Expression of HIF-1alpha increased in parallel with secretion of VEGF following LCEC stimulation. VEGF-induced cell proliferation was inhibited by the MEK inhibitor U1026 and by ERK1/2 small interfering RNA. CONCLUSIONS The PKA-ERK1/2-VEGF signaling pathway promotes growth of liver cysts in mice. In Pkd2-defective LCECs, PKA-dependent ERK1/2 signaling controls HIF-1alpha-dependent VEGF secretion and VEGFR-2 signaling. Autocrine and paracrine VEGF signaling promotes the growth of liver cysts in Pkd2KO mice. VEGF inhibitors might be used to treat patients with polycystic liver disease.
Collapse
Affiliation(s)
- Carlo Spirli
- Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Lemaigre FP. Molecular mechanisms of biliary development. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 97:103-26. [PMID: 21074731 DOI: 10.1016/b978-0-12-385233-5.00004-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The biliary tree drains the bile produced by hepatocytes to the duodenum via a network of intrahepatic and extrahepatic ducts. In the embryo, the intrahepatic ducts are formed near the branches of the portal vein and derive from the liver precursor cells of the hepatic bud, whereas the extrahepatic ducts directly emerge from the primitive gut. Despite this dual origin, intrahepatic and extrahepatic ducts are lined by a common cell type, the cholangiocyte. In this chapter, we describe how bile ducts are formed and cholangiocytes differentiate, and focus on the regulation of these processes by intercellular signaling pathways and by transcriptional and posttranscriptional mechanisms.
Collapse
|