1
|
Rizvi F, Lee YR, Diaz-Aragon R, Bawa PS, So J, Florentino RM, Wu S, Sarjoo A, Truong E, Smith AR, Wang F, Everton E, Ostrowska A, Jung K, Tam Y, Muramatsu H, Pardi N, Weissman D, Soto-Gutierrez A, Shin D, Gouon-Evans V. VEGFA mRNA-LNP promotes biliary epithelial cell-to-hepatocyte conversion in acute and chronic liver diseases and reverses steatosis and fibrosis. Cell Stem Cell 2023; 30:1640-1657.e8. [PMID: 38029740 PMCID: PMC10843608 DOI: 10.1016/j.stem.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/07/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023]
Abstract
The liver is known for its remarkable regenerative ability through proliferation of hepatocytes. Yet, during chronic injury or severe hepatocyte death, proliferation of hepatocytes is exhausted. To overcome this hurdle, we propose vascular-endothelial-growth-factor A (VEGFA) as a therapeutic means to accelerate biliary epithelial-cell (BEC)-to-hepatocyte conversion. Investigation in zebrafish establishes that blocking VEGF receptors abrogates BEC-driven liver repair, while VEGFA overexpression promotes it. Delivery of VEGFA via nonintegrative and safe nucleoside-modified mRNA encapsulated into lipid nanoparticles (mRNA-LNPs) in acutely or chronically injured mouse livers induces robust BEC-to-hepatocyte conversion and elimination of steatosis and fibrosis. In human and murine diseased livers, we further identified VEGFA-receptor KDR-expressing BECs associated with KDR-expressing cell-derived hepatocytes. This work defines KDR-expressing cells, most likely being BECs, as facultative progenitors. This study reveals unexpected therapeutic benefits of VEGFA delivered via nucleoside-modified mRNA-LNP, whose safety is widely validated with COVID-19 vaccines, for harnessing BEC-driven repair to potentially treat liver diseases.
Collapse
Affiliation(s)
- Fatima Rizvi
- Center for Regenerative Medicine, Department of Medicine, Section of Gastroenterology, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Yu-Ri Lee
- Department of Developmental Biology, Pittsburgh Liver Research Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Ricardo Diaz-Aragon
- Department of Pathology, Center for Transcriptional Medicine, Pittsburgh Liver Research Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Pushpinder S Bawa
- Center for Regenerative Medicine, Department of Medicine, Section of Gastroenterology, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Juhoon So
- Department of Developmental Biology, Pittsburgh Liver Research Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Rodrigo M Florentino
- Department of Pathology, Center for Transcriptional Medicine, Pittsburgh Liver Research Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Susan Wu
- Center for Regenerative Medicine, Department of Medicine, Section of Gastroenterology, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Arianna Sarjoo
- Center for Regenerative Medicine, Department of Medicine, Section of Gastroenterology, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Emily Truong
- Center for Regenerative Medicine, Department of Medicine, Section of Gastroenterology, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Anna R Smith
- Center for Regenerative Medicine, Department of Medicine, Section of Gastroenterology, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Feiya Wang
- Center for Regenerative Medicine, Department of Medicine, Section of Gastroenterology, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Elissa Everton
- Center for Regenerative Medicine, Department of Medicine, Section of Gastroenterology, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Alina Ostrowska
- Department of Pathology, Center for Transcriptional Medicine, Pittsburgh Liver Research Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Kyounghwa Jung
- Department of Developmental Biology, Pittsburgh Liver Research Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Ying Tam
- Acuitas Therapeutics, Vancouver, BC V6T 1Z3, Canada
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Drew Weissman
- Department of Medicine, Infectious Diseases Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 10104, USA
| | - Alejandro Soto-Gutierrez
- Department of Pathology, Center for Transcriptional Medicine, Pittsburgh Liver Research Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Donghun Shin
- Department of Developmental Biology, Pittsburgh Liver Research Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Valerie Gouon-Evans
- Center for Regenerative Medicine, Department of Medicine, Section of Gastroenterology, Boston University and Boston Medical Center, Boston, MA 02118, USA.
| |
Collapse
|
2
|
Hu Y, Hu X, Luo J, Huang J, Sun Y, Li H, Qiao Y, Wu H, Li J, Zhou L, Zheng S. Liver organoid culture methods. Cell Biosci 2023; 13:197. [PMID: 37915043 PMCID: PMC10619312 DOI: 10.1186/s13578-023-01136-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/20/2023] [Indexed: 11/03/2023] Open
Abstract
Organoids, three-dimensional structures cultured in vitro, can recapitulate the microenvironment, complex architecture, and cellular functions of in vivo organs or tissues. In recent decades, liver organoids have been developed rapidly, and their applications in biomedicine, such as drug screening, disease modeling, and regenerative medicine, have been widely recognized. However, the lack of repeatability and consistency, including the lack of standardized culture conditions, has been a major obstacle to the development and clinical application of liver organoids. It is time-consuming for researchers to identify an appropriate medium component scheme, and the usage of some ingredients remains controversial. In this review, we summarized and compared different methods for liver organoid cultivation that have been published in recent years, focusing on controversial medium components and discussing their advantages and drawbacks. We aimed to provide an effective reference for the development and standardization of liver organoid cultivation.
Collapse
Affiliation(s)
- Yiqing Hu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Xiaoyi Hu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jia Luo
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jiacheng Huang
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Yaohan Sun
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Haoyu Li
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Yinbiao Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Hao Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jianhui Li
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, 310015, China
- The Organ Repair and Regeneration Medicine Institute of Hangzhou, Hangzhou, 310003, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, China.
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, 310015, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, China.
| |
Collapse
|
3
|
Kocas-Kilicarslan ZN, Cetin Z, Faccioli LAP, Motomura T, Amirneni S, Diaz-Aragon R, Florentino RM, Sun Y, Pla-Palacin I, Xia M, Miedel MT, Kurihara T, Hu Z, Ostrowska A, Wang Z, Constantine R, Li A, Taylor DL, Behari J, Soto-Gutierrez A, Tafaleng EN. Polymorphisms Associated With Metabolic Dysfunction-Associated Steatotic Liver Disease Influence the Progression of End-Stage Liver Disease. GASTRO HEP ADVANCES 2023; 3:67-77. [PMID: 38292457 PMCID: PMC10827334 DOI: 10.1016/j.gastha.2023.09.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
BACKGROUND AND AIMS Chronic liver injury that results in cirrhosis and end-stage liver disease (ESLD) causes more than 1 million deaths annually worldwide. Although the impact of genetic factors on the severity of metabolic dysfunction-associated steatotic liver disease (MASLD) and alcohol-related liver disease (ALD) has been previously studied, their contribution to the development of ESLD remains largely unexplored. METHODS We genotyped 6 MASLD-associated polymorphisms in healthy (n = 123), metabolic dysfunction-associated steatohepatitis (MASH) (n = 145), MASLD-associated ESLD (n = 72), and ALD-associated ESLD (n = 57) cohorts and performed multinomial logistic regression to determine the combined contribution of genetic, demographic, and clinical factors to the progression of ESLD. RESULTS Distinct sets of factors are associated with the progression to ESLD. The PNPLA3 rs738409:G and TM6SF2 rs58542926:T alleles, body mass index (BMI), age, and female sex were positively associated with progression from a healthy state to MASH. The PNPLA3 rs738409:G allele, age, male sex, and having type 2 diabetes mellitus were positively associated, while BMI was negatively associated with progression from MASH to MASLD-associated ESLD. The PNPLA3 rs738409:G and GCKR rs780094:T alleles, age, and male sex were positively associated, while BMI was negatively associated with progression from a healthy state to ALD-associated ESLD. The findings indicate that the PNPLA3 rs738409:G allele increases susceptibility to ESLD regardless of etiology, the TM6SF2 rs58542926:T allele increases susceptibility to MASH, and the GCKR rs780094:T allele increases susceptibility to ALD-associated ESLD. CONCLUSION The PNPLA3, TM6SF2, and GCKR minor alleles influence the progression of MASLD-associated or ALD-associated ESLD. Genotyping for these variants in MASLD and ALD patients can enhance risk assessment, prompting early interventions to prevent ESLD.
Collapse
Affiliation(s)
- Zehra N. Kocas-Kilicarslan
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Zeliha Cetin
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Lanuza A. P. Faccioli
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Takashi Motomura
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sriram Amirneni
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ricardo Diaz-Aragon
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Rodrigo M. Florentino
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yiyue Sun
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- School of Medicine, Tsinghua University, Beijing, China
| | - Iris Pla-Palacin
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mengying Xia
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mark T. Miedel
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Takeshi Kurihara
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Zhiping Hu
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Alina Ostrowska
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zi Wang
- Department of Statistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Albert Li
- Discovery Life Sciences, Huntsville, Alabama
| | - D. Lansing Taylor
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jaideep Behari
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Alejandro Soto-Gutierrez
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania
| | - Edgar N. Tafaleng
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
4
|
Rizvi F, Lee YR, Diaz-Aragon R, So J, Florentino RM, Smith AR, Everton E, Ostrowska A, Jung K, Tam Y, Muramatsu H, Pardi N, Weissman D, Soto-Gutierrez A, Shin D, Gouon-Evans V. VEGFA mRNA-LNP promotes biliary epithelial cell-to-hepatocyte conversion in acute and chronic liver diseases and reverses steatosis and fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.537186. [PMID: 37131823 PMCID: PMC10153196 DOI: 10.1101/2023.04.17.537186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The liver is known for its remarkable regenerative ability through proliferation of hepatocytes. Yet, during chronic injury or severe hepatocyte death, proliferation of hepatocytes is exhausted. To overcome this hurdle, we propose vascular-endothelial-growth-factor A (VEGFA) as a therapeutic means to accelerate biliary epithelial cell (BEC)-to-hepatocyte conversion. Investigation in zebrafish establishes that blocking VEGF receptors abrogates BEC-driven liver repair, while VEGFA overexpression promotes it. Delivery of VEGFA via non-integrative and safe nucleoside-modified mRNA encapsulated into lipid-nanoparticles (mRNA-LNP) in acutely or chronically injured mouse livers induces robust BEC-to-hepatocyte conversion and reversion of steatosis and fibrosis. In human and murine diseased livers, we further identified VEGFA-receptor KDR-expressing BECs associated with KDR-expressing cell-derived hepatocytes. This defines KDR-expressing cells, most likely being BECs, as facultative progenitors. This study reveals novel therapeutic benefits of VEGFA delivered via nucleoside-modified mRNA-LNP, whose safety is widely validated with COVID-19 vaccines, for harnessing BEC-driven repair to potentially treat liver diseases. Highlights Complementary mouse and zebrafish models of liver injury demonstrate the therapeutic impact of VEGFA-KDR axis activation to harness BEC-driven liver regeneration.VEGFA mRNA LNPs restore two key features of the chronic liver disease in humans such as steatosis and fibrosis.Identification in human cirrhotic ESLD livers of KDR-expressing BECs adjacent to clusters of KDR+ hepatocytes suggesting their BEC origin.KDR-expressing BECs may represent facultative adult progenitor cells, a unique BEC population that has yet been uncovered.
Collapse
|
5
|
Zhou S, Rao Z, Xia Y, Wang Q, Liu Z, Wang P, Cheng F, Zhou H. CCAAT/Enhancer-binding Protein Homologous Protein Promotes ROS-mediated Liver Ischemia and Reperfusion Injury by Inhibiting Mitophagy in Hepatocytes. Transplantation 2023; 107:129-139. [PMID: 35821597 PMCID: PMC9746334 DOI: 10.1097/tp.0000000000004244] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/28/2022] [Accepted: 05/19/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Liver ischemia and reperfusion (IR) injury represent a major risk factor in both partial hepatectomy and liver transplantation. CCAAT/enhancer-binding protein homologous protein (CHOP) is a key regulator of cell death, its precise molecular basis in regulating hepatocyte death during liver IR has not been delineated. METHODS Hepatocellular CHOP deficient mice were generated by bone marrow chimera models using global CHOP knockout mice. Liver partial warm ischemia model and hypoxia/reoxygenation model of primary hepatocytes were applied. Liver injury and mitophagy-related signaling pathways were investigated. IR-stressed patient liver tissues and serum samples were analyzed as well. RESULTS Mice with hepatocellular CHOP deficiency exhibited alleviated cell death, decreased reactive oxygen species (ROS) expression, and enhanced mitophagy in hepatocytes after IR, confirmed by in vitro studies of hepatocytes after hypoxia/reoxygenation. Mitochondria ROS scavenge by Mito TEMPO effectively attenuated hepatocyte death and liver IR injury of wild-type mice, whereas no significant effects were observed in hepatocellular CHOP -deficient mice. CHOP depletion upregulated dynamin-related protein 1 and Beclin-1 activation in the mitochondria of hepatocytes leading to enhanced mitophagy. Following IR, increased CHOP expression and impaired mitophagy activation were observed in the livers of patients undergoing hepatectomy. N-acetyl cysteine pretreatment significantly improved the liver function of patients after surgery. CONCLUSIONS IR-induced CHOP activation exacerbates ROS-mediated hepatocyte death by inhibiting dynamin-related protein 1-Beclin-1-dependent mitophagy.
Collapse
Affiliation(s)
- Shun Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Zhuqing Rao
- Department of Anesthesiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yongxiang Xia
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Qi Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- School of Medical, Southeast University, Nanjing, China
| | - Zheng Liu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Ping Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Feng Cheng
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Haoming Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| |
Collapse
|
6
|
Effects of Pro-Inflammatory Cytokines on Hepatic Metabolism in Primary Human Hepatocytes. Int J Mol Sci 2022; 23:ijms232314880. [PMID: 36499207 PMCID: PMC9740548 DOI: 10.3390/ijms232314880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Three decades of hepatocyte transplantation have confirmed such a cell-based approach as an adjunct or alternative treatment to solid organ transplantation. Donor cell survival and engraftment were indirectly measured by hepatospecific secretive or released metabolites, such as ammonia metabolism in urea cycle defects. In cases of sepsis or viral infection, ammonia levels can significantly and abruptly increase in these recipients, erroneously implying rejection. Pro-inflammatory cytokines associated with viral or bacterial infections are known to affect many liver functions, including drug-metabolizing enzymes and hepatic transport activities. We examined the influence of pro-inflammatory cytokines in primary human hepatocytes, isolated from both normal donors or patients with metabolic liver diseases. Different measures of hepatocyte functions, including ammonia metabolism and phase 1-3 metabolism, were performed. All the hepatic functions were profoundly and significantly suppressed after exposure to concentrations of from 0.1 to 10 ng/mL of different inflammatory cytokines, alone and in combination. Our data indicate that, like phase I metabolism, suppression of phase II/III and ammonia metabolism occurs in hepatocytes exposed to pro-inflammatory cytokines in the absence of cell death. Such inflammatory events do not necessarily indicate a rejection response or loss of the cell graft, and these systemic inflammatory signals should be carefully considered when the immunosuppressant regiment is reduced or relieved in a hepatocyte transplantation recipient in response to such alleged rejection.
Collapse
|
7
|
Pozzobon M, D’Agostino S, Roubelakis MG, Cargnoni A, Gramignoli R, Wolbank S, Gindraux F, Bollini S, Kerdjoudj H, Fenelon M, Di Pietro R, Basile M, Borutinskaitė V, Piva R, Schoeberlein A, Eissner G, Giebel B, Ponsaerts P. General consensus on multimodal functions and validation analysis of perinatal derivatives for regenerative medicine applications. Front Bioeng Biotechnol 2022; 10:961987. [PMID: 36263355 PMCID: PMC9574482 DOI: 10.3389/fbioe.2022.961987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 07/01/2022] [Indexed: 11/26/2022] Open
Abstract
Perinatal tissues, such as placenta and umbilical cord contain a variety of somatic stem cell types, spanning from the largely used hematopoietic stem and progenitor cells to the most recently described broadly multipotent epithelial and stromal cells. As perinatal derivatives (PnD), several of these cell types and related products provide an interesting regenerative potential for a variety of diseases. Within COST SPRINT Action, we continue our review series, revising and summarizing the modalities of action and proposed medical approaches using PnD products: cells, secretome, extracellular vesicles, and decellularized tissues. Focusing on the brain, bone, skeletal muscle, heart, intestinal, liver, and lung pathologies, we discuss the importance of potency testing in validating PnD therapeutics, and critically evaluate the concept of PnD application in the field of tissue regeneration. Hereby we aim to shed light on the actual therapeutic properties of PnD, with an open eye for future clinical application. This review is part of a quadrinomial series on functional/potency assays for validation of PnD, spanning biological functions, such as immunomodulation, anti-microbial/anti-cancer, anti-inflammation, wound healing, angiogenesis, and regeneration.
Collapse
Affiliation(s)
- Michela Pozzobon
- Department of Women’s and Children’s Health, University of Padova, Padova, Italy
| | - Stefania D’Agostino
- Department of Women’s and Children’s Health, University of Padova, Padova, Italy
| | - Maria G. Roubelakis
- Laboratory of Biology, Medical School of Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Cargnoni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, The Research Center in Cooperation with AUVA Trauma Research Center, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Florelle Gindraux
- Service de Chirurgie Orthopédique, Traumatologique et plastique, CHU Besançon, Laboratoire de Nanomédecine, Imagerie, Thérapeutique EA 4662, University Bourgogne Franche-Comté, Besançon, France
| | - Sveva Bollini
- Department of Experimental Medicine (DIMES), School of Medical and Pharmaceutical Sciences, University of Genova, Genova, Italy
| | - Halima Kerdjoudj
- University of Reims Champagne Ardenne, EA 4691 BIOS “Biomatériaux et Inflammation en Site Osseux”, UFR d’Odontologie, Reims, France
| | | | - Roberta Di Pietro
- Department of Medicine and Ageing Sciences, Section of Biomorphology, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Mariangela Basile
- Department of Medicine and Ageing Sciences, Section of Biomorphology, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Veronika Borutinskaitė
- Department of Molecular Cell Biology, Institute of Biochemistry, Vilnius University, Vilnius, Lithuania
| | - Roberta Piva
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Andreina Schoeberlein
- Department of Obstetrics and Feto-maternal Medicine, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Guenther Eissner
- Systems Biology Ireland, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
8
|
Bluhme E, Henckel E, Gramignoli R, Kjellin T, Hammarstedt C, Nowak G, Karadagi A, Johansson H, Jynge Ö, Söderström M, Fischler B, Strom S, Ellis E, Hallberg B, Jorns C. Procurement and Evaluation of Hepatocytes for Transplantation From Neonatal Donors After Circulatory Death. Cell Transplant 2022; 31:9636897211069900. [PMID: 35094608 PMCID: PMC8811420 DOI: 10.1177/09636897211069900] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hepatocyte transplantation is a promising treatment for liver failure and inborn metabolic liver diseases, but progress has been hampered by a scarcity of available organs. Here, hepatocytes isolated from livers procured for a neonatal hepatocyte donation program within a research setting were assessed for metabolic function and suitability for transplantation. Organ donation was considered for infants who died in neonatal intensive care in the Stockholm region during 2015–2021. Inclusion was assessed when a decision to discontinue life-sustaining treatment had been made and hepatectomy performed after declaration of death. Hepatocyte isolation was performed by three-step collagenase perfusion. Hepatocyte viability, yield, and function were assessed using fresh and cryopreserved cells. Engraftment and maturation of cryopreserved neonatal hepatocytes were assessed by transplantation into an immunodeficient mouse model and analysis of the gene expression of phase I, phase II, and liver-specific enzymes and proteins. Twelve livers were procured. Median warm ischemia time (WIT) was 190 [interquartile range (IQR): 80–210] minutes. Median viability was 86% (IQR: 71%–91%). Median yield was 6.9 (IQR: 3.4–12.8) x106 viable hepatocytes/g. Transplantation into immunodeficient mice resulted in good engraftment and maturation of hepatocyte-specific proteins and enzymes. A neonatal organ donation program including preterm born infants was found to be feasible. Hepatocytes isolated from neonatal donors had good viability, function, and engraftment despite prolonged WIT. Therefore, neonatal livers should be considered as a donor source for clinical hepatocyte transplantation, even in cases with extended WIT.
Collapse
Affiliation(s)
- Emil Bluhme
- Department of Clinical Science, Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Transplantation, Karolinska University Hospital, Stockholm, Sweden
| | - Ewa Henckel
- Department of Clinical Science, Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Neonatology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Roberto Gramignoli
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Therese Kjellin
- Department of Neonatology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Christina Hammarstedt
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Greg Nowak
- Department of Clinical Science, Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Transplantation, Karolinska University Hospital, Stockholm, Sweden
| | - Ahmad Karadagi
- Department of Clinical Science, Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Transplantation, Karolinska University Hospital, Stockholm, Sweden
| | - Helene Johansson
- Department of Clinical Science, Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Öystein Jynge
- Organisation for Organ Donation in Central Sweden, Stockholm, Sweden
| | - Maria Söderström
- Organisation for Organ Donation in Central Sweden, Stockholm, Sweden
| | - Björn Fischler
- Department of Clinical Science, Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Pediatric Gastroenterology, Hepatology and Nutrition, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Stephen Strom
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Ewa Ellis
- Department of Clinical Science, Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Boubou Hallberg
- Department of Clinical Science, Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Carl Jorns
- Department of Clinical Science, Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Transplantation, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
9
|
Selvam AK, Jawad R, Gramignoli R, Achour A, Salter H, Björnstedt M. A Novel mRNA-Mediated and MicroRNA-Guided Approach to Specifically Eradicate Drug-Resistant Hepatocellular Carcinoma Cell Lines by Se-Methylselenocysteine. Antioxidants (Basel) 2021; 10:1094. [PMID: 34356326 PMCID: PMC8301172 DOI: 10.3390/antiox10071094] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 01/25/2023] Open
Abstract
Despite progress in the treatment of non-visceral malignancies, the prognosis remains poor for malignancies of visceral organs and novel therapeutic approaches are urgently required. We evaluated a novel therapeutic regimen based on treatment with Se-methylselenocysteine (MSC) and concomitant tumor-specific induction of Kynurenine aminotransferase 1 (KYAT1) in hepatocellular carcinoma (HCC) cell lines, using either vector-based and/or lipid nanoparticle-mediated delivery of mRNA. Supplementation of MSC in KYAT1 overexpressed cells resulted in significantly increased cytotoxicity, due to ROS formation, as compared to MSC alone. Furthermore, microRNA antisense-targeted sites for miR122, known to be widely expressed in normal hepatocytes while downregulated in hepatocellular carcinoma, were added to specifically limit cytotoxicity in HCC cells, thereby limiting the off-target effects. KYAT1 expression was significantly reduced in cells with high levels of miR122 supporting the concept of miR-guided induction of tumor-specific cytotoxicity. The addition of alpha-ketoacid favored the production of methylselenol, enhancing the cytotoxic efficacy of MSC in HCC cells, with no effects on primary human hepatocytes. Altogether, the proposed regimen offers great potential to safely and specifically target hepatic tumors that are currently untreatable.
Collapse
Affiliation(s)
- Arun Kumar Selvam
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Karolinska University Hospital, S-141 86 Stockholm, Sweden; (A.K.S.); (R.J.); (R.G.); (H.S.)
| | - Rim Jawad
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Karolinska University Hospital, S-141 86 Stockholm, Sweden; (A.K.S.); (R.J.); (R.G.); (H.S.)
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Karolinska University Hospital, S-141 86 Stockholm, Sweden; (A.K.S.); (R.J.); (R.G.); (H.S.)
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, & Division of Infectious Diseases, Karolinska University Hospital, SE-171 77 Solna, Sweden;
| | - Hugh Salter
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Karolinska University Hospital, S-141 86 Stockholm, Sweden; (A.K.S.); (R.J.); (R.G.); (H.S.)
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | - Mikael Björnstedt
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Karolinska University Hospital, S-141 86 Stockholm, Sweden; (A.K.S.); (R.J.); (R.G.); (H.S.)
| |
Collapse
|
10
|
Gene Editing Correction of a Urea Cycle Defect in Organoid Stem Cell Derived Hepatocyte-like Cells. Int J Mol Sci 2021; 22:ijms22031217. [PMID: 33530582 PMCID: PMC7865883 DOI: 10.3390/ijms22031217] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 12/17/2022] Open
Abstract
Urea cycle disorders are enzymopathies resulting from inherited deficiencies in any genes of the cycle. In severe cases, currently available therapies are marginally effective, with liver transplantation being the only definitive treatment. Donor liver availability can limit even this therapy. Identification of novel therapeutics for genetic-based liver diseases requires models that provide measurable hepatic functions and phenotypes. Advances in stem cell and genome editing technologies could provide models for the investigation of cell-based genetic diseases, as well as the platforms for drug discovery. This report demonstrates a practical, and widely applicable, approach that includes the successful reprogramming of somatic cells from a patient with a urea cycle defect, their genetic correction and differentiation into hepatic organoids, and the subsequent demonstration of genetic and phenotypic change in the edited cells consistent with the correction of the defect. While individually rare, there is a large number of other genetic-based liver diseases. The approach described here could be applied to a broad range and a large number of patients with these hepatic diseases where it could serve as an in vitro model, as well as identify successful strategies for corrective cell-based therapy.
Collapse
|
11
|
Fontes P, Komori J, Lopez R, Marsh W, Lagasse E. Development of Ectopic Livers by Hepatocyte Transplantation Into Swine Lymph Nodes. Liver Transpl 2020; 26:1629-1643. [PMID: 32810371 PMCID: PMC7756213 DOI: 10.1002/lt.25872] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 08/01/2020] [Accepted: 08/09/2020] [Indexed: 12/16/2022]
Abstract
Orthotopic liver transplantation continues to be the only effective therapy for patients with end-stage liver disease. Unfortunately, many of these patients are not considered transplant candidates, lacking effective therapeutic options that would address both the irreversible progression of their hepatic failure and the control of their portal hypertension. In this prospective study, a swine model was exploited to induce subacute liver failure. Autologous hepatocytes, isolated from the left hepatic lobe, were transplanted into the mesenteric lymph nodes (LNs) by direct cell injection. At 30-60 days after transplantation, hepatocyte engraftment in LNs was successfully identified in all transplanted animals with the degree of ectopic liver mass detected being proportional to the induced native liver injury. These ectopic livers developed within the LNs showed remarkable histologic features of swine hepatic lobules, including the formation of sinusoids and bile ducts. On the basis of our previous tyrosinemic mouse model and the present pig models of induced subacute liver failure, the generation of auxiliary liver tissue using the LNs as hepatocyte engraftment sites represents a potential therapeutic approach to supplement declining hepatic function in the treatment of liver disease.
Collapse
Affiliation(s)
- Paulo Fontes
- WVU MedicineDepartment of SurgerySchool of MedicineWest Virginia UniversityMorgantownWV,LyGenesis, Inc.PittsburghPA
| | - Junji Komori
- McGowan Institute for Regenerative MedicineDepartment of PathologySchool of MedicineUniversity of PittsburghPittsburghPA,Department of SurgeryTakamatsu Red Cross HospitalKagawaJapan
| | - Roberto Lopez
- WVU MedicineDepartment of SurgerySchool of MedicineWest Virginia UniversityMorgantownWV,LyGenesis, Inc.PittsburghPA
| | - Wallis Marsh
- WVU MedicineDepartment of SurgerySchool of MedicineWest Virginia UniversityMorgantownWV
| | - Eric Lagasse
- LyGenesis, Inc.PittsburghPA,McGowan Institute for Regenerative MedicineDepartment of PathologySchool of MedicineUniversity of PittsburghPittsburghPA
| |
Collapse
|
12
|
Hendrawan S, Bono E, Hutter A, Weber U, Lheman J, Baer HU. Evaluation of 3D PLLA scaffolds coated with nano-thick collagen as carrier for hepatocytes. J Biomed Mater Res B Appl Biomater 2020; 109:723-732. [PMID: 33063448 DOI: 10.1002/jbm.b.34738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 09/09/2020] [Accepted: 09/30/2020] [Indexed: 12/30/2022]
Abstract
Orthotopic liver transplantation is presently the most effectual method for the treatment of end-stage liver diseases. Though, one major issue is the restricted number of donor organs that are accessible. Hence, liver tissue engineering is under investigation with the goal of restoring liver functions. In this study, we investigated 3D porous scaffolds made of PLLA coated with a nano thick collagen layer (matrices). Primary rat dermal fibroblasts were used in a first study phase to check matrices' cytocompatibility. More than 70% of seeded cells could adhere and remain viable 24 and 48 hours after the seeding. To test the suitability of the matrices for human primary hepatocytes, HepaRG cells were seeded and analyzed for viability, adhesion rate, and functionality such as albumin secretion. About 80% of seeded HepaRG adhered to the scaffolds remaining viable up to 72 hours. Cells were homogeneously distributed in the entire scaffold with albumin secretion increasing with time. Our results indicate that PLLA collagen-coated matrices allow hepatocytes attachment and distribution throughout the 3D structure, as well as support cell functionality. Such matrices have been applied in our clinical phase II trial. Functional hepatocytes were successfully implanted in patients suffering from liver-cirrhosis with higher cell numbers and adhesions rate compared to our previous trial with the first matrix type and a general improvement in clinical condition.
Collapse
Affiliation(s)
- Siufui Hendrawan
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Tarumanagara University, Jakarta, Indonesia.,Tarumanagara Human Cell Technology Laboratory, Jakarta, Indonesia
| | - Epifania Bono
- Zürich University of Applied Sciences, Institute of Chemistry and Biotechnology, Wädenswil, Switzerland
| | - Albert Hutter
- Zürich University of Applied Sciences, Institute of Chemistry and Biotechnology, Wädenswil, Switzerland
| | - Ursula Weber
- Tarumanagara Human Cell Technology Laboratory, Jakarta, Indonesia.,Baermed, Centre of Abdominal Surgery, Hirslanden Clinic, Zürich, Switzerland.,Department of Visceral and Transplantation Surgery, University of Bern, Bern, Switzerland
| | - Jennifer Lheman
- Tarumanagara Human Cell Technology Laboratory, Jakarta, Indonesia
| | - Hans U Baer
- Baermed, Centre of Abdominal Surgery, Hirslanden Clinic, Zürich, Switzerland.,Department of Visceral and Transplantation Surgery, University of Bern, Bern, Switzerland.,Rumah Sakit Gading Pluit, Jakarta, Indonesia
| |
Collapse
|
13
|
Systemic modified messenger RNA for replacement therapy in alpha 1-antitrypsin deficiency. Sci Rep 2020; 10:7052. [PMID: 32341402 PMCID: PMC7184591 DOI: 10.1038/s41598-020-64017-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 04/09/2020] [Indexed: 12/31/2022] Open
Abstract
Alpha 1-antitrypsin (AAT) deficiency arises from an inherited mutation in the SERPINA1 gene. The disease causes damage in the liver where the majority of the AAT protein is produced. Lack of functioning circulating AAT protein also causes uninhibited elastolytic activity in the lungs leading to AAT deficiency-related emphysema. The only therapy apart from liver transplantation is augmentation with human AAT protein pooled from sera, which is only reserved for patients with advanced lung disease caused by severe AAT deficiency. We tested modified mRNA encoding human AAT in primary human hepatocytes in culture, including hepatocytes from AAT deficient patients. Both expression and functional activity were investigated. Secreted AAT protein increased from 1,14 to 3,43 µg/ml in media from primary human hepatocytes following mRNA treatment as investigated by ELISA and western blot. The translated protein showed activity and protease inhibitory function as measured by elastase activity assay. Also, mRNA formulation in lipid nanoparticles was assessed for systemic delivery in both wild type mice and the NSG-PiZ transgenic mouse model of AAT deficiency. Systemic intravenous delivery of modified mRNA led to hepatic uptake and translation into a functioning protein in mice. These data support the use of systemic mRNA therapy as a potential treatment for AAT deficiency.
Collapse
|
14
|
Florentino RM, Fraunhoffer NA, Morita K, Takeishi K, Ostrowska A, Achreja A, Animasahun O, Haep N, Arazov S, Agarwal N, Collin de l'Hortet A, Guzman-Lepe J, Tafaleng EN, Mukherjee A, Troy K, Banerjee S, Paranjpe S, Michalopoulos GK, Bell A, Nagrath D, Hainer SJ, Fox IJ, Soto-Gutierrez A. Cellular Location of HNF4α is Linked With Terminal Liver Failure in Humans. Hepatol Commun 2020; 4:859-875. [PMID: 32490322 PMCID: PMC7262291 DOI: 10.1002/hep4.1505] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/18/2020] [Accepted: 02/25/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatocyte nuclear factor 4 alpha (HNF4α) is a transcription factor that plays a critical role in hepatocyte function, and HNF4α‐based reprogramming corrects terminal liver failure in rats with chronic liver disease. In the livers of patients with advanced cirrhosis, HNF4α RNA expression levels decrease as hepatic function deteriorates, and protein expression is found in the cytoplasm. These findings could explain impaired hepatic function in patients with degenerative liver disease. In this study, we analyzed HNF4α localization and the pathways involved in post‐translational modification of HNF4α in human hepatocytes from patients with decompensated liver function. RNA‐sequencing analysis revealed that AKT‐related pathways, specifically phospho‐AKT, is down‐regulated in cirrhotic hepatocytes from patients with terminal failure, in whom nuclear levels of HNF4α were significantly reduced, and cytoplasmic expression of HNF4α was increased. cMET was also significantly reduced in failing hepatocytes. Moreover, metabolic profiling showed a glycolytic phenotype in failing human hepatocytes. The contribution of cMET and phospho‐AKT to nuclear localization of HNF4α was confirmed using Spearman's rank correlation test and pathway analysis, and further correlated with hepatic dysfunction by principal component analysis. HNF4α acetylation, a posttranslational modification important for nuclear retention, was also significantly reduced in failing human hepatocytes when compared with normal controls. Conclusion: These results suggest that the alterations in the cMET‐AKT pathway directly correlate with HNF4α localization and level of hepatocyte dysfunction. This study suggests that manipulation of HNF4α and pathways involved in HNF4α posttranslational modification may restore hepatocyte function in patients with terminal liver failure.
Collapse
Affiliation(s)
- Rodrigo M Florentino
- Department of Pathology University of Pittsburgh Pittsburgh PA.,Department of Physiology and Biophysics Universidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Nicolas A Fraunhoffer
- Department of Pathology University of Pittsburgh Pittsburgh PA.,Facultad de Ciencias de la Salud Carrera de Medicina Universidad Maimónides Buenos Aires Argentina.,Centro de Estudios Farmacológicos y Botánicos-CONICET Buenos Aires Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas Buenos Aires Argentina
| | - Kazutoyo Morita
- Department of Pathology University of Pittsburgh Pittsburgh PA
| | - Kazuki Takeishi
- Department of Pathology University of Pittsburgh Pittsburgh PA.,Department of Surgery and Science Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Alina Ostrowska
- Department of Surgery Children's Hospital of Pittsburgh of UPMC University of Pittsburgh Pittsburgh PA
| | - Abhinav Achreja
- Laboratory for Systems Biology of Human Diseases Department of Biomedical Engineering Biointerfaces Institute University of Michigan Ann Arbor MI
| | - Olamide Animasahun
- Laboratory for Systems Biology of Human Diseases Department of Biomedical Engineering Biointerfaces Institute University of Michigan Ann Arbor MI
| | - Nils Haep
- Department of Pathology University of Pittsburgh Pittsburgh PA
| | - Shohrat Arazov
- Department of Pathology University of Pittsburgh Pittsburgh PA
| | - Nandini Agarwal
- Department of Pathology University of Pittsburgh Pittsburgh PA.,School of Bioscience and Technology Vellore Institute of Technology Vellore India
| | | | | | - Edgar N Tafaleng
- Department of Surgery Children's Hospital of Pittsburgh of UPMC University of Pittsburgh Pittsburgh PA
| | - Amitava Mukherjee
- Department of Surgery Children's Hospital of Pittsburgh of UPMC University of Pittsburgh Pittsburgh PA
| | - Kris Troy
- Department of Biological Sciences University of Pittsburgh Pittsburgh PA
| | - Swati Banerjee
- Department of Pathology University of Pittsburgh Pittsburgh PA
| | | | | | - Aaron Bell
- Department of Pathology University of Pittsburgh Pittsburgh PA
| | - Deepak Nagrath
- Laboratory for Systems Biology of Human Diseases Department of Biomedical Engineering Biointerfaces Institute University of Michigan Ann Arbor MI.,Department of Chemical Engineering and Rogel Cancer Center University of Michigan Ann Arbor MI
| | - Sarah J Hainer
- Department of Biological Sciences University of Pittsburgh Pittsburgh PA
| | - Ira J Fox
- Department of Surgery Children's Hospital of Pittsburgh of UPMC University of Pittsburgh Pittsburgh PA
| | | |
Collapse
|
15
|
Tanaka K, Okitsu T, Teramura N, Iijima K, Hayashida O, Teramae H, Hattori S. Recombinant collagenase from Grimontia hollisae as a tissue dissociation enzyme for isolating primary cells. Sci Rep 2020; 10:3927. [PMID: 32127566 PMCID: PMC7054364 DOI: 10.1038/s41598-020-60802-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 02/17/2020] [Indexed: 11/09/2022] Open
Abstract
Collagenase products are crucial to isolate primary cells in basic research and clinical therapies, where their stability in collagenolytic activity is required. However, currently standard collagenase products from Clostridium histolyticum lack such stability. Previously, we produced a recombinant 74-kDa collagenase from Grimontia hollisae, which spontaneously became truncated to ~60 kDa and possessed no stability. In this study, to generate G. hollisae collagenase useful as a collagenase product, we designed recombinant 62-kDa collagenase consisting only of the catalytic domain, which exhibits high production efficiency. We demonstrated that this recombinant collagenase is stable and active under physiological conditions. Moreover, it possesses higher specific activity against collagen and cleaves a wider variety of collagens than a standard collagenase product from C. histolyticum. Furthermore, it dissociated murine pancreata by digesting the collagens within the pancreata in a dose-dependent manner, and this dissociation facilitated isolation of pancreatic islets with masses and numbers comparable to those isolated using the standard collagenase from C. histolyticum. Implantation of these isolated islets into five diabetic mice led to normalisation of the blood glucose concentrations of all the recipients. These findings suggest that recombinant 62-kDa collagenase from G. hollisae can be used as a collagenase product to isolate primary cells.
Collapse
Affiliation(s)
- Keisuke Tanaka
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, 302-0017, Japan.
| | - Teru Okitsu
- Institute of Industrial Science, The University of Tokyo, Meguro, Tokyo, 153-8904, Japan.
| | - Naoko Teramura
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, 302-0017, Japan
| | - Katsumasa Iijima
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, 302-0017, Japan
| | - Osamu Hayashida
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, 302-0017, Japan
| | - Hiroki Teramae
- Faculty of Teacher Education, Shumei University, Yachiyo, Chiba, 276-0003, Japan
| | - Shunji Hattori
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, 302-0017, Japan
| |
Collapse
|
16
|
Aging and Caloric Restriction Modulate the DNA Methylation Profile of the Ribosomal RNA Locus in Human and Rat Liver. Nutrients 2020; 12:nu12020277. [PMID: 31973116 PMCID: PMC7070571 DOI: 10.3390/nu12020277] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/13/2020] [Accepted: 01/17/2020] [Indexed: 12/21/2022] Open
Abstract
A growing amount of evidence suggests that the downregulation of protein synthesis is an adaptive response during physiological aging, which positively contributes to longevity and can be modulated by nutritional interventions like caloric restriction (CR). The expression of ribosomal RNA (rRNA) is one of the main determinants of translational rate, and epigenetic modifications finely contribute to its regulation. Previous reports suggest that hypermethylation of ribosomal DNA (rDNA) locus occurs with aging, although with some species- and tissue- specificity. In the present study, we experimentally measured DNA methylation of three regions (the promoter, the 5′ of the 18S and the 5′ of 28S sequences) in the rDNA locus in liver tissues from rats at two, four, 10, and 18 months. We confirm previous findings, showing age-related hypermethylation, and describe, for the first time, that this gain in methylation also occurs in human hepatocytes. Furthermore, we show that age-related hypermethylation is enhanced in livers of rat upon CR at two and 10 months, and that at two months a trend towards the reduction of rRNA expression occurs. Collectively, our results suggest that CR modulates age-related regulation of methylation at the rDNA locus, thus providing an epigenetic readout of the pro-longevity effects of CR.
Collapse
|
17
|
Tokodai K, Kumagai-Braesch M, Karadagi A, Johansson H, Ågren N, Jorns C, Ericzon BG, Ellis E. Blood Group Antigen Expression in Isolated Human Liver Cells in Preparation for Implementing Clinical ABO-Incompatible Hepatocyte Transplantation. J Clin Exp Hepatol 2020; 10:106-113. [PMID: 32189925 PMCID: PMC7068001 DOI: 10.1016/j.jceh.2019.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 07/05/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND ABO blood group antigens in the liver are expressed mainly on endothelial cells or biliary epithelial cells but not on hepatocytes. This suggests that ABO-incompatible hepatocyte transplantation (ABOi-HTx) is theoretically feasible. However, the effects of stress on ABO blood group antigen expression caused by isolation and intraportal infusion require thorough investigation before ABOi-HTx can be implemented in clinical settings. METHODS Human hepatocytes were isolated from liver tissue obtained from liver resection or deceased donor livers. The expression of blood group antigens on cryopreserved human liver tissues and isolated hepatocyte smear specimens were examined by immunofluorescent staining. The effect of proinflammatory cytokines on blood group antigen expression of hepatocytes was evaluated by flow cytometry. Instant blood-mediated inflammatory reaction after hepatocyte incubation with ABO-incompatible whole blood was examined using the tubing loop model. RESULTS Blood group antigens were mainly expressed on vessels in the portal area. In hepatocyte smear specimens, isolated hepatocytes did not express blood group antigens. In contrast, a subset of cells in the smear specimens of nonparenchymal liver cells stained positive. In the flow cytometry analysis, isolated hepatocytes were negative for blood group antigens, even after 4-h incubation with cytokines. Platelet counts and complement activation were not significantly different in ABO-identical versus ABO-incompatible settings in the tubing loop model. CONCLUSION Our study showed that blood group antigens were not expressed on hepatocytes, even after isolation procedures or subsequent incubation with cytokines. This finding is an important step toward removing the restriction of ABO matching in hepatocyte transplantation. Our results suggest that ABOi-HTx is a feasible therapeutic option, especially in patients who require urgent treatment with freshly isolated hepatocytes, such as those with acute liver failure.
Collapse
Key Words
- ABO-incompatible transplantation
- ABOi, ABO-incompatible
- ABOi-HTx, ABO-incompatible hepatocyte transplantation
- C3a, Complement 3a
- CCA, Cholangiocarcinoma
- CRC, Colorectal cancer metastasis
- DCD, Donation after death
- DSA, Donor-specific anti-HLA antibody
- HCC, Hepatocellular carcinoma
- HTx, Hepatocyte transplantation
- IBMIR, Instant blood-mediated inflammatory reaction
- ICH, Intracranial hemorrhage
- IQR, Interquartile range
- MSCs, Mesenchymal stem cells
- blood group antigens
- hepatocytes
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ewa Ellis
- Address for correspondence: Ewa Ellis Ph.D., Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Karolinska Institute, SE-141 86, Stockholm, Sweden.
| |
Collapse
|
18
|
Heibel SK, McGuire PJ, Haskins N, Datta Majumdar H, Rayavarapu S, Nagaraju K, Hathout Y, Brown K, Tuchman M, Caldovic L. AMP-activated protein kinase signaling regulated expression of urea cycle enzymes in response to changes in dietary protein intake. J Inherit Metab Dis 2019; 42:1088-1096. [PMID: 31177541 PMCID: PMC7385982 DOI: 10.1002/jimd.12133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 04/02/2019] [Accepted: 06/05/2019] [Indexed: 12/30/2022]
Abstract
Abundance of urea cycle enzymes in the liver is regulated by dietary protein intake. Although urea cycle enzyme levels rise in response to a high-protein (HP) diet, signaling networks that sense dietary protein intake and trigger changes in expression of urea cycle genes have not been identified. The aim of this study was to identify signaling pathway(s) that respond to changes in protein intake and regulate expression of urea cycle genes in mice and human hepatocytes. Mice were adapted to either HP or low-protein diets followed by isolation of liver protein and mRNA and integrated analysis of the proteomic and transcriptomic data. HP diet led to increased expression of mRNA and enzymes in amino acid degradation pathways and decreased expression of mRNA and enzymes in carbohydrate and fat metabolism, which implicated adenosine monophosphate-activated protein kinase (AMPK) as a possible regulator. Primary human hepatocytes, treated with 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) an activator of AMPK, were used to test whether AMPK regulates expression of urea cycle genes. The abundance of carbamoylphosphate synthetase 1 and ornithine transcarbamylase mRNA increased in hepatocytes treated with AICAR, which supports a role for AMPK signaling in regulation of the urea cycle. Because AMPK is either a target of drugs used to treat type-2 diabetes, these drugs might increase the expression of urea cycle enzymes in patients with partial urea cycle disorders, which could be the basis of a new therapeutic approach.
Collapse
Affiliation(s)
- Sandra Kirsch Heibel
- Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Ave NW, Washington DC, USA
| | | | - Nantaporn Haskins
- Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Ave NW, Washington DC, USA
| | - Himani Datta Majumdar
- Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Ave NW, Washington DC, USA
| | - Sree Rayavarapu
- Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Ave NW, Washington DC, USA
| | - Kanneboyina Nagaraju
- Department of Pharmaceutical Sciences, Binghamton University, Binghamton NY, USA
| | - Yetrib Hathout
- Department of Pharmaceutical Sciences, Binghamton University, Binghamton NY, USA
| | - Kristy Brown
- Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Ave NW, Washington DC, USA
| | - Mendel Tuchman
- Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Ave NW, Washington DC, USA
| | - Ljubica Caldovic
- Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Ave NW, Washington DC, USA
| |
Collapse
|
19
|
Srinivasan RC, Zabulica M, Hammarstedt C, Wu T, Gramignoli R, Kannisto K, Ellis E, Karadagi A, Fingerhut R, Allegri G, Rüfenacht V, Thöny B, Häberle J, Nuoffer JM, Strom SC. A liver-humanized mouse model of carbamoyl phosphate synthetase 1-deficiency. J Inherit Metab Dis 2019; 42:1054-1063. [PMID: 30843237 DOI: 10.1002/jimd.12067] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/25/2019] [Indexed: 12/31/2022]
Abstract
A liver-humanized mouse model for CPS1-deficiency was generated by the high-level repopulation of the mouse liver with CPS1-deficient human hepatocytes. When compared with mice that are highly repopulated with CPS1-proficient human hepatocytes, mice that are repopulated with CPS1-deficient human hepatocytes exhibited characteristic symptoms of human CPS1 deficiency including an 80% reduction in CPS1 metabolic activity, delayed clearance of an ammonium chloride infusion, elevated glutamine and glutamate levels, and impaired metabolism of [15 N]ammonium chloride into urea, with no other obvious phenotypic differences. Because most metabolic liver diseases result from mutations that alter critical pathways in hepatocytes, a model that incorporates actual disease-affected, mutant human hepatocytes is useful for the investigation of the molecular, biochemical, and phenotypic differences induced by that mutation. The model is also expected to be useful for investigations of modified RNA, gene, and cellular and small molecule therapies for CPS1-deficiency. Liver-humanized models for this and other monogenic liver diseases afford the ability to assess the therapy on actual disease-affected human hepatocytes, in vivo, for long periods of time and will provide data that are highly relevant for investigations of the safety and efficacy of gene-editing technologies directed to human hepatocytes and the translation of gene-editing technology to the clinic.
Collapse
Affiliation(s)
- Raghuraman C Srinivasan
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Mihaela Zabulica
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Christina Hammarstedt
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Tingting Wu
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Kristina Kannisto
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Ewa Ellis
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Ahmad Karadagi
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Ralph Fingerhut
- Division of Metabolism and Children's Research Centre (CRC), University Children's Hospital Zurich, Zurich, Switzerland
- Swiss Newborn Screening Laboratory, University Children's Hospital Zurich, Zurich, Switzerland
| | - Gabriella Allegri
- Division of Metabolism and Children's Research Centre (CRC), University Children's Hospital Zurich, Zurich, Switzerland
| | - Véronique Rüfenacht
- Division of Metabolism and Children's Research Centre (CRC), University Children's Hospital Zurich, Zurich, Switzerland
| | - Beat Thöny
- Division of Metabolism and Children's Research Centre (CRC), University Children's Hospital Zurich, Zurich, Switzerland
- Swiss Newborn Screening Laboratory, University Children's Hospital Zurich, Zurich, Switzerland
| | - Johannes Häberle
- Division of Metabolism and Children's Research Centre (CRC), University Children's Hospital Zurich, Zurich, Switzerland
- Zurich Centre for Integrative Human Physiology (ZIHP) and, Neuroscience Centre Zurich (ZNZ), Zurich, Switzerland
| | - Jean-Marc Nuoffer
- Institute for Clinical Chemistry and University Children's Hospital, Bern, Switzerland
| | - Stephen C Strom
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
Bacalini MG, Franceschi C, Gentilini D, Ravaioli F, Zhou X, Remondini D, Pirazzini C, Giuliani C, Marasco E, Gensous N, Di Blasio AM, Ellis E, Gramignoli R, Castellani G, Capri M, Strom S, Nardini C, Cescon M, Grazi GL, Garagnani P. Molecular Aging of Human Liver: An Epigenetic/Transcriptomic Signature. J Gerontol A Biol Sci Med Sci 2019; 74:1-8. [PMID: 29554203 DOI: 10.1093/gerona/gly048] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Indexed: 12/12/2022] Open
Abstract
The feasibility of liver transplantation from old healthy donors suggests that this organ is able to preserve its functionality during aging. To explore the biological basis of this phenomenon, we characterized the epigenetic profile of liver biopsies collected from 45 healthy liver donors ranging from 13 to 90 years old using the Infinium HumanMethylation450 BeadChip. The analysis indicates that a large remodeling in DNA methylation patterns occurs, with 8,823 age-associated differentially methylated CpG probes. Notably, these age-associated changes tended to level off after the age of 60, as confirmed by Horvath's clock. Using stringent selection criteria, we further identified a DNA methylation signature of aging liver including 75 genomic regions. We demonstrated that this signature is specific for liver compared to other tissues and that it is able to detect biological age-acceleration effects associated with obesity. Finally, we combined DNA methylation measurements with available expression data. Although the intersection between the two omic characterizations was low, both approaches suggested a previously unappreciated role of epithelial-mesenchymal transition and Wnt-signaling pathways in the aging of human liver.
Collapse
Affiliation(s)
| | - Claudio Franceschi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Italy.,DIMES-Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, Bologna, Italy.,CIG, Interdepartmental Center 'L. Galvani', Alma Mater Studiorum, Bologna, Italy
| | - Davide Gentilini
- Department of Brain and Behavioral Sciences, University of Pavia, Italy
| | - Francesco Ravaioli
- DIMES-Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, Bologna, Italy
| | - Xiaoyuan Zhou
- Group of Clinical Genomic Networks, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, PR China.,University of Chinese Academy of Sciences, Beijing, PR China.,Department of Neurology, University of San Francisco, California
| | - Daniel Remondini
- Department of Physics and Astronomy (DIFA) and INFN Sez. Bologna, Alma Mater Studiorum, Italy
| | | | - Cristina Giuliani
- Department of Biological Geological and Environmental Sciences, Laboratory of Molecular Anthropology and Centre for Genome Biology, University of Bologna, Italy
| | - Elena Marasco
- CIG, Interdepartmental Center 'L. Galvani', Alma Mater Studiorum, Bologna, Italy
| | - Noémie Gensous
- DIMES-Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, Bologna, Italy
| | | | - Ewa Ellis
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Gastone Castellani
- CIG, Interdepartmental Center 'L. Galvani', Alma Mater Studiorum, Bologna, Italy.,Department of Biological Geological and Environmental Sciences, Laboratory of Molecular Anthropology and Centre for Genome Biology, University of Bologna, Italy
| | - Miriam Capri
- DIMES-Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, Bologna, Italy.,CIG, Interdepartmental Center 'L. Galvani', Alma Mater Studiorum, Bologna, Italy
| | - Stephen Strom
- Department of Laboratory Medicine, Karolinska Institute and Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Christine Nardini
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,CNR IAC "Mauro Picone", Roma, Italy.,Personal Genomics S.r.l., Verona, Italy
| | - Matteo Cescon
- General Surgery and Transplant Unit, Department of Medical and Surgical Sciences, Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | | | - Paolo Garagnani
- DIMES-Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, Bologna, Italy.,CIG, Interdepartmental Center 'L. Galvani', Alma Mater Studiorum, Bologna, Italy.,Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Applied Biomedical Research Center, S. Orsola-Malpighi Polyclinic, Bologna, Italy.,Institute of Molecular Genetics (IGM)-CNR, Unit of Bologna, Italy.,Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopaedic Institute, Bologna, Italy
| |
Collapse
|
21
|
Czaja W, Nakamura YK, Li N, Eldridge JA, DeAvila DM, Thompson TB, Rodgers BD. Myostatin regulates pituitary development and hepatic IGF1. Am J Physiol Endocrinol Metab 2019; 316:E1036-E1049. [PMID: 30888862 PMCID: PMC6620572 DOI: 10.1152/ajpendo.00001.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Circulating myostatin-attenuating agents are being developed to treat muscle-wasting disease despite their potential to produce serious off-target effects, as myostatin/activin receptors are widely distributed among many nonmuscle tissues. Our studies suggest that the myokine not only inhibits striated muscle growth but also regulates pituitary development and growth hormone (GH) action in the liver. Using a novel myostatin-null label-retaining model (Jekyll mice), we determined that the heterogeneous pool of pituitary stem, transit-amplifying, and progenitor cells in Jekyll mice depletes more rapidly after birth than the pool in wild-type mice. This correlated with increased levels of GH, prolactin, and the cells that secrete these hormones, somatotropes and lactotropes, respectively, in Jekyll pituitaries. Recombinant myostatin also stimulated GH release and gene expression in pituitary cell cultures although inhibiting prolactin release. In primary hepatocytes, recombinant myostatin blocked GH-stimulated expression of two key mediators of growth, insulin-like growth factor (IGF)1 and the acid labile subunit and increased expression of an inhibitor, IGF-binding protein-1. The significance of these findings was demonstrated by smaller muscle fiber size in a model lacking myostatin and liver IGF1 expression (LID-o-Mighty mice) compared with that in myostatin-null (Mighty) mice. These data together suggest that myostatin may regulate pituitary development and function and that its inhibitory actions in muscle may be partly mediated by attenuating GH action in the liver. They also suggest that circulating pharmacological inhibitors of myostatin could produce unintended consequences in these and possibly other tissues.
Collapse
Affiliation(s)
- Wioletta Czaja
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
- Department of Biochemistry and Molecular Biology, University of Georgia , Athens, Georgia
| | - Yukiko K Nakamura
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
| | - Naisi Li
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
| | - Jennifer A Eldridge
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
| | - David M DeAvila
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
| | - Thomas B Thompson
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati , Cincinnati, Ohio
| | - Buel D Rodgers
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
- AAVogen, Incorporated, Rockville, Maryland
| |
Collapse
|
22
|
Chen D, Ni HM, Wang L, Ma X, Yu J, Ding WX, Zhang L. p53 Up-regulated Modulator of Apoptosis Induction Mediates Acetaminophen-Induced Necrosis and Liver Injury in Mice. Hepatology 2019; 69:2164-2179. [PMID: 30552702 PMCID: PMC6461480 DOI: 10.1002/hep.30422] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 12/03/2018] [Indexed: 12/28/2022]
Abstract
Acetaminophen (APAP) overdose is one of the leading causes of hepatotoxicity and acute liver failure in the United States. Accumulating evidence suggests that hepatocyte necrosis plays a critical role in APAP-induced liver injury (AILI). However, the mechanisms of APAP-induced necrosis and liver injury are not fully understood. In this study, we found that p53 up-regulated modulator of apoptosis (PUMA), a B-cell lymphoma-2 (Bcl-2) homology domain 3 (BH3)-only Bcl-2 family member, was markedly induced by APAP in mouse livers and in isolated human and mouse hepatocytes. PUMA deficiency suppressed APAP-induced mitochondrial dysfunction and release of cell death factors from mitochondria, and protected against APAP-induced hepatocyte necrosis and liver injury in mice. PUMA induction by APAP was p53 independent, and required receptor-interacting protein kinase 1 (RIP1) and c-Jun N-terminal kinase (JNK) by transcriptional activation. Furthermore, a small-molecule PUMA inhibitor, administered after APAP treatment, mitigated APAP-induced hepatocyte necrosis and liver injury. Conclusion: Our results demonstrate that RIP1/JNK-dependent PUMA induction mediates AILI by promoting hepatocyte mitochondrial dysfunction and necrosis, and suggest that PUMA inhibition is useful for alleviating acute hepatotoxicity attributed to APAP overdose.
Collapse
Affiliation(s)
- Dongshi Chen
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Lei Wang
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Xiaowen Ma
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jian Yu
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA.,Corresponding authors
| | - Lin Zhang
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Corresponding authors
| |
Collapse
|
23
|
Matsumura M, Imura T, Inagaki A, Ogasawara H, Fukuoka K, Fathi I, Miyagi S, Ohashi K, Unno M, Kamei T, Satomi S, Goto M. A Simple and Useful Predictive Assay for Evaluating the Quality of Isolated Hepatocytes for Hepatocyte Transplantation. Sci Rep 2019; 9:6166. [PMID: 30992529 PMCID: PMC6467914 DOI: 10.1038/s41598-019-42720-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/08/2019] [Indexed: 12/13/2022] Open
Abstract
No optimal assay for assessing isolated hepatocytes before hepatocyte transplantation (HTx) has been established, therefore reliable and rapid assays are warranted. Isolated rat hepatocytes were dipped in a water bath (necrosis model), and were also cultured with Okadaic acid (apoptosis model) or vehicle, followed by cellular assessment including trypan blue exclusion (TBE) viability, ADP /ATP ratio, plating efficiency (PE), DNA quantity and ammonia elimination. Hepatocytes were transplanted into the liver of analbuminemic rats, subsequently engraftment was assessed by serum albumin and the histology of transplanted grafts. In the necrosis model, the ADP/ATP ratio was strongly and negatively correlated with the TBE (R2 = 0.559, P < 0.001). In the apoptosis model, the ADP/ATP ratio assay, PE, DNA quantification and an ammonia elimination test clearly distinguished the groups (P < 0.001, respectively). The ADP/ATP ratio, PE and DNA quantity were well-correlated and the ammonia elimination was slightly correlated with the transplant outcome. TBE could not distinguish the groups and was not correlated with the outcome. The ADP/ATP ratio assay predicted the transplant outcome. PE and DNA quantification may improve the accuracy of the retrospective (evaluations require several days) quality assessment of hepatocytes. The ADP/ATP ratio assay, alone or with a short-term metabolic assay could improve the efficiency of HTx.
Collapse
Affiliation(s)
- Muneyuki Matsumura
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan
| | - Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, 980-0872, Sendai, Japan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, 980-0872, Sendai, Japan
| | - Hiroyuki Ogasawara
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan
| | - Kengo Fukuoka
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan
| | - Ibrahim Fathi
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, 980-0872, Sendai, Japan
| | - Shigehito Miyagi
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan
| | - Kazuo Ohashi
- Graduate School of Pharmaceutical Sciences, Osaka University, 565-0871, Osaka, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan
| | - Susumu Satomi
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan
| | - Masafumi Goto
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan. .,Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, 980-0872, Sendai, Japan.
| |
Collapse
|
24
|
Kammerer S, Küpper JH. Human hepatocyte systems for in vitro toxicology analysis. ACTA ACUST UNITED AC 2018. [DOI: 10.3233/jcb-179012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Sarah Kammerer
- Institute of Biotechnology, Brandenburg University of Technology, Cottbus-Senftenberg, Germany
| | - Jan-Heiner Küpper
- Institute of Biotechnology, Brandenburg University of Technology, Cottbus-Senftenberg, Germany
| |
Collapse
|
25
|
Ng SS, Saeb-Parsy K, Blackford SJI, Segal JM, Serra MP, Horcas-Lopez M, No DY, Mastoridis S, Jassem W, Frank CW, Cho NJ, Nakauchi H, Glenn JS, Rashid ST. Human iPS derived progenitors bioengineered into liver organoids using an inverted colloidal crystal poly (ethylene glycol) scaffold. Biomaterials 2018; 182:299-311. [PMID: 30149262 PMCID: PMC6131727 DOI: 10.1016/j.biomaterials.2018.07.043] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 07/25/2018] [Indexed: 12/29/2022]
Abstract
Generation of human organoids from induced pluripotent stem cells (iPSCs) offers exciting possibilities for developmental biology, disease modelling and cell therapy. Significant advances towards those goals have been hampered by dependence on animal derived matrices (e.g. Matrigel), immortalized cell lines and resultant structures that are difficult to control or scale. To address these challenges, we aimed to develop a fully defined liver organoid platform using inverted colloid crystal (ICC) whose 3-dimensional mechanical properties could be engineered to recapitulate the extracellular niche sensed by hepatic progenitors during human development. iPSC derived hepatic progenitors (IH) formed organoids most optimally in ICC scaffolds constructed with 140 μm diameter pores coated with type I collagen in a two-step process mimicking liver bud formation. The resultant organoids were closer to adult tissue, compared to 2D and 3D controls, with respect to morphology, gene expression, protein secretion, drug metabolism and viral infection and could integrate, vascularise and function following implantation into livers of immune-deficient mice. Preliminary interrogation of the underpinning mechanisms highlighted the importance of TGFβ and hedgehog signalling pathways. The combination of functional relevance with tuneable mechanical properties leads us to propose this bioengineered platform to be ideally suited for a range of future mechanistic and clinical organoid related applications.
Collapse
Affiliation(s)
- Soon Seng Ng
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, England, UK; Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge and the Cambridge NIHR Biomedical Research Centre, Cambridge, UK
| | - Samuel J I Blackford
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, England, UK
| | - Joe M Segal
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, England, UK
| | - Maria Paola Serra
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, England, UK
| | - Marta Horcas-Lopez
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, England, UK
| | - Da Yoon No
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Sotiris Mastoridis
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, England, UK
| | - Wayel Jassem
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, England, UK
| | - Curtis W Frank
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Nam Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Hiromitsu Nakauchi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeffrey S Glenn
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| | - S Tamir Rashid
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, England, UK; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
26
|
Comparative study on the gut microbiotas of four economically important Asian carp species. SCIENCE CHINA-LIFE SCIENCES 2018; 61:696-705. [DOI: 10.1007/s11427-016-9296-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/11/2018] [Indexed: 12/24/2022]
|
27
|
Ong J, Serra MP, Segal J, Cujba AM, Ng SS, Butler R, Millar V, Hatch S, Zimri S, Koike H, Chan K, Bonham A, Walk M, Voss T, Heaton N, Mitry R, Dhawan A, Ebner D, Danovi D, Nakauchi H, Rashid ST. Imaging-Based Screen Identifies Laminin 411 as a Physiologically Relevant Niche Factor with Importance for i-Hep Applications. Stem Cell Reports 2018; 10:693-702. [PMID: 29478892 PMCID: PMC5919292 DOI: 10.1016/j.stemcr.2018.01.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/21/2018] [Accepted: 01/22/2018] [Indexed: 12/29/2022] Open
Abstract
Use of hepatocytes derived from induced pluripotent stem cells (i-Heps) is limited by their functional differences in comparison with primary cells. Extracellular niche factors likely play a critical role in bridging this gap. Using image-based characterization (high content analysis; HCA) of freshly isolated hepatocytes from 17 human donors, we devised and validated an algorithm (Hepatocyte Likeness Index; HLI) for comparing the hepatic properties of cells against a physiological gold standard. The HLI was then applied in a targeted screen of extracellular niche factors to identify substrates driving i-Heps closer to the standard. Laminin 411, the top hit, was validated in two additional induced pluripotent stem cell (iPSC) lines, primary tissue, and an in vitro model of α1-antitrypsin deficiency. Cumulatively, these data provide a reference method to control and screen for i-Hep differentiation, identify Laminin 411 as a key niche protein, and underscore the importance of combining substrates, soluble factors, and HCA when developing iPSC applications. iPSC-derived hepatocytes (i-Heps) are functionally limited compared with primary cells Factors within the extracellular niche likely play a role in bridging this gap Laminin 411 was shown to be an important niche factor for i-Heps High content image analysis (HCA) can help development of i-Hep applications
Collapse
Affiliation(s)
- John Ong
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, London SE1 9RT, UK
| | - Maria Paola Serra
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, London SE1 9RT, UK
| | - Joe Segal
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, London SE1 9RT, UK
| | - Ana-Maria Cujba
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, London SE1 9RT, UK
| | - Soon Seng Ng
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, London SE1 9RT, UK
| | - Richard Butler
- The Gurdon Institute Imaging Facility, Cambridge University, Cambridge CB2 1QN, UK
| | - Val Millar
- Target Discovery Institute, Oxford University, Oxford OX3 7FZ, UK
| | - Stephanie Hatch
- Target Discovery Institute, Oxford University, Oxford OX3 7FZ, UK
| | - Salman Zimri
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, London SE1 9RT, UK
| | - Hiroyuki Koike
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Karen Chan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andrew Bonham
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Ty Voss
- Perkin Elmer, Houston, TX 77055, USA
| | - Nigel Heaton
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, London SE1 9RT, UK
| | - Ragai Mitry
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, London SE1 9RT, UK
| | - Anil Dhawan
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, London SE1 9RT, UK
| | - Daniel Ebner
- Target Discovery Institute, Oxford University, Oxford OX3 7FZ, UK
| | - Davide Danovi
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, London SE1 9RT, UK
| | - Hiromitsu Nakauchi
- The Gurdon Institute Imaging Facility, Cambridge University, Cambridge CB2 1QN, UK
| | - S Tamir Rashid
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, London SE1 9RT, UK; The Gurdon Institute Imaging Facility, Cambridge University, Cambridge CB2 1QN, UK.
| |
Collapse
|
28
|
Major RD, Kluge M, Jara M, Nösser M, Horner R, Gassner J, Struecker B, Tang P, Lippert S, Reutzel-Selke A, Geisel D, Denecke T, Stockmann M, Pratschke J, Sauer IM, Raschzok N. The Predictive Value of the Maximal Liver Function Capacity Test for the Isolation of Primary Human Hepatocytes. Tissue Eng Part C Methods 2018; 24:179-186. [PMID: 29382276 DOI: 10.1089/ten.tec.2017.0369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
The need for primary human hepatocytes is constantly growing for basic research, as well as for therapeutic applications. However, the isolation outcome strongly depends on the quality of liver tissue, and we are still lacking a preoperative test that allows the prediction of the hepatocyte isolation outcome. In this study, we evaluated the "maximal liver function capacity test" (LiMAx) as predictive test for the quantitative and qualitative outcome of hepatocyte isolation. This test is already used in clinical routine to measure preoperative and to predict postoperative liver function. The patient's preoperative mean LiMAx was obtained from the patient records, and preoperative computed tomography and magnetic resonance images were used to calculate the whole liver volume to adjust the mean LiMAx. The outcome parameters of the hepatocyte isolation procedures were analyzed in correlation with the adjusted mean LiMAx. Primary human hepatocytes were isolated from partial hepatectomies (n = 64). From these 64 hepatectomies we included 48 to our study and correlated their isolation outcome parameters with volume corrected LiMAx values. From a total of 11 hepatocyte isolation procedures, metabolic parameters (albumin, urea, and aspartate aminotransferase or AST) were assessed during the hepatocyte cultivation period of 5 days. The volume adjusted mean LiMAx showed a significant positive correlation with the total cell yield (p = 0.049; r = 0.242; n = 48). The correlations of volume adjusted LiMAx values with viable cell yield and cell viability did not reach statistical significance. To create a more homogenous study group regarding tumor entities, subgroup analyses were performed. A subgroup analysis of isolations from patients with colorectal metastasis revealed a significant correlation between volume adjusted mean LiMAx and total cell yield (p = 0.012; r = 0.488; n = 21) and viable cell yield (p = 0.034; r = 0.405; n = 21), whereas a subgroup analysis of isolations of patients with carcinoma of the biliary tree showed significant correlations of volume adjusted mean LiMAx with cell viability (r = 0.387; p = 0.046; n = 20) and lacked significant correlations with total cell yield (r = -0.060; p = 0.401; n = 20) and viable cell yield (r = 0.012; p = 0.480; n = 20). The volume-adjusted mean LiMAx did not show a significant correlation with any of the metabolic parameters. In conclusion, the LiMAx test might be a useful tool to predict the quantitative outcome of hepatocyte isolation, as long as underlying liver disease is taken into consideration.
Collapse
Affiliation(s)
- Rebeka D Major
- 1 Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité-Universitatsmedizin Berlin , Berlin, Germany
| | - Martin Kluge
- 1 Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité-Universitatsmedizin Berlin , Berlin, Germany
| | - Maximilian Jara
- 1 Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité-Universitatsmedizin Berlin , Berlin, Germany
| | - Maximilian Nösser
- 1 Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité-Universitatsmedizin Berlin , Berlin, Germany
| | - Rosa Horner
- 1 Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité-Universitatsmedizin Berlin , Berlin, Germany
| | - Joseph Gassner
- 1 Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité-Universitatsmedizin Berlin , Berlin, Germany
| | - Benjamin Struecker
- 1 Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité-Universitatsmedizin Berlin , Berlin, Germany
| | - Peter Tang
- 1 Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité-Universitatsmedizin Berlin , Berlin, Germany
| | - Steffen Lippert
- 1 Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité-Universitatsmedizin Berlin , Berlin, Germany
| | - Anja Reutzel-Selke
- 1 Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité-Universitatsmedizin Berlin , Berlin, Germany
| | - Dominik Geisel
- 2 Department of Diagnostic and Interventional Radiology, Campus Virchow-Klinikum, Charité-Universitatsmedizin Berlin , Berlin, Germany
| | - Timm Denecke
- 2 Department of Diagnostic and Interventional Radiology, Campus Virchow-Klinikum, Charité-Universitatsmedizin Berlin , Berlin, Germany
| | - Martin Stockmann
- 1 Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité-Universitatsmedizin Berlin , Berlin, Germany .,3 Department of Surgery, Evangelisches Krankenhaus Paul Gerhardt Stift , Lutherstadt Wittenberg, Germany
| | - Johann Pratschke
- 1 Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité-Universitatsmedizin Berlin , Berlin, Germany
| | - Igor M Sauer
- 1 Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité-Universitatsmedizin Berlin , Berlin, Germany
| | - Nathanael Raschzok
- 1 Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité-Universitatsmedizin Berlin , Berlin, Germany .,4 BIH Charité Clinican Scientist Program, Berlin Institute of Health (BIH) , Berlin, Germany
| |
Collapse
|
29
|
Cusimano A, Balasus D, Azzolina A, Augello G, Emma MR, Di Sano C, Gramignoli R, Strom SC, McCubrey JA, Montalto G, Cervello M. Oleocanthal exerts antitumor effects on human liver and colon cancer cells through ROS generation. Int J Oncol 2017; 51:533-544. [PMID: 28656311 DOI: 10.3892/ijo.2017.4049] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 05/04/2017] [Indexed: 11/05/2022] Open
Abstract
The beneficial health properties of the Mediter-ranean diet are well recognized. The principle source of fat in Mediterranean diet is extra-virgin olive oil (EVOO). Oleocanthal (OC) is a naturally occurring minor phenolic compound isolated from EVOO, which has shown a potent anti-inflammatory activity, by means of its ability to inhibit the cyclooxygenase (COX) enzymes COX-1 and COX-2. A large body of evidence indicates that phenols exhibit anticancer activities. The aim of the present study was to evaluate the potential anticancer effects of OC in hepatocellular carcinoma (HCC) and colorectal carcinoma (CRC) models. A panel of human HCC (HepG2, Huh7, Hep3B and PLC/PRF/5) and CRC (HT29, SW480) cell lines was used. Cells were treated with OC, and cell viability and apoptosis were evaluated. Compared with classical commercially available COX inhibitors (ibuprofen, indomethacin, nimesulide), OC was more effective in inducing cell growth inhibition in HCC and CRC cells. Moreover, OC inhibited colony formation and induced apoptosis, as confirmed by PARP cleavage, activation of caspases 3/7 and chromatin condensation. OC treatment in a dose dependent-manner induced expression of γH2AX, a marker of DNA damage, increased intracellular ROS production and caused mitochondrial depolarization. Moreover, the effects of OC were suppressed by the ROS scavenger N-acetyl-L-cysteine. Finally, OC was not toxic in primary normal human hepatocytes. In conclusion, OC treatment was found to exert a potent anticancer activity against HCC and CRC cells. Taken together, our findings provide preclinical support of the chemotherapeutic potential of EVOO against cancer.
Collapse
Affiliation(s)
- Antonella Cusimano
- Institute of Biomedicine and Molecular Immunology 'Alberto Monroy', National Research Council (CNR), Palermo, Italy
| | - Daniele Balasus
- Institute of Biomedicine and Molecular Immunology 'Alberto Monroy', National Research Council (CNR), Palermo, Italy
| | - Antonina Azzolina
- Institute of Biomedicine and Molecular Immunology 'Alberto Monroy', National Research Council (CNR), Palermo, Italy
| | - Giuseppa Augello
- Institute of Biomedicine and Molecular Immunology 'Alberto Monroy', National Research Council (CNR), Palermo, Italy
| | - Maria R Emma
- Institute of Biomedicine and Molecular Immunology 'Alberto Monroy', National Research Council (CNR), Palermo, Italy
| | - Caterina Di Sano
- Institute of Biomedicine and Molecular Immunology 'Alberto Monroy', National Research Council (CNR), Palermo, Italy
| | - Roberto Gramignoli
- Division of Pathology, Department of Laboratory Medicine, Cell Transplantation and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Stephen C Strom
- Division of Pathology, Department of Laboratory Medicine, Cell Transplantation and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Giuseppe Montalto
- Institute of Biomedicine and Molecular Immunology 'Alberto Monroy', National Research Council (CNR), Palermo, Italy
| | - Melchiorre Cervello
- Institute of Biomedicine and Molecular Immunology 'Alberto Monroy', National Research Council (CNR), Palermo, Italy
| |
Collapse
|
30
|
Brandhorst D, Brandhorst H, Johnson PRV. Enzyme Development for Human Islet Isolation: Five Decades of Progress or Stagnation? Rev Diabet Stud 2017. [PMID: 28632819 DOI: 10.1900/rds.2017.14.22] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
In comparison to procedures used for the separation of individual cell types from other organs, the process of human pancreatic islet isolation aims to digest the pancreatic exocrine matrix completely without dispersing the individual cells within the endocrine cell cluster. This objective is unique within the field of tissue separation, and outlines the challenge of islet isolation to balance two opposing priorities. Although significant progress has been made in the characterization and production of enzyme blends for islet isolation, there are still numerous areas which require improvement. The ultimate goal of enzyme production, namely the routine production of a consistent and standardized enzyme blend, has still not been realized. This seems to be mainly the result of a lack of detailed knowledge regarding the structure of the pancreatic extracellular matrix and the synergistic interplay between collagenase and different supplementary proteases during the degradation of the extracellular matrix. Furthermore, the activation of intrinsic proteolytic enzymes produced by the pancreatic acinar cells, also impacts on the chance of a successful outcome of human islet isolation. This overview discusses the challenges of pancreatic enzymatic digestion during human islet isolation, and outlines the developments in this field over the past 5 decades.
Collapse
Affiliation(s)
- Daniel Brandhorst
- Nuffield Department of Surgical Sciences, University of Oxford, United Kingdom
| | - Heide Brandhorst
- Nuffield Department of Surgical Sciences, University of Oxford, United Kingdom
| | - Paul R V Johnson
- Nuffield Department of Surgical Sciences, University of Oxford, United Kingdom
| |
Collapse
|
31
|
Karadagi A, Johansson H, Zemack H, Salipalli S, Mörk LM, Kannisto K, Jorns C, Gramignoli R, Strom S, Stokkeland K, Ericzon BG, Jonigk D, Janciauskiene S, Nowak G, Ellis ECS. Exogenous alpha 1-antitrypsin down-regulates SERPINA1 expression. PLoS One 2017; 12:e0177279. [PMID: 28486562 PMCID: PMC5423693 DOI: 10.1371/journal.pone.0177279] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/01/2017] [Indexed: 11/19/2022] Open
Abstract
The main goal of the therapy with purified human plasma alpha1-antitrypsin (A1AT) is to increase A1AT levels and to prevent lungs from elastolytic activity in patients with PiZZ (Glu342Lys) A1AT deficiency-related emphysema. Potential hepatic gains of this therapy are unknown. Herein, we investigated the effect of A1AT therapy on SERPINA1 (gene encoding A1AT) expression. The expression of SERPINA1 was determined in A1AT or A1AT plus Oncostatin M (OSM) treated primary human hepatocytes isolated from liver tissues from A1AT deficient patients and control liver tissues. In addition, SERPINA1 mRNA was assessed in lung tissues from PiZZ emphysema patients with and without A1AT therapy, and in adherent human peripheral blood mononuclear cells (PBMC) isolated from healthy PiMM donors. In a dose-dependent manner purified A1AT lowered SERPINA1 expression in hepatocytes. This latter effect was more prominent in hepatocytes stimulated with OSM. Although it did not reach statistical significance (P = 0.0539)-analysis of lung tissues showed lower SERPINA1 expression in PiZZ emphysema patients receiving augmentation therapy relative to those without therapy. Finally, exogenously added purified A1AT (1mg/ml) reduced SERPINA1 expression in naïve as well as in lipopolysaccharide (LPS)-stimulated human adherent PBMCs. Exogenous A1AT protein reduces its own endogenous expression. Hence, augmentation with native M-A1AT protein and a parallel reduction in expression of dysfunctional mutant Z-A1AT may be beneficial for PiZZ liver, and this motivates further studies.
Collapse
Affiliation(s)
- Ahmad Karadagi
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Helene Johansson
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Helen Zemack
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Sandeep Salipalli
- Department of Respiratory Medicine, Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Lisa-Mari Mörk
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Kristina Kannisto
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Carl Jorns
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Roberto Gramignoli
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Stephen Strom
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Knut Stokkeland
- Department of Medicine, Visby Hospital, Visby, Sweden
- Department of Medicine, Gastroenterology and Hepatology Unit, Karolinska Institute, Stockholm, Sweden
| | - Bo-Göran Ericzon
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Sabina Janciauskiene
- Department of Respiratory Medicine, Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Greg Nowak
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Ewa C S Ellis
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
32
|
Wang S, Ni HM, Dorko K, Kumer SC, Schmitt TM, Nawabi A, Komatsu M, Huang H, Ding WX. Increased hepatic receptor interacting protein kinase 3 expression due to impaired proteasomal functions contributes to alcohol-induced steatosis and liver injury. Oncotarget 2017; 7:17681-98. [PMID: 26769846 PMCID: PMC4951242 DOI: 10.18632/oncotarget.6893] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 12/23/2015] [Indexed: 12/13/2022] Open
Abstract
Chronic alcohol exposure increased hepatic receptor-interacting protein kinase (RIP) 3 expression and necroptosis in the liver but its mechanisms are unclear. In the present study, we demonstrated that chronic alcohol feeding plus binge (Gao-binge) increased RIP3 but not RIP1 protein levels in mouse livers. RIP3 knockout mice had decreased serum alanine amino transferase activity and hepatic steatosis but had no effect on hepatic neutrophil infiltration compared with wild type mice after Gao-binge alcohol treatment. The hepatic mRNA levels of RIP3 did not change between Gao-binge and control mice, suggesting that alcohol-induced hepatic RIP3 proteins are regulated at the posttranslational level. We found that Gao-binge treatment decreased the levels of proteasome subunit alpha type-2 (PSMA2) and proteasome 26S subunit, ATPase 1 (PSMC1) and impaired hepatic proteasome function. Pharmacological or genetic inhibition of proteasome resulted in the accumulation of RIP3 in mouse livers. More importantly, human alcoholics had decreased expression of PSMA2 and PSMC1 but increased protein levels of RIP3 compared with healthy human livers. Moreover, pharmacological inhibition of RIP1 decreased Gao-binge-induced hepatic inflammation, neutrophil infiltration and NF-κB subunit (p65) nuclear translocation but failed to protect against steatosis and liver injury induced by Gao-binge alcohol. In conclusion, results from this study suggest that impaired hepatic proteasome function by alcohol exposure may contribute to hepatic accumulation of RIP3 resulting in necroptosis and steatosis while RIP1 kinase activity is important for alcohol-induced inflammation.
Collapse
Affiliation(s)
- Shaogui Wang
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.,Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Kenneth Dorko
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sean C Kumer
- Department of General Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Timothy M Schmitt
- Department of General Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Atta Nawabi
- Department of General Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Masaaki Komatsu
- Department of Biochemistry, School of Medicine Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Heqing Huang
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
33
|
Watanabe M, Zemack H, Johansson H, Hagbard L, Jorns C, Li M, Ellis E. Maintenance of Hepatic Functions in Primary Human Hepatocytes Cultured on Xeno-Free and Chemical Defined Human Recombinant Laminins. PLoS One 2016; 11:e0161383. [PMID: 27598296 PMCID: PMC5012698 DOI: 10.1371/journal.pone.0161383] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 08/04/2016] [Indexed: 02/04/2023] Open
Abstract
Refined methods for maintaining specific functions of isolated hepatocytes under xeno-free and chemical defined conditions is of great importance for the development of hepatocyte research and regenerative therapy. Laminins, a large family of heterotrimeric basement membrane adhesion proteins, are highly cell and tissue type specific components of the extracellular matrix and strongly influence the behavior and function of associated cells and/or tissues. However, detailed biological functions of many laminin isoforms are still to be evaluated. In this study, we determined the distribution of laminin isoforms in human liver tissue and isolated primary human hepatocytes by western blot analysis, and investigated the efficacy of different human recombinant laminin isoforms on hepatic functions during culture. Protein expressions of laminin-chain α2, α3, α4, β1, β3, γ1, and γ2 were detected in both isolated human hepatocytes and liver tissue. No α1 and α5 expression could be detected in liver tissue or hepatocytes. Hepatocytes were isolated from five different individual livers, and cultured on human recombinant laminin isoforms -111, -211, -221, -332, -411, -421, -511, and -521 (Biolamina AB), matrigel (extracted from Engelbreth-Holm-Swarm sarcoma), or collagen type IV (Collagen). Hepatocytes cultured on laminin showed characteristic hexagonal shape in a flat cell monolayer. Viability, double stranded DNA concentration, and Ki67 expression for hepatocytes cultured for six days on laminin were comparable to those cultured on EHS and Collagen. Hepatocytes cultured on laminin also displayed production of human albumin, alpha-1-antitrypsin, bile acids, and gene expression of liver-enriched factors, such as hepatocyte nuclear factor 4 alpha, glucose-6-phosphate, cytochrome P450 3A4, and multidrug resistance-associated protein 2. We conclude that all forms of human recombinant laminin tested maintain cell viability and liver-specific functions of primary human hepatocytes, and that recombinant laminin is a promising xeno-free and chemical defined strategy for preservation of hepatocyte specific function in vitro.
Collapse
Affiliation(s)
- Masaaki Watanabe
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| | - Helen Zemack
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Helene Johansson
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska Institutet, Stockholm, Sweden
| | | | - Carl Jorns
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Meng Li
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ewa Ellis
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Horner R, Kluge M, Gassner J, Nösser M, Major RD, Reutzel-Selke A, Leder AK, Struecker B, Morgul MH, Pratschke J, Sauer IM, Raschzok N. Hepatocyte Isolation After Laparoscopic Liver Resection. Tissue Eng Part C Methods 2016; 22:839-46. [DOI: 10.1089/ten.tec.2016.0187] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Rosa Horner
- Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité—Universitätsmedizin, Berlin, Germany
| | - Martin Kluge
- Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité—Universitätsmedizin, Berlin, Germany
| | - Joseph Gassner
- Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité—Universitätsmedizin, Berlin, Germany
| | - Maximilian Nösser
- Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité—Universitätsmedizin, Berlin, Germany
| | - Rebeka Dalma Major
- Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité—Universitätsmedizin, Berlin, Germany
| | - Anja Reutzel-Selke
- Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité—Universitätsmedizin, Berlin, Germany
| | - Annekatrin K. Leder
- Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité—Universitätsmedizin, Berlin, Germany
| | - Benjamin Struecker
- Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité—Universitätsmedizin, Berlin, Germany
| | - Mehmet H. Morgul
- Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité—Universitätsmedizin, Berlin, Germany
| | - Johann Pratschke
- Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité—Universitätsmedizin, Berlin, Germany
| | - Igor M. Sauer
- Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité—Universitätsmedizin, Berlin, Germany
| | - Nathanael Raschzok
- Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Experimental Surgery and Regenerative Medicine, Charité—Universitätsmedizin, Berlin, Germany
- BIH Charité Clinican Scientist Program, Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
35
|
Giebel NL, Shadley JD, McCarver DG, Dorko K, Gramignoli R, Strom SC, Yan K, Simpson PM, Hines RN. Role of Chromatin Structural Changes in Regulating Human CYP3A Ontogeny. ACTA ACUST UNITED AC 2016; 44:1027-37. [PMID: 26921389 DOI: 10.1124/dmd.116.069344] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/25/2016] [Indexed: 11/22/2022]
Abstract
Variability in drug-metabolizing enzyme developmental trajectories contributes to interindividual differences in susceptibility to chemical toxicity and adverse drug reactions, particularly in the first years of life. Factors linked to these interindividual differences are largely unknown, but molecular mechanisms regulating ontogeny are likely involved. To evaluate chromatin structure dynamics as a likely contributing mechanism, age-dependent changes in modified and variant histone occupancy were evaluated within known CYP3A4 and 3A7 regulatory domains. Chromatin immunoprecipitation using fetal or postnatal human hepatocyte chromatin pools followed by quantitative polymerase chain reaction DNA amplification was used to determine relative chromatin occupancy by modified and variant histones. Chromatin structure representing a poised transcriptional state (bivalent chromatin), indicated by the occupancy by modified histones associated with both active and repressed transcription, was observed for CYP3A4 and most 3A7 regulatory regions in both postnatal and fetal livers. However, the CYP3A4 regulatory regions had significantly greater occupancy by modified histones associated with repressed transcription in the fetal liver. Conversely, some modified histones associated with active transcription exhibited greater occupancy in the postnatal liver. CYP3A7 regulatory regions also had significantly greater occupancy by modified histones associated with repressed transcription in the fetus. The observed occupancy by modified histones is consistent with chromatin structural dynamics contributing to CYP3A4 ontogeny, although the data are less conclusive regarding CYP3A7. Interpretation of the latter data may be confounded by cell-type heterogeneity in the fetal liver.
Collapse
Affiliation(s)
- Nicholas L Giebel
- Departments of Pediatrics and Pharmacology and Toxicology, Medical College of Wisconsin, and Children's Research Institute, Children's Hospital and Health Systems, Milwaukee, Wisconsin (N.L.G., J.D.S., D.G.M., K.Y., P.M.S., R.N.H.); and Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (K.D., R.G., S.C.S.)
| | - Jeffrey D Shadley
- Departments of Pediatrics and Pharmacology and Toxicology, Medical College of Wisconsin, and Children's Research Institute, Children's Hospital and Health Systems, Milwaukee, Wisconsin (N.L.G., J.D.S., D.G.M., K.Y., P.M.S., R.N.H.); and Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (K.D., R.G., S.C.S.)
| | - D Gail McCarver
- Departments of Pediatrics and Pharmacology and Toxicology, Medical College of Wisconsin, and Children's Research Institute, Children's Hospital and Health Systems, Milwaukee, Wisconsin (N.L.G., J.D.S., D.G.M., K.Y., P.M.S., R.N.H.); and Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (K.D., R.G., S.C.S.)
| | - Kenneth Dorko
- Departments of Pediatrics and Pharmacology and Toxicology, Medical College of Wisconsin, and Children's Research Institute, Children's Hospital and Health Systems, Milwaukee, Wisconsin (N.L.G., J.D.S., D.G.M., K.Y., P.M.S., R.N.H.); and Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (K.D., R.G., S.C.S.)
| | - Roberto Gramignoli
- Departments of Pediatrics and Pharmacology and Toxicology, Medical College of Wisconsin, and Children's Research Institute, Children's Hospital and Health Systems, Milwaukee, Wisconsin (N.L.G., J.D.S., D.G.M., K.Y., P.M.S., R.N.H.); and Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (K.D., R.G., S.C.S.)
| | - Stephen C Strom
- Departments of Pediatrics and Pharmacology and Toxicology, Medical College of Wisconsin, and Children's Research Institute, Children's Hospital and Health Systems, Milwaukee, Wisconsin (N.L.G., J.D.S., D.G.M., K.Y., P.M.S., R.N.H.); and Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (K.D., R.G., S.C.S.)
| | - Ke Yan
- Departments of Pediatrics and Pharmacology and Toxicology, Medical College of Wisconsin, and Children's Research Institute, Children's Hospital and Health Systems, Milwaukee, Wisconsin (N.L.G., J.D.S., D.G.M., K.Y., P.M.S., R.N.H.); and Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (K.D., R.G., S.C.S.)
| | - Pippa M Simpson
- Departments of Pediatrics and Pharmacology and Toxicology, Medical College of Wisconsin, and Children's Research Institute, Children's Hospital and Health Systems, Milwaukee, Wisconsin (N.L.G., J.D.S., D.G.M., K.Y., P.M.S., R.N.H.); and Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (K.D., R.G., S.C.S.)
| | - Ronald N Hines
- Departments of Pediatrics and Pharmacology and Toxicology, Medical College of Wisconsin, and Children's Research Institute, Children's Hospital and Health Systems, Milwaukee, Wisconsin (N.L.G., J.D.S., D.G.M., K.Y., P.M.S., R.N.H.); and Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (K.D., R.G., S.C.S.)
| |
Collapse
|
36
|
Chinnici CM, Timoneri F, Amico G, Pietrosi G, Vizzini G, Spada M, Pagano D, Gridelli B, Conaldi PG. Characterization of Liver-Specific Functions of Human Fetal Hepatocytes in Culture. Cell Transplant 2015; 24:1139-53. [DOI: 10.3727/096368914x680082] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
This study was designed to assess liver-specific functions of human fetal liver cells proposed as a potential source for hepatocyte transplantation. Fetal liver cells were isolated from livers of different gestational ages (16-22 weeks), and the functions of cell preparations were evaluated by establishing primary cultures. We observed that 20- to 22-week-gestation fetal liver cell cultures contained a predominance of cells with hepatocytic traits that did not divide in vitro but were functionally competent. Fetal hepatocytes performed liver-specific functions at levels comparable to those of their adult counterpart. Moreover, exposure to dexamethasone in combination with oncostatin M promptly induced further maturation of the cells through the acquisition of additional functions (i.e., ability to store glycogen and uptake of indocyanine green). In some cases, particularly in cultures obtained from fetuses of earlier gestational ages (16-18 weeks gestation), cells with mature hepatocytic traits proved to be sporadic, and the primary cultures were mainly populated by clusters of proliferating cells. Consequently, the values of liver-specific functions detected in these cultures were low. We observed that a low cell density culture system rapidly prompted loss of the mature hepatocytic phenotype with downregulations of all the liver-specific functions. We found that human fetal liver cells can be cryopreserved without significant loss of viability and function and evaluated up to 1 year in storage in liquid nitrogen. They might, therefore, be suitable for cell banking and allow for the transplantation of large numbers of cells, thus improving clinical outcomes. Overall, our results indicate that fetal hepatocytes could be used as a cell source for hepatocyte transplantation. Fetal liver cells have been used so far to treat end-stage liver disease. Additional studies are needed to include these cells in cell-based therapies aimed to treat liver failure and inborn errors of metabolism.
Collapse
Affiliation(s)
- Cinzia Maria Chinnici
- Fondazione Ri.MED, Regenerative Medicine and Biomedical Technologies Unit, Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT, Palermo, Italy
| | - Francesca Timoneri
- Fondazione Ri.MED, Regenerative Medicine and Biomedical Technologies Unit, Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT, Palermo, Italy
| | - Giandomenico Amico
- Fondazione Ri.MED, Regenerative Medicine and Biomedical Technologies Unit, Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT, Palermo, Italy
| | - Giada Pietrosi
- Hepatology Unit, Department of Medicine, IRCCS-ISMETT, Palermo, Italy
| | - Giovanni Vizzini
- Hepatology Unit, Department of Medicine, IRCCS-ISMETT, Palermo, Italy
| | - Marco Spada
- Department of Surgery, IRCCS-ISMETT, Palermo, Italy
| | | | - Bruno Gridelli
- Fondazione Ri.MED, Regenerative Medicine and Biomedical Technologies Unit, Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT, Palermo, Italy
- Department of Surgery, IRCCS-ISMETT, Palermo, Italy
| | - Pier Giulio Conaldi
- Fondazione Ri.MED, Regenerative Medicine and Biomedical Technologies Unit, Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT, Palermo, Italy
| |
Collapse
|
37
|
Jorns C, Gramignoli R, Saliem M, Zemack H, Mörk LM, Isaksson B, Nowak G, Ericzon BG, Strom S, Ellis E. Strategies for short-term storage of hepatocytes for repeated clinical infusions. Cell Transplant 2015; 23:1009-18. [PMID: 25199147 DOI: 10.3727/096368913x667484] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Hepatocyte transplantation is an upcoming treatment for patients with metabolic liver diseases. Repeated cell infusions over 1-2 days improve clinical outcome. Isolated hepatocytes are usually cold stored in preservation solutions between repeated infusions. However, during cold storage isolated hepatocytes undergo cell death. We investigated if tissue preservation and repeated isolations are better than storage of isolated hepatocytes when cold preserving human hepatocytes. Liver tissue obtained from liver surgery or organ donors was divided into two pieces. Hepatocytes were isolated by collagenase digestion. Hepatocytes were analyzed directly after isolation (fresh) or after storage for 48 h at 4°C in University of Wisconsin solution (UW cells). Liver tissue from the same donor was stored at 4°C in UW and hepatocytes were isolated after 48 h (UW tissue cells). Hepatocyte viability and function was evaluated by trypan blue exclusion, plating efficiency, ammonia metabolism, CYP 1A1/2, 2C9, 3A7, and 3A4 activities, phase II conjugation, and apoptosis evaluation by TUNEL assay and caspase-3/7 activities. Hepatocytes stored in UW showed a significantly lower viability compared to fresh cells or hepatocytes isolated from tissue stored for 48 h (54% vs. 71% vs. 79%). Plating efficiency was significantly decreased for cells stored in UW (40%) compared to fresh and UW tissue cells (63% vs. 55%). No significant differences between UW cells and UW tissue cells could be shown for CYP activities or ammonia metabolism. Hepatocytes stored in UW showed a strong increase in TUNEL-positive cells, whereas TUNEL staining in cold-stored liver tissue and hepatocytes isolated after 48 h was unchanged. This observation was confirmed by increased caspase-3/7 activities in UW cells. Although preservation of isolated hepatocytes in UW maintains function, cold storage of liver tissue and repeated hepatocyte isolations is superior to cold storage of isolated hepatocytes in preserving hepatocyte viability and function.
Collapse
Affiliation(s)
- Carl Jorns
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Gramignoli R, Dorko K, Tahan V, Skvorak KJ, Ellis E, Jorns C, Ericzon BG, Fox IJ, Strom SC. Hypothermic storage of human hepatocytes for transplantation. Cell Transplant 2015; 23:1143-51. [PMID: 23768881 DOI: 10.3727/096368913x668627] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Transplantation of human hepatocytes is gaining recognition as a bridge or an alternative to orthotopic liver transplantation for patients with acute liver failure and genetic defects. Since most patients require multiple cell infusions over an extended period of time, we investigated hepatic functions in cells maintained in University of Wisconsin solution at 4°C up to 72 h. Eleven different assessments of hepatic viability and function were investigated both pre- and posthypothermic storage, including plating efficiency, caspase-3/7 activity, ammonia metabolism, and drug-metabolizing capacity of isolated hepatocytes. Long-term function, basal, and induced cytochrome P450 activities were measured after exposure to prototypical inducing agents. Cells from 47 different human liver specimens were analyzed. Viability significantly decreased in cells cold stored in UW solution, while apoptosis level and plating efficiency were not significantly different from fresh cells. Luminescent and fluorescent methods assessed phases I and II activities both pre- and post-24-72 h of cold preservation. A robust induction (up to 200-fold) of phase I enzymes was observed in cultured cells. Phase II and ammonia metabolism remained stable during hypothermic storage, although the inductive effect of culture on each metabolic activity was eventually lost. Using techniques that characterize 11 measurements of hepatic viability and function from plating efficiency, to ammonia metabolism, to phases I and II drug metabolism, it was determined that while viability decreased, the remaining viable cells in cold-stored suspensions retained critical hepatic functions for up to 48 h at levels not significantly different from those observed in freshly isolated cells.
Collapse
Affiliation(s)
- Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Gramignoli R, Vosough M, Kannisto K, Srinivasan RC, Strom SC. Clinical Hepatocyte Transplantation: Practical Limits and Possible Solutions. Eur Surg Res 2015; 54:162-77. [DOI: 10.1159/000369552] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 11/04/2014] [Indexed: 01/07/2023]
Abstract
Since the first human hepatocyte transplants (HTx) in 1992, clinical studies have clearly established proof of principle for this therapy as a treatment for patients with acquired or inherited liver disease. Although major accomplishments have been made, there are still some specific limitations to this technology, which, if overcome, could greatly enhance the efficacy and implementation of this therapy. Here, we describe what in our view are the most significant obstacles to the clinical application of HTx and review the solutions currently proposed. The obstacles of significance include the limited number and quality of liver tissues as a cell source, the lack of clinical grade reagents, quality control evaluation of hepatocytes prior to transplantation, hypothermic storage of cells prior to transplantation, preconditioning treatments to enhance engraftment and proliferation of donor cells, tracking or monitoring cells after transplantation, and the optimal immunosuppression protocols for transplant recipients.
Collapse
|
40
|
Duret C, Moreno D, Balasiddaiah A, Roux S, Briolotti P, Raulet E, Herrero A, Ramet H, Biron-Andreani C, Gerbal-Chaloin S, Ramos J, Navarro F, Hardwigsen J, Maurel P, Aldabe R, Daujat-Chavanieu M. Cold Preservation of Human Adult Hepatocytes for Liver Cell Therapy. Cell Transplant 2015; 24:2541-55. [PMID: 25622096 DOI: 10.3727/096368915x687020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hepatocyte transplantation is a promising alternative therapy for the treatment of hepatic failure, hepatocellular deficiency, and genetic metabolic disorders. Hypothermic preservation of isolated human hepatocytes is potentially a simple and convenient strategy to provide on-demand hepatocytes in sufficient quantity and of the quality required for biotherapy. In this study, first we assessed how cold storage in three clinically safe preservative solutions (UW, HTS-FRS, and IGL-1) affects the viability and in vitro functionality of human hepatocytes. Then we evaluated whether such cold-preserved human hepatocytes could engraft and repopulate damaged livers in a mouse model of liver failure. Human hepatocytes showed comparable viabilities after cold preservation in the three solutions. The ability of fresh and cold-stored hepatocytes to attach to a collagen substratum and to synthesize and secrete albumin, coagulation factor VII, and urea in the medium after 3 days in culture was also equally preserved. Cold-stored hepatocytes were then transplanted in the spleen of immunodeficient mice previously infected with adenoviruses containing a thymidine kinase construct and treated with a single dose of ganciclovir to induce liver injury. Engraftment and liver repopulation were monitored over time by measuring the blood level of human albumin and by assessing the expression of specific human hepatic mRNAs and proteins in the recipient livers by RT-PCR and immunohistochemistry, respectively. Our findings show that cold-stored human hepatocytes in IGL-1 and HTS-FRS preservative solutions can survive, engraft, and proliferate in a damaged mouse liver. These results demonstrate the usefulness of human hepatocyte hypothermic preservation for cell transplantation.
Collapse
Affiliation(s)
- Cedric Duret
- INSERM, U1040, Institut de Recherche en Biothérapie, F-34295 Montpellier, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Huch M, Gehart H, van Boxtel R, Hamer K, Blokzijl F, Verstegen MMA, Ellis E, van Wenum M, Fuchs SA, de Ligt J, van de Wetering M, Sasaki N, Boers SJ, Kemperman H, de Jonge J, Ijzermans JNM, Nieuwenhuis EES, Hoekstra R, Strom S, Vries RRG, van der Laan LJW, Cuppen E, Clevers H. Long-term culture of genome-stable bipotent stem cells from adult human liver. Cell 2015; 160:299-312. [PMID: 25533785 PMCID: PMC4313365 DOI: 10.1016/j.cell.2014.11.050] [Citation(s) in RCA: 1036] [Impact Index Per Article: 115.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 09/21/2014] [Accepted: 11/21/2014] [Indexed: 02/08/2023]
Abstract
Despite the enormous replication potential of the human liver, there are currently no culture systems available that sustain hepatocyte replication and/or function in vitro. We have shown previously that single mouse Lgr5+ liver stem cells can be expanded as epithelial organoids in vitro and can be differentiated into functional hepatocytes in vitro and in vivo. We now describe conditions allowing long-term expansion of adult bile duct-derived bipotent progenitor cells from human liver. The expanded cells are highly stable at the chromosome and structural level, while single base changes occur at very low rates. The cells can readily be converted into functional hepatocytes in vitro and upon transplantation in vivo. Organoids from α1-antitrypsin deficiency and Alagille syndrome patients mirror the in vivo pathology. Clonal long-term expansion of primary adult liver stem cells opens up experimental avenues for disease modeling, toxicology studies, regenerative medicine, and gene therapy.
Collapse
Affiliation(s)
- Meritxell Huch
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands.
| | - Helmuth Gehart
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Ruben van Boxtel
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Karien Hamer
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Francis Blokzijl
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC-University Medical Center, Postbus 2040, 3000 CA Rotterdam, the Netherlands
| | - Ewa Ellis
- Unit for Transplantation Surgery, Department of CLINTEC, Karolinska Institute, Karolinska University Hospital Huddinge, Hälsovägen, Flemingsberg, SE-141 86 Stockholm, Sweden
| | - Martien van Wenum
- Surgical Laboratory, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Sabine A Fuchs
- Division of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Joep de Ligt
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Marc van de Wetering
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands; Hubrecht Organoid Technology (HUB), Uppsalalaan 8, 3584CT, Utrecht, the Netherlands
| | - Nobuo Sasaki
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Susanne J Boers
- Division of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Hans Kemperman
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Jeroen de Jonge
- Department of Surgery, Erasmus MC-University Medical Center, Postbus 2040, 3000 CA Rotterdam, the Netherlands
| | - Jan N M Ijzermans
- Department of Surgery, Erasmus MC-University Medical Center, Postbus 2040, 3000 CA Rotterdam, the Netherlands
| | - Edward E S Nieuwenhuis
- Division of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Ruurdtje Hoekstra
- Surgical Laboratory, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Stephen Strom
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institute, Alfred Nobels Alle 8, F 56 141-86 Stockholm, Sweden
| | - Robert R G Vries
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands; Hubrecht Organoid Technology (HUB), Uppsalalaan 8, 3584CT, Utrecht, the Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center, Postbus 2040, 3000 CA Rotterdam, the Netherlands
| | - Edwin Cuppen
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Hans Clevers
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands.
| |
Collapse
|
42
|
Hansel MC, Gramignoli R, Skvorak KJ, Dorko K, Marongiu F, Blake W, Davila J, Strom SC. The history and use of human hepatocytes for the treatment of liver diseases: the first 100 patients. CURRENT PROTOCOLS IN TOXICOLOGY 2014; 62:14.12.1-23. [PMID: 25378242 PMCID: PMC4343212 DOI: 10.1002/0471140856.tx1412s62] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Orthotopic liver transplantation remains the only curative treatment for many end-stage liver diseases, yet the number of patients receiving liver transplants remains limited by the number of organs available for transplant. There is a need for alternative therapies for liver diseases. The transplantation of isolated hepatocytes (liver cells) has been used as an experimental therapy for liver disease in a limited number of cases. Recently, the 100th case of hepatocyte transplantation was reported. This review discusses the history of the hepatocyte transplant field, the major discoveries that supported and enabled the first hepatocyte transplants, and reviews the cases and outcomes of the first 100 clinical transplants. Some of the problems that limit the application or efficacy of hepatocyte transplantation are discussed, as are possible solutions to these problems. In conclusion, hepatocyte transplants have proven effective particularly in cases of metabolic liver disease where reversal or amelioration of the characteristic symptoms of the disease is easily quantified. However, no patients have been completely corrected of a metabolic liver disease for a significant amount of time by hepatocyte transplantation alone. It is likely that future developments in new sources of cells for transplantation will be required before this cellular therapy can be fully implemented and available for large numbers of patients.
Collapse
Affiliation(s)
- Marc C Hansel
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania
| | | | | | | | | | | | | | | |
Collapse
|
43
|
An algorithm that predicts the viability and the yield of human hepatocytes isolated from remnant liver pieces obtained from liver resections. PLoS One 2014; 9:e107567. [PMID: 25313881 PMCID: PMC4196847 DOI: 10.1371/journal.pone.0107567] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 08/12/2014] [Indexed: 01/09/2023] Open
Abstract
Isolated human primary hepatocytes are an essential in vitro model for basic and clinical research. For successful application as a model, isolated hepatocytes need to have a good viability and be available in sufficient yield. Therefore, this study aims to identify donor characteristics, intra-operative factors, tissue processing and cell isolation parameters that affect the viability and yield of human hepatocytes. Remnant liver pieces from tissue designated as surgical waste were collected from 1034 donors with informed consent. Human hepatocytes were isolated by a two-step collagenase perfusion technique with modifications and hepatocyte yield and viability were subsequently determined. The accompanying patient data was collected and entered into a database. Univariate analyses found that the viability and the yield of hepatocytes were affected by many of the variables examined. Multivariate analyses were then carried out to confirm the factors that have a significant relationship with the viability and the yield. It was found that the viability of hepatocytes was significantly decreased by the presence of fibrosis, liver fat and with increasing gamma-glutamyltranspeptidase activity and bilirubin content. Yield was significantly decreased by the presence of liver fat, septal fibrosis, with increasing aspartate aminotransferase activity, cold ischemia times and weight of perfused liver. However, yield was significantly increased by chemotherapy treatment. In conclusion, this study determined the variables that have a significant effect on the viability and the yield of isolated human hepatocytes. These variables have been used to generate an algorithm that can calculate projected viability and yield of isolated human hepatocytes. In this way, projected viability can be determined even before isolation of hepatocytes, so that donors that result in high viability and yield can be identified. Further, if the viability and yield of the isolated hepatocytes is lower than expected, this will highlight a methodological problem that can be addressed.
Collapse
|
44
|
Gramignoli R, Tahan V, Dorko K, Venkataramanan R, Fox IJ, Ellis ECS, Vosough M, Strom SC. Rapid and sensitive assessment of human hepatocyte functions. Cell Transplant 2014; 23:1545-56. [PMID: 24702711 DOI: 10.3727/096368914x680064] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Transplantation of human hepatocytes (HTx) has gained recognition as a bridge to, or an alternative to, orthotopic liver transplantation for patients with acute liver failure or genetic defects in liver function. Although the quality of the hepatocytes used for cell transplantation is critical, no consensus exists on protocols to assess the function of hepatocytes prior to HTx. Application of this cell therapy in clinical practice could be aided by fast and reliable assays to evaluate the functional competence of isolated hepatocytes prior to clinical transplantation. Traditional assays for measuring metabolic functions in primary hepatocytes frequently involve highly technical equipment, time-consuming methods, and large numbers of cells. We describe a novel approach for the rapid assessment of the metabolic capabilities of human hepatocytes. This report details simple procedures to evaluate 11 endpoints from cells isolated from human liver that can be performed by a single operator within approximately 2 h of isolation. Longer term cultured hepatocytes were also analyzed to determine if the results from the 2-h tests were predictive of long-term hepatic function. The assays simultaneously measure five cytochrome P450 activities, one phase II activity, plating efficiency, and ammonia metabolism in addition to viability and cell yield. The assays require fewer than 20 million cells and can be completed using commonly available and inexpensive laboratory equipment. The protocol details methods that can be used in a time frame that would allow analysis of hepatic functions in freshly isolated hepatocytes prior to their use for clinical transplantation.
Collapse
Affiliation(s)
- Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Yoshida O, Kimura S, Jackson EK, Robson SC, Geller DA, Murase N, Thomson AW. CD39 expression by hepatic myeloid dendritic cells attenuates inflammation in liver transplant ischemia-reperfusion injury in mice. Hepatology 2013; 58:2163-75. [PMID: 23813862 PMCID: PMC3844081 DOI: 10.1002/hep.26593] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 06/12/2013] [Indexed: 12/11/2022]
Abstract
UNLABELLED Hepatic innate immune cells, in particular, interstitial dendritic cells (DCs), regulate inflammatory responses and may promote inherent liver tolerogenicity. After tissue injury, adenosine triphosphate (ATP) is released and acts as a damage-associated molecular pattern that activates innate immune cells by pattern recognition receptors. CD39 (ectonucleoside triphosphate diphosphohydrolase-1) rapidly hydrolyzes extracellular ATP to maintain physiological levels. We hypothesized that CD39 expression on liver DCs might contribute to regulation of their innate immune functions. Mouse liver conventional myeloid DCs (mDCs) were hyporesponsive to ATP, compared with their splenic counterparts. This disparity was ascribed to more efficient hydrolysis of ATP by higher expression of CD39 on liver mDCs. Human liver mDCs expressed greater levels of CD39 than those from peripheral blood. The comparatively high expression of CD39 on liver mDCs correlated strongly with both ATP hydrolysis and adenosine production. Notably, CD39(-/-) mouse liver mDCs exhibited a more mature phenotype, greater responsiveness to Toll-like receptor 4 ligation, and stronger proinflammatory and immunostimulatory activity than wild-type (WT) liver mDCs. To investigate the role of CD39 on liver mDCs in vivo, we performed orthotopic liver transplantation with extended cold preservation using CD39(-/-) or WT donor mouse livers. Compared to WT liver grafts, CD39(-/-) grafts exhibited enhanced interstitial DC activation, elevated proinflammatory cytokine levels, and more-severe tissue injury. Moreover, portal venous delivery of WT, but not CD39(-/-) liver mDCs, to donor livers immediately post-transplant exerted a protective effect against graft injury in CD39(-/-) to CD39(-/-) liver transplantation. CONCLUSIONS These data reveal that CD39 expression on conventional liver mDCs limits their proinflammatory activity and confers protective properties on these important innate immune cells against liver transplant ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Osamu Yoshida
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Shoko Kimura
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Edwin K. Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Simon C. Robson
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02215, USA
| | - David A. Geller
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Noriko Murase
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Angus W. Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
46
|
Ni HM, Du K, You M, Ding WX. Critical role of FoxO3a in alcohol-induced autophagy and hepatotoxicity. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1815-1825. [PMID: 24095927 DOI: 10.1016/j.ajpath.2013.08.011] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/31/2013] [Accepted: 08/19/2013] [Indexed: 02/07/2023]
Abstract
Autophagy is a lysosomal degradation process that degrades long-lived cellular proteins and damaged organelles as a critical cell survival mechanism in response to stress. We recently reported that acute ethanol induces autophagy, which then reduces ethanol-induced liver injury. However, the mechanisms by which ethanol induces autophagy are not known. In the present study, ethanol treatment significantly increased both mRNA and protein levels of various essential autophagy-related genes in primary cultured mouse hepatocytes and in mouse liver. Both nuclear translocation of FoxO3a and expression of FoxO3a target genes were increased in ethanol-treated primary hepatocytes and mouse liver. Overexpression of a dominant negative form of FoxO3a inhibited ethanol-induced autophagy-related gene expression and enhanced ethanol-induced cell death in primary hepatocytes, which suggests that FoxO3a is a key factor in regulating ethanol-induced autophagy and cell survival. Resveratrol, a well-known SIRT1 agonist, further enhanced ethanol-induced expression of autophagy-related genes, likely via increased deacetylation of FoxO3a. Moreover, acute ethanol-treated Foxo3a(-/-) mice exhibited decreased autophagy-related gene expression, but enhanced steatosis and liver injury, compared with wild-type mice. FoxO3a thus plays a critical role in ethanol-induced autophagy in mouse liver. Modulating the FoxO3a autophagy pathway may offer novel therapeutic approaches for treating alcoholic liver pathogenesis.
Collapse
Affiliation(s)
- Hong-Min Ni
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Kuo Du
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Min You
- Department of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, Tampa, Florida
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
47
|
Komori J, DeWard AD, Gramignoli R, Strom SC, Fontes P, Lagasse E. Potential barriers to human hepatocyte transplantation in MUP-uPAtg(⁺/⁺)Rag2⁻/⁻γC⁻/⁻ mice. Cell Transplant 2013; 23:1537-44. [PMID: 23998208 DOI: 10.3727/096368913x672046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Primary human fetal and adult hepatocytes have been considered feasible donor cell sources for cell transplantation. We compared the engraftment efficiencies between adult human, fetal human, and adult porcine hepatocytes after transplantation into MUP-uPA(tg(+/+))Rag2(-/-)γC(-/-)mice. Transplantation of adult human hepatocytes yielded a 1,000-fold higher serum albumin level compared to transplantation of fetal human hepatocytes, while transplantation of adult porcine hepatocytes resulted in a 100-fold higher serum albumin level than adult human hepatocytes. These results suggest that adult liver cells are superior to fetal liver cells for transplantation, and caution should be applied if porcine hepatocytes are used for preclinical studies as a proof of concept for human hepatocytes.
Collapse
Affiliation(s)
- Junji Komori
- McGowan Institute for Regenerative Medicine, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | | | | | | |
Collapse
|
48
|
Carchman EH, Whelan S, Loughran P, Mollen K, Stratamirovic S, Shiva S, Rosengart MR, Zuckerbraun BS. Experimental sepsis-induced mitochondrial biogenesis is dependent on autophagy, TLR4, and TLR9 signaling in liver. FASEB J 2013; 27:4703-11. [PMID: 23982147 DOI: 10.1096/fj.13-229476] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Organ injury in sepsis is initially characterized by dysfunction without cell death and structural damage, and thus with the ability to recover organ function. Adaptive metabolic responses to sepsis can prevent bioenergetic failure and death. These studies were aimed at investigating the influence of sepsis on mitochondrial homeostasis, focusing on removal of dysfunctional mitochondria and restitution of a healthy mitochondrial population. These data demonstrate decreased hepatic oxidative phosphorylation by 31 ± 11% following murine cecal ligation and puncture (CLP) at 8 h and 34 ± 9% following LPS treatment in vitro at 12 h (P<0.05). In addition, there was a loss of mitochondrial membrane potential. Mitochondrial density and number initially decreased (relative area per micrograph of 64±10% at baseline vs. 39±13% at 8 h following LPS; P<0.05) and was associated with an increase in autophagy and mitophagy. CLP-induced markers of mitochondrial biogenesis and mitochondrial number and density recovered over time. Furthermore, these data suggest that mitochondrial biogenesis was dependent on an autophagy and mitochondrial DNA/Toll-like receptor 9 (TLR9) signaling pathway. These results suggest that hepatocyte survival and maintenance of function in sepsis is dependent on a mitochondrial homeostasis pathway marked by mitophagy and biogenesis.
Collapse
|
49
|
Pillai VC, Venkataramanan R, Parise RA, Christner SM, Gramignoli R, Strom SC, Rudek MA, Beumer JH. Ritonavir and efavirenz significantly alter the metabolism of erlotinib--an observation in primary cultures of human hepatocytes that is relevant to HIV patients with cancer. Drug Metab Dispos 2013; 41:1843-51. [PMID: 23913028 DOI: 10.1124/dmd.113.052100] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Erlotinib is approved for the treatment of non-small cell lung and pancreatic cancers, and is metabolized by CYP3A4. Inducers and inhibitors of CYP3A enzymes such as ritonavir and efavirenz, respectively, may be used as part of the highly active antiretroviral therapy drugs to treat patients with human immunodeficiency virus (HIV). When HIV patients with a malignancy need treatment with erlotinib, there is a potential of as-yet-undefined drug-drug interaction. We evaluated these interactions using human hepatocytes benchmarked against the interaction of erlotinib with ketoconazole and rifampin, the archetype cytochrome P450 inhibitor and inducer, respectively. Hepatocytes were treated with vehicle [0.1% dimethylsulfoxide, ritonavir (10 μM)], ketoconazole (10 μM), efavirenz (10 μM), or rifampin (10 μM) for 4 days. On day 5, erlotinib (5 μM) was incubated with the above agents for another 24-48 hours. Concentrations of erlotinib and O-desmethyl erlotinib were quantitated in collected samples (combined lysate and medium) using liquid chromatography and tandem mass spectrometry. The half-life (t(½)) of erlotinib increased from 10.6 ± 2.6 to 153 ± 80 and 23.9 ± 4.8 hours, respectively, upon treatment with ritonavir and ketoconazole. The apparent intrinsic clearance (C(Lint, app)) of erlotinib was lowered 16-fold by ritonavir and 1.9-fold by ketoconazole. Efavirenz and rifampin decreased t1/2 of erlotinib from 10.3 ± 1.1 to 5.0 ± 1.5 and 3.4 ± 0.2 hours, respectively. Efavirenz and rifampin increased the C(Lint, app) of erlotinib by 2.2- and 2-fold, respectively. Our results suggest that to achieve desired drug exposure, the clinically used dose (150 mg daily) of erlotinib may have to be significantly reduced (25 mg every other day) or increased (300 mg daily), respectively, when ritonavir or efavirenz is coadministered.
Collapse
Affiliation(s)
- Venkateswaran C Pillai
- Department of Pharmaceutical Sciences and Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (V.C.P., R.V.); Molecular Therapeutics Drug Discovery program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania (R.A.P., S.M.C., J.H.B.); Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet and Hospital, Stockholm, Sweden (R.G., S.C.S.); and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland (M.A.R.)
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Gramignoli R, Tahan V, Dorko K, Skvorak KJ, Hansel MC, Zhao W, Venkataramanan R, Ellis ECS, Jorns C, Ericzon BG, Rosenborg S, Kuiper R, Soltys KA, Mazariegos GV, Fox IJ, Wilson EM, Grompe M, Strom SC. New potential cell source for hepatocyte transplantation: discarded livers from metabolic disease liver transplants. Stem Cell Res 2013; 11:563-73. [PMID: 23644508 DOI: 10.1016/j.scr.2013.03.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 03/16/2013] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED Domino liver transplantation is a method used to increase the number of liver grafts available for orthotopic liver transplantation (OLT). Reports indicate that livers from patients with metabolic liver disease can be safely transplanted into select recipients if the donor's defect and the recipient's metabolic needs are carefully considered. The liver of patients with many types of metabolic liver disease is morphologically and biochemically normal, except for the mutation that characterizes that disease. Other biochemical functions normally performed by the liver are present and presumably "normal" in these hepatocytes. Hepatocytes were isolated from the liver of 35 organ donors and 35 liver tissues taken at OLT from patients with liver disease were analyzed for 9 different measures of viability and function. The data indicate that cells isolated from some diseased livers performed as well or better than those isolated from organ donors with respect to viability, cell yield, plating efficiency and in assays of liver function, including drug metabolism, conjugation reactions and ammonia metabolism. Cells from metabolic diseased livers rapidly and efficiently repopulated a mouse liver upon transplantation. CONCLUSIONS As with domino liver transplantation, domino cell transplantation deserves consideration as method to extend the pool of available organs and cells for transplantation.
Collapse
Affiliation(s)
- Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|