1
|
Igarashi R, Oda M, Okada R, Yano T, Takahashi S, Pastuhov S, Matano M, Masuda N, Togasaki K, Ohta Y, Sato S, Hishiki T, Suematsu M, Itoh M, Fujii M, Sato T. Generation of human adult hepatocyte organoids with metabolic functions. Nature 2025:10.1038/s41586-025-08861-y. [PMID: 40240606 DOI: 10.1038/s41586-025-08861-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/04/2025] [Indexed: 04/18/2025]
Abstract
Proliferating hepatocytes often undergo ductal metaplasia to balance the energy trade-off between cellular functions and replication, hindering the expansion of human adult hepatocytes with functional competency1. Here we demonstrate that the combined activation of Wnt and STAT3 signalling enables long-term self-renewal of human adult hepatocyte organoids. YAP activation facilitates hepatocyte proliferation but commits it towards the biliary duct lineage. By contrast, STAT3 activation by oncostatin M induces hepatocyte proliferation while counteracting ductal metaplasia and maintaining the hepatic identity. Xenotransplanted hepatocyte organoids repopulate the recipient mouse liver and reconstitute the metabolic zonation structure. Upon niche factor removal and hormone supplementation, hepatocyte organoids form cord-like structures with bile canalicular networks and exhibit major liver metabolic functions comparable to those of in vivo hepatocytes. Hepatocyte organoids are amenable to gene editing, prompting functional modelling of inherent metabolic liver diseases. The new culture system offers a promising avenue for developing therapeutic strategies against human liver diseases.
Collapse
Affiliation(s)
- Ryo Igarashi
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mayumi Oda
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Ryo Okada
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Tomoki Yano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Sirirat Takahashi
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Strahil Pastuhov
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mami Matano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Norio Masuda
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Kazuhiro Togasaki
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Yuki Ohta
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Saeko Sato
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Takako Hishiki
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Suematsu
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Manabu Itoh
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Masayuki Fujii
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Toshiro Sato
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan.
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
2
|
Zhang Y, Hao H, Li H, Duan Q, Zheng X, Feng Y, Yang K, Shen S. Cellular Metabolomics Reveals Differences in the Scope of Liver Protection Between Ammonium-Based Glycyrrhizinate and Magnesium Isoglycyrrhizinate. Metabolites 2025; 15:263. [PMID: 40278392 PMCID: PMC12029898 DOI: 10.3390/metabo15040263] [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: 01/05/2025] [Revised: 03/14/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
Background: Despite the well-established liver-protective efficacy of monoammonium glycyrrhizinate (MONO), diammonium glycyrrhizinate (DIAM), and magnesium isoglycyrrhizinate (MAGN), which has been translated into clinical practice, their clinical differentiation remains elusive owing to their structural similarities and overlapping therapeutic effects. Methods: The present study delves into the pharmacokinetics, cellular-level liver-protective potencies, and underlying mechanisms of action of these three compounds through a comprehensive analysis. Results: The findings reveal that both DIAM and MAGN exhibit superior bioavailability and hepatoprotective profiles compared to MONO. Notably, an investigation of the metabolic pathways mediating liver protection in normal human liver cells (LO2), utilizing an ultra-performance liquid chromatography-time of flight tandem mass spectrometry (UPLC-TOF-MS/MSe) platform, demonstrated that MAGN augments antioxidant components, thereby favoring its application in drug-induced liver injury (DILI). Conversely, DIAM appears to be a more suitable candidate for addressing non-alcoholic fatty liver disease (NAFLD) and viral hepatitis. Conclusion: This study contributes novel perspectives on the mechanisms of action and potential clinical utilities of DIAM and MAGN in liver disease prevention and management.
Collapse
Affiliation(s)
- Yihua Zhang
- Key Laboratory of Analytical Science and Technology of Hebei Province, College of Chemistry and Materials Science, Hebei University, Baoding 071002, China;
- NDMA Key Laboratory for Quality Control and Evaluation of Generic Drug, Hebei Institute for Drug and Medical Device Control, Shijiazhuang 050200, China; (H.L.); (Q.D.); (X.Z.); (Y.F.)
| | - Han Hao
- School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China;
| | - Hui Li
- NDMA Key Laboratory for Quality Control and Evaluation of Generic Drug, Hebei Institute for Drug and Medical Device Control, Shijiazhuang 050200, China; (H.L.); (Q.D.); (X.Z.); (Y.F.)
| | - Qiong Duan
- NDMA Key Laboratory for Quality Control and Evaluation of Generic Drug, Hebei Institute for Drug and Medical Device Control, Shijiazhuang 050200, China; (H.L.); (Q.D.); (X.Z.); (Y.F.)
| | - Xiaoming Zheng
- NDMA Key Laboratory for Quality Control and Evaluation of Generic Drug, Hebei Institute for Drug and Medical Device Control, Shijiazhuang 050200, China; (H.L.); (Q.D.); (X.Z.); (Y.F.)
| | - Yan Feng
- NDMA Key Laboratory for Quality Control and Evaluation of Generic Drug, Hebei Institute for Drug and Medical Device Control, Shijiazhuang 050200, China; (H.L.); (Q.D.); (X.Z.); (Y.F.)
| | - Kun Yang
- School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China;
| | - Shigang Shen
- Key Laboratory of Analytical Science and Technology of Hebei Province, College of Chemistry and Materials Science, Hebei University, Baoding 071002, China;
| |
Collapse
|
3
|
Malakmahmoudi N, Pisu R, Laconi E, Marongiu F. Dietary Rhythms and MASLD-Related Hepatocellular Carcinoma. Cancers (Basel) 2024; 16:3481. [PMID: 39456575 PMCID: PMC11505995 DOI: 10.3390/cancers16203481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Dietary rhythms have emerged as a relevant variable in the equation relating nutrition and health. Both experimental and epidemiological studies point to potential beneficial effects of adequate fasting intervals between meals on the evolution of chronic diseases associated with aging. Metabolic dysfunction-associated steatotic liver disease (MASLD) is eminently related to diet and unsurprisingly, diet-based approaches are a mainstay in countering its long-term clinical evolution, including the emergence of hepatocellular carcinoma (HCC). We briefly discuss current evidence linking fasting intervals, MASLD, and HCC and propose a working hypothesis to reconcile some of the apparently conflicting results. This hypothesis relates the beneficial effects of time-restricted eating schedules to the quantity and quality of food, and it is easily amenable to testing.
Collapse
Affiliation(s)
| | | | - Ezio Laconi
- Department of Biomedical Science, University of Cagliari, 09124 Cagliari, Italy; (N.M.); (R.P.); (F.M.)
| | | |
Collapse
|
4
|
Sande-Melon M, Bergemann D, Fernández-Lajarín M, González-Rosa JM, Cox AG. Development of a hepatic cryoinjury model to study liver regeneration. Development 2024; 151:dev203124. [PMID: 38975841 PMCID: PMC11318111 DOI: 10.1242/dev.203124] [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: 05/29/2024] [Accepted: 06/20/2024] [Indexed: 07/09/2024]
Abstract
The liver is a remarkable organ that can regenerate in response to injury. Depending on the extent of injury, the liver can undergo compensatory hyperplasia or fibrosis. Despite decades of research, the molecular mechanisms underlying these processes are poorly understood. Here, we developed a new model to study liver regeneration based on cryoinjury. To visualise liver regeneration at cellular resolution, we adapted the CUBIC tissue-clearing approach. Hepatic cryoinjury induced a localised necrotic and apoptotic lesion characterised by inflammation and infiltration of innate immune cells. After this initial phase, we observed fibrosis, which resolved as regeneration re-established homeostasis in 30 days. Importantly, this approach enables the comparison of healthy and injured parenchyma within an individual animal, providing unique advantages to previous models. In summary, the hepatic cryoinjury model provides a fast and reproducible method for studying the cellular and molecular pathways underpinning fibrosis and liver regeneration.
Collapse
Affiliation(s)
- Marcos Sande-Melon
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - David Bergemann
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13th Street, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Miriam Fernández-Lajarín
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13th Street, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA 02467, USA
| | - Juan Manuel González-Rosa
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13th Street, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA 02467, USA
| | - Andrew G. Cox
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3000, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, Victoria 3000, Australia
| |
Collapse
|
5
|
Lotfi M, Maharati A, Hamidi AA, Taghehchian N, Moghbeli M. MicroRNA-532 as a probable diagnostic and therapeutic marker in cancer patients. Mutat Res 2024; 829:111874. [PMID: 38986233 DOI: 10.1016/j.mrfmmm.2024.111874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/12/2024]
Abstract
The high mortality rate in cancer patients is always one of the main challenges of the health systems globally. Several factors are involved in the high rate of cancer related mortality, including late diagnosis and drug resistance. Cancer is mainly diagnosed in the advanced stages of tumor progression that causes the failure of therapeutic strategies and increases the death rate in these patients. Therefore, assessment of the molecular mechanisms associated with the occurrence of cancer can be effective to introduce early tumor diagnostic markers. MicroRNAs (miRNAs) as the stable non-coding RNAs in the biological body fluids are involved in regulation of cell proliferation, migration, and apoptosis. MiR-532 deregulation has been reported in different tumor types. Therefore, in the present review we discussed the role of miR-532 during tumor growth. It has been shown that miR-532 has mainly a tumor suppressor role through the regulation of transcription factors, chemokines, and signaling pathways such as NF-kB, MAPK, PI3K/AKT, and WNT. In addition to the independent role of miR-532 in regulation of cellular processes, it also functions as a mediator of lncRNAs and circRNAs. Therefore, miR-532 can be considered as a non-invasive diagnostic/prognostic marker as well as a therapeutic target in cancer patients.
Collapse
Affiliation(s)
- Malihe Lotfi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Abbas Hamidi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Sande-Melon M, Bergemann D, Fernández-Lajarín M, González-Rosa JM, Cox AG. Development of a hepatic cryoinjury model to study liver regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.24.550437. [PMID: 38948752 PMCID: PMC11212901 DOI: 10.1101/2023.07.24.550437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The liver is a remarkable organ that can regenerate in response to injury. Depending on the extent of injury, the liver can undergo compensatory hyperplasia or fibrosis. Despite decades of research, the molecular mechanisms underlying these processes are poorly understood. Here, we developed a new model to study liver regeneration based on cryoinjury. To visualise liver regeneration at cellular resolution, we adapted the CUBIC tissue-clearing approach. Hepatic cryoinjury induced a localised necrotic and apoptotic lesion characterised by inflammation and infiltration of innate immune cells. Following this initial phase, we observed fibrosis, which resolved as regeneration re-established homeostasis in 30 days. Importantly, this approach enables the comparison of healthy and injured parenchyma with an individual animal, providing unique advantages to previous models. In summary, the hepatic cryoinjury model provides a fast and reproducible method for studying the cellular and molecular pathways underpinning fibrosis and liver regeneration.
Collapse
Affiliation(s)
- Marcos Sande-Melon
- Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, 3000, Australia
| | - David Bergemann
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13 Street, 02129 MA, USA
- Harvard Medical School
| | - Miriam Fernández-Lajarín
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13 Street, 02129 MA, USA
- Harvard Medical School
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA 02467
| | - Juan Manuel González-Rosa
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13 Street, 02129 MA, USA
- Harvard Medical School
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA 02467
| | - Andrew G. Cox
- Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, 3000, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, Victoria, 3000, Australia
| |
Collapse
|
7
|
Li G, Park HJ, Suh JH, Choi HS. 7-Ketocholesterol plays a key role in cholesterol-induced hepatitis via macrophage and neutrophil infiltration. J Nutr Biochem 2024; 125:109552. [PMID: 38134972 DOI: 10.1016/j.jnutbio.2023.109552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023]
Abstract
This study sought to explore the role of 7-ketocholesterol (7-KC) in liver damage caused by high cholesterol intake and its potential pathological mechanism in mice. Our in vivo findings indicated that mice fed a high-cholesterol diet had elevated serum levels of 7-KC, accompanied by liver injury and inflammation, similar to human nonalcoholic steatohepatitis. Furthermore, the high-cholesterol diet induced neutrophil infiltration, which played a critical role in liver damage through myeloperoxidase (MPO) activity. Upon stimulation with 7-KC, macrophages exhibited increased expression of C-X-C motif chemokine ligand 1 (CXCL1) and CXCL2, as well as ATP-binding cassette transporter A1 (ABCA1) and ABCG1. Hepatocytes, on the other hand, exhibited increased expression of CXCL2 and ABCG1. The infiltration of neutrophils in the liver was primarily caused by CXCL1 and CXCL2, resulting in hepatocyte cell death due to elevated MPO activity. Our data also revealed that the activation of macrophages by 7-KC via ABCA1 or ABCG1 was not associated with lipid accumulation. Collectively, these findings suggest that high cholesterol-induced hepatitis in mice involves, at least partially, the recruitment of neutrophils to the liver by 7-KC-activated macrophages. This is mediated by increased expression of CXCL1 and CXCL2 through ABCA1 or ABCG1, which act as 7-KC efflux transporters. Additionally, hepatocytes contribute to this process by increased expression of CXCL2 through ABCG1. Therefore, our findings suggest that 7-KC may play a role in high cholesterol-induced hepatitis in mice by activating macrophages and hepatocytes, ultimately leading to neutrophil infiltration.
Collapse
Affiliation(s)
- Guoen Li
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
| | - Hyun-Jung Park
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
| | - Jae-Hee Suh
- Department of Pathology, Ulsan University Hospital, Ulsan, Korea
| | - Hye-Seon Choi
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea.
| |
Collapse
|
8
|
Guillard J, Untereiner V, Garnotel R, Boulagnon-Rombi C, Gobinet C, Proult I, Sockalingum GD, Thiéfin G. Longitudinal Study of Cirrhosis Development in STAM and carbon tetrachloride Mouse Models Using Fourier Transform Infrared Spectral Imaging. J Transl Med 2023; 103:100231. [PMID: 37544611 DOI: 10.1016/j.labinv.2023.100231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023] Open
Abstract
Animal models of cirrhosis are of great interest to investigate the pathological process leading to the final stage of cirrhosis. The aim of this study was to analyze the different steps involved in the progressive development of cirrhosis using Fourier transform infrared spectral histology in 2 mouse models of cirrhosis, the STAM model of metabolic cirrhosis, and the carbon tetrachloride-induced cirrhosis model. Formalin-fixed, paraffin-embedded liver samples were obtained from 3 mice at 5 time points in each model to analyze the course of hepatic lesions up to the formation of cirrhosis. For each time point, adjacent 3-μm-thick liver sections were obtained for histologic stains and spectral histology. Fourier transform infrared acquisitions of liver sections were performed at projected pixel sizes of 25 μm × 25 μm and 6.25 μm × 6.25 μm. Spectral images were then preprocessed with an extended multiplicative signal correction and analyzed with common k-means clustering, including all stages in each model. In both models, the 2- and 4-class common k-means clustering in the 1000 to 1350 cm-1 range showed that spectral classes characterized by higher absorbance peaks of glycogen were predominant at baseline, then decreased markedly in early stages of hepatic damage, and almost disappeared in cirrhotic tissues. Concomitantly, spectral classes characterized by higher absorbance peaks of nucleic acids became progressively predominant during the course of hepatic lesions. These results were confirmed using k-means clustering on the peaks of interest identified for glycogen and nucleic acid content. Our study showed that the glycogen depletion previously described at the stage of cirrhosis is an early event in the pathological process, independently of the cause of cirrhosis. In addition, there was a progressive increase in the nucleic acid content, which may be linked to increased proliferation and polyploidy in response to cellular lesions.
Collapse
Affiliation(s)
- Julien Guillard
- Université de Reims Champagne-Ardenne, BioSpecT, Reims, France
| | - Valérie Untereiner
- Université de Reims Champagne-Ardenne, Plateforme en Imagerie Cellulaire et Tissulaire, Reims, France
| | - Roselyne Garnotel
- Université de Reims Champagne-Ardenne, BioSpecT, Reims, France; Laboratoire de Biochimie-Pharmacologie-Toxicologie, Pôle de Biologie Territoriale, Centre Hospitalo-Universitaire de Reims, Reims, France
| | - Camille Boulagnon-Rombi
- Laboratoire de Biopathologie, Pôle de Biologie Territoriale, Centre Hospitalo-Universitaire de Reims, Reims, France
| | - Cyril Gobinet
- Université de Reims Champagne-Ardenne, BioSpecT, Reims, France
| | - Isabelle Proult
- Université de Reims Champagne-Ardenne, Centre National de la Recherche Scientifique, MEDyC, Reims, France
| | | | - Gérard Thiéfin
- Université de Reims Champagne-Ardenne, BioSpecT, Reims, France; Service d'Hépato-Gastroentérologie et de Cancérologie Digestive, Centre Hospitalo-Universitaire de Reims, Reims, France.
| |
Collapse
|
9
|
Chiu YC, Huang KW, Lin YH, Yin WR, Hou YT. Development of a decellularized liver matrix-based nanocarrier for liver regeneration after partial hepatectomy. JOURNAL OF MATERIALS SCIENCE 2023; 58:15162-15180. [DOI: 10.1007/s10853-023-08971-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/18/2023] [Indexed: 10/08/2024]
|
10
|
Su H, Haque M, Becker S, Edlund K, Duda J, Wang Q, Reißing J, Marschall HU, Candels LS, Mohamed M, Sjöland W, Liao L, Drexler SA, Strowig T, Rahnenführer J, Hengstler JG, Hatting M, Trautwein C. Long-term hypercaloric diet exacerbates metabolic liver disease in PNPLA3 I148M animals. Liver Int 2023; 43:1699-1713. [PMID: 37073116 DOI: 10.1111/liv.15587] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/22/2023] [Accepted: 04/10/2023] [Indexed: 04/20/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic fatty liver disease (NAFLD) is a major health burden associated with the metabolic syndrome leading to liver fibrosis, cirrhosis and ultimately liver cancer. In humans, the PNPLA3 I148M polymorphism of the phospholipase patatin-like phospholipid domain containing protein 3 (PNPLA3) has a well-documented impact on metabolic liver disease. In this study, we used a mouse model mimicking the human PNPLA3 I148M polymorphism in a long-term high fat diet (HFD) experiment to better define its role for NAFLD progression. METHODS Male mice bearing wild-type Pnpla3 (Pnpla3WT ), or the human polymorphism PNPLA3 I148M (Pnpla3148M/M ) were subjected to HFD feeding for 24 and 52 weeks. Further analysis concerning basic phenotype, inflammation, proliferation and cell death, fibrosis and microbiota were performed in each time point. RESULTS After 52 weeks HFD Pnpla3148M/M animals had more liver fibrosis, enhanced numbers of inflammatory cells as well as increased Kupffer cell activity. Increased hepatocyte cell turnover and ductular proliferation were evident in HFD Pnpla3148M/M livers. Microbiome diversity was decreased after HFD feeding, changes were influenced by HFD feeding (36%) and the PNPLA3 I148M genotype (12%). Pnpla3148M/M mice had more faecal bile acids. RNA-sequencing of liver tissue defined an HFD-associated signature, and a Pnpla3148M/M specific pattern, which suggests Kupffer cell and monocytes-derived macrophages as significant drivers of liver disease progression in Pnpla3148M/M animals. CONCLUSION With long-term HFD feeding, mice with the PNPLA3 I148M genotype show exacerbated NAFLD. This finding is linked to PNPLA3 I148M-specific changes in microbiota composition and liver gene expression showing a stronger inflammatory response leading to enhanced liver fibrosis progression.
Collapse
Affiliation(s)
- Huan Su
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Madhuri Haque
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Svea Becker
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Karolina Edlund
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Dortmund, Germany
| | - Julia Duda
- Department of Statistics, TU Dortmund University, Dortmund, Germany
| | - Qingbi Wang
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Johanna Reißing
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Lena S Candels
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Mohamed Mohamed
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Wilhelm Sjöland
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Lijun Liao
- Department of Pain Management, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Stephan A Drexler
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Till Strowig
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Jan G Hengstler
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Dortmund, Germany
| | - Maximilian Hatting
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Christian Trautwein
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
11
|
Korbecki J, Bosiacki M, Barczak K, Łagocka R, Chlubek D, Baranowska-Bosiacka I. The Clinical Significance and Role of CXCL1 Chemokine in Gastrointestinal Cancers. Cells 2023; 12:1406. [PMID: 37408240 DOI: 10.3390/cells12101406] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/09/2023] [Accepted: 05/15/2023] [Indexed: 07/07/2023] Open
Abstract
One area of cancer research is the interaction between cancer cells and immune cells, in which chemokines play a vital role. Despite this, a comprehensive summary of the involvement of C-X-C motif ligand 1 (CXCL1) chemokine (also known as growth-regulated gene-α (GRO-α), melanoma growth-stimulatory activity (MGSA)) in cancer processes is lacking. To address this gap, this review provides a detailed analysis of CXCL1's role in gastrointestinal cancers, including head and neck cancer, esophageal cancer, gastric cancer, liver cancer (hepatocellular carcinoma (HCC)), cholangiocarcinoma, pancreatic cancer (pancreatic ductal adenocarcinoma), and colorectal cancer (colon cancer and rectal cancer). This paper presents the impact of CXCL1 on various molecular cancer processes, such as cancer cell proliferation, migration, and invasion, lymph node metastasis, angiogenesis, recruitment to the tumor microenvironment, and its effect on immune system cells, such as tumor-associated neutrophils (TAN), regulatory T (Treg) cells, myeloid-derived suppressor cells (MDSCs), and macrophages. Furthermore, this review discusses the association of CXCL1 with clinical aspects of gastrointestinal cancers, including its correlation with tumor size, cancer grade, tumor-node-metastasis (TNM) stage, and patient prognosis. This paper concludes by exploring CXCL1's potential as a therapeutic target in anticancer therapy.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28 St., 65-046 Zielona Góra, Poland
| | - Mateusz Bosiacki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
- Department of Functional Diagnostics and Physical Medicine, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Żołnierska 54 Str., 71-210 Szczecin, Poland
| | - Katarzyna Barczak
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Ryta Łagocka
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| |
Collapse
|
12
|
Cabrera-Galván JJ, Araujo E, de Mirecki-Garrido M, Pérez-Rodríguez D, Guerra B, Aranda-Tavío H, Guerra-Rodríguez M, Brito-Casillas Y, Melián C, Martínez-Martín MS, Fernández-Pérez L, Recio C. SOCS2 protects against chemical-induced hepatocellular carcinoma progression by modulating inflammation and cell proliferation in the liver. Biomed Pharmacother 2023; 157:114060. [PMID: 36455458 DOI: 10.1016/j.biopha.2022.114060] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/03/2022] [Accepted: 11/27/2022] [Indexed: 11/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent and lethal cancers worldwide, but the precise intracellular mechanisms underlying the progression of this inflammation associated cancer are not well established. SOCS2 protein plays an important role in the carcinogenesis of different tumors by regulating cytokine signalling through the JAK/STAT axis. However, its role in HCC is unclear. Here, we investigate the role of SOCS2 in HCC progression and its potential as HCC biomarker. The effects of SOCS2 in HCC progression were evaluated in an experimental model of diethylnitrosamine (DEN)-induced HCC in C57BL/6 and SOCS2 deficient mice, in cultured hepatic cells, and in liver samples from HCC patients. Mice lacking SOCS2 showed higher liver tumor burden with increased malignancy grade, inflammation, fibrosis, and proliferation than their controls. Protein and gene expression analysis reported higher pSTAT5 and pSTAT3 activation, upregulation of different proteins involved in survival and proliferation, and increased levels of proinflammatory and pro-tumoral mediators in the absence of SOCS2. Clinically relevant, downregulated expression of SOCS2 was found in neoplasia from HCC patients compared to healthy liver tissue, correlating with the malignancy grade. In summary, our data show that lack of SOCS2 increases susceptibility to chemical-induced HCC and suggest the tumor suppressor role of this protein by regulating the oncogenic and inflammatory responses mediated by STAT5 and STAT3 in the liver. Hence, SOCS2 emerges as an attractive target molecule and potential biomarker to deepen in the study of HCC treatment.
Collapse
Affiliation(s)
- Juan José Cabrera-Galván
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain; Departamento Morfología, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain
| | - Eduardo Araujo
- Departamento Morfología, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain
| | - Mercedes de Mirecki-Garrido
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain
| | - David Pérez-Rodríguez
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain
| | - Borja Guerra
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain; Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain and Instituto de Investigaciones Biomédicas "Alberto Sols" Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Haidée Aranda-Tavío
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain
| | - Miguel Guerra-Rodríguez
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain
| | - Yeray Brito-Casillas
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain
| | - Carlos Melián
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain
| | - María Soledad Martínez-Martín
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain; Departamento Morfología, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain; Servicio Anatomía Patológica, Complejo Hospitalario Universitario Insular - Materno Infantil, Las Palmas de Gran Canaria, Spain
| | - Leandro Fernández-Pérez
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain; Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain and Instituto de Investigaciones Biomédicas "Alberto Sols" Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlota Recio
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain.
| |
Collapse
|
13
|
Ohkubo R, Mu WC, Wang CL, Song Z, Barthez M, Wang Y, Mitchener N, Abdullayev R, Lee YR, Ma Y, Curtin M, Srinivasan S, Zhang X, Yang F, Sudmant PH, Pisco AO, Neff N, Haynes CM, Chen D. The hepatic integrated stress response suppresses the somatotroph axis to control liver damage in nonalcoholic fatty liver disease. Cell Rep 2022; 41:111803. [PMID: 36516757 PMCID: PMC9825120 DOI: 10.1016/j.celrep.2022.111803] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 10/14/2022] [Accepted: 11/18/2022] [Indexed: 12/15/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) can be ameliorated by calorie restriction, which leads to the suppressed somatotroph axis. Paradoxically, the suppressed somatotroph axis is associated with patients with NAFLD and is correlated with the severity of fibrosis. How the somatotroph axis becomes dysregulated and whether the repressed somatotroph axis impacts liver damage during the progression of NAFLD are unclear. Here, we identify a regulatory branch of the hepatic integrated stress response (ISR), which represses the somatotroph axis in hepatocytes through ATF3, resulting in enhanced cell survival and reduced cell proliferation. In mouse models of NAFLD, the ISR represses the somatotroph axis, leading to reduced apoptosis and inflammation but decreased hepatocyte proliferation and exacerbated fibrosis in the liver. NAD+ repletion reduces the ISR, rescues the dysregulated somatotroph axis, and alleviates NAFLD. These results establish that the hepatic ISR suppresses the somatotroph axis to control cell fate decisions and liver damage in NAFLD.
Collapse
Affiliation(s)
- Rika Ohkubo
- Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Wei-Chieh Mu
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chih-Ling Wang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Zehan Song
- Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Marine Barthez
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yifei Wang
- Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Nathaniel Mitchener
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Rasul Abdullayev
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yeong Rim Lee
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yuze Ma
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Megan Curtin
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Suraj Srinivasan
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Xingjia Zhang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Fanghan Yang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Peter H Sudmant
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Center for Computational Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Norma Neff
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Cole M Haynes
- Department of Molecular, Cell and Cancer Biology, UMass-Chan Medical School, Worcester, MA 01605, USA
| | - Danica Chen
- Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
14
|
Zou Z, Sun Y, Wang L, Ma S, Sun C, Zhou Y, Yang G. GIV is a promising novel poor prognostic factor in liver hepatocellular carcinoma. Medicine (Baltimore) 2022; 101:e29645. [PMID: 35960100 PMCID: PMC9371552 DOI: 10.1097/md.0000000000029645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Numerous studies have implicated Gα-interacting, vesicle-associated protein (GIV) in the development and metastasis of various cancers. However, its role remains unclear in liver hepatocellular carcinoma (LIHC). We aimed to demonstrate the relationship between GIV and LIHC based on The Cancer Genome Atlas database. We use the Gene Expression Profiling Interactive Analysis and UALCAN to explore the expression of GIV and the survive analysis of GIV in patients with LIHC, genetic alteration analysis, immune infiltration analysis, functional enrichment, protein-protein interaction network analyses, and transcription factor targets of GIV-correlated genes and GIV-interacting genes were performed this study. GIV expression was significantly elevated in LIHC tissues. Remarkable correlation was established between GIV expression and LIHC pathological stage. Low expression of GIV in tumor tissues had a better prognosis than GIV-high expression. GIV alteration frequency was 1.44% in patients with LIHC. GIV-unaltered patients had better survival than GIV-altered ones. Moreover, GIV expression level in LIHC significantly correlated with the infiltration level of immune cells and cancer-associated fibroblasts. The functions of differentially expressed GIVs are associated with the cell cycle pathway. Our data imply that E2F4, E2F1, MYC, and MYCN are key transcription factors for GIV-correlated genes and GIV-interacted genes. GIV may be an adverse prognostic factor for patients with LIHC; it also can be a potential therapeutic target against LIHC. Further studies are required to validate our findings.
Collapse
Affiliation(s)
- Zhenzhen Zou
- Department of Laboratory, Dushuhu Public Hospital Affiliated to Soochow University, Suzhou, Jiangsu Province, China
| | - Yibin Sun
- Department of Gastroenterology, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Lin Wang
- Department of Laboratory, Dushuhu Public Hospital Affiliated to Soochow University, Suzhou, Jiangsu Province, China
| | - Sai Ma
- Department of Central Laboratory, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Chunrong Sun
- Department of General Surgery, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Yu Zhou
- Department of General Surgery, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Souzhou Jiangsu, China
- * Correspondence: Yu Zhou, Department of General Surgery, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Rd., Suzhou, Jiangsu Province 215000, China (e-mail: )
| | - Guorong Yang
- Department of General Surgery, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
| |
Collapse
|
15
|
The Importance of CXCL1 in the Physiological State and in Noncancer Diseases of the Oral Cavity and Abdominal Organs. Int J Mol Sci 2022; 23:ijms23137151. [PMID: 35806156 PMCID: PMC9266754 DOI: 10.3390/ijms23137151] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/23/2022] [Accepted: 06/25/2022] [Indexed: 02/06/2023] Open
Abstract
CXCL1 is a CXC chemokine, CXCR2 ligand and chemotactic factor for neutrophils. In this paper, we present a review of the role of the chemokine CXCL1 in physiology and in selected major non-cancer diseases of the oral cavity and abdominal organs (gingiva, salivary glands, stomach, liver, pancreas, intestines, and kidneys). We focus on the importance of CXCL1 on implantation and placentation as well as on human pluripotent stem cells. We also show the significance of CXCL1 in selected diseases of the abdominal organs, including the gastrointestinal tract and oral cavity (periodontal diseases, periodontitis, Sjögren syndrome, Helicobacter pylori infection, diabetes, liver cirrhosis, alcoholic liver disease (ALD), non-alcoholic fatty liver disease (NAFLD), HBV and HCV infection, liver ischemia and reperfusion injury, inflammatory bowel disease (Crohn’s disease and ulcerative colitis), obesity and overweight, kidney transplantation and ischemic-reperfusion injury, endometriosis and adenomyosis).
Collapse
|
16
|
Heinke P, Rost F, Rode J, Trus P, Simonova I, Lázár E, Feddema J, Welsch T, Alkass K, Salehpour M, Zimmermann A, Seehofer D, Possnert G, Damm G, Druid H, Brusch L, Bergmann O. Diploid hepatocytes drive physiological liver renewal in adult humans. Cell Syst 2022; 13:499-507.e12. [PMID: 35649419 DOI: 10.1016/j.cels.2022.05.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/19/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022]
Abstract
Physiological liver cell replacement is central to maintaining the organ's high metabolic activity, although its characteristics are difficult to study in humans. Using retrospective radiocarbon (14C) birth dating of cells, we report that human hepatocytes show continuous and lifelong turnover, allowing the liver to remain a young organ (average age <3 years). Hepatocyte renewal is highly dependent on the ploidy level. Diploid hepatocytes show more than 7-fold higher annual birth rates than polyploid hepatocytes. These observations support the view that physiological liver cell renewal in humans is mainly dependent on diploid hepatocytes, whereas polyploid cells are compromised in their ability to divide. Moreover, cellular transitions between diploid and polyploid hepatocytes are limited under homeostatic conditions. With these findings, we present an integrated model of homeostatic liver cell generation in humans that provides fundamental insights into liver cell turnover dynamics.
Collapse
Affiliation(s)
- Paula Heinke
- Center for Regenerative Therapies, Technische Universität Dresden, 01307 Dresden, Germany
| | - Fabian Rost
- Center for Regenerative Therapies, Technische Universität Dresden, 01307 Dresden, Germany; Max Planck Institute for the Physics of Complex Systems, 01187 Dresden, Germany; Centre for Information Services and High Performance Computing, Technische Universität Dresden, 01187 Dresden, Germany
| | - Julian Rode
- Centre for Information Services and High Performance Computing, Technische Universität Dresden, 01187 Dresden, Germany
| | - Palina Trus
- Center for Regenerative Therapies, Technische Universität Dresden, 01307 Dresden, Germany
| | - Irina Simonova
- Center for Regenerative Therapies, Technische Universität Dresden, 01307 Dresden, Germany
| | - Enikő Lázár
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Joshua Feddema
- Center for Regenerative Therapies, Technische Universität Dresden, 01307 Dresden, Germany
| | - Thilo Welsch
- Visceral-, Thoracic- and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Kanar Alkass
- Department of Oncology-Pathology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Mehran Salehpour
- Department of Physics and Astronomy, Applied Nuclear Physics, Ion Physics, Uppsala University, 75120 Uppsala, Sweden
| | - Andrea Zimmermann
- Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, 04103 Leipzig, Germany; Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, 04103 Leipzig, Germany; Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - Göran Possnert
- Department of Physics and Astronomy, Applied Nuclear Physics, Ion Physics, Uppsala University, 75120 Uppsala, Sweden
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, 04103 Leipzig, Germany; Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - Henrik Druid
- Department of Oncology-Pathology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Lutz Brusch
- Centre for Information Services and High Performance Computing, Technische Universität Dresden, 01187 Dresden, Germany
| | - Olaf Bergmann
- Center for Regenerative Therapies, Technische Universität Dresden, 01307 Dresden, Germany; Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden.
| |
Collapse
|
17
|
Mahmoud GS, Hosny G, Sayed SA. Hepatoprotective effect of trypsin/chymotrypsin against olanzapine-induced non-alcoholic steatohepatitis in rats. Can J Physiol Pharmacol 2021; 99:1088-1096. [PMID: 34473596 DOI: 10.1139/cjpp-2021-0075] [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: 11/22/2022]
Abstract
Metabolic side effects of atypical antipsychotics are an important cause of deterioration of cognitive function and failure of drug adherence. The antifatty effect trypsin/chymotrypsin (T/C) and their mechanisms of action remain unclear. To investigate possible therapeutic effect of T/C in rat model of chronic olanzapine (OLZ) - induced hepatic steatosis. Twenty rats were divided into two groups: control (C), given distilled water, and O, given 1 mg/kg of OLZ orally daily for 7 weeks. Then, both groups were given T/C 3 enzyme activity unit (EAU)/kg orally as an add-on treatment daily for the next 5 weeks and were named T/C or T/C+O groups. Rat performance in radial arm water maze was tested twice before and after T/C treatment. We measured liver enzymes, alpha-1 antitrypsin, albumin, total protein, direct and total bilirubin, inflammatory cytokines, and lipoprotein serum levels. Liver samples were collected for histopathology and Ki67 expression. The T/C add-on caused significant reduction in OLZ-induced elevation of alanine transaminase (ALT; P < 0.01), aspartate transaminase (AST; P < 0.001), alkaline phosphatase (ALP; P < 0.05), total cholesterol (Tc; P < 0.01), low-density lipoproteins (LDL-c; P < 0.05), steatosis score (P < 0.001), hepatocyte necrosis (P < 0.01), and significantly increased Ki67 expression (P < 0.01). The T/C add-on to OLZ provided protection against hepatic steatosis, elevated enzymes, and disturbed lipid profile and increased Ki67 without disturbing memory function.
Collapse
Affiliation(s)
- Ghada S Mahmoud
- Departments of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ghada Hosny
- Department of Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Sally A Sayed
- Departments of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
18
|
Wang J, Zhang C, Chen X, Li Y, Li A, Liu D, Li F, Luo T. Functions of CXC chemokines as biomarkers and potential therapeutic targets in the hepatocellular carcinoma microenvironment. Transl Cancer Res 2021; 10:2169-2187. [PMID: 35116536 PMCID: PMC8797652 DOI: 10.21037/tcr-21-127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/26/2021] [Indexed: 12/16/2022]
Abstract
Background Several studies have indicated that CXC chemokines influence the prognosis and therapy in patients with hepatocellular carcinoma (HCC). However, there are limited studies on the roles of CXC chemokines in HCC based on data acquired from various databases. This study aimed to conduct an in-depth and comprehensive bioinformatic analysis of the expression and functions of CXC chemokines in HCC. Methods Data was obtained from various databases including ONCOMINE, UALCAN, STRING, GeneMinia, DAVID, Kaplan-Meier plotter, TIMER, GSCALite and NetworkAnalyst for the analysis of the expression and functions of the CXC chemokines in HCC. Results Analysis of the differential expression levels of CXC chemokines between HCC and adjacent normal tissues revealed that the mRNA expression levels of CXCL1/2/5/6/7/12/14 were significantly lower in HCC tissues than those in adjacent normal tissues, whereas the mRNA expression levels of CXCL9/16/17 were significantly higher in HCC tissues. Analysis of the relationship between CXC chemokines and overall survival revealed that high mRNA expression levels of CXCL1/3/5/6/8 were associated with poor overall survival, whereas high mRNA expression levels of CXCL2/4/7/9/10/12 were associated with better overall survival. The functions of CXC chemokines and related genes were associated with cytokine-cytokine receptor interactions and chemokine signaling pathway. Analysis of the association between CXC chemokines and activity of cancer pathways indicated that the DNA damage response and hormone androgen receptor (AR) signaling pathways were inhibited, whereas apoptosis, epithelial-mesenchymal transition (EMT) and Ras/mitogen-activated protein kinase (MAPK) signaling pathways were activated. The expression of CXC chemokines was positively correlated with the infiltration of six types of immune cells (B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils and dendritic cells). Conclusions This study has demonstrated that CXC chemokines can influence survival of patients with HCC by recruiting different types of immune cells into the tumor microenvironment.
Collapse
Affiliation(s)
- Jukun Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chao Zhang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xin Chen
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yu Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ang Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Dongbin Liu
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tao Luo
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Dalwadi DA, Torrens L, Abril-Fornaguera J, Pinyol R, Willoughby C, Posey J, Llovet JM, Lanciault C, Russell DW, Grompe M, Naugler WE. Liver Injury Increases the Incidence of HCC following AAV Gene Therapy in Mice. Mol Ther 2021; 29:680-690. [PMID: 33554867 PMCID: PMC7854305 DOI: 10.1016/j.ymthe.2020.10.018] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/10/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Adeno-associated virus (AAV) integrates into host genomes at low frequency, but when integration occurs in oncogenic hotspots it can cause hepatocellular carcinoma (HCC). Given the possibility of recombinant AAV (rAAV) integration leading to HCC, common causes of liver inflammation like non-alcoholic fatty liver disease (NAFLD) may increase the risk of rAAV-induced HCC. A rAAV targeting the oncogenic mouse Rian locus was used, and as expected led to HCC in all mice infected as neonates, likely due to growth-related hepatocyte proliferation in young mice. Mice infected with rAAV as adults did not develop HCC unless they were fed a diet leading to NAFLD, with increased inflammation and hepatocyte proliferation. Female mice were less susceptible to rAAV-induced HCC, and male mice with NAFLD treated with estrogen exhibited less inflammation and immune exhaustion associated with oncogenesis compared to those without estrogen. Adult NAFLD mice infected with a non-targeted control rAAV also developed HCC, though only half as frequently as those exposed to the Rian targeted rAAV. This study shows that adult mice exposed to rAAV gene therapy in the context of chronic liver disease developed HCC at high frequency, and thus warrants further study in humans given the high prevalence of NAFLD in the population.
Collapse
Affiliation(s)
- Dhwanil A Dalwadi
- Department of Medicine, Division of Gastroenterology and Hepatology, Oregon Health and Science University, Portland, OR 97239, USA; Papé Family, Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Laura Torrens
- Translational Research in Hepatic Oncology, Liver Unit, IDIBAPS-Hospital Clínic, University of Barcelona, Catalonia, Spain
| | - Jordi Abril-Fornaguera
- Translational Research in Hepatic Oncology, Liver Unit, IDIBAPS-Hospital Clínic, University of Barcelona, Catalonia, Spain
| | - Roser Pinyol
- Translational Research in Hepatic Oncology, Liver Unit, IDIBAPS-Hospital Clínic, University of Barcelona, Catalonia, Spain
| | - Catherine Willoughby
- Translational Research in Hepatic Oncology, Liver Unit, IDIBAPS-Hospital Clínic, University of Barcelona, Catalonia, Spain
| | - Jeffrey Posey
- Papé Family, Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Josep M Llovet
- Translational Research in Hepatic Oncology, Liver Unit, IDIBAPS-Hospital Clínic, University of Barcelona, Catalonia, Spain; Mount Sinai Liver Cancer Program, Divisions of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, NY, USA; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
| | - Christian Lanciault
- Department of Pathology, Oregon Health and Science University, Portland, OR 97239, USA
| | - David W Russell
- Department of Medicine, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Markus Grompe
- Papé Family, Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Willscott E Naugler
- Department of Medicine, Division of Gastroenterology and Hepatology, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
20
|
Muñoz-Durango N, Arrese M, Hernández A, Jara E, Kalergis AM, Cabrera D. A Mineralocorticoid Receptor Deficiency in Myeloid Cells Reduces Liver Steatosis by Impairing Activation of CD8 + T Cells in a Nonalcoholic Steatohepatitis Mouse Model. Front Immunol 2020; 11:563434. [PMID: 33391254 PMCID: PMC7772468 DOI: 10.3389/fimmu.2020.563434] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/10/2020] [Indexed: 01/05/2023] Open
Abstract
Background and Aims The mineralocorticoid receptor (MR) and renin-angiotensin-aldosterone system (RAAS) are implicated in non-alcoholic liver fatty disease (NALFD). However, inflammatory mechanisms linking MR and RAAS with disease pathology remain unclear. Here we aimed to evaluate the contribution of myeloid MR to the inflammatory response in an animal model of non-alcoholic steatohepatitis (NASH), induced with a methionine-choline deficient diet (MCD). Methods Mice with a conditional deficiency of MR in myeloid cells (MyMRKO) and their counterpart floxed control mice (FC) were fed for 18 days with MCD or chow diet, respectively. Serum levels of aminotransferases and aldosterone levels were measured and hepatic steatosis, inflammation and fibrosis scored histologically. Hepatic triglyceride content (HTC) and hepatic mRNA levels of pro-inflammatory pro-fibrotic-associated genes were also assessed. Deep flow cytometric analysis was used to dissect the immune response during NASH development. Results MyMRKO mice fed with an MCD diet exhibited reduced hepatic inflammation and lower HTC than controls. Absolute number and percentage of liver inflammatory infiltrate cells (except for CD8+ T lymphocytes) were similar in both MyMRKO and control mice fed with an MCD diet but expression of the costimulatory molecule CD86 by dendritic cells and the CD25 activation marker in CD8+ T cells were significantly reduced in MyMRKO. Conclusions Proinflammatory cells are functionally suppressed in the absence of MR. We hypothesized that loss of MR in myeloid cells reduces lipid accumulation in the liver, in part through modulating the adaptive immune response, which is pivotal for the development of steatosis.
Collapse
Affiliation(s)
- Natalia Muñoz-Durango
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marco Arrese
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile.,Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra Hernández
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Evelyn Jara
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomás, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniel Cabrera
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile.,Facultad de Ciencias Médicas, Universidad Bernardo O Higgins, Santiago, Chile
| |
Collapse
|
21
|
Unraveling the Role of Leptin in Liver Function and Its Relationship with Liver Diseases. Int J Mol Sci 2020; 21:ijms21249368. [PMID: 33316927 PMCID: PMC7764544 DOI: 10.3390/ijms21249368] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/19/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Since its discovery twenty-five years ago, the fat-derived hormone leptin has provided a revolutionary framework for studying the physiological role of adipose tissue as an endocrine organ. Leptin exerts pleiotropic effects on many metabolic pathways and is tightly connected with the liver, the major player in systemic metabolism. As a consequence, understanding the metabolic and hormonal interplay between the liver and adipose tissue could provide us with new therapeutic targets for some chronic liver diseases, an increasing problem worldwide. In this review, we assess relevant literature regarding the main metabolic effects of leptin on the liver, by direct regulation or through the central nervous system (CNS). We draw special attention to the contribution of leptin to the non-alcoholic fatty liver disease (NAFLD) pathogenesis and its progression to more advanced stages of the disease as non-alcoholic steatohepatitis (NASH). Likewise, we describe the contribution of leptin to the liver regeneration process after partial hepatectomy, the mainstay of treatment for certain hepatic malignant tumors.
Collapse
|
22
|
Fourman LT, Billingsley JM, Agyapong G, Ho Sui SJ, Feldpausch MN, Purdy J, Zheng I, Pan CS, Corey KE, Torriani M, Kleiner DE, Hadigan CM, Stanley TL, Chung RT, Grinspoon SK. Effects of tesamorelin on hepatic transcriptomic signatures in HIV-associated NAFLD. JCI Insight 2020; 5:140134. [PMID: 32701508 PMCID: PMC7455119 DOI: 10.1172/jci.insight.140134] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common comorbidity among people living with HIV that has a more aggressive course than NAFLD among the general population. In a recent randomized placebo-controlled trial, we demonstrated that the growth hormone-releasing hormone analog tesamorelin reduced liver fat and prevented fibrosis progression in HIV-associated NAFLD over 1 year. As such, tesamorelin is the first strategy that has shown to be effective against NAFLD among the population with HIV. The current study leveraged paired liver biopsy specimens from this trial to identify hepatic gene pathways that are differentially modulated by tesamorelin versus placebo. Using gene set enrichment analysis, we found that tesamorelin increased hepatic expression of hallmark gene sets involved in oxidative phosphorylation and decreased hepatic expression of gene sets contributing to inflammation, tissue repair, and cell division. Tesamorelin also reciprocally up- and downregulated curated gene sets associated with favorable and poor hepatocellular carcinoma prognosis, respectively. Notably, among tesamorelin-treated participants, these changes in hepatic expression correlated with improved fibrosis-related gene score. Our findings inform our knowledge of the biology of pulsatile growth hormone action and provide a mechanistic basis for the observed clinical effects of tesamorelin on the liver.
Collapse
Affiliation(s)
- Lindsay T Fourman
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - James M Billingsley
- Harvard Chan Bioinformatics Core, Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - George Agyapong
- Liver Center, Digestive Healthcare Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Shannan J Ho Sui
- Harvard Chan Bioinformatics Core, Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Meghan N Feldpausch
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Julia Purdy
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Isabel Zheng
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Chelsea S Pan
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kathleen E Corey
- Liver Center, Digestive Healthcare Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Martin Torriani
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - David E Kleiner
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Colleen M Hadigan
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Takara L Stanley
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Raymond T Chung
- Liver Center, Digestive Healthcare Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Coia H, Ma N, Hou Y, Permaul E, Berry DL, Cruz MI, Pannkuk E, Girgis M, Zhu Z, Lee Y, Rodriquez O, Cheema A, Chung FL. Theaphenon E prevents fatty liver disease and increases CD4+ T cell survival in mice fed a high-fat diet. Clin Nutr 2020; 40:110-119. [PMID: 32439267 DOI: 10.1016/j.clnu.2020.04.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/06/2020] [Accepted: 04/24/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND & AIMS Obesity is a major cause of non-alcoholic fatty liver disease (NAFLD). NAFLD is an epidemic affecting nearly 34% of the adult population in the US. As a chronic inflammatory disease, NAFLD influences the immune system by dysregulating T-cell activity. Remedies for the adverse effects on the immune system are urgently needed. We studied Theaphenon E (TE), a standardized formulation of green tea extract, on the adverse effects of NAFLD in C57BL/6J mice fed a high fat diet (HFD). METHODS Mice received HFD, low fat diet (LFD) or HFD+2% TE for 35 weeks. Hepatic lipid accumulation, cell proliferation, apoptosis and CD4+T lymphocytes were measured throughout the bioassay. The hepatic composition of fatty acids was determined. The effects of epigallocatechin gallate (EGCG) metabolites on lipid accumulation in mouse and primary human liver cells were studied. RESULTS Unlike mice receiving HFD, mice on HFD+2% TE maintained normal liver to body weight ratios with low levels of alanine and aspartate aminotransferase (ALT and AST). Hepatic lipid accumulation was observed in HFD mice, accompanied by increased proliferation, reduced apoptosis and loss of CD4+ T lymphocytes. TE significantly inhibited lipid accumulation, decreased proliferation, induced apoptosis and increased CD4+ T cell survival in HFD mice. It was found that the EGCG metabolite EGC-M3 reduced lipid accumulation in mouse and human hepatocytes. Linoleic acid showed the largest increase (2.5-fold) in livers of mice on a HFD and this increase was significantly suppressed by TE. CONCLUSIONS Livers of HFD-fed mice showed lipid accumulation, increased proliferation, reduced apoptosis, elevated linoleic acid and loss of CD4+ T cells. TE effectively ameliorated all of these adverse effects.
Collapse
Affiliation(s)
- Heidi Coia
- Department of Biochemistry & Molecular Biology, Georgetown University Medical Center, Washington, DC 20057, USA.
| | - Ning Ma
- Department of Biochemistry & Molecular Biology, Georgetown University Medical Center, Washington, DC 20057, USA.
| | - Yanqi Hou
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.
| | - Eva Permaul
- Department of Pathology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.
| | - Deborah L Berry
- Department of Pathology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.
| | - M Idalia Cruz
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.
| | - Evan Pannkuk
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.
| | - Michael Girgis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.
| | - Zizhao Zhu
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.
| | - Yichen Lee
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.
| | - Olga Rodriquez
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.
| | - Amrita Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.
| | - Fung-Lung Chung
- Department of Biochemistry & Molecular Biology, Georgetown University Medical Center, Washington, DC 20057, USA; Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.
| |
Collapse
|
24
|
Miyazoe Y, Miuma S, Miyaaki H, Kanda Y, Nakashiki S, Sasaki R, Haraguchi M, Shibata H, Honda T, Taura N, Nakao K. Extracellular vesicles from senescent hepatic stellate cells promote cell viability of hepatoma cells through increasing EGF secretion from differentiated THP-1 cells. Biomed Rep 2020; 12:163-170. [PMID: 32190304 PMCID: PMC7054706 DOI: 10.3892/br.2020.1279] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 01/17/2020] [Indexed: 12/28/2022] Open
Abstract
Since the discovery of the senescence-associated secretory phenotype, the role of senescent hepatic stellate cells (HSCs) in hepatocellular carcinoma (HCC) development has gained increasing attention. Similar to cytokines, extracellular vesicles (EVs) are essential for intercellular communication. However, the function of EVs derived from senescent HSCs in HCC progression has not been extensively studied. The aims of the present study were to characterize the EVs derived from senescent HSCs and determine their role in the tumor microenvironment. Cellular senescence was induced in human hepatic stellate cells (HHSteCs) with various concentrations of etoposide. Induction was confirmed using EdU staining and 53BP1 and p21 immunostaining. EVs were isolated by ultracentrifugation and analyzed by nanoparticle tracking analysis. Multiplex immunoassays were used to compare the levels of growth factors secreted from hepatoma cell lines and macrophage cells pretreated with EVs derived from senescent HHSteCs (senescent EVs) with those pretreated with EVs derived from normal cultured HHSteCs (normal EVs). Treatment with 25 µM etoposide for 3 days was the most effective at inducing senescence in HHSteCs. This finding was confirmed by induction of irreversible cell-cycle arrest, upregulation of 53BP1 and p21 expression, and increased SA-β-gal staining. Senescent HHSteCs released increased quantities of EV particles compared with normally cultured HHSteCs. Multiplex analysis revealed that there was no difference between hepatoma cell lines treated with normal EVs and those treated with senescent EVs in growth factor secretion. In contrast, the secretion of epidermal growth factor (EGF) was increased by macrophage cells treated with senescent EVs compared with those treated with normal EVs. Furthermore, senescent EVs did not affect the viability of hepatoma cells but increased the viability of hepatoma cells co-cultured with macrophage cells. In conclusion, the release of EVs from senescent HSCs was higher compared with normal HSCs. Furthermore, senescent EVs promoted HCC development by upregulating EGF secretion from macrophages.
Collapse
Affiliation(s)
- Yuri Miyazoe
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Satoshi Miuma
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Hisamitsu Miyaaki
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yasuko Kanda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Suguru Nakashiki
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Ryu Sasaki
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Masafumi Haraguchi
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Hidetaka Shibata
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Takuya Honda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Naota Taura
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Kazuhiko Nakao
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| |
Collapse
|
25
|
Usal M, Regnault C, Veyrenc S, Couturier K, Batandier C, Bulteau AL, Lejon D, Combourieu B, Lafond T, Raveton M, Reynaud S. Concomitant exposure to benzo[a]pyrene and triclosan at environmentally relevant concentrations induces metabolic syndrome with multigenerational consequences in Silurana (Xenopus) tropicalis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 689:149-159. [PMID: 31271984 DOI: 10.1016/j.scitotenv.2019.06.386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/18/2019] [Accepted: 06/23/2019] [Indexed: 06/09/2023]
Abstract
Numerous studies suggest that amphibians are highly sensitive to endocrine disruptors (ED) but their precise role in population decline remains unknown. This study shows that frogs exposed to a mixture of ED throughout their life cycle, at environmentally relevant concentrations, developed an unexpected metabolic syndrome. Female Silurana (Xenopus) tropicalis exposed to a mixture of benzo[a]pyrene and triclosan (50 ng·L-1 each) from the tadpole stage developed liver steatosis and transcriptomic signature associated with glucose intolerance syndrome, and pancreatic insulin hyper secretion typical of pre-diabetes. These metabolic disorders were associated with delayed metamorphosis and developmental mortality in their progeny, both of which have been linked to reduced adult recruitment and reproductive success. Indeed, F1 females were smaller and lighter and presented reduced reproductive capacities, demonstrating a reduced fitness of ED-exposed Xenopus. Our results confirm that amphibians are highly sensitive to ED even at concentrations considered to be safe for other animals. This study demonstrates that ED might be considered as direct contributing factors to amphibian population decline, due to their disruption of energetic metabolism.
Collapse
Affiliation(s)
- Marie Usal
- Univ. Grenoble-Alpes, Univ. Savoie Mont Blanc, CNRS, LECA, 38000 Grenoble, France
| | - Christophe Regnault
- Univ. Grenoble-Alpes, Univ. Savoie Mont Blanc, CNRS, LECA, 38000 Grenoble, France
| | - Sylvie Veyrenc
- Univ. Grenoble-Alpes, Univ. Savoie Mont Blanc, CNRS, LECA, 38000 Grenoble, France.
| | | | | | - Anne-Laure Bulteau
- Institut de Génomique Fonctionnelle de Lyon, Univ. Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 69000 Lyon, France.
| | - David Lejon
- Rovaltain Research Company, F26300 Alixan, France.
| | | | - Thomas Lafond
- Centre de Ressources Biologiques Xénopes, Univ. Rennes 1, CNRS, UMS 3387 Rennes, France.
| | - Muriel Raveton
- Univ. Grenoble-Alpes, Univ. Savoie Mont Blanc, CNRS, LECA, 38000 Grenoble, France.
| | - Stéphane Reynaud
- Univ. Grenoble-Alpes, Univ. Savoie Mont Blanc, CNRS, LECA, 38000 Grenoble, France.
| |
Collapse
|
26
|
Li Y, Xu A, Jia S, Huang J. Recent advances in the molecular mechanism of sex disparity in hepatocellular carcinoma. Oncol Lett 2019; 17:4222-4228. [PMID: 30988804 PMCID: PMC6447942 DOI: 10.3892/ol.2019.10127] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 01/25/2019] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is more frequently observed and aggressive in men compared with women. Increasing evidence demonstrates that the sex disparity appears to be mediated by the stimulatory effects of androgens and the protective effects of estrogen in the development and progression of HCC. In the past few decades, studies on the sex difference of HCC mainly focused on the effect of sex hormones on the transactivation of hepatitis B virus X protein and the release of inflammatory cytokines, and these studies have further intensified in recent years. Sex hormones are also involved in genetic alterations and DNA damage repair in hepatocytes through binding to their specific cellular receptors and affecting the corresponding signaling pathways. Furthermore, the theory of sex chromosomes participating in HCC has been considered. The present review discussed the recent advances in the molecular mechanisms of sex disparity in HCC, with the aim of improving the understanding of the underlying critical factors and exploring more effective methods for the prevention and treatment of HCC.
Collapse
Affiliation(s)
- Yanmeng Li
- Experimental Center, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
- National Clinical Research Center for Digestive Disease, Beijing 100050, P.R. China
| | - Anjian Xu
- Experimental Center, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
- National Clinical Research Center for Digestive Disease, Beijing 100050, P.R. China
| | - Siyu Jia
- Experimental Center, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
- National Clinical Research Center for Digestive Disease, Beijing 100050, P.R. China
| | - Jian Huang
- Experimental Center, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
- National Clinical Research Center for Digestive Disease, Beijing 100050, P.R. China
| |
Collapse
|
27
|
Li X, Wang Z, Klaunig JE. Modulation of xenobiotic nuclear receptors in high-fat diet induced non-alcoholic fatty liver disease. Toxicology 2018; 410:199-213. [DOI: 10.1016/j.tox.2018.08.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/06/2018] [Accepted: 08/13/2018] [Indexed: 02/07/2023]
|
28
|
Wan S, Zhang L, Quan Y, Wei K. Resveratrol-loaded PLGA nanoparticles: enhanced stability, solubility and bioactivity of resveratrol for non-alcoholic fatty liver disease therapy. ROYAL SOCIETY OPEN SCIENCE 2018; 5:181457. [PMID: 30564426 PMCID: PMC6281916 DOI: 10.1098/rsos.181457] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 10/16/2018] [Indexed: 05/25/2023]
Abstract
Resveratrol (3, 4', 5-trihydroxy-trans-stilbene, RSV), a nutraceutical, has recently attracted lots of attention because of its outstanding pharmacological potential. The effects of RSV on non-alcoholic fatty liver disease (NAFLD) remain inconclusive, although a wealth of research has been done. The major obstacle presented was RSV's poor bioavailability due to its poor aqueous solubility, chemical instability and intestinal metabolism. In this study, nanotechnology was used to encapsulate RSV to enhance its stability, water solubility and bioactivity, which can be used to treat NAFLD by HepG2 hepatocytes-induced in vitro. RSV-loaded poly (d, l-lactide-co-glycolide acid) (PLGA) nanoparticles (RSV-PLGA-NPs) were prepared according to an oil/water (O/W) emulsion technique. The RSV-PLGA-NPs were of spherical morphology with an average size of 176.1 nm and a negative charge of -22.6 mV. These nanoparticles exhibited remarkable encapsulation efficiency (EE%) (97.25%) and drug loading (14.9%) for RSV. A sustained RSV release from RSV-PLGA-NPs could be achieved especially in acidic conditions when simulating transporting through the gastrointestinal tract. In addition, these nanoparticles were stable enough to store at 4°C for a least six months with unchanged EE%. Moreover, RSV-PLGA-NPs were more efficient in alleviating lipogenesis, promoting lipolysis and reducing hepatocellular proliferation than free RSV due to its improved stability, water solubility and bioactivity. These findings indicated that the RSV-PLGA-NPs provided superb and stable drug delivery with small particle size, high capsulation efficiency, well-controlled drug release, which greatly enhanced the stability, water solubility and bioactivity. Besides, the discovery that the inhibitory effect of RSV-PLGA-NPs on hepatocellular proliferation and lipid accumulation in steatotic HepG2 cells may provide a new way to study the mechanism of NAFLD. Therefore, RSV-PLGA-NPs have a promising potential for NAFLD therapy.
Collapse
Affiliation(s)
- Shuqian Wan
- School of Biological Science and Engineering, South China University of Technology, Guangzhou 510640, People's Republic of China
| | - Long Zhang
- Wenzhou Institute of Biomaterials and Engineering, CAS, Wenzhou, Zhejiang 325011, People's Republic of China
| | - Yunyun Quan
- Wenzhou Institute of Biomaterials and Engineering, CAS, Wenzhou, Zhejiang 325011, People's Republic of China
| | - Kun Wei
- School of Biological Science and Engineering, South China University of Technology, Guangzhou 510640, People's Republic of China
- Wenzhou Institute of Biomaterials and Engineering, CAS, Wenzhou, Zhejiang 325011, People's Republic of China
| |
Collapse
|
29
|
Coia H, Ma N, Hou Y, Dyba MD, Fu Y, Cruz MI, Benitez C, Graham GT, McCutcheon JN, Zheng YL, Sun B, Kallakury BV, Ma J, Fang HB, Berry DL, Muralidaran V, Chung FL. Prevention of Lipid Peroxidation-derived Cyclic DNA Adduct and Mutation in High-Fat Diet-induced Hepatocarcinogenesis by Theaphenon E. Cancer Prev Res (Phila) 2018; 11:665-676. [PMID: 30131435 PMCID: PMC6171362 DOI: 10.1158/1940-6207.capr-18-0160] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/20/2018] [Accepted: 08/17/2018] [Indexed: 12/21/2022]
Abstract
Obesity is associated with cancer risk and its link with liver cancer is particularly strong. Obesity causes non-alcoholic fatty liver disease (NAFLD) that could progress to hepatocellular carcinoma (HCC). Chronic inflammation likely plays a key role. We carried out a bioassay in the high-fat diet (HFD)-fed C57BL/6J mice to provide insight into the mechanisms of obesity-related HCC by studying γ-OHPdG, a mutagenic DNA adduct derived from lipid peroxidation. In an 80-week bioassay, mice received a low-fat diet (LFD), high-fat diet (HFD), and HFD with 2% Theaphenon E (TE) (HFD+TE). HFD mice developed a 42% incidence of HCC and LFD mice a 16%. Remarkably, TE, a standardized green tea extract formulation, completely blocked HCC in HFD mice with a 0% incidence. γ-OHPdG measured in the hepatic DNA of mice fed HFD and HFD+TE showed its levels increased during the early stages of NAFLD in HFD mice and the increases were significantly suppressed by TE, correlating with the tumor data. Whole-exome sequencing showed an increased mutation load in the liver tumors of HFD mice with G>A and G>T as the predominant mutations, consistent with the report that γ-OHPdG induces G>A and G>T. Furthermore, the mutation loads were significantly reduced in HFD+TE mice, particularly G>T, the most common mutation in human HCC. These results demonstrate in a relevant model of obesity-induced HCC that γ-OHPdG formation during fatty liver disease may be an initiating event for accumulated mutations that leads to HCC and this process can be effectively inhibited by TE. Cancer Prev Res; 11(10); 665-76. ©2018 AACR.
Collapse
MESH Headings
- Animals
- Carcinogenesis/drug effects
- Carcinogenesis/genetics
- Carcinoma, Hepatocellular/epidemiology
- Carcinoma, Hepatocellular/etiology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/prevention & control
- DNA Adducts/drug effects
- Diet, High-Fat/adverse effects
- Drug Screening Assays, Antitumor
- Incidence
- Lipid Peroxidation/drug effects
- Liver/drug effects
- Liver/pathology
- Liver Neoplasms, Experimental/epidemiology
- Liver Neoplasms, Experimental/etiology
- Liver Neoplasms, Experimental/pathology
- Liver Neoplasms, Experimental/prevention & control
- Male
- Mice
- Mice, Inbred C57BL
- Mutation Rate
- Obesity/complications
- Obesity/etiology
- Obesity/pathology
- Plant Extracts/administration & dosage
- Plant Extracts/chemistry
- Polyphenols/administration & dosage
- Tea/chemistry
- Exome Sequencing
Collapse
Affiliation(s)
- Heidi Coia
- Department of Biochemistry & Molecular Biology, Georgetown University Medical Center, Washington DC
| | - Ning Ma
- Department of Biochemistry & Molecular Biology, Georgetown University Medical Center, Washington DC
| | - Yanqi Hou
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC
| | - Marcin D Dyba
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC
| | - Ying Fu
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC
| | - M Idalia Cruz
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC
| | - Carlos Benitez
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC
| | - Garrett T Graham
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC
| | - Justine N McCutcheon
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC
| | - Yun-Ling Zheng
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC
| | - Bing Sun
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC
| | - Bhaskar V Kallakury
- Department of Pathology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington
| | - Junfeng Ma
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC
| | - Hong-Bin Fang
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University, Washington DC
| | - Deborah L Berry
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC
| | - Vinona Muralidaran
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC
| | - Fung-Lung Chung
- Department of Biochemistry & Molecular Biology, Georgetown University Medical Center, Washington DC.
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC
| |
Collapse
|
30
|
Ma KL, Wu Y, Zhang Y, Wang GH, Hu ZB, Ruan XZ. Activation of the CXCL16/CXCR6 pathway promotes lipid deposition in fatty livers of apolipoprotein E knockout mice and HepG2 cells. Am J Transl Res 2018; 10:1802-1816. [PMID: 30018721 PMCID: PMC6038063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 05/19/2018] [Indexed: 06/08/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD), characterised by early lipid accumulation and subsequent inflammation in the liver, is becoming a worldwide challenge due to its increasing prevalence in developing and developed countries. This study aimed to investigate the role of CXC chemokine ligand 16 (CXCL16) and its receptor CXC chemokine receptor 6 (CXCR6) in NAFLD under inflammation. We used IL-1β stimulation in human hepatoblastoma cell line (HepG2) for in vitro studies and casein injection in apolipoprotein E knockout mice in vivo to induce inflammatory stress. The effects of inflammation on cholesterol accumulation were examined by histochemical staining and a quantitative intracellular cholesterol assay. The gene and protein expression of molecules involved in CXCL16/CXCR6 pathway and extracellular matrix (ECM) were examined by real-time polymerase chain reaction (PCR) and Western blotting. The fluorescence intensity of reactive oxygen species (ROS) was assessed by flow cytometry. Results showed that significantly elevated levels of serum amyloid protein A in casein-injected mice confirmed the successful induction of inflamed NAFLD model. Inflammation significantly increased lipid accumulation in livers compared with the high-fat diet group and the controls. Furthermore, inflammation increased the expression of CXCL16, CXCR6, and adisintegrin and metalloproteinase domain-containing protein 10 (ADAM10) in livers, accompanied with increased ECM expression and ROS production. These effects were further confirmed by in vitro studies. Interestingly, CXCL16 gene knockdown in HepG2 cells induced by CXCL16 siRNA resulted in decreased lipid accumulation, ECM excretion, and ROS production. These findings demonstrated that inflammation-mediated activation of CXCL16/CXCR6 is involved in the progression of NAFLD.
Collapse
Affiliation(s)
- Kun Ling Ma
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast UniversityNanjing, Jiangsu Province, China
| | - Yu Wu
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast UniversityNanjing, Jiangsu Province, China
| | - Yang Zhang
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast UniversityNanjing, Jiangsu Province, China
| | - Gui Hua Wang
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast UniversityNanjing, Jiangsu Province, China
| | - Ze Bo Hu
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast UniversityNanjing, Jiangsu Province, China
| | - Xiong Zhong Ruan
- Centre for Nephrology, University College London (UCL) Medical School, Royal Free CampusUK
| |
Collapse
|
31
|
Fu H, Xu J, Chen J, Li G, Zhao X, Chen P. Microarray analysis reveals Tmub1 as a cell cycle-associated protein in rat hepatocytes. Mol Med Rep 2018; 17:4337-4344. [PMID: 29344642 PMCID: PMC5802207 DOI: 10.3892/mmr.2018.8451] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 01/11/2018] [Indexed: 12/18/2022] Open
Abstract
Transmembrane and ubiquitin-like domain containing protein 1 (Tmub1), formerly known as hepatocyte odd protein shuttling (HOPS) has been recognized as a ubiquitously expressed shuttling protein that moves between the nucleus and cytoplasm in hepatocytes. Tmub1 is involved in liver regeneration and functions as a bridging protein in tumor cell proliferation. To investigate the transcriptional profile and potential biological processes affected by Tmub1 expression in normal rat hepatocytes, microarray and bioinformatics experiments were used to identify 127 mRNAs differentially expressed between Tmub1-overexpression, Tmub1-knockdown and normal BRL-3A cells (fold-change ≥2.5). The expression levels of 17 key node genes associated with the cell cycle were confirmed by reverse transcription-quantitative polymerase chain reaction analysis. Flow cytometry, 5-Ethynyl-20-deoxyuridine, Cell Counting Kit-8 and western blotting experiments revealed the effects on the cell cycle and the inhibition of proliferation in BRL-3A cells overexpressing Tmub1. Further co-immunoprecipitation assays demonstrated that Tmub1 interacts with cyclin A2 during the cell cycle and that the overexpression of Tmub1 may postpone cyclin A2 and cyclin B1 degradation in the M phase. The results of the present study indicated that Tmub1 functions as a cell proliferation inhibitor and cell cycle-associated protein.
Collapse
Affiliation(s)
- Hangwei Fu
- Department of Hepatobiliary Surgery, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Jianhua Xu
- Department of Hepatobiliary Surgery, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Jian Chen
- Department of Hepatobiliary Surgery, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Guangyao Li
- Department of Hepatobiliary Surgery, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Xiaobiao Zhao
- Department of Hepatobiliary Surgery, 187 Military Hospital, Haikou, Hainan 571159, P.R. China
| | - Ping Chen
- Department of Hepatobiliary Surgery, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| |
Collapse
|
32
|
Khadge S, Sharp JG, Thiele GM, McGuire TR, Klassen LW, Duryee MJ, Britton HC, Dafferner AJ, Beck J, Black PN, DiRusso CC, Talmadge J. Dietary omega-3 and omega-6 polyunsaturated fatty acids modulate hepatic pathology. J Nutr Biochem 2017; 52:92-102. [PMID: 29175671 DOI: 10.1016/j.jnutbio.2017.09.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 09/06/2017] [Accepted: 09/13/2017] [Indexed: 12/13/2022]
Abstract
Recent evidence has suggested that dietary polyunsaturated fatty acids (PUFAs) modulate inflammation; however, few studies have focused on the pathobiology of PUFA using isocaloric and isolipidic diets and it is unclear if the associated pathologies are due to dietary PUFA composition, lipid metabolism or obesity, as most studies compare diets fed ad libitum. Our studies used isocaloric and isolipidic liquid diets (35% of calories from fat), with differing compositions of omega (ω)-6 or long chain (Lc) ω-3 PUFA that were pair-fed and assessed hepatic pathology, inflammation and lipid metabolism. Consistent with an isocaloric, pair-fed model we observed no significant difference in diet consumption between the groups. In contrast, the body and liver weight, total lipid level and abdominal fat deposits were significantly higher in mice fed an ω-6 diet. An analysis of the fatty acid profile in plasma and liver showed that mice on the ω-6 diet had significantly more arachidonic acid (AA) in the plasma and liver, whereas, in these mice ω-3 fatty acids such as eicosapentaenoic acid (EPA) were not detected and docosahexaenoic acid (DHA) was significantly lower. Histopathologic analyses documented that mice on the ω-6 diet had a significant increase in macrovesicular steatosis, extramedullary myelopoiesis (EMM), apoptotic hepatocytes and decreased glycogen storage in lobular hepatocytes, and hepatocyte proliferation relative to mice fed the Lc ω-3 diet. Together, these results support PUFA dietary regulation of hepatic pathology and inflammation with implications for enteral feeding regulation of steatosis and other hepatic lesions.
Collapse
Affiliation(s)
- Saraswoti Khadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | - John Graham Sharp
- Depatment of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE
| | - Geoffrey M Thiele
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE; Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE
| | - Timothy R McGuire
- Department of Pharmacy Practice, University of Nebraska Medical Center, Omaha, NE
| | - Lynell W Klassen
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE; Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE
| | - Michael J Duryee
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Holly C Britton
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | - Alicia J Dafferner
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | - Jordan Beck
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE
| | - Paul N Black
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE
| | | | - James Talmadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE.
| |
Collapse
|
33
|
Salazar GA, Peñaloza HF, Pardo-Roa C, Schultz BM, Muñoz-Durango N, Gómez RS, Salazar FJ, Pizarro DP, Riedel CA, González PA, Alvarez-Lobos M, Kalergis AM, Bueno SM. Interleukin-10 Production by T and B Cells Is a Key Factor to Promote Systemic Salmonella enterica Serovar Typhimurium Infection in Mice. Front Immunol 2017; 8:889. [PMID: 28824622 PMCID: PMC5539121 DOI: 10.3389/fimmu.2017.00889] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/12/2017] [Indexed: 12/16/2022] Open
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is a Gram-negative bacterium that produces disease in numerous hosts. In mice, oral inoculation is followed by intestinal colonization and subsequent systemic dissemination, which leads to severe pathogenesis without the activation of an efficient anti-Salmonella immune response. This feature suggests that the infection caused by S. Typhimurium may promote the production of anti-inflammatory molecules by the host that prevent efficient T cell activation and bacterial clearance. In this study, we describe the contribution of immune cells producing the anti-inflammatory cytokine interleukin-10 (IL-10) to the systemic infection caused by S. Typhimurium in mice. We observed that the production of IL-10 was required by S. Typhimurium to cause a systemic disease, since mice lacking IL-10 (IL-10-/-) were significantly more resistant to die after an infection as compared to wild-type (WT) mice. IL-10-/- mice had reduced bacterial loads in internal organs and increased levels of pro-inflammatory cytokines in serum at 5 days of infection. Importantly, WT mice showed high bacterial loads in tissues and no increase of cytokines in serum after 5 days of S. Typhimurium infection, except for IL-10. In WT mice, we observed a peak of il-10 messenger RNA production in ileum, spleen, and liver after 5 days of infection. Importantly, the adoptive transfer of T or B cells from WT mice restored the susceptibility of IL-10-/- mice to systemic S. Typhimurium infection, suggesting that the generation of regulatory cells in vivo is required to sustain a systemic infection by S. Typhimurium. These findings support the notion that IL-10 production from lymphoid cells is a key process in the infective cycle of S. Typhimurium in mice due to generation of a tolerogenic immune response that prevents bacterial clearance and supports systemic dissemination.
Collapse
Affiliation(s)
- Geraldyne A. Salazar
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de, Chile Santiago, Chile
| | - Hernán F. Peñaloza
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de, Chile Santiago, Chile
| | - Catalina Pardo-Roa
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de, Chile Santiago, Chile
| | - Bárbara M. Schultz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de, Chile Santiago, Chile
| | - Natalia Muñoz-Durango
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de, Chile Santiago, Chile
| | - Roberto S. Gómez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de, Chile Santiago, Chile
| | - Francisco J. Salazar
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de, Chile Santiago, Chile
| | - Daniela P. Pizarro
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de, Chile Santiago, Chile
| | - Claudia A. Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas y Medicina, Universidad Andrés Bello, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de, Chile Santiago, Chile
| | - Manuel Alvarez-Lobos
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de, Chile Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de, Chile Santiago, Chile
| |
Collapse
|
34
|
Adipokines and Non-Alcoholic Fatty Liver Disease: Multiple Interactions. Int J Mol Sci 2017; 18:ijms18081649. [PMID: 28758929 PMCID: PMC5578039 DOI: 10.3390/ijms18081649] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 07/18/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence links obesity with low-grade inflammation which may originate from adipose tissue that secretes a plethora of pro- and anti-inflammatory cytokines termed adipokines. Adiponectin and leptin have evolved as crucial signals in many obesity-related pathologies including non-alcoholic fatty liver disease (NAFLD). Whereas adiponectin deficiency might be critically involved in the pro-inflammatory state associated with obesity and related disorders, overproduction of leptin, a rather pro-inflammatory mediator, is considered of equal relevance. An imbalanced adipokine profile in obesity consecutively contributes to metabolic inflammation in NAFLD, which is associated with a substantial risk for developing hepatocellular carcinoma (HCC) also in the non-cirrhotic stage of disease. Both adiponectin and leptin have been related to liver tumorigenesis especially in preclinical models. This review covers recent advances in our understanding of some adipokines in NAFLD and associated HCC.
Collapse
|
35
|
Haga H, Yan IK, Borrelli DA, Matsuda A, Parasramka M, Shukla N, Lee DD, Patel T. Extracellular vesicles from bone marrow-derived mesenchymal stem cells protect against murine hepatic ischemia/reperfusion injury. Liver Transpl 2017; 23:791-803. [PMID: 28407355 PMCID: PMC5495137 DOI: 10.1002/lt.24770] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 03/01/2017] [Indexed: 12/20/2022]
Abstract
Hepatic ischemia/reperfusion injury (IRI) and associated inflammation contributes to liver dysfunction and complications after liver surgery and transplantation. Mesenchymal stem cells (MSCs) have been reported to reduce hepatic IRI because of their reparative immunomodulatory effects in injured tissues. Recent studies have highlighted beneficial effects of extracellular vesicles from mesenchymal stem cells (MSC-EV) on tissue injury. The effects of systemically administered mouse bone marrow-derived MSC-EV were evaluated in an experimental murine model of hepatic IRI induced by cross-clamping the hepatic artery and portal vein for 90 minutes followed by reperfusion for periods of up to 6 hours. Compared with controls, intravenous administration of MSC-EV 30 minutes prior to IRI dramatically reduced the extent of tissue necrosis, decreased caspase 3-positive and apoptotic cells, and reduced serum aminotransferase levels. MSC-EV increased hepatic messenger RNA (mRNA) expression of NACHT, LRR, and PYD domains-containing protein 12, and the chemokine (C-X-C motif) ligand 1, and reduced mRNA expression of several inflammatory cytokines such as interleukin 6 during IRI. MSC-EV increased cell viability and suppressed both oxidative injury and nuclear factor kappa B activity in murine hepatocytes in vitro. In conclusion, the administration of extracellular vesicles derived from bone marrow-derived MSCs may ameliorate hepatic IRI by reducing hepatic injury through modulation of the inflammatory response.Liver Transplantation 23 791-803 2017 AASLD.
Collapse
Affiliation(s)
- Hiroaki Haga
- Departments of Transplantation and Cancer Biology; Mayo Clinic; Jacksonville Florida
| | - Irene K. Yan
- Departments of Transplantation and Cancer Biology; Mayo Clinic; Jacksonville Florida
| | - David A. Borrelli
- Departments of Transplantation and Cancer Biology; Mayo Clinic; Jacksonville Florida
| | - Akiko Matsuda
- Departments of Transplantation and Cancer Biology; Mayo Clinic; Jacksonville Florida
| | - Mansi Parasramka
- Departments of Transplantation and Cancer Biology; Mayo Clinic; Jacksonville Florida
| | - Neha Shukla
- Departments of Transplantation and Cancer Biology; Mayo Clinic; Jacksonville Florida
| | - David D. Lee
- Departments of Transplantation and Cancer Biology; Mayo Clinic; Jacksonville Florida
| | - Tushar Patel
- Departments of Transplantation and Cancer Biology; Mayo Clinic; Jacksonville Florida
| |
Collapse
|
36
|
Characterization of long non-coding RNA transcriptome in high-energy diet induced nonalcoholic steatohepatitis minipigs. Sci Rep 2016; 6:30709. [PMID: 27466003 PMCID: PMC4964571 DOI: 10.1038/srep30709] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 07/07/2016] [Indexed: 12/17/2022] Open
Abstract
Today, obesity and nonalcoholic steatohepatitis are a worldwide epidemic, although how these syndromes are regulated with respect to lncRNAs remains largely unknown. Our previous studies have revealed important pathological features and molecular characteristics of nonalcoholic steatohepatitis in the minipig model, and in this study, we analyze the features of lncRNAs and their potential target genes. Minipig samples only from liver were analyzed using next-generation deep sequencing. In total, we obtained 585 million raw reads approximately 70.4 Gb of high quality data. After a strict five-step filtering process, 1,179 lncRNAs were identified, including 89 differentially expressed lncRNAs (P < 0.05) in the experiment group relative to the control group. The cis and trans analysis identified target genes that were enriched for specific GO terms (P < 0.01), including immune processes, chemokine activity, cytokine activity, and G-protein coupled receptor binding, which are closely related to nonalcoholic steatohepatitis. The predicted protein-coding targets of the differentially expressed lncRNAs were further analyzed, such as PPAR, FADS2, DGAT2, ACAA2, CYP2E1, ADH4, and Fos. This study reveals a wealth of candidate lncRNAs involved in NASH and their regulated pathways, which should facilitate further research into the molecular mechanisms of this disorder.
Collapse
|
37
|
Abstract
The recent recognition of the clinical association between type 2 diabetes (T2D) and several types of human cancer has been further highlighted by reports of antidiabetic drugs treating or promoting cancer. At the cellular level, a plethora of molecules operating within distinct signaling pathways suggests cross-talk between the multiple pathways at the interface of the diabetes–cancer link. Additionally, a growing body of emerging evidence implicates homeostatic pathways that may become imbalanced during the pathogenesis of T2D or cancer or that become chronically deregulated by prolonged drug administration, leading to the development of cancer in diabetes and vice versa. This notion underscores the importance of combining clinical and basic mechanistic studies not only to unravel mechanisms of disease development but also to understand mechanisms of drug action. In turn, this may help the development of personalized strategies in which drug doses and administration durations are tailored to individual cases at different stages of the disease progression to achieve more efficacious treatments that undermine the diabetes–cancer association.
Collapse
Affiliation(s)
- Slavica Tudzarova
- Wolfson Institute for Biomedical Research, University College London, London WC1E6BT, UK
| | - Mahasin A Osman
- Department of Molecular Physiology, Pharmacology and Biotechnology, Division of Biology and Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912 Department of Chemistry and Forensic Sciences, College of Sciences and Technology, Savannah State University, Savannah, GA 41404
| |
Collapse
|
38
|
Jin J, Valanejad L, Nguyen TP, Lewis K, Wright M, Cast A, Stock L, Timchenko L, Timchenko NA. Activation of CDK4 Triggers Development of Non-alcoholic Fatty Liver Disease. Cell Rep 2016; 16:744-56. [PMID: 27373160 DOI: 10.1016/j.celrep.2016.06.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/18/2016] [Accepted: 05/31/2016] [Indexed: 01/13/2023] Open
Abstract
The development of non-alcoholic fatty liver disease (NAFLD) is a multiple step process. Here, we show that activation of cdk4 triggers the development of NAFLD. We found that cdk4 protein levels are elevated in mouse models of NAFLD and in patients with fatty livers. This increase leads to C/EBPα phosphorylation on Ser193 and formation of C/EBPα-p300 complexes, resulting in hepatic steatosis, fibrosis, and hepatocellular carcinoma (HCC). The disruption of this pathway in cdk4-resistant C/EBPα-S193A mice dramatically reduces development of high-fat diet (HFD)-mediated NAFLD. In addition, inhibition of cdk4 by flavopiridol or PD-0332991 significantly reduces development of hepatic steatosis, the first step of NAFLD. Thus, this study reveals that activation of cdk4 triggers NAFLD and that inhibitors of cdk4 may be used for the prevention/treatment of NAFLD.
Collapse
Affiliation(s)
- Jingling Jin
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Leila Valanejad
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue ML 7015, Cincinnati, OH 45229, USA
| | - Thuy Phuong Nguyen
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue ML 7015, Cincinnati, OH 45229, USA
| | - Kyle Lewis
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue ML 7015, Cincinnati, OH 45229, USA; Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Mary Wright
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue ML 7015, Cincinnati, OH 45229, USA
| | - Ashley Cast
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue ML 7015, Cincinnati, OH 45229, USA
| | - Lauren Stock
- Department of Neurology, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue ML 7015, Cincinnati, OH 45229, USA
| | - Lubov Timchenko
- Department of Neurology, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue ML 7015, Cincinnati, OH 45229, USA
| | - Nikolai A Timchenko
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue ML 7015, Cincinnati, OH 45229, USA; Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
39
|
Matter MS, Marquardt JU, Andersen JB, Quintavalle C, Korokhov N, Stauffer JK, Kaji K, Decaens T, Quagliata L, Elloumi F, Hoang T, Molinolo A, Conner EA, Weber A, Heikenwalder M, Factor VM, Thorgeirsson SS. Oncogenic driver genes and the inflammatory microenvironment dictate liver tumor phenotype. Hepatology 2016; 63:1888-99. [PMID: 26844528 PMCID: PMC4874846 DOI: 10.1002/hep.28487] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 01/29/2016] [Accepted: 02/01/2016] [Indexed: 12/12/2022]
Abstract
UNLABELLED The majority of hepatocellular carcinoma develops in the background of chronic liver inflammation caused by viral hepatitis and alcoholic or nonalcoholic steatohepatitis. However, the impact of different types of chronic inflammatory microenvironments on the phenotypes of tumors generated by distinct oncogenes is largely unresolved. To address this issue, we generated murine liver tumors by constitutively active AKT-1 (AKT) and β-catenin (CAT), followed by induction of chronic liver inflammation by 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) and carbon tetrachloride. Also, the impact of DDC-induced chronic liver inflammation was compared between two liver tumor models using a combination of AKT-CAT or AKT-NRAS(G12V) . Treatment with DDC and carbon tetrachloride significantly facilitated the adenoma-to-carcinoma conversion and accelerated the growth of AKT-CAT tumors. Furthermore, DDC treatment altered the morphology of AKT-CAT tumors and caused loss of lipid droplets. Transcriptome analysis of AKT-CAT tumors revealed that cellular growth and proliferation were mainly affected by chronic inflammation and caused up-regulation of Cxcl16, Galectin-3, and Nedd9, among others. Integration with transcriptome profiles from human hepatocellular carcinomas further demonstrated that AKT-CAT tumors generated in the context of chronic liver inflammation showed enrichment of poor prognosis gene sets or decrease of good prognosis gene sets. In contrast, DDC had a more subtle effect on AKT-NRAS(G12V) tumors and primarily enhanced already existent tumor characteristics as supported by transcriptome analysis. However, it also reduced lipid droplets in AKT-NRAS(G12V) tumors. CONCLUSION Our study suggests that liver tumor phenotype is defined by a combination of driving oncogenes but also the nature of chronic liver inflammation. (Hepatology 2016;63:1888-1899).
Collapse
Affiliation(s)
- Matthias S. Matter
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, NCI-NIH, Bethesda, Maryland,Institute of Pathology, University Hospital of Basel, Basel, Switzerland
| | - Jens U. Marquardt
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, NCI-NIH, Bethesda, Maryland,Department of Medicine I, Johannes Gutenberg University, Mainz, Germany
| | - Jesper B. Andersen
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, NCI-NIH, Bethesda, Maryland,Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | | | - Nikolay Korokhov
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, NCI-NIH, Bethesda, Maryland
| | - Jim K. Stauffer
- Cancer and Inflammation Program, NCI-Frederick, Frederick, Maryland
| | - Kosuke Kaji
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, NCI-NIH, Bethesda, Maryland
| | - Thomas Decaens
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, NCI-NIH, Bethesda, Maryland
| | - Luca Quagliata
- Institute of Pathology, University Hospital of Basel, Basel, Switzerland
| | - Fathi Elloumi
- National Cancer Institute, CCR at Leidos Inc. NIH, Bethesda, Maryland
| | - Tanya Hoang
- Institute of Pathology, University Hospital of Basel, Basel, Switzerland
| | - Alfredo Molinolo
- Oral and Pharyngeal Cancer Branch, NIDCR-NIH, Bethesda, Maryland
| | - Elizabeth A. Conner
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, NCI-NIH, Bethesda, Maryland
| | - Achim Weber
- Institute of Surgical Pathology, University and University Hospital Zurich, 8091 Zurich, Switzerland
| | - Mathias Heikenwalder
- Institute of Virology, Technische Universität München (TUM)/Helmholtz Zentrum München (HMGU), Munich, Germany,Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Neuenheimer Feld, Heidelberg, Germany
| | - Valentina M. Factor
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, NCI-NIH, Bethesda, Maryland
| | - Snorri S. Thorgeirsson
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, NCI-NIH, Bethesda, Maryland
| |
Collapse
|
40
|
Shen M, Shi H. Estradiol and Estrogen Receptor Agonists Oppose Oncogenic Actions of Leptin in HepG2 Cells. PLoS One 2016; 11:e0151455. [PMID: 26982332 PMCID: PMC4794158 DOI: 10.1371/journal.pone.0151455] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 02/28/2016] [Indexed: 11/19/2022] Open
Abstract
Obesity is a significant risk factor for certain cancers, including hepatocellular carcinoma (HCC). Leptin, a hormone secreted by white adipose tissue, precipitates HCC development. Epidemiology data show that men have a much higher incidence of HCC than women, suggesting that estrogens and its receptors may inhibit HCC development and progression. Whether estrogens antagonize oncogenic action of leptin is uncertain. To investigate potential inhibitory effects of estrogens on leptin-induced HCC development, HCC cell line HepG2 cells were treated with leptin in combination with 17 β-estradiol (E2), estrogen receptor-α (ER-α) selective agonist PPT, ER-β selective agonist DPN, or G protein-coupled ER (GPER) selective agonist G-1. Cell number, proliferation, and apoptosis were determined, and leptin- and estrogen-related intracellular signaling pathways were analyzed. HepG2 cells expressed a low level of ER-β mRNA, and leptin treatment increased ER-β expression. E2 suppressed leptin-induced HepG2 cell proliferation and promoted cell apoptosis in a dose-dependent manner. Additionally E2 reversed leptin-induced STAT3 and leptin-suppressed SOCS3, which was mainly achieved by activation of ER-β. E2 also enhanced ERK via activating ER-α and GPER and activated p38/MAPK via activating ER-β. To conclude, E2 and its receptors antagonize the oncogenic actions of leptin in HepG2 cells by inhibiting cell proliferation and stimulating cell apoptosis, which was associated with reversing leptin-induced changes in SOCS3/STAT3 and increasing p38/MAPK by activating ER-β, and increasing ERK by activating ER-α and GPER. Identifying roles of different estrogen receptors would provide comprehensive understanding of estrogenic mechanisms in HCC development and shed light on potential treatment for HCC patients.
Collapse
Affiliation(s)
- Minqian Shen
- Department of Biology, Cell, Molecular, and Structural Biology, Miami University, Oxford, Ohio, United States of America
| | - Haifei Shi
- Department of Biology, Cell, Molecular, and Structural Biology, Miami University, Oxford, Ohio, United States of America
- * E-mail:
| |
Collapse
|
41
|
Tarantino G, Finelli C. Lipids, Low-Grade Chronic Inflammation and NAFLD. HANDBOOK OF LIPIDS IN HUMAN FUNCTION 2016:731-759. [DOI: 10.1016/b978-1-63067-036-8.00028-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
42
|
Song X, Wang Z, Jin Y, Wang Y, Duan W. Loss of miR-532-5p in vitro promotes cell proliferation and metastasis by influencing CXCL2 expression in HCC. Am J Transl Res 2015; 7:2254-2261. [PMID: 26807173 PMCID: PMC4697705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 07/31/2015] [Indexed: 06/05/2023]
Abstract
MicroRNAs (miRNAs) have been widely reported, which play important roles in cancer development. CXCL2 acts as an oncogene, however, its regulation by miRNAs is not clear in hepatocellular carcinoma (HCC). In our research, it is aimed to study the role of CXCL2 in HCC and the regulation of its expression by miRNAs. Firstly, we found that CXCL2 was up-regulated in the blood of patients with HCC and cell lines compared with the normal controls. CXCL2 could enhance HCC cell proliferation and metastasis. miR-532-5p was predicted as a regulatory miRNA of CXCL2 in HCC, and negatively associated with CXCL2 in HCC samples. It was also verified that miR-532-5p inhibited cell proliferation and metastasis of HCC cells by inhibition CXCL2. Collectively, our findings suggested that miR-532-5p may function as a tumor suppressor in HCC by targeting CXCL2.
Collapse
Affiliation(s)
- Xiaofei Song
- Department of Clinical Laboratory, Shandong Provincial Hospital Affliated to Shandong Uiniversty Jinan, China
| | - Zie Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affliated to Shandong Uiniversty Jinan, China
| | - Yan Jin
- Department of Clinical Laboratory, Shandong Provincial Hospital Affliated to Shandong Uiniversty Jinan, China
| | - Yong Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affliated to Shandong Uiniversty Jinan, China
| | - Wenbing Duan
- Department of Clinical Laboratory, Shandong Provincial Hospital Affliated to Shandong Uiniversty Jinan, China
| |
Collapse
|
43
|
The Roles of CXCL16 and CXCR6 in Liver Inflammation and Fibrosis. CURRENT PATHOBIOLOGY REPORTS 2015. [DOI: 10.1007/s40139-015-0090-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
44
|
Nguyen-Lefebvre AT, Horuzsko A. Kupffer Cell Metabolism and Function. JOURNAL OF ENZYMOLOGY AND METABOLISM 2015; 1:101. [PMID: 26937490 PMCID: PMC4771376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Kupffer cells are resident liver macrophages and play a critical role in maintaining liver functions. Under physiological conditions, they are the first innate immune cells and protect the liver from bacterial infections. Under pathological conditions, they are activated by different components and can differentiate into M1-like (classical) or M2-like (alternative) macrophages. The metabolism of classical or alternative activated Kupffer cells will determine their functions in liver damage. Special functions and metabolism of Kupffer cells suggest that they are an attractive target for therapy of liver inflammation and related diseases, including cancer and infectious diseases. Here we review the different types of Kupffer cells and their metabolism and functions in physiological and pathological conditions.
Collapse
Affiliation(s)
- Anh Thu Nguyen-Lefebvre
- Cancer Center, Molecular Oncology Program, Georgia Regents University, Augusta, GA, 30912, USA
| | - Anatolij Horuzsko
- Cancer Center, Molecular Oncology Program, Georgia Regents University, Augusta, GA, 30912, USA
- Cancer Center, Molecular Oncology Program, Department of Medicine, Georgia Regents University, 1410 Laney Walker Blvd., Augusta, GA, 30912, USA
| |
Collapse
|
45
|
Orci LA, Berney T, Majno PE, Lacotte S, Oldani G, Morel P, Mentha G, Toso C. Donor characteristics and risk of hepatocellular carcinoma recurrence after liver transplantation. Br J Surg 2015; 102:1250-7. [PMID: 26098966 DOI: 10.1002/bjs.9868] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 04/05/2015] [Accepted: 05/07/2015] [Indexed: 01/27/2023]
Abstract
BACKGROUND To date, studies assessing the risk of post-transplant hepatocellular carcinoma (HCC) recurrence have focused on tumour characteristics. This study investigated the impact of donor characteristics and graft quality on post-transplant HCC recurrence. METHODS Using the Scientific Registry of Transplant Recipients patients with HCC who received a liver transplant between 2004 and 2011 were included, and post-transplant HCC recurrence was assessed. A multivariable competing risk regression model was fitted, adjusting for confounders such as recipient sex, age, tumour volume, α-fetoprotein, time on the waiting list and transplant centre. RESULTS A total of 9724 liver transplant recipients were included. Patients receiving a graft procured from a donor older than 60 years (adjusted hazard ratio (HR) 1.38, 95 per cent c.i. 1.10 to 1.73; P = 0.006), a donor with a history of diabetes (adjusted HR 1.43, 1.11 to 1.83; P = 0.006) and a donor with a body mass index of 35 kg/m(2) or more (adjusted HR 1.36, 1.04 to 1.77; P = 0.023) had an increased rate of post-transplant HCC recurrence. In 3007 patients with documented steatosis, severe graft steatosis (more than 60 per cent) was also linked to an increased risk of recurrence (adjusted HR 1.65, 1.03 to 2.64; P = 0.037). Recipients of organs from donation after cardiac death donors with prolonged warm ischaemia had higher recurrence rates (adjusted HR 4.26, 1.20 to 15.1; P = 0.025). CONCLUSION Donor-related factors such as donor age, body mass index, diabetes and steatosis are associated with an increased rate of HCC recurrence after liver transplantation.
Collapse
Affiliation(s)
- L A Orci
- Division of Abdominal and Transplantation Surgery, Department of Surgery, University of Geneva, Geneva, Switzerland.,Hepato-Pancreato-Biliary Centre, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - T Berney
- Division of Abdominal and Transplantation Surgery, Department of Surgery, University of Geneva, Geneva, Switzerland.,Hepato-Pancreato-Biliary Centre, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - P E Majno
- Division of Abdominal and Transplantation Surgery, Department of Surgery, University of Geneva, Geneva, Switzerland.,Hepato-Pancreato-Biliary Centre, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - S Lacotte
- Division of Abdominal and Transplantation Surgery, Department of Surgery, University of Geneva, Geneva, Switzerland
| | - G Oldani
- Division of Abdominal and Transplantation Surgery, Department of Surgery, University of Geneva, Geneva, Switzerland.,Hepato-Pancreato-Biliary Centre, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - P Morel
- Division of Abdominal and Transplantation Surgery, Department of Surgery, University of Geneva, Geneva, Switzerland.,Hepato-Pancreato-Biliary Centre, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - G Mentha
- Division of Abdominal and Transplantation Surgery, Department of Surgery, University of Geneva, Geneva, Switzerland.,Hepato-Pancreato-Biliary Centre, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - C Toso
- Division of Abdominal and Transplantation Surgery, Department of Surgery, University of Geneva, Geneva, Switzerland.,Hepato-Pancreato-Biliary Centre, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
46
|
Liang CM, Chen L, Hu H, Ma HY, Gao LL, Qin J, Zhong CP. Chemokines and their receptors play important roles in the development of hepatocellular carcinoma. World J Hepatol 2015; 7:1390-1402. [PMID: 26052384 PMCID: PMC4450202 DOI: 10.4254/wjh.v7.i10.1390] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 11/08/2014] [Accepted: 04/07/2015] [Indexed: 02/06/2023] Open
Abstract
The chemokine system consists of four different subclasses with over 50 chemokines and 19 receptors. Their functions in the immune system have been well elucidated and research during the last decades unveils their new roles in hepatocellular carcinoma (HCC). The chemokines and their receptors in the microenvironment influence the development of HCC by several aspects including: inflammation, effects on immune cells, angiogenesis, and direct effects on HCC cells. Regarding these aspects, pre-clinical research by targeting the chemokine system has yielded promising data, and these findings bring us new clues in the chemokine-based therapies for HCC.
Collapse
|
47
|
20 years of leptin: Role of leptin in cardiomyocyte physiology and physiopathology. Life Sci 2015; 140:10-8. [PMID: 25748420 DOI: 10.1016/j.lfs.2015.02.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 02/14/2015] [Indexed: 02/08/2023]
Abstract
Since the discovery of leptin in 1994 by Zhang et al., there have been a number of reports showing its implication in the development of a wide range of cardiovascular diseases. However, there exists some controversy about how leptin can induce or preserve cardiovascular function, as different authors have found contradictory results about leptin beneficial or detrimental effects in leptin deficient/resistant murine models and in wild type tissue and cardiomyocytes. Here, we will focus on the main discoveries about the leptin functions at cardiac level within the last two decades, focusing on its role in cardiac metabolism, remodeling and contractile function.
Collapse
|
48
|
Mendonsa AM, VanSaun MN, Ustione A, Piston DW, Fingleton BM, Gorden DL. Host and tumor derived MMP13 regulate extravasation and establishment of colorectal metastases in the liver. Mol Cancer 2015; 14:49. [PMID: 25880591 PMCID: PMC4351934 DOI: 10.1186/s12943-014-0282-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/22/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Non alcoholic fatty liver disease (NAFLD) is one of the most common liver diseases in the United States and worldwide. Our studies have previously shown an increase in metastatic burden in steatotic vs. normal livers using a mouse model of diet induced steatosis. In the present study we aim to identify and evaluate the molecular factors responsible for this increase in tumor burden. METHODS We assessed changes in expression of a panel of matrix metalloproteinases (MMPs) using qRT-PCR between normal and steatotic livers and validated them with western blot analysis of protein levels. To evaluate the role of MMP13 on tumor development, we utilized a splenic injection model of liver metastasis in Wildtype and Mmp13 deficient mice, using either parental or stable Mmp13 knockdown cell lines. Further, to evaluate changes in the ability of tumor cells to extravasate we utilized whole organ confocal microscopy to identify individual tumor cells relative to the vasculature. MTT, migration and invasion assays were performed to evaluate the role of tumor derived MMP13 on hallmarks of cancer in vitro. RESULTS We found that MMP13 was significantly upregulated in the steatotic liver both in mice as well as human patients with NAFLD. We showed a decrease in metastatic tumor burden in Mmp13-/- mice compared to wildtype mice, explained in part by a reduction in the number of tumor cells extravasating from the hepatic vasculature in the Mmp13-/- mice compared to wildtype mice. Additionally, loss of tumor derived MMP13 through stable knockdown in tumor cell lines lead to decreased migratory and invasive properties in vitro and metastatic burden in vivo. CONCLUSIONS This study demonstrates that stromal as well as tumor derived MMP13 contribute to tumor cell extravasation and establishment of metastases in the liver microenvironment.
Collapse
Affiliation(s)
- Alisha M Mendonsa
- Department of Cancer Biology, Vanderbilt University, 2220 Pierce Ave S, Nashville, TN, 37232, USA.
| | - Michael N VanSaun
- Department of Cancer Biology, Vanderbilt University, 2220 Pierce Ave S, Nashville, TN, 37232, USA. .,Department of Surgery, Vanderbilt University, 801 Oxford House, 1313 21st Ave. S, Nashville, TN, 37212, USA.
| | - Alessandro Ustione
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall 21st Avenue South, Nashville, TN, 37232, USA.
| | - David W Piston
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall 21st Avenue South, Nashville, TN, 37232, USA.
| | - Barbara M Fingleton
- Department of Cancer Biology, Vanderbilt University, 2220 Pierce Ave S, Nashville, TN, 37232, USA.
| | - David Lee Gorden
- Department of Cancer Biology, Vanderbilt University, 2220 Pierce Ave S, Nashville, TN, 37232, USA. .,Department of Surgery, Vanderbilt University, 801 Oxford House, 1313 21st Ave. S, Nashville, TN, 37212, USA.
| |
Collapse
|
49
|
Kramer JA, Grindley J, Crowell AM, Makaron L, Kohli R, Kirby M, Mansfield KG, Wachtman LM. The common marmoset as a model for the study of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Vet Pathol 2014; 52:404-13. [PMID: 24913270 DOI: 10.1177/0300985814537839] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is considered the hepatic manifestation of metabolic syndrome. The more clinically concerning form of the disease, nonalcoholic steatohepatitis (NASH), is characterized by steatosis, lobular inflammation, and ballooning degeneration. Here we describe a naturally occurring syndrome in the common marmoset that recapitulates the pathologic findings associated with NAFLD/NASH in humans. Hepatomegaly determined to result from NAFLD was observed in 33 of 183 marmosets. A comprehensive histopathologic assessment performed in 31 marmosets demonstrated that NAFLD was characterized by variably sized, Oil Red O staining cytoplasmic vacuoles and observed primarily in animals with evidence of obesity and insulin resistance. A subset of marmosets (16 of 31) also demonstrated evidence of NASH characterized by multifocal inflammation combined with ballooning hepatocellular degeneration. Marmosets with NASH demonstrated an increase in immunostaining with an antibody targeted against the human leukocyte antigens (HLA)-DP, HLA-DQ, and HLA-DR compared with marmosets without NASH (38.89 cells/10× field vs 12.05 cells/10× field, P = .05). In addition, marmosets with NASH demonstrated increased Ki-67 immunopositive cellular proliferation compared with those without (5.95 cells/10× field vs 1.53 cells/10× field, P = .0002). Finally, animals with NASH demonstrated significantly increased mean circulating serum iron levels (160.47 μg/dl, P = .008) and an increase in numbers of Prussian blue-positive Kupffer cells (9.28 cells/40× field, P = .005) relative to marmosets without NASH (97.75 μg/dl and 1.87 cells/40×, respectively). This study further characterizes the histopathology of NAFLD/NASH and suggests that the marmoset may be a valuable animal model with which to investigate the host and environmental factors contributing to the progression of NAFLD/ NASH.
Collapse
Affiliation(s)
- J A Kramer
- Department of Veterinary Resources, New England Primate Research Center, Southborough, MA, USA
| | - J Grindley
- Cummings School of Veterinary Medicine, Tufts University, N. Grafton, MA, USA
| | - A M Crowell
- University of Notre Dame, South Bend, IN, USA
| | - L Makaron
- Department of Veterinary Resources, New England Primate Research Center, Southborough, MA, USA
| | - R Kohli
- Division of Gastroentrology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - M Kirby
- Division of Gastroentrology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - K G Mansfield
- Novartis Institute for Biomedical Research, Cambridge, MA, USA
| | - L M Wachtman
- Department of Veterinary Resources, New England Primate Research Center, Southborough, MA, USA
| |
Collapse
|
50
|
Liu XY, Xu JF. Liver resection for young patients with large hepatocellular carcinoma: a single center experience from China. World J Surg Oncol 2014; 12:175. [PMID: 24894964 PMCID: PMC4049471 DOI: 10.1186/1477-7819-12-175] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 05/19/2014] [Indexed: 12/15/2022] Open
Abstract
Background To investigate retrospectively the clinicopathological characteristics and outcomes of young patients with large hepatocellular carcinoma after hepatectomy. Methods From January 2003 to December 2012, a total of 153 patients with large hepatocellular carcinoma (HCC) who received liver resection were included in the study. The clinicopathological features were analyzed retrospectively. The perioperative data were compared between those aged <40 years (the young group) and those aged >40 years (the older group). Prognostic factors and long-term survival were evaluated. Results The young group had more hepatitis B virus-related HCC than the older group (87.2% vs 66.3%, P = 0.031). In the young group, 15 patients (21.5%) were overweight (body mass index 25 to 29.9 kg/m2) or obese (body mass index ≥30 kg/m2), and 38 patients (45.8%) were overweight or obese in the older group (P = 0.032). Other clinicopathological characteristics were similar between the two groups. The perioperative data showed that the older group had more pulmonary infection after hepatectomy. Vascular invasion and high Edmondson-Steiner grade were the independent prognostic factors for long-term survival. There was no statistical difference between the young group and the older group in overall survival and disease-free survival (P = 0.109 and P = 0.087, respectively). Conclusions Liver resection for young patients with large HCC was safe and efficacious and should be recommended.
Collapse
Affiliation(s)
| | - Jiang-feng Xu
- Department of surgery, Yiwu Affiliated Hospital of Zhejiang University School of Medicine, east building in huajiachi campus,kaixuan road 268, 310020 Hangzhou, Zhejiang, China.
| |
Collapse
|