1
|
Ahmadini AAH, Raghav YS, Mahnashi AM, Islam Rather KU, Ali I. Neural networks to model COVID-19 dynamics and allocate healthcare resources. Sci Rep 2025; 15:15326. [PMID: 40312406 PMCID: PMC12045988 DOI: 10.1038/s41598-025-00153-9] [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: 01/15/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025] Open
Abstract
This study presents a neural network-based framework for COVID-19 transmission prediction and healthcare resource optimization. The model achieves high prediction accuracy by integrating epidemiological, mobility, vaccination, and environmental data and enables dynamic resource allocation. The results demonstrate significant improvements in forecasting performance and healthcare preparedness compared to traditional models. This work enhances decision-making in pandemic management by leveraging machine learning for real-time operational efficiency.
Collapse
Affiliation(s)
- Abdullah Ali H Ahmadini
- Department of Mathematics, College of Science, Jazan University, P.O. Box. 114, Jazan, 45142, Kingdom of Saudi Arabia
| | - Yashpal Singh Raghav
- Department of Mathematics, College of Science, Jazan University, P.O. Box. 114, Jazan, 45142, Kingdom of Saudi Arabia
| | - Ali M Mahnashi
- Department of Mathematics, College of Science, Jazan University, P.O. Box. 114, Jazan, 45142, Kingdom of Saudi Arabia
| | | | - Irfan Ali
- Department of Statistics and Operations Research, Aligarh Muslim University, Aligarh, 202002, India.
| |
Collapse
|
2
|
Pavinato L, Baggiolini A. Oncogenic competence: balancing mutations, cellular state, and microenvironment. Trends Cancer 2025; 11:276-285. [PMID: 39875306 DOI: 10.1016/j.trecan.2025.01.002] [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: 10/01/2024] [Revised: 12/18/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025]
Abstract
Cancer development is driven by mutations, yet tumor-causing mutations only lead to tumor formation within specific cellular contexts. The reasons why certain mutations trigger malignant transformation in some contexts but not others remain often unclear. Both intrinsic and extrinsic factors play a key role in driving carcinogenesis by leading the cells toward a state of 'oncogenic competence'. This state is shaped by the transcriptional and epigenetic programs that define a specific cell in time and space. These programs arise from the interplay between genetic mutations, cellular lineage, differentiation state, and microenvironment. A deeper understanding of oncogenic competence is essential to uncover the mechanisms behind tumor initiation and, ultimately, advance the development of novel targeted therapies for cancer treatment and prevention.
Collapse
Affiliation(s)
- Lisa Pavinato
- Institute of Oncology Research (IOR), Bellinzona Institutes of Science (BIOS+), Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Arianna Baggiolini
- Institute of Oncology Research (IOR), Bellinzona Institutes of Science (BIOS+), Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland.
| |
Collapse
|
3
|
Chiu V, Yee C, Main N, Stevanovski I, Watt M, Wilson T, Angus P, Roberts T, Shackel N, Herath C. Oncogenic plasmid DNA and liver injury agent dictates liver cancer development in a mouse model. Clin Sci (Lond) 2024; 138:1227-1248. [PMID: 39254423 PMCID: PMC11427747 DOI: 10.1042/cs20240560] [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: 04/07/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/11/2024]
Abstract
Primary liver cancer is an increasing problem worldwide and is associated with significant mortality. A popular method of modeling liver cancer in mice is plasmid hydrodynamic tail vein injection (HTVI). However, plasmid-HTVI models rarely recapitulate the chronic liver injury which precedes the development of most human liver cancer. We sought to investigate how liver injury using thioacetamide contributes to the pathogenesis and progression of liver cancer in two oncogenic plasmid-HTVI-induced mouse liver cancer models. Fourteen-week-old male mice received double-oncogene plasmid-HTVI (SB/AKT/c-Met and SB/AKT/NRas) and then twice-weekly intraperitoneal injections of thioacetamide for 6 weeks. Liver tissue was examined for histopathological changes, including fibrosis and steatosis. Further characterization of fibrosis and inflammation was performed with immunostaining and real-time quantitative PCR. RNA sequencing with pathway analysis was used to explore novel pathways altered in the cancer models. Hepatocellular and cholangiocellular tumors were observed in mice injected with double-oncogene plasmid-HTVI models (SB/AKT/c-Met and SB/AKT/NRas). Thioacetamide induced mild fibrosis and increased alpha smooth muscle actin-expressing cells. However, the combination of plasmids and thioacetamide did not significantly increase tumor size, but increased multiplicity of small neoplastic lesions. Cancer and/or liver injury up-regulated profibrotic and proinflammatory genes while metabolic pathway genes were mostly down-regulated. We conclude that the liver injury microenvironment can interact with liver cancer and alter its presentation. However, the effects on cancer development vary depending on the genetic drivers with differing active oncogenic pathways. Therefore, the choice of plasmid-HTVI model and injury agent may influence the extent to which injury promotes liver cancer development.
Collapse
Affiliation(s)
- Vincent Chiu
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Christine Yee
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Nathan Main
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Igor Stevanovski
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Matthew Watt
- School of Biomedical Sciences, University of Melbourne, Victoria, Australia
| | - Trevor Wilson
- Hudson Institute of Medical Research, Monash University, Victoria, Australia
| | - Peter Angus
- Department of Gastroenterology and Hepatology, Austin Health, Heidelberg, Victoria, Australia
| | - Tara Roberts
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Nicholas Shackel
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Chandana Herath
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
- Department of Medicine, Austin Health, University of Melbourne, Victoria, Australia
| |
Collapse
|
4
|
Singh AK, Chaube B, Citrin KM, Fowler JW, Lee S, Catarino J, Knight J, Lowery S, Shree S, Boutagy N, Ruz-Maldonado I, Harry K, Shanabrough M, Ross TT, Malaker S, Suárez Y, Fernández-Hernando C, Grabinska K, Sessa WC. Loss of cis-PTase function in the liver promotes a highly penetrant form of fatty liver disease that rapidly transitions to hepatocellular carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.13.566870. [PMID: 38014178 PMCID: PMC10680637 DOI: 10.1101/2023.11.13.566870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Obesity-linked fatty liver is a significant risk factor for hepatocellular carcinoma (HCC)1,2; however, the molecular mechanisms underlying the transition from non-alcoholic fatty liver disease (NAFLD) to HCC remains unclear. The present study explores the role of the endoplasmic reticulum (ER)-associated protein NgBR, an essential component of the cis-prenyltransferases (cis-PTase) enzyme3, in chronic liver disease. Here we show that genetic depletion of NgBR in hepatocytes of mice (N-LKO) intensifies triacylglycerol (TAG) accumulation, inflammatory responses, ER/oxidative stress, and liver fibrosis, ultimately resulting in HCC development with 100% penetrance after four months on a high-fat diet. Comprehensive genomic and single cell transcriptomic atlas from affected livers provides a detailed molecular analysis of the transition from liver pathophysiology to HCC development. Importantly, pharmacological inhibition of diacylglycerol acyltransferase-2 (DGAT2), a key enzyme in hepatic TAG synthesis, abrogates diet-induced liver damage and HCC burden in N-LKO mice. Overall, our findings establish NgBR/cis-PTase as a critical suppressor of NAFLD-HCC conversion and suggests that DGAT2 inhibition may serve as a promising therapeutic approach to delay HCC formation in patients with advanced non-alcoholic steatohepatitis (NASH).
Collapse
Affiliation(s)
- Abhishek K. Singh
- Department of Pharmacology, and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Balkrishna Chaube
- Department of Comparative Medicine, Yale Center for Molecular and Systems Metabolism and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Kathryn M Citrin
- Department of Comparative Medicine, Yale Center for Molecular and Systems Metabolism and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Joseph Wayne Fowler
- Department of Pharmacology, and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Sungwoon Lee
- Department of Pharmacology, and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Jonatas Catarino
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - James Knight
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Sarah Lowery
- Chemistry Research Building, Yale University, New Haven, CT, USA
| | - Sonal Shree
- Molecular Biophysics & Biochemistry, Yale University, New Haven, CT, USA
| | - Nabil Boutagy
- Department of Pharmacology, and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Inmaculada Ruz-Maldonado
- Department of Comparative Medicine, Yale Center for Molecular and Systems Metabolism and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Kathy Harry
- Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Marya Shanabrough
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | - Stacy Malaker
- Chemistry Research Building, Yale University, New Haven, CT, USA
| | - Yajaira Suárez
- Department of Comparative Medicine, Yale Center for Molecular and Systems Metabolism and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Carlos Fernández-Hernando
- Department of Comparative Medicine, Yale Center for Molecular and Systems Metabolism and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Kariona Grabinska
- Department of Pharmacology, and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - William C. Sessa
- Department of Pharmacology, and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| |
Collapse
|
5
|
Feng J, Zeng Y, Li S. Single-cell characterization of tumor evolution from precursor diseases. Trends Cancer 2023; 9:775-776. [PMID: 37544795 DOI: 10.1016/j.trecan.2023.07.012] [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: 07/05/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 08/08/2023]
Abstract
Elucidating the characteristics of precursor diseases is important to the understanding of malignant transformation during tumor progression. In a recent study, Dang et al. characterized the cellular and molecular evolution from precursor conditions to multiple myeloma (MM) by integrating single-cell RNA sequencing (scRNA-seq) and B cell-receptor sequencing (scBCR-seq).
Collapse
Affiliation(s)
- Jiling Feng
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Yu Zeng
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Shengli Li
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China.
| |
Collapse
|
6
|
Ha S, Gujrati H, Wang BD. Aberrant PI3Kδ splice isoform as a potential biomarker and novel therapeutic target for endocrine cancers. Front Endocrinol (Lausanne) 2023; 14:1190479. [PMID: 37670888 PMCID: PMC10475954 DOI: 10.3389/fendo.2023.1190479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/27/2023] [Indexed: 09/07/2023] Open
Abstract
Introduction PI3K/AKT signaling pathway is upregulated in a broad spectrum of cancers. Among the class I PI3Ks (PI3Kδ/β/δ isoforms), PI3Kδ has been implicated in hematologic cancers and solid tumors. Alternative splicing is a post-transcriptional process for acquiring proteomic diversity in eukaryotic cells. Emerging evidence has highlighted the involvement of aberrant mRNA splicing in cancer development/progression. Methods Our previous studies revealed that PIK3CD-S is an oncogenic splice variant that promotes tumor aggressiveness and drug resistance in prostate cancer (PCa). To further evaluate the potential of utilizing PI3Kδ-S (encoded from PIK3CD-S) as a cancer biomarker and/or drug target, comprehensive analyses were performed in a series of patient samples and cell lines derived from endocrine/solid tumors. Specifically, IHC, immunofluorescence, western blot and RT-PCR assay results have demonstrated that PI3Kδ isoforms were highly expressed in endocrine/solid tumor patient specimens and cell lines. Results Differential PIK3CD-S/PIK3CD-L expression profiles were identified in a panel of endocrine/solid tumor cells. SiRNA knockdown of PIK3CD-L or PIK3CD-S differentially inhibits AKT/mTOR signaling in PCa, breast, colon and lung cancer cell lines. Moreover, siRNA knockdown of PTEN increased PI3Kδ levels and activated AKT/mTOR signaling, while overexpression of PTEN reduced PI3Kδ levels and inhibited AKT/mTOR signaling in cancer cells. Intriguingly, PI3Kδ-S levels remained unchanged upon either siRNA knockdown or overexpression of PTEN. Taken together, these results suggested that PTEN negatively regulates PI3Kδ-L and its downstream AKT/mTOR signaling, while PI3Kδ-S promotes AKT/mTOR signaling without regulation by PTEN. Lastly, PI3Kδ inhibitor Idelalisib and SRPK1/2 inhibitor SRPIN340 were employed to assess their efficacies on inhibiting the PI3Kδ-expressing endocrine/solid tumors. Our results have shown that Idelalisib effectively inhibited PI3Kδ-L (but not PI3Kδ-S) mediated AKT/mTOR signaling. In contrast, SRPIN340 reversed the aberrant mRNA splicing, thereby inhibiting AKT/mTOR signaling. In-vitro functional assays have further demonstrated that a combination of Idelalisib and SRPIN340 achieved a synergistic drug effect (with drastically reduced cell viabilities/growths of tumor spheroids) in inhibiting the advanced tumor cells. Conclusion In summary, our study has suggested a promising potential of utilizing PI3Kδ-S (an oncogenic isoform conferring drug resistance and exempt from PTEN regulation) as a prognostic biomarker and drug target in advanced endocrine cancers.
Collapse
Affiliation(s)
- Siyoung Ha
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD, United States
| | - Himali Gujrati
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD, United States
| | - Bi-Dar Wang
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD, United States
- Hormone Related Cancers Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| |
Collapse
|
7
|
CAI TIANYING, BAI JUNJIE, TAN PENG, HUANG ZHIWEI, LIU CHEN, WU ZIMING, CHENG YONGLANG, LI TONGXI, CHEN YIFAN, RUAN JIAN, GAO LIN, DU YICHAO, FU WENGUANG. Zyxin promotes hepatocellular carcinoma progression via the activation of AKT/mTOR signaling pathway. Oncol Res 2023; 31:805-817. [PMID: 37547758 PMCID: PMC10398406 DOI: 10.32604/or.2023.029549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 05/17/2023] [Indexed: 08/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy that is driven by multiple genes and pathways. The aim of this study was to investigate the role and specific mechanism of the actin-interacting protein zyxin (ZYX) in HCC. We found that the expression of ZYX was significantly higher in HCC tissues compared to that in normal liver tissues. In addition, overexpression of ZYX in hepatoma cell lines (PLC/PRF/5, HCCLM3) enhanced their proliferation, migration and invasion, whereas ZYX knockdown had the opposite effects (SK HEP-1, Huh-7). Furthermore, the change in the expression levels of ZYX also altered that of proteins related to cell cycle, migration and invasion. Similar results were obtained with xenograft models. The AKT/mTOR signaling pathway is one of the key mediators of cancer development. While ZYX overexpression upregulated the levels of phosphorylated AKT/mTOR proteins, its knockdown had the opposite effect. In addition, the AKT inhibitor MK2206 neutralized the pro-oncogenic effects of ZYX on the HCC cells, whereas the AKT activator SC79 restored the proliferation, migration and invasion of HCC cells with ZYX knockdown. Taken together, ZYX promotes the malignant progression of HCC by activating AKT/mTOR signaling pathway, and is a potential therapeutic target in HCC.
Collapse
Affiliation(s)
- TIANYING CAI
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Biobank, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - JUNJIE BAI
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - PENG TAN
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - ZHIWEI HUANG
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - CHEN LIU
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - ZIMING WU
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - YONGLANG CHENG
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - TONGXI LI
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - YIFAN CHEN
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - JIAN RUAN
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - LIN GAO
- Department of Health Management, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - YICHAO DU
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - WENGUANG FU
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| |
Collapse
|
8
|
Schrenk D, Bignami M, Bodin L, Chipman JK, del Mazo J, Hogstrand C, (Ron) Hoogenboom L, Leblanc J, Nebbia CS, Nielsen E, Ntzani E, Petersen A, Sand S, Schwerdtle T, Vleminckx C, Wallace H, Romualdo B, Cristina F, Stephen H, Marco I, Mosbach‐Schulz O, Riolo F, Christodoulidou A, Grasl‐Kraupp B. Risk assessment of N-nitrosamines in food. EFSA J 2023; 21:e07884. [PMID: 36999063 PMCID: PMC10043641 DOI: 10.2903/j.efsa.2023.7884] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
EFSA was asked for a scientific opinion on the risks to public health related to the presence of N-nitrosamines (N-NAs) in food. The risk assessment was confined to those 10 carcinogenic N-NAs occurring in food (TCNAs), i.e. NDMA, NMEA, NDEA, NDPA, NDBA, NMA, NSAR, NMOR, NPIP and NPYR. N-NAs are genotoxic and induce liver tumours in rodents. The in vivo data available to derive potency factors are limited, and therefore, equal potency of TCNAs was assumed. The lower confidence limit of the benchmark dose at 10% (BMDL10) was 10 μg/kg body weight (bw) per day, derived from the incidence of rat liver tumours (benign and malignant) induced by NDEA and used in a margin of exposure (MOE) approach. Analytical results on the occurrence of N-NAs were extracted from the EFSA occurrence database (n = 2,817) and the literature (n = 4,003). Occurrence data were available for five food categories across TCNAs. Dietary exposure was assessed for two scenarios, excluding (scenario 1) and including (scenario 2) cooked unprocessed meat and fish. TCNAs exposure ranged from 0 to 208.9 ng/kg bw per day across surveys, age groups and scenarios. 'Meat and meat products' is the main food category contributing to TCNA exposure. MOEs ranged from 3,337 to 48 at the P95 exposure excluding some infant surveys with P95 exposure equal to zero. Two major uncertainties were (i) the high number of left censored data and (ii) the lack of data on important food categories. The CONTAM Panel concluded that the MOE for TCNAs at the P95 exposure is highly likely (98-100% certain) to be less than 10,000 for all age groups, which raises a health concern.
Collapse
|
9
|
Ghelfi J, Decaens T, Macek Jilkova Z. Rat Model of Hepatocellular Carcinoma for Better Understanding Immune Effects of Transarterial Chemoembolization. Radiology 2023; 306:E1. [PMID: 36066362 DOI: 10.1148/radiol.220179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Julien Ghelfi
- Université Grenoble Alpes, Saint-Martin-d'Hères, France.,Departments of Radiology, CHU Grenoble Alpes, Boulevard de la Chantourne, 38700 La Tronche, France.,Institute for Advanced Biosciences, INSERM U1209/CNRS UMR 5309, University of Grenoble-Alpes, La Tronche, France
| | - Thomas Decaens
- Université Grenoble Alpes, Saint-Martin-d'Hères, France.,Hepatology and Gastrointestinal Medical Oncology, CHU Grenoble Alpes, Boulevard de la Chantourne, 38700 La Tronche, France.,Institute for Advanced Biosciences, INSERM U1209/CNRS UMR 5309, University of Grenoble-Alpes, La Tronche, France
| | - Zuzana Macek Jilkova
- Hepatology and Gastrointestinal Medical Oncology, CHU Grenoble Alpes, Boulevard de la Chantourne, 38700 La Tronche, France.,Institute for Advanced Biosciences, INSERM U1209/CNRS UMR 5309, University of Grenoble-Alpes, La Tronche, France
| |
Collapse
|
10
|
McLaughlin KL, Nelson MAM, Coalson HS, Hagen JT, Montgomery MM, Wooten AR, Zeczycki TN, Vohra NA, Fisher-Wellman KH. Bioenergetic Phenotyping of DEN-Induced Hepatocellular Carcinoma Reveals a Link Between Adenylate Kinase Isoform Expression and Reduced Complex I-Supported Respiration. Front Oncol 2022; 12:919880. [PMID: 35756609 PMCID: PMC9213884 DOI: 10.3389/fonc.2022.919880] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/16/2022] [Indexed: 11/21/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common form of liver cancer worldwide. Increasing evidence suggests that mitochondria play a central role in malignant metabolic reprogramming in HCC, which may promote disease progression. To comprehensively evaluate the mitochondrial phenotype present in HCC, we applied a recently developed diagnostic workflow that combines high-resolution respirometry, fluorometry, and mitochondrial-targeted nLC-MS/MS proteomics to cell culture (AML12 and Hepa 1-6 cells) and diethylnitrosamine (DEN)-induced mouse models of HCC. Across both model systems, CI-linked respiration was significantly decreased in HCC compared to nontumor, though this did not alter ATP production rates. Interestingly, CI-linked respiration was found to be restored in DEN-induced tumor mitochondria through acute in vitro treatment with P1, P5-di(adenosine-5′) pentaphosphate (Ap5A), a broad inhibitor of adenylate kinases. Mass spectrometry-based proteomics revealed that DEN-induced tumor mitochondria had increased expression of adenylate kinase isoform 4 (AK4), which may account for this response to Ap5A. Tumor mitochondria also displayed a reduced ability to retain calcium and generate membrane potential across a physiological span of ATP demand states compared to DEN-treated nontumor or saline-treated liver mitochondria. We validated these findings in flash-frozen human primary HCC samples, which similarly displayed a decrease in mitochondrial respiratory capacity that disproportionately affected CI. Our findings support the utility of mitochondrial phenotyping in identifying novel regulatory mechanisms governing cancer bioenergetics.
Collapse
Affiliation(s)
- Kelsey L McLaughlin
- Brody School of Medicine, Department of Physiology, East Carolina University, Greenville, NC, United States.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
| | - Margaret A M Nelson
- Brody School of Medicine, Department of Physiology, East Carolina University, Greenville, NC, United States.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
| | - Hannah S Coalson
- Brody School of Medicine, Department of Physiology, East Carolina University, Greenville, NC, United States.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
| | - James T Hagen
- Brody School of Medicine, Department of Physiology, East Carolina University, Greenville, NC, United States.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
| | - McLane M Montgomery
- Brody School of Medicine, Department of Physiology, East Carolina University, Greenville, NC, United States.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
| | - Ashley R Wooten
- Brody School of Medicine, Department of Biochemistry and Molecular Biology, East Carolina University, Greenville, NC, United States
| | - Tonya N Zeczycki
- Brody School of Medicine, Department of Biochemistry and Molecular Biology, East Carolina University, Greenville, NC, United States
| | - Nasreen A Vohra
- Brody School of Medicine, Department of Surgery, East Carolina University, Greenville, NC, United States
| | - Kelsey H Fisher-Wellman
- Brody School of Medicine, Department of Physiology, East Carolina University, Greenville, NC, United States.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States.,UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| |
Collapse
|
11
|
Tischfield DJ, Gurevich A, Johnson O, Gatmaytan I, Nadolski GJ, Soulen MC, Kaplan DE, Furth E, Hunt SJ, Gade TPF. Transarterial Embolization Modulates the Immune Response within Target and Nontarget Hepatocellular Carcinomas in a Rat Model. Radiology 2022; 303:215-225. [PMID: 35014906 PMCID: PMC8962821 DOI: 10.1148/radiol.211028] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 10/12/2021] [Accepted: 10/28/2021] [Indexed: 12/24/2022]
Abstract
Background Transarterial embolization (TAE) is the most common treatment for hepatocellular carcinoma (HCC); however, there remain limited data describing the influence of TAE on the tumor immune microenvironment. Purpose To characterize TAE-induced modulation of the tumor immune microenvironment in a rat model of HCC and identify factors that modulate this response. Materials and Methods TAE was performed on autochthonous HCCs induced in rats with use of diethylnitrosamine. CD3, CD4, CD8, and FOXP3 lymphocytes, as well as programmed cell death protein ligand-1 (PD-L1) expression, were examined in three cohorts: tumors from rats that did not undergo embolization (control), embolized tumors (target), and nonembolized tumors from rats that had a different target tumor embolized (nontarget). Differences in immune cell recruitment associated with embolic agent type (tris-acryl gelatin microspheres [TAGM] vs hydrogel embolics) and vascular location were examined in rat and human tissues. A generalized estimating equation model and t, Mann-Whitney U, and χ2 tests were used to compare groups. Results Cirrhosis-induced alterations in CD8, CD4, and CD25/CD4 lymphocytes were partially normalized following TAE (CD8: 38.4%, CD4: 57.6%, and CD25/CD4: 21.1% in embolized liver vs 47.7% [P = .02], 47.0% [P = .01], and 34.9% [P = .03], respectively, in cirrhotic liver [36.1%, 59.6%, and 4.6% in normal liver]). Embolized tumors had a greater number of CD3, CD4, and CD8 tumor-infiltrating lymphocytes relative to controls (191.4 cells/mm2 vs 106.7 cells/mm2 [P = .03]; 127.8 cells/mm2 vs 53.8 cells/mm2 [P < .001]; and 131.4 cells/mm2 vs 78.3 cells/mm2 [P = .01]) as well as a higher PD-L1 expression score (4.1 au vs 1.9 au [P < .001]). A greater number of CD3, CD4, and CD8 lymphocytes were found near TAGM versus hydrogel embolics (4.1 vs 2.0 [P = .003]; 3.7 vs 2.0 [P = .01]; and 2.2 vs 1.1 [P = .03], respectively). The number of lymphocytes adjacent to embolics differed based on vascular location (17.9 extravascular CD68+ peri-TAGM cells vs 7.0 intravascular [P < .001]; 6.4 extravascular CD68+ peri-hydrogel embolic cells vs 3.4 intravascular [P < .001]). Conclusion Transarterial embolization-induced dynamic alterations of the tumor immune microenvironment are influenced by underlying liver disease, embolic agent type, and vascular location. © RSNA, 2022 Online supplemental material is available for this article. See also the editorials by Kennedy et al and by White in this issue.
Collapse
Affiliation(s)
| | | | - Omar Johnson
- From the Penn Image-Guided Interventions Laboratory (D.J.T., A.G.,
O.J., I.G., G.J.N., S.J.H., T.P.F.G.), Department of Radiology (D.J.T., O.J.,
G.J.N., M.C.S., S.J.H., T.P.F.G.), and Department of Pathology (E.F.), Hospital
of the University of Pennsylvania, 3400 Spruce St, Philadelphia, PA 19104;
Division of Gastroenterology and Hepatology (D.E.K.) and Department of Cancer
Biology (T.P.F.G.), Perelman School of Medicine at the University of
Pennsylvania, Philadelphia, Pa; and Gastroenterology Section, Corporal Michael
J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pa (D.E.K.)
| | - Isabela Gatmaytan
- From the Penn Image-Guided Interventions Laboratory (D.J.T., A.G.,
O.J., I.G., G.J.N., S.J.H., T.P.F.G.), Department of Radiology (D.J.T., O.J.,
G.J.N., M.C.S., S.J.H., T.P.F.G.), and Department of Pathology (E.F.), Hospital
of the University of Pennsylvania, 3400 Spruce St, Philadelphia, PA 19104;
Division of Gastroenterology and Hepatology (D.E.K.) and Department of Cancer
Biology (T.P.F.G.), Perelman School of Medicine at the University of
Pennsylvania, Philadelphia, Pa; and Gastroenterology Section, Corporal Michael
J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pa (D.E.K.)
| | - Gregory J. Nadolski
- From the Penn Image-Guided Interventions Laboratory (D.J.T., A.G.,
O.J., I.G., G.J.N., S.J.H., T.P.F.G.), Department of Radiology (D.J.T., O.J.,
G.J.N., M.C.S., S.J.H., T.P.F.G.), and Department of Pathology (E.F.), Hospital
of the University of Pennsylvania, 3400 Spruce St, Philadelphia, PA 19104;
Division of Gastroenterology and Hepatology (D.E.K.) and Department of Cancer
Biology (T.P.F.G.), Perelman School of Medicine at the University of
Pennsylvania, Philadelphia, Pa; and Gastroenterology Section, Corporal Michael
J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pa (D.E.K.)
| | - Michael C. Soulen
- From the Penn Image-Guided Interventions Laboratory (D.J.T., A.G.,
O.J., I.G., G.J.N., S.J.H., T.P.F.G.), Department of Radiology (D.J.T., O.J.,
G.J.N., M.C.S., S.J.H., T.P.F.G.), and Department of Pathology (E.F.), Hospital
of the University of Pennsylvania, 3400 Spruce St, Philadelphia, PA 19104;
Division of Gastroenterology and Hepatology (D.E.K.) and Department of Cancer
Biology (T.P.F.G.), Perelman School of Medicine at the University of
Pennsylvania, Philadelphia, Pa; and Gastroenterology Section, Corporal Michael
J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pa (D.E.K.)
| | - David E. Kaplan
- From the Penn Image-Guided Interventions Laboratory (D.J.T., A.G.,
O.J., I.G., G.J.N., S.J.H., T.P.F.G.), Department of Radiology (D.J.T., O.J.,
G.J.N., M.C.S., S.J.H., T.P.F.G.), and Department of Pathology (E.F.), Hospital
of the University of Pennsylvania, 3400 Spruce St, Philadelphia, PA 19104;
Division of Gastroenterology and Hepatology (D.E.K.) and Department of Cancer
Biology (T.P.F.G.), Perelman School of Medicine at the University of
Pennsylvania, Philadelphia, Pa; and Gastroenterology Section, Corporal Michael
J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pa (D.E.K.)
| | - Emma Furth
- From the Penn Image-Guided Interventions Laboratory (D.J.T., A.G.,
O.J., I.G., G.J.N., S.J.H., T.P.F.G.), Department of Radiology (D.J.T., O.J.,
G.J.N., M.C.S., S.J.H., T.P.F.G.), and Department of Pathology (E.F.), Hospital
of the University of Pennsylvania, 3400 Spruce St, Philadelphia, PA 19104;
Division of Gastroenterology and Hepatology (D.E.K.) and Department of Cancer
Biology (T.P.F.G.), Perelman School of Medicine at the University of
Pennsylvania, Philadelphia, Pa; and Gastroenterology Section, Corporal Michael
J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pa (D.E.K.)
| | - Stephen J. Hunt
- From the Penn Image-Guided Interventions Laboratory (D.J.T., A.G.,
O.J., I.G., G.J.N., S.J.H., T.P.F.G.), Department of Radiology (D.J.T., O.J.,
G.J.N., M.C.S., S.J.H., T.P.F.G.), and Department of Pathology (E.F.), Hospital
of the University of Pennsylvania, 3400 Spruce St, Philadelphia, PA 19104;
Division of Gastroenterology and Hepatology (D.E.K.) and Department of Cancer
Biology (T.P.F.G.), Perelman School of Medicine at the University of
Pennsylvania, Philadelphia, Pa; and Gastroenterology Section, Corporal Michael
J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pa (D.E.K.)
| | - Terence P. F. Gade
- From the Penn Image-Guided Interventions Laboratory (D.J.T., A.G.,
O.J., I.G., G.J.N., S.J.H., T.P.F.G.), Department of Radiology (D.J.T., O.J.,
G.J.N., M.C.S., S.J.H., T.P.F.G.), and Department of Pathology (E.F.), Hospital
of the University of Pennsylvania, 3400 Spruce St, Philadelphia, PA 19104;
Division of Gastroenterology and Hepatology (D.E.K.) and Department of Cancer
Biology (T.P.F.G.), Perelman School of Medicine at the University of
Pennsylvania, Philadelphia, Pa; and Gastroenterology Section, Corporal Michael
J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pa (D.E.K.)
| |
Collapse
|
12
|
Kerdput V, Kanjanapongkul K, Itharat A, Pramong R, Lamers WH, Hakvoort TBM, Jongejan A, Pradidarcheep W. Molecular Changes Following Induction of Hepatocellular Carcinoma by Diethylnitrosamine and Thioacetamide, and Subsequent Treatment with Dioscorea membranacea Extract. Int J Med Sci 2022; 19:1806-1815. [PMID: 36313224 PMCID: PMC9608040 DOI: 10.7150/ijms.72987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 09/07/2022] [Indexed: 12/02/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a primary liver cancer commonly found in adults. Previously, we showed the anticancer effects of Thai herbal plant extract, Dioscorea membranacea Pierre (DM), in HCC-bearing rats. In the present study, we further examined the proposed mechanism of DM, including apoptosis and antioxidant activity. Moreover, we used RNA sequencing (RNA-seq) to analyze molecular pathways in the rat model in which HCC was induced by diethylnitrosamine (DEN) and thioacetamide (TAA). The HCC-bearing rats were then treated with 40 mg/kg of DM for 8 weeks, after which experimental and control rats were sacrificed and liver tissues were collected. The RNA-seq data of DEN/TAA-treated rats exhibited upregulation of 16 hallmark pathways, including epithelial mesenchymal transition, inflammatory responses, and angiogenesis (p<0.01). DM extract expanded the Bax protein-positive pericentral zone in the tumor areas and decreased hepatic malondialdehyde levels, implying a decrease in lipid peroxidation in liver. However, DM treatment did not ameliorate the molecular pathways induced in DEN/TAA-treated livers. Our findings indicate that DM extract has antioxidant activity and exerts its pro-apoptotic effect on rat HCCs in vivo at the (post-)translational level.
Collapse
Affiliation(s)
- Vichununt Kerdput
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand
| | - Kritsakorn Kanjanapongkul
- Department of Science, Mahidol University International College, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Arunporn Itharat
- Department of Applied Thai Traditional Medicine, Faculty of Medicine, Thammasat University, Pathumthani 10120, Thailand
| | - Ratchadaporn Pramong
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand
| | - Wouter H Lamers
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Location AMC, 1105 BK Amsterdam, The Netherlands
| | - Theodorus B M Hakvoort
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Location AMC, 1105 BK Amsterdam, The Netherlands
| | - Aldo Jongejan
- Bioinformatics Laboratory, Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Wisuit Pradidarcheep
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand
| |
Collapse
|
13
|
DEN-Induced Rat Model Reproduces Key Features of Human Hepatocellular Carcinoma. Cancers (Basel) 2021; 13:cancers13194981. [PMID: 34638465 PMCID: PMC8508319 DOI: 10.3390/cancers13194981] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 01/15/2023] Open
Abstract
Simple Summary Hepatocellular carcinoma is the most frequent form of primary liver cancer, characterized by increasing incidence and high mortality. Animal models of hepatocellular carcinoma are widely used to study the biology of cancer and to test potential therapies. Herein, we describe how the rat model of DEN-induced hepatocellular carcinoma mimics the pathogenesis of hepatocellular carcinoma seen in humans, including liver damage, chronic inflammation, hepatocytes proliferation, liver fibrosis and cirrhosis, disorganized vasculature, and modulations of the liver’s immune microenvironment. Our results should help the hepatocellular carcinoma field to better tailor the use of the DEN-induced rat liver cancer model for testing specific experimental hypotheses or to perform preclinical testing. Abstract Hepatocellular carcinoma (HCC) is the most common type of liver cancer. The majority of HCC cases are associated with liver fibrosis or cirrhosis developing from chronic liver injuries. The immune system of the liver contributes to the severity of tissue damage, the establishment of fibrosis and the disease’s progression towards HCC. Herein, we provide a detailed characterization of the DEN-induced HCC rat model during fibrosis progression and HCC development with a special focus on the liver’s inflammatory microenvironment. Fischer 344 male rats were treated weekly for 14 weeks with intra-peritoneal injections of 50 mg/kg DEN. The rats were sacrificed before starting DEN-injections at 0 weeks, after 8 weeks, 14 weeks and 20 weeks after the start of DEN-injections. We performed histopathological, immunohistochemical, RT-qPCR, RNA-seq and flow cytometry analysis. Data were compared between tumor and non-tumor samples from the DEN-treated versus untreated rats, as well as versus human HCCs. Chronic DEN injections lead to liver damage, hepatocytes proliferation, liver fibrosis and cirrhosis, disorganized vasculature, and a modulated immune microenvironment that mimics the usual events observed during human HCC development. The RNA-seq results showed that DEN-induced liver tumors in the rat model shared remarkable molecular characteristics with human HCC, especially with HCC associated with high proliferation. In conclusion, our study provides detailed insight into hepatocarcinogenesis in a commonly used model of HCC, facilitating the future use of this model for preclinical testing.
Collapse
|
14
|
Optimized protocol for an inducible rat model of liver tumor with chronic hepatocellular injury, inflammation, fibrosis, and cirrhosis. STAR Protoc 2021; 2:100353. [PMID: 33665633 PMCID: PMC7905469 DOI: 10.1016/j.xpro.2021.100353] [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] [Indexed: 11/21/2022] Open
Abstract
Animal models of liver cancer are instrumental in the study of hepatocarcinogenesis and development of novel therapeutic approaches. Here, we describe steps to establish liver cancer in a rat model, via chronic administration of diethylnitrosamine. This causes liver tumors with a sequential progression of hepatitis, cirrhosis, and tumor formation, which closely mimics the development of human liver cancer. This protocol was optimized to significantly increase the incidence of liver tumor formation and reduce the duration of the procedure. For complete details on the use and execution of this protocol, please refer to Chen et al. (2020). Detailed protocol for a carcinogen-induced rat hepatocellular carcinoma model Undergoes a sequential progression of hepatitis, cirrhosis, and tumor formation Optimized to significantly increase the incidence of liver tumor formation
Collapse
|