1
|
Xu X, Cheng Y, Yang Z, Yin Y, Qian Y, Yang H, Zhu S, Tian H, Zhuang Y, Zhu S, Yang P, Qin S, Shen W. Wogonin potentiates the irradiation effect on hepatocellular carcinoma by activating the Hippo-Yes-associated protein/transcriptional co-activator with PDZ-binding motif pathway. Int Immunopharmacol 2025; 157:114740. [PMID: 40318272 DOI: 10.1016/j.intimp.2025.114740] [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: 11/26/2024] [Revised: 04/11/2025] [Accepted: 04/23/2025] [Indexed: 05/07/2025]
Abstract
OBJECTIVE To investigate whether wogonin increases the radiosensitivity of hepatocellular carcinoma (HCC) cells by activating Hippo-Yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif (TAZ) signaling. METHODS HCC cells were treated with irradiation and wogonin; their proliferation and apoptosis were evaluated. Xenograft models were established to assess the radio-synergistic effects of wogonin; we evaluated whether wogonin influences the efficacy of radiotherapy in HCC cells by activating Hippo-YAP/TAZ signaling. RESULTS Fifty micromolar wogonin enhanced the radiosensitivity of HCC cells; 4-Gy X-rays promoted apoptosis in HCC cells. Wogonin pretreatment significantly increased radiosensitivity. In xenograft models, tumor weight and volume in the 100 mg/kg wogonin plus irradiation group were significantly reduced; pYAP and pTAZ levels were downregulated in HCC cells treated with radiotherapy. Following treatment with 4-Gy X-rays and 100 μM wogonin, the relative pYAP/total YAP and pTAZ/total TAZ ratios increased. We identified the possible target genes of YAP/TAZ: AXL, CCN1, and CCN2. WB results revealed the upregulation of AXL, CCN1, and CCN2 in the irradiation group. However, in the group receiving irradiation and wogonin, the protein expression levels of AXL, CCN1, and CCN2 were downregulated. XMU-MP-1 inhibited pYAP and pTAZ expression in the combination treatment group, thereby promoting AXL, CCN1, and CCN2 expression. The proliferative ability of HCC cells in the wogonin plus irradiation group was partially recovered following treatment with XMU-MP-1. Apoptosis in HCC cells was reversed after pretreatment with 2 μM XMU-MP-1 in the wogonin plus irradiation group. CONCLUSION Wogonin may modulate Hippo-YAP/TAZ signaling and enhance the radiosensitivity of HCCs.
Collapse
Affiliation(s)
- Xiao Xu
- Department of Radiotherapy, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu, China; Department of Radiotherapy, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Yan Cheng
- School of Computer Science and Engineering, Taizhou Institute of Science & Technology, Taizhou 225300, Jiangsu, China
| | - Zeyu Yang
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou 215006, Jiangsu, China
| | - Yong Yin
- Department of Science and Technology, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu, China
| | - Yonghong Qian
- Department of Radiotherapy, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu, China
| | - Haiyu Yang
- Department of Clinical Laboratory, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu, China
| | - Shusheng Zhu
- Department of Thoracic Surgery, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu, China
| | - Hu Tian
- Department of Science and Technology, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu, China
| | - Yanshuang Zhuang
- Department of Science and Technology, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu, China
| | - Shimin Zhu
- Department of Radiotherapy, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu, China
| | - Pingjin Yang
- Department of Clinical Laboratory, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu, China
| | - Songbing Qin
- Department of Radiotherapy, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China.
| | - Weigan Shen
- Department of Cell Biology, Yangzhou University Medical College, Yangzhou 225100, Jiangsu, China.
| |
Collapse
|
2
|
Custode BM, Annunziata F, Dos Santos Matos F, Schiano V, Maffia V, Lillo M, Colonna R, De Cegli R, Ballabio A, Pastore N. Folliculin depletion results in liver cell damage and cholangiocarcinoma through MiT/TFE activation. Cell Death Differ 2025:10.1038/s41418-025-01486-8. [PMID: 40189703 DOI: 10.1038/s41418-025-01486-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/11/2025] [Accepted: 03/17/2025] [Indexed: 05/11/2025] Open
Abstract
Mutations in the tumor suppressor gene Folliculin (FLCN) are responsible for Birt-Hogg-Dube' (BHD) syndrome, a rare inherited condition that predisposes affected individuals to skin tumors, pulmonary cysts, and kidney tumors. FLCN regulates key cellular pathways, including TFEB, TFE3, and mTORC1, which are critical for maintaining cell homeostasis. Loss of FLCN leads to both hyperactivation of mTORC1 and constitutive activation of TFEB and TFE3, contributing to tumorigenesis. While previous studies showed that Flcn liver-specific conditional knockout (FlcnLiKO) mice are protected from developing liver fibrosis and damage upon high-fat diet exposure, the potential role of FLCN loss in liver carcinogenesis remained unexplored. Here, we demonstrate that hepatic loss of FLCN in mice results in cancer associated with inflammation and fibrosis with features of cholangiocarcinoma (CCA). This phenotype emerges in mice over 90-week-old, with a male predominance. Moreover, FlcnLiKO mice are more prone to develop diethylnitrosamine (DEN)- or 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)- induced liver tumors with heterogenous histological features. Notably, depletion of TFE3, but not TFEB, in the liver of FlcnLiKO mice fully rescues the cancer phenotype and normalized mTORC1 signaling, highlighting TFE3 as the primary driver of liver cancer and mTORC1 hyperactivity in the absence of FLCN.
Collapse
Affiliation(s)
| | | | | | - Valentina Schiano
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Veronica Maffia
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Milena Lillo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Rita Colonna
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Rossella De Cegli
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Department of Translational Medicine, Medical Genetics, Federico II University, Naples, Italy
| | - Nunzia Pastore
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy.
- Department of Translational Medicine, Medical Genetics, Federico II University, Naples, Italy.
| |
Collapse
|
3
|
Nuri A, Jeilu S, Teklu Y, Abdu K, Muhye A, Getachew MS, Dagne I, Yigezu M, Adugna SA, Nambiar VS, Oumer A. Magnitude, patterns, and factors associated with liver disease among clinically suspected clients in Eastern Ethiopia: hidden public health tragedy. BMC Gastroenterol 2025; 25:36. [PMID: 39856553 PMCID: PMC11760694 DOI: 10.1186/s12876-025-03628-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 01/19/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Liver diseases are a public health problem in developing regions, and the majority of them are asymptomatic. Evidence on the current burden of liver disease and associated context-specific risk factors is scarce in the study area, where various risk factors for liver disease, including khat chewing, smoking, and aflatoxin exposure, are common. This study was to assess the magnitude, patterns, and factors associated with liver disease among patients visiting governmental hospitals in eastern Ethiopia. METHODS A facility-based cross-sectional study was conducted among randomly selected 388 clinically suspected adults for liver disease visiting a public hospital in Dire Dawa, Ethiopia. Data was collected using a combination of structured questionnaires, physical examinations, and investigations through interviews, medical chart reviews, liver enzymes, and the results of ultrasound examinations. Bivariable and multivariable logistic regression analyses were employed to determine the factors associated with liver disease. Candidate variables in bivariable analysis were selected based on p-values < 0.25, biological plausibility, and evidence of associations from previous studies. The level of significance was considered at a P-value < 0.05, and the adjusted odds ratio with a 95% confidence interval was reported. RESULTS A total of 388 (92%) adults were included and liver disease was diagnosed among 295 (76%; 71.5-80.2) of the study participants, where chronic liver disease due to non-viral (65.1%) and viral causes (34.9%) were prevalent. The risk of liver disease was higher among females (AOR = 3.43; 1.80-6.55) and under grade twelve (AOR = 3.90; 1.95-7.82) compared to counterparts. Furthermore, alcohol consumption (AOR = 3.14; 1.33-7.32), khat chewing (AOR = 1.69; 0.91-3.15), a history of hospitalization (AOR = 4.20; 2.29-7.11), and a history of intestinal parasite infection (AOR = 1.17; 0.55-2.49) were found to be positively associated with an increased risk of liver disease among adults. CONCLUSIONS Liver disease detection rate was found to be high and could be associated with substance use (alcohol and khat), history of hospitalization, and worm infection, which can be incorporated into the prevention strategies of liver diseases.
Collapse
Affiliation(s)
- Aliya Nuri
- Department of Public Health, College of Medicine and Health Sciences, Dire Dawa University, Dire Dawa, Ethiopia
| | - Sufian Jeilu
- Department of Internal Medicine, Sabian General Hospital, Dire Dawa, Ethiopia
| | - Yared Teklu
- School of Medicine, College of Medicine and Health Sciences, Dire Dawa University, Dire Dawa, Ethiopia
| | - Kadir Abdu
- Department of Nursing, College of Health Sciences, Oda Bultum University, Chiro, Ethiopia
| | - Ahmed Muhye
- Department of Public Health, College of Medicine and Health Sciences, Dire Dawa University, Dire Dawa, Ethiopia
| | - Milkiyas Solomon Getachew
- Department of Public Health, College of Medicine and Health Sciences, Dire Dawa University, Dire Dawa, Ethiopia
| | - Imam Dagne
- Department of Public Health, College of Medicine and Health Sciences, Dire Dawa University, Dire Dawa, Ethiopia
| | - Muluken Yigezu
- Department of Public Health, College of Medicine and Health Sciences, Dire Dawa University, Dire Dawa, Ethiopia
| | - Sewmehon Amsalu Adugna
- Department of Midwifery, College of Health Sciences, Oda Bultum University, Chiro, Ethiopia
| | - Vanisha S Nambiar
- Department of Foods and Nutrition, Faculty of Family and Community Sciences, the Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Abdu Oumer
- Department of Public Health, College of Medicine and Health Sciences, Dire Dawa University, Dire Dawa, Ethiopia.
| |
Collapse
|
4
|
Yousif MA. Aflatoxins in liver disease. TREATMENT AND MANAGEMENT OF TROPICAL LIVER DISEASE 2025:176-181. [DOI: 10.1016/b978-0-323-87031-3.00030-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Ma H, Mustafa MA, Maashi MS, Menon SV, Sivaprasad GV, Hjazi A, Ibrahim FM, Jabbar HS, Meng X. Maternal and cord blood levels of metals and fetal liver function. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125305. [PMID: 39542167 DOI: 10.1016/j.envpol.2024.125305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/24/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Exposure to metals during pregnancy has been associated with adverse birth outcomes, but its impact on fetal liver function remains poorly understood. This study for the first time aimed to investigate the association between maternal and umbilical cord blood Metals levels and umbilical liver enzymes. A comprehensive analysis was conducted on 450 mother-newborn pairs in 2022, measuring 13 Metals in serum samples from pregnant women during the third trimester and umbilical cord blood samples. Regression analyses were utilized to examine the relationship between levels of maternal and umbilical cord blood Metals and the levels of gamma-glutamyl transferase (GGT), umbilical alkaline phosphatase (ALP), alanine aminotransferase (ALT), and aspartate aminotransferase (AST). G-comp analyses evaluated the combined effect of metals exposure on umbilical liver enzymes. Elevated levels of certain Metals in cord blood and maternal samples were positively linked with increased umbilical GGT, ALP, ALT, and AST levels. Notably, zinc (Zn) levels in cord blood exhibited an inverse correlation with umbilical liver enzyme levels. Furthermore, g-comp analyses revealed significant positive associations between exposure to metals mixtures and umbilical liver enzyme levels. An increase of one quartile in the mixture of maternal and umbilical Metals was linked with 99.45 U/L (95% CI:37.72, 161.19, p < 0.01), 2.79 (95% CI: 0.92, 4.65, p < 0.01), and 87.17 (95% CI: 53.96, 120.38, p < 0.01) increase in ALP, ALT and GGT levels. Further examination of the weight of Metals revealed As, Cd, Ni, Pb, Hg and Cr with the highest positive effects and Zn with the highest negative effect in the mixture effect on the umbilical liver enzyme. In summary, our results underscore the potential influence of prenatal heavy metal exposure on fetal liver function.
Collapse
Affiliation(s)
- Haowei Ma
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | - Marwah Suliman Maashi
- Medical Laboratory Sciences Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia; Regenerative Medicine Unit at King Fahad Medical Research Centre, Jeddah, Saudi Arabia
| | - Soumya V Menon
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - G V Sivaprasad
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Ahmed Hjazi
- Clinical Medicine, Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Fatma Magdi Ibrahim
- Community Health Nursing, RAK Medical and Health Sciences University, United Arab Emirates; Geriatric nursing, Mansoura University, Egypt
| | - Hijran Sanaan Jabbar
- Department of Chemistry, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Xuan Meng
- Hepatobiliary Surgery Department, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; Zibo City Fourth People's Hospital, Zibo, 255067, China.
| |
Collapse
|
6
|
Dai C, Li D, Velkov T, Shen J, Hao Z. The Detoxification Effects of Melatonin on Aflatoxin-Caused Toxic Effects and Underlying Molecular Mechanisms. Antioxidants (Basel) 2024; 13:1528. [PMID: 39765856 PMCID: PMC11726890 DOI: 10.3390/antiox13121528] [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: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 01/15/2025] Open
Abstract
Aflatoxins (AFTs) are a form of mycotoxins mainly produced by Aspergillus flavus and Aspergillus parasiticus, which are common contaminants in various agricultural sources such as feed, milk, food, and grain crops. Aflatoxin B1 (AFB1) is the most toxic one among all AFTs. AFB1 undergoes bioactivation into AFB1-8,9-epoxide, then leads to diverse harmful effects such as neurotoxicity, carcinogenicity, hepatotoxicity, reproductive toxicity, nephrotoxicity, and immunotoxicity, with specific molecular mechanisms varying in different pathologies. The detoxification of AFB1 is of great importance for safeguarding the health of animals and humans and has increasingly attracted global attention. Recent research has shown that melatonin supplementation can effectively mitigate AFB1-induced multiple toxic effects. The protection mechanisms of melatonin involve the inhibition of oxidative stress, the upregulation of antioxidant enzyme activity, the reduction of mitochondrial dysfunction, the inactivation of the mitochondrial apoptotic pathway, the blockade of inflammatory responses, and the attenuation of cytochrome P450 enzymes' expression and activities. In summary, this review sheds new light on the potential role of melatonin as a potential detoxifying agent against AFB1. Further exploration of the precise molecular mechanisms and clinical efficacy of this promising treatment is urgently needed.
Collapse
Affiliation(s)
- Chongshan Dai
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, China
| | - Daowen Li
- College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300392, China
| | - Tony Velkov
- Department of Pharmacology, Biodiscovery Institute, Monash University, Parkville, VIC 3052, Australia
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, China
| | - Zhihui Hao
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, China
| |
Collapse
|
7
|
Barole ND, Kirnake V. Investigation of Environmental Factors as a Key Progression in the Treatment of Fatty Liver Disease: A Study Protocol. Cureus 2024; 16:e69144. [PMID: 39398757 PMCID: PMC11467620 DOI: 10.7759/cureus.69144] [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: 07/25/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
Background Fatty liver disease (FLD) is currently a global health problem associated with environmental and metabolic diseases. In addition to air pollution, chemicals, and dietary choices, metabolic problems can also contribute to the development of FLD. However, in order to understand this situation, environmental conditions need to be investigated comprehensively. Materials and methods This study used a scientific method to assess the environmental factors that play a role in FLD. Individuals from different ethnic backgrounds will be recruited as participants to increase diversity in the sample. The survey will include questions on food, exposure to air pollution, finances, and cultural practices. Statistical analysis will be conducted to further reveal environmental changes and factors that affect FLD, leading to a better understanding of environmental factors that cause FLD in the population. Results The study will significantly identify the environmental factors, such as diet, physical activity, exposure to pollutants, etc., that influence the progression and treatment outcomes of FLD. Conclusion This study will demonstrate that environmental factors influence the occurrence of FLD.
Collapse
Affiliation(s)
- Nisha D Barole
- Clinical Research, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Vijendra Kirnake
- Gastroenterology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
8
|
Buttler L, Tiede A, Griemsmann M, Rieland H, Mauz J, Kahlhöfer J, Wedemeyer H, Cornberg M, Tergast TL, Maasoumy B, Hupa-Breier KL. Folic acid supplementation is associated with a decreased mortality and reduced hospital readmission in patients with decompensated alcohol-related liver cirrhosis. Clin Nutr 2024; 43:1719-1727. [PMID: 38909513 DOI: 10.1016/j.clnu.2024.05.044] [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: 03/25/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND AND AIMS Thiamine and folic acid malnutrition is highly frequent in patients with decompensated alcohol-related liver cirrhosis (aLC). Current guidelines therefore recommend vitamin supplementation in these patients. However, implementation and its impact on the clinical outcome remains unknown. Therefore, we aimed to analyze the use of thiamine and folic acid and their effects on mortality and morbidity in patients with decompensated aLC. METHODS A number of 289 consecutive patients with decompensated aLC who received a paracentesis at Hannover Medical School between 2011 and 2023 were retrospectively investigated. The use of folic acid and thiamine-containing supplements was assessed in the discharge medication. Patients were followed for up to one year regarding liver transplant (LTx)-free survival and the incidence of hepatic encephalopathy, infections and hepatic decompensation requiring rehospitalization. RESULTS Median baseline MELD was 15, median age 56.6 years. 73.0% (n = 211) were male patients. At hospital discharge, thiamine-containing supplements and folic acid were prescribed to 48.1% (n = 139) and 18.0% (n = 52) patients, respectively. Neither thiamine nor folic acid prescription were linked to improved clinical outcomes within 90 days. However, folic acid intake was associated with a higher one-year LTx-free survival (HR = 0.48; p = 0.04) in the multivariable analysis. Furthermore, folic acid substitution was linked to a decreased risk of rehospitalization within one year (HR = 0.55; p = 0.01) in the multivariable competing risk model. In contrast, thiamine prescription did neither affect LTx-free survival nor the here investigated liver-related complications. CONCLUSION Folic acid, but not thiamine substitution was linked to an improved outcome in patients with decompensated aLC.
Collapse
Affiliation(s)
- Laura Buttler
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany.
| | - Anja Tiede
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; German Centre for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany.
| | - Marie Griemsmann
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany.
| | - Hannah Rieland
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany.
| | - Jim Mauz
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany.
| | - Julia Kahlhöfer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; German Centre for Infection Research (DZIF), HepNet Study-House/ German Liver Foundation, Hannover, Germany.
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; German Centre for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany; RESIST Cluster of Excellence, Hannover Medical School, Hannover, Germany; Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.
| | - Markus Cornberg
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; German Centre for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany; Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany; TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany.
| | - Tammo L Tergast
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany.
| | - Benjamin Maasoumy
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; German Centre for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany.
| | - Katharina L Hupa-Breier
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
9
|
Singh D, Khan MA, Mishra D, Goel A, Ansari MA, Akhtar K, Siddique HR. Apigenin enhances sorafenib anti-tumour efficacy in hepatocellular carcinoma. Transl Oncol 2024; 43:101920. [PMID: 38394865 PMCID: PMC10899070 DOI: 10.1016/j.tranon.2024.101920] [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: 01/12/2024] [Revised: 02/10/2024] [Accepted: 02/19/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND The "one drug-one target" paradigm has various limitations affecting drug efficacy, such as resistance profiles and adverse effects. Combinational therapies help reduce unexpected off-target effects and accelerate therapeutic efficacy. Sorafenib- an FDA-approved drug for liver cancer, has multiple limitations. Therefore, it is recommended to identify an agent that increases its effectiveness and reduces toxicity. In this regard, Apigenin, a plant flavone, would be an excellent option to explore. METHODS We used in silico, in vitro, and animal models to explore our hypothesis. For the in vitro study, HepG2 and Huh7 cells were exposed to Apigenin (12-96 μM) and Sorafenib (1-10 μM). For the in vivo study, Diethylnitrosamine (DEN) (25 mg/kg) induced tumor-bearing animals were given Apigenin (50 mg/kg) or Sorafenib (10 mg/kg) alone and combined. Apigenin's bioavailability was checked by UPLC. Tumor nodules were studied macroscopically and by Scanning Electron Microscopy (SEM). Biochemical analysis, histopathology, immunohistochemistry, and qRT-PCR were done. RESULTS The results revealed Apigenin's good bioavailability. In silico study showed binding affinity of both chemicals with p53, NANOG, ß-Catenin, c-MYC, and TLR4. We consistently observed a better therapeutic efficacy in combination than alone treatment. Combination treatment showed i) better cytotoxicity, apoptosis induction, and cell cycle arrest of tumor cells, ii) tumor growth reduction, iii) increased expression of p53 and decreased Cd10, Nanog, ß-Catenin, c-Myc, Afp, and Tlr4. CONCLUSIONS In conclusion, Apigenin could enhance the therapeutic efficacy of Sorafenib against liver cancer and may be a promising therapeutic approach for treating HCC. However, further research is imperative to gain more in-depth mechanistic insights.
Collapse
Affiliation(s)
- Deepti Singh
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Mohammad Afsar Khan
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Dhruv Mishra
- Department of Zoology, DAV College (PG), Maa Shakumbhari University, Muzaffarnagar-251001, India
| | - Aditya Goel
- Department of Biotechnology, SCLS, Jamia Hamdard University, New Delhi 110062, India
| | - Mairaj Ahmed Ansari
- Department of Biotechnology, SCLS, Jamia Hamdard University, New Delhi 110062, India
| | - Kafil Akhtar
- Department of Pathology, JN Medical College, Aligarh Muslim University, Aligarh 202002, India
| | - Hifzur R Siddique
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
10
|
Ozturk NB, Pham HN, Mouhaffel R, Ibrahim R, Alsaqa M, Gurakar A, Saberi B. A Longitudinal Analysis of Mortality Related to Chronic Viral Hepatitis and Hepatocellular Carcinoma in the United States. Viruses 2024; 16:694. [PMID: 38793576 PMCID: PMC11125803 DOI: 10.3390/v16050694] [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: 03/19/2024] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/26/2024] Open
Abstract
(1) Background: Hepatocellular carcinoma (HCC) contributes to the significant burden of cancer mortality in the United States (US). Despite highly efficacious antivirals, chronic viral hepatitis (CVH) remains an important cause of HCC. With advancements in therapeutic modalities, along with the aging of the population, we aimed to assess the contribution of CVH in HCC-related mortality in the US between 1999-2020. (2) Methods: We queried all deaths related to CVH and HCC in the multiple-causes-of-death files from the CDC Wide-ranging Online Data for Epidemiologic Research (WONDER) database between 1999-2020. Using the direct method of standardization, we adjusted all mortality information for age and compared the age-adjusted mortality rates (AAMRs) across demographic populations and by percentile rankings of social vulnerability. Temporal shifts in mortality were quantified using log-linear regression models. (3) Results: A total of 35,030 deaths were identified between 1999-2020. The overall crude mortality increased from 0.27 in 1999 to 8.32 in 2016, followed by a slight reduction to 7.04 in 2020. The cumulative AAMR during the study period was 4.43 (95% CI, 4.39-4.48). Males (AAMR 7.70) had higher mortality rates compared to females (AAMR 1.44). Mortality was higher among Hispanic populations (AAMR 6.72) compared to non-Hispanic populations (AAMR 4.18). Higher mortality was observed in US counties categorized as the most socially vulnerable (AAMR 5.20) compared to counties that are the least socially vulnerable (AAMR 2.53), with social vulnerability accounting for 2.67 excess deaths per 1,000,000 person-years. (4) Conclusions: Our epidemiological analysis revealed an overall increase in CVH-related HCC mortality between 1999-2008, followed by a stagnation period until 2020. CVH-related HCC mortality disproportionately affected males, Hispanic populations, and Black/African American populations, Western US regions, and socially vulnerable counties. These insights can help aid in the development of strategies to target vulnerable patients, focus on preventive efforts, and allocate resources to decrease HCC-related mortality.
Collapse
Affiliation(s)
- N. Begum Ozturk
- Department of Medicine, Beaumont Hospital, Royal Oak, MI 48073, USA
| | - Hoang Nhat Pham
- Department of Medicine, University of Arizona Tucson, Tucson, AZ 85721, USA
| | - Rama Mouhaffel
- Department of Medicine, University of Arizona Tucson, Tucson, AZ 85721, USA
| | - Ramzi Ibrahim
- Department of Medicine, University of Arizona Tucson, Tucson, AZ 85721, USA
| | - Marwan Alsaqa
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02130, USA
| | - Ahmet Gurakar
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Research Building, Suite 918, Baltimore, MD 21205, USA
| | - Behnam Saberi
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02130, USA
| |
Collapse
|
11
|
Chen T, Jiang H, He Y, Shen Y, Huang Z, Gu Y, Wei Q, Zhao J, Chen X. Nanoplastics and chrysene pollution: Potential new triggers for nonalcoholic fatty liver disease and hepatitis, insights from juvenile Siniperca chuatsi. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 922:171125. [PMID: 38382600 DOI: 10.1016/j.scitotenv.2024.171125] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 02/13/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024]
Abstract
Nanopolystyrene (NP) and chrysene (CHR) are ubiquitous contaminants in the natural environment; however, research on their hepatotoxicity and associated adverse effects remains relatively inadequate. The present study aimed to investigate the hepatotoxic effects of NP and/or CHR at environmentally relevant concentrations, as well as the underlying molecular mechanisms, in juvenile Siniperca chuatsi (mandarin fish). After a 21-day exposure period, the livers of exposed S. chuatsi exhibited macrostructural and microstructural damage accompanied by oxidative stress. Importantly, our study provides the first evidence that NP exposure leads to the development of nonalcoholic fatty liver disease (NAFLD) and hepatitis in S. chuatsi. Similarly, CHR exposure has also been found, for the first time, to cause hepatic sinusoidal dilatation (HSD) and hepatitis. Exposure to the combination of NP and CHR alleviated the symptoms of NAFLD, HSD, and hepatitis. Furthermore, our comprehensive multi-omic analysis revealed that the pathogenesis of NP-induced NAFLD was mainly due to induction of the triglyceride synthesis pathway and inhibition of the very-low-density lipoprotein secretion process. CHR induced HSD primarily through a reduction in vasoprotective ability and smooth muscle contractility. Hepatitis was induced by activation of the JAK-STAT/NF-kappa B signaling pathways, which upregulated the expression of inflammation-specific genes. Collectively, results of this study offer novel insight into the multiple hepatotoxicity endpoints of NP and/or CHR exposure at environmentally relevant concentrations in organisms, and highlight the importance of nanoplastic/CHR pollution for liver health.
Collapse
Affiliation(s)
- Tiantian Chen
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Hewei Jiang
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai 201306, China
| | - Yaoji He
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai 201306, China
| | - Yawei Shen
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Zequn Huang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Yifeng Gu
- Department of Surgical Oncology, Institute of Clinical Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Qun Wei
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Jinliang Zhao
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Xiaowu Chen
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
12
|
Contreras-Mancilla J, Cerapio JP, Ruiz E, Fernández R, Casavilca-Zambrano S, Machicado C, Fournié JJ, Pineau P, Bertani S. Hepatocellular carcinoma in Peru: A molecular description of an unconventional clinical presentation. REVISTA DE GASTROENTEROLOGIA DE MEXICO (ENGLISH) 2024; 89:194-204. [PMID: 37164797 DOI: 10.1016/j.rgmxen.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 01/17/2023] [Indexed: 05/12/2023]
Abstract
INTRODUCTION AND AIM Hepatocellular carcinoma (HCC) is the third most frequent cancer of digestive tract tumors in Peru, with a high mortality rate of 17.7 per 100,000 inhabitants. A significant number of HCC cases in Peru do not follow the classic clinical epidemiology of the disease described in other parts of the world. Those patients present with a distinct transcriptome profile and a singular tumor process, suggesting a particular type of hepatocarcinogenesis in a portion of the Peruvian population. Our aim was to understand the clinical and biologic involvement of the epigenetic profile (methylation) and gene expression (transcriptome) of HCC in Peruvian patients. METHODS HCC and liver transcriptome and DNA methylation profiles were evaluated in 74 Peruvian patients. RESULTS When grouped by age, there was greater DNA methylation in younger patients with HCC but no differences with respect to the transcriptomic profile. A high prevalence of the hepatitis B virus (HBV) (>90%) was also observed in the younger patients with HCC. Enrichment analyses in both molecular profiles pinpointed PRC2 as an important molecular effector of that liver tumor process in Peruvian patients. CONCLUSION HCC in Peruvian patients has a unique molecular profile, associated with the presence of HBV, as well as overall DNA hypermethylation related to undifferentiated liver cells or cellular reprogramming.
Collapse
Affiliation(s)
- J Contreras-Mancilla
- Laboratorio de Investigación Traslacional y Biología Computacional, Facultad de Ciencias y Filosofía - LID, Universidad Peruana Cayetano Heredia, Lima, Peru; Laboratorio Mixto Internacional de Oncología Antropológica Molecular (LOAM), IRD, INEN, Lima, Peru
| | - J P Cerapio
- Laboratorio Mixto Internacional de Oncología Antropológica Molecular (LOAM), IRD, INEN, Lima, Peru; Université de Toulouse, UMR 1037 CRCT, INSERM, CNRS, UPS, Toulouse, France; Laboratorio de Excelencia Toulouse-Cáncer (TOUCAN), Toulouse, France
| | - E Ruiz
- Laboratorio Mixto Internacional de Oncología Antropológica Molecular (LOAM), IRD, INEN, Lima, Peru; Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru
| | - R Fernández
- Laboratorio Mixto Internacional de Oncología Antropológica Molecular (LOAM), IRD, INEN, Lima, Peru; Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru
| | - S Casavilca-Zambrano
- Laboratorio Mixto Internacional de Oncología Antropológica Molecular (LOAM), IRD, INEN, Lima, Peru; Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru
| | - C Machicado
- Laboratorio de Investigación Traslacional y Biología Computacional, Facultad de Ciencias y Filosofía - LID, Universidad Peruana Cayetano Heredia, Lima, Peru; Instituto de Biocomputación y Sistemas Complejos (BIFI), Universidad de Zaragoza, Zaragoza, Spain
| | - J J Fournié
- Laboratorio Mixto Internacional de Oncología Antropológica Molecular (LOAM), IRD, INEN, Lima, Peru; Université de Toulouse, UMR 1037 CRCT, INSERM, CNRS, UPS, Toulouse, France; Laboratorio de Excelencia Toulouse-Cáncer (TOUCAN), Toulouse, France
| | - P Pineau
- Laboratorio Mixto Internacional de Oncología Antropológica Molecular (LOAM), IRD, INEN, Lima, Peru; Institut Pasteur, U 993, INSERM, Paris, France
| | - S Bertani
- Laboratorio Mixto Internacional de Oncología Antropológica Molecular (LOAM), IRD, INEN, Lima, Peru; Université de Toulouse, UMR 152 PHARMADEV, IRD, UPS, Toulouse, France.
| |
Collapse
|
13
|
Niemeijer M, Więcek W, Fu S, Huppelschoten S, Bouwman P, Baze A, Parmentier C, Richert L, Paules RS, Bois FY, van de Water B. Mapping Interindividual Variability of Toxicodynamics Using High-Throughput Transcriptomics and Primary Human Hepatocytes from Fifty Donors. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:37005. [PMID: 38498338 PMCID: PMC10947137 DOI: 10.1289/ehp11891] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Understanding the variability across the human population with respect to toxicodynamic responses after exposure to chemicals, such as environmental toxicants or drugs, is essential to define safety factors for risk assessment to protect the entire population. Activation of cellular stress response pathways are early adverse outcome pathway (AOP) key events of chemical-induced toxicity and would elucidate the estimation of population variability of toxicodynamic responses. OBJECTIVES We aimed to map the variability in cellular stress response activation in a large panel of primary human hepatocyte (PHH) donors to aid in the quantification of toxicodynamic interindividual variability to derive safety uncertainty factors. METHODS High-throughput transcriptomics of over 8,000 samples in total was performed covering a panel of 50 individual PHH donors upon 8 to 24 h exposure to broad concentration ranges of four different toxicological relevant stimuli: tunicamycin for the unfolded protein response (UPR), diethyl maleate for the oxidative stress response (OSR), cisplatin for the DNA damage response (DDR), and tumor necrosis factor alpha (TNF α ) for NF- κ B signaling. Using a population mixed-effect framework, the distribution of benchmark concentrations (BMCs) and maximum fold change were modeled to evaluate the influence of PHH donor panel size on the correct estimation of interindividual variability for the various stimuli. RESULTS Transcriptome mapping allowed the investigation of the interindividual variability in concentration-dependent stress response activation, where the average of BMCs had a maximum difference of 864-, 13-, 13-, and 259-fold between different PHHs for UPR, OSR, DDR, and NF- κ B signaling-related genes, respectively. Population modeling revealed that small PHH panel sizes systematically underestimated the variance and gave low probabilities in estimating the correct human population variance. Estimated toxicodynamic variability factors of stress response activation in PHHs based on this dataset ranged between 1.6 and 6.3. DISCUSSION Overall, by combining high-throughput transcriptomics and population modeling, improved understanding of interindividual variability in chemical-induced activation of toxicity relevant stress pathways across the human population using a large panel of plated cryopreserved PHHs was established, thereby contributing toward increasing the confidence of in vitro-based prediction of adverse responses, in particular hepatotoxicity. https://doi.org/10.1289/EHP11891.
Collapse
Affiliation(s)
- Marije Niemeijer
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, The Netherlands
| | | | - Shuai Fu
- Simcyp Division, CERTARA, Sheffield, UK
| | - Suzanna Huppelschoten
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, The Netherlands
| | - Peter Bouwman
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, The Netherlands
| | | | | | | | - Richard S. Paules
- Division of the National Toxicology Program, NIEHS, NIH, Research Triangle Park, North Carolina, USA
| | | | - Bob van de Water
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, The Netherlands
| |
Collapse
|
14
|
Zhao L, Clay-Gilmour A, Zhang J, Zhang X, Steck SE. Higher ultra-processed food intake is associated with adverse liver outcomes: a prospective cohort study of UK Biobank participants. Am J Clin Nutr 2024; 119:49-57. [PMID: 37871746 DOI: 10.1016/j.ajcnut.2023.10.014] [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: 07/18/2023] [Revised: 10/11/2023] [Accepted: 10/18/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND Ultra-processed food (UPF) intake has been positively associated with obesity and diabetes. The relationship between UPF intake and liver health has been scarcely studied. OBJECTIVES We aimed to evaluate the association of UPF intake with risk of adverse liver outcomes including nonalcoholic fatty liver disease (NAFLD), liver fibrosis/cirrhosis, liver cancer, severe liver disease, and serum biomarkers of liver health. METHODS A total of 173,889 participants aged 40 to 69 y from the UK Biobank were included. UPF intake was defined using 24-h dietary recalls and NOVA classification. Liver outcome data were obtained from cancer registry, in-hospital records, and death registries. Serum biomarkers were measured at baseline. We used Cox proportional hazards models to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for associations between UPF and adverse liver outcomes adjusting for demographics, lifestyle factors, body mass index, and diabetes. We used multinomial logistic regression to evaluate associations between UPF and liver function biomarkers. RESULTS After a median follow-up of 8.9 y, we documented 1108 NAFLD, 350 liver fibrosis/cirrhosis, 134 liver cancer, and 550 severe liver disease cases. Higher UPF intake was associated with increased risk of NAFLD (HRQuartile 4 vs. Quartile 1: 1.43; 95% CI: 1.21, 1.70; Ptrend < 0.001), liver fibrosis/cirrhosis (HR: 1.18; 95% CI: 0.87, 1.59; Ptrend = 0.009), and severe liver disease (HR: 1.50; 95% CI: 1.19, 1.90; Ptrend < 0.001) but not with liver cancer (HR: 1.00; 95% CI: 0.63, 1.58; Ptrend = 0.88). Higher UPF intake was associated with elevated levels of C-reactive protein, alkaline phosphatase, aspartate aminotransferase, γ-glutamyltransferase, and triglycerides and lower cholesterols (all Ptrend < 0.001). CONCLUSIONS Higher UPF intake is associated with an increased risk of NAFLD, liver fibrosis and cirrhosis, and severe liver disease and adverse levels of multiple clinical biomarkers, suggesting the potential importance of reducing UPF intake to improve liver health.
Collapse
Affiliation(s)
- Longgang Zhao
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Alyssa Clay-Gilmour
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States
| | - Jiajia Zhang
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States
| | - Xuehong Zhang
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Susan E Steck
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States.
| |
Collapse
|
15
|
Alotaibi KS, Almalki DA. Hepatoprotective Effect of Moringa Oil on Rats under Fungicide Toxicity. DOKL BIOCHEM BIOPHYS 2023; 513:S53-S59. [PMID: 38379081 DOI: 10.1134/s1607672923600367] [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: 11/15/2023] [Revised: 12/10/2023] [Accepted: 12/11/2023] [Indexed: 02/22/2024]
Abstract
The present study is designed to evaluate whether pretreatment with moringa would have a protective effect on thioacetamide (TAA)-induced liver fibrosis, assessing biochemical and histopathological changes in Wistar male rats. Exposure to TAA induced notable biochemical and histopathological alterations. Liver fibrosis induced by TAA, along with associated biochemical and histological damage, has not been previously investigated in male rats supplemented with moringa oil. The experiment involved forty male rats distributed across four groups, each comprising ten rats. Group 1 served as controls and received intraperitoneal injections of saline solution twice weekly for six weeks. Group 2 rats were injected with 300 mg/kg body weight of TAA (Sigma-Aldrich Corp.) twice weekly for the same duration. Group 3 rats were orally supplemented with moringa oil at 800 mg/kg body weight/day and received intraperitoneal injections of TAA at the same dosage as Group 2 for six weeks. Finally, Group 4 rats were injected with saline solution twice weekly and orally supplemented with moringa oil at 800 mg/kg body weight/day for the same period. At the end of the experiment, we determined body weight and performed liver function analysis. Additionally, we examined the liver histology of the different groups. Results showed that moringa oil treatment protected rat livers from TAA toxicity by improving liver function analysis and preventing liver fibrosis. Moringa oil can be considered a promising agent for protection against TAA toxicity.
Collapse
Affiliation(s)
- Khalid S Alotaibi
- General Science and English Language Department, College of Applied Sciences, AlMaarefa University, Riyadh, Saudi Arabia
| | - Daklallah A Almalki
- Department of Biology, Faculty of Sciences and Arts in Al-Mikhwah, Al-Baha University, Al-Mikhwah, Saudi Arabia.
| |
Collapse
|
16
|
Pauletto M, Giantin M, Tolosi R, Bassan I, Bardhi A, Barbarossa A, Montanucci L, Zaghini A, Dacasto M. Discovering the Protective Effects of Quercetin on Aflatoxin B1-Induced Toxicity in Bovine Foetal Hepatocyte-Derived Cells (BFH12). Toxins (Basel) 2023; 15:555. [PMID: 37755981 PMCID: PMC10534839 DOI: 10.3390/toxins15090555] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 09/28/2023] Open
Abstract
Aflatoxin B1 (AFB1) induces lipid peroxidation and mortality in bovine foetal hepatocyte-derived cells (BFH12), with underlying transcriptional perturbations associated mainly with cancer, cellular damage, inflammation, bioactivation, and detoxification pathways. In this cell line, curcumin and resveratrol have proven to be effective in mitigating AFB1-induced toxicity. In this paper, we preliminarily assessed the potential anti-AFB1 activity of a natural polyphenol, quercetin (QUE), in BFH12 cells. To this end, we primarily measured QUE cytotoxicity using a WST-1 reagent. Then, we pre-treated the cells with QUE and exposed them to AFB1. The protective role of QUE was evaluated by measuring cytotoxicity, transcriptional changes (RNA-sequencing), lipid peroxidation (malondialdehyde production), and targeted post-transcriptional modifications (NQO1 and CYP3A enzymatic activity). The results demonstrated that QUE, like curcumin and resveratrol, reduced AFB1-induced cytotoxicity and lipid peroxidation and caused larger transcriptional variations than AFB1 alone. Most of the differentially expressed genes were involved in lipid homeostasis, inflammatory and immune processes, and carcinogenesis. As for enzymatic activities, QUE significantly reverted CYP3A variations induced by AFB1, but not those of NQO1. This study provides new knowledge about key molecular mechanisms involved in QUE-mediated protection against AFB1 toxicity and encourages in vivo studies to assess QUE's bioavailability and beneficial effects on aflatoxicosis.
Collapse
Affiliation(s)
- Marianna Pauletto
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, I-35020 Legnaro, Italy; (M.G.); (R.T.); (I.B.); (M.D.)
| | - Mery Giantin
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, I-35020 Legnaro, Italy; (M.G.); (R.T.); (I.B.); (M.D.)
| | - Roberta Tolosi
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, I-35020 Legnaro, Italy; (M.G.); (R.T.); (I.B.); (M.D.)
| | - Irene Bassan
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, I-35020 Legnaro, Italy; (M.G.); (R.T.); (I.B.); (M.D.)
| | - Anisa Bardhi
- Department of Veterinary Medical Sciences, Alma Mater Studiorum—University of Bologna, Via Tolara di Sopra 50, Ozzano dell’Emilia, I-40064 Bologna, Italy; (A.B.); (A.B.); (A.Z.)
| | - Andrea Barbarossa
- Department of Veterinary Medical Sciences, Alma Mater Studiorum—University of Bologna, Via Tolara di Sopra 50, Ozzano dell’Emilia, I-40064 Bologna, Italy; (A.B.); (A.B.); (A.Z.)
| | - Ludovica Montanucci
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA;
| | - Anna Zaghini
- Department of Veterinary Medical Sciences, Alma Mater Studiorum—University of Bologna, Via Tolara di Sopra 50, Ozzano dell’Emilia, I-40064 Bologna, Italy; (A.B.); (A.B.); (A.Z.)
| | - Mauro Dacasto
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, I-35020 Legnaro, Italy; (M.G.); (R.T.); (I.B.); (M.D.)
| |
Collapse
|
17
|
Barouki R, Samson M, Blanc EB, Colombo M, Zucman-Rossi J, Lazaridis KN, Miller GW, Coumoul X. The exposome and liver disease - how environmental factors affect liver health. J Hepatol 2023; 79:492-505. [PMID: 36889360 PMCID: PMC10448911 DOI: 10.1016/j.jhep.2023.02.034] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 03/10/2023]
Abstract
Since the initial development of the exposome concept, much effort has been devoted to the characterisation of the exposome through analytical, epidemiological, and toxicological/mechanistic studies. There is now an urgent need to link the exposome to human diseases and to include exposomics in the characterisation of environment-linked pathologies together with genomics and other omics. Liver diseases are particularly well suited for such studies since major functions of the liver include the detection, detoxification, and elimination of xenobiotics, as well as inflammatory responses. It is well known that several liver diseases are associated with i) addictive behaviours such as alcohol consumption, smoking, and to a certain extent dietary imbalance and obesity, ii) viral and parasitic infections, and iii) exposure to toxins and occupational chemicals. Recent studies indicate that environmental exposures are also significantly associated with liver diseases, and these include air pollution (particulate matter and volatile chemicals), contaminants such as polyaromatic hydrocarbons, bisphenol A and per-and poly-fluorinated substances, and physical stressors such as radiation. Furthermore, microbial metabolites and the "gut-liver" axis play a major role in liver diseases. Exposomics is poised to play a major role in the field of liver pathology. Methodological advances such as the exposomics-metabolomics framework, the determination of risk factors' genomic and epigenomic signatures, and cross-species biological pathway analysis should further delineate the impact of the exposome on the liver, opening the way for improved prevention, as well as the identification of new biomarkers of exposure and effects, and additional therapeutic targets.
Collapse
Affiliation(s)
| | - Michel Samson
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | | | - Massimo Colombo
- San Raffaele Hospital, Liver Center, Via Olgettina 60, 20132, Milan, Italy
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Inserm, AP-HP, Hôpital Européen Georges Pompidou, Institut du Cancer Paris CARPEM, F-75006, Paris, France
| | | | - Gary W Miller
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, 10032, USA
| | | |
Collapse
|
18
|
Abstract
Hepatocyte nuclear factor 4 α (HNF4α) is a highly conserved member of the nuclear receptor superfamily expressed at high levels in the liver, kidney, pancreas, and gut. In the liver, HNF4α is exclusively expressed in hepatocytes, where it is indispensable for embryonic and postnatal liver development and for normal liver function in adults. It is considered a master regulator of hepatic differentiation because it regulates a significant number of genes involved in hepatocyte-specific functions. Loss of HNF4α expression and function is associated with the progression of chronic liver disease. Further, HNF4α is a target of chemical-induced liver injury. In this review, we discuss the role of HNF4α in liver pathophysiology and highlight its potential use as a therapeutic target for liver diseases.
Collapse
Affiliation(s)
- Manasi Kotulkar
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Dakota R Robarts
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Udayan Apte
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
19
|
Herren OM, Gillman AS, Marshall VJ, Das R. Understanding the Changing Landscape of Health Disparities in Chronic Liver Diseases and Liver Cancer. GASTRO HEP ADVANCES 2022; 2:505-520. [PMID: 37347072 PMCID: PMC10281758 DOI: 10.1016/j.gastha.2022.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/23/2023]
Abstract
Liver disease and liver cancer disparities in the U.S. are reflective of complex multiple determinants of health. This review describes the disproportionate burden of liver disease and liver cancer among racial, ethnic, sexual, and gender minority, rural, low socioeconomic status (SES) populations, and place-based contexts. The contributions of traditional and lifestyle-related risk factors (e.g., alcohol consumption, evitable toxin exposure, nutrition quality) and comorbid conditions (e.g., viral hepatitis, obesity, type II diabetes) to disparities is also explored. Biopsychosocial mechanisms defining the physiological consequences of inequities underlying these health disparities, including inflammation, allostatic load, genetics, epigenetics, and social epigenomics are described. Guided by the National Institute on Minority Health and Health Disparities (NIMHD) framework, integrative research of unexplored social and biological mechanisms of health disparities, appropriate methods and measures for early screening, diagnosis, assessment, and strategies for timely treatment and maintaining multidisciplinary care should be actively pursued. We review emerging research on adverse social determinants of liver health, such as structural racism, discrimination, stigma, SES, rising care-related costs, food insecurity, healthcare access, health literacy, and environmental exposures to pollutants. Limited research on protective factors of liver health is also described. Research from effective, multilevel, community-based interventions indicate a need for further intervention efforts that target both risk and protective factors to address health disparities. Policy-level impacts are also needed to reduce disparities. These insights are important, as the social contexts and inequities that influence determinants of liver disease/cancer have been worsened by the coronavirus disease-2019 pandemic and are forecasted to amplify disparities.
Collapse
Affiliation(s)
- Olga M. Herren
- Extramural Scientific Programs, Division of Integrative Biological and Behavioral Sciences
| | - Arielle S. Gillman
- Extramural Scientific Programs, Division of Integrative Biological and Behavioral Sciences
| | - Vanessa J. Marshall
- Office of the Director National Institute on Minority Health and Health Disparities (NIMHD), Bethesda, MD
| | - Rina Das
- Extramural Scientific Programs, Division of Integrative Biological and Behavioral Sciences
| |
Collapse
|
20
|
Han X, Long W, Liu Y, Xu J. Prognostic value and immunological role of BAIAP2L2 in liver hepatocellular carcinoma: A pan-cancer analysis. Front Surg 2022; 9:985034. [PMID: 36338652 PMCID: PMC9634486 DOI: 10.3389/fsurg.2022.985034] [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: 07/03/2022] [Accepted: 09/28/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND In recent years, the role of BAI1-associated protein 2-like 2 (BAIAP2L2) in the prognosis and immune microenvironment of various cancers has attracted increasing attention. However, its clinical value and immune infiltration in liver hepatocellular carcinoma (LIHC) remain unclear. OBJECTIVE To investigate the prognostic value of BAIAP2L2 and its correlation with immune infiltration in LIHC, we conducted corresponding data mining. METHODS In this study, The Cancer Genome Atlas, GTEx, StarBase, UALCAN, TIMER, GEPIA, Human Protein Atlas, Kaplan-Meier Plotter, cBioPortal, LinkedOmics, STRING and BioGPS databases were used to analyze BAIAP2L2 in cancers. Logistic regression and Cox regression were performed to analyze the correlation between clinical features and BAIAP2L2 expression in LIHC. In addition, the diagnostic and prognostic values of BAIAP2L2 in LIHC were determined by receiver operating characteristic (ROC) curves and nomograms. Single-sample gene set enrichment analysis (ssGSEA), BioGPS and TIMER were used to analyze the correlation between BAIAP2L2 and immune infiltration. More importantly, quantitative real-time polymerase chain reaction was used to verify BAIAP2L2 expression in a liver cancer cell line and a normal cell line. Visualization of data was mostly achieved using R language, version 3.6.3. RESULTS High BAIAP2L2 levels indicated poor overall survival (OS) and disease-free survival (DFS) of patients with LIHC. Abnormally increased expression of BAIAP2L2 in LIHC may be the result of both genetic alterations and lower DNA methylation levels. Furthermore, Cox regression analysis showed that high BAIAP2L2 expression was an independent risk factor for OS and DFS in patients with liver cancer. ROC curves and nomograms also confirmed the diagnostic and prognostic values of BAIAP2L2 in LIHC. Additionally, a PPI network of BAIAP2L2 was established and results implyed that BAIAP2L2 interacts with MTSS1, AMPH, FCHO1, SYT9, PDK2, MTSS1L, PM20D1, CHST4 and PALM3. ssGSEA showed that BAIAP2L2 was associated with T cells and natural killer cells. Simultaneously, the TIMER database showed that the expression of BAIAP2L2 in LIHC was positively correlated with tumor infiltrating cells, including B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils and dendritic cells. CONCLUSIONS Through pan-cancer analysis, prognostic and immunological value of BAIAP2L2 in LIHC was identified. This is the first report on the potential of BAIAP2L2 as a prognostic biomarker and its correlation with immune infiltration in LIHC.
Collapse
Affiliation(s)
- Xiudan Han
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China,Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, China,Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang, China
| | - Wei Long
- Department of Rheumatology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ying Liu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China,Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, China,Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang, China
| | - Jixiong Xu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China,Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, China,Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang, China,Correspondence: Jixiong Xu
| |
Collapse
|
21
|
Lee JH, Im SS. Function of gaseous hydrogen sulfide in liver fibrosis. BMB Rep 2022; 55:481-487. [PMID: 36195563 PMCID: PMC9623240 DOI: 10.5483/bmbrep.2022.55.10.124] [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] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/07/2022] [Accepted: 09/21/2022] [Indexed: 11/06/2022] Open
Abstract
Over the past few years, hydrogen sulfide (H2S) has been shown to exert several biological functions in mammalian. The endogenous production of H2S is mainly mediated by cystathione β-synthase, cystathione γ-lyase and 3-mercaptopyruvate sulfur transferase. These enzymes are broadly expressed in liver tissue and regulates liver function by working on a variety of molecular targets. As an important regulator of liver function, H2S is critically involved in the pathogenesis of various liver diseases, such as non-alcoholic steatohepatitis and liver cancer. Targeting H2S-generating enzymes may be a therapeutic strategy for controlling liver diseases. This review described the function of H2S in liver disease and summarized recent characterized role of H2S in several cellular process of the liver. [BMB Reports 2022; 55(10): 481-487].
Collapse
Affiliation(s)
- Jae-Ho Lee
- Department of Physiology, Keimyung University School of Medicine, Daegu 42601, Korea
| | - Seung-Soon Im
- Department of Physiology, Keimyung University School of Medicine, Daegu 42601, Korea
| |
Collapse
|
22
|
Hepatitis Viruses Control Host Immune Responses by Modifying the Exosomal Biogenesis Pathway and Cargo. Int J Mol Sci 2022; 23:ijms231810862. [PMID: 36142773 PMCID: PMC9505460 DOI: 10.3390/ijms231810862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
The development of smart immune evasion mechanisms is crucial for the establishment of acute and chronic viral hepatitis. Hepatitis is a major health problem worldwide arising from different causes, such as pathogens, metabolic disorders, and xenotoxins, with the five hepatitis viruses A, B, C, D, and E (HAV, HBV, HCV, HDV, and HEV) representing the majority of the cases. Most of the hepatitis viruses are considered enveloped. Recently, it was reported that the non-enveloped HAV and HEV are, in reality, quasi-enveloped viruses exploiting exosomal-like biogenesis mechanisms for budding. Regardless, all hepatitis viruses use exosomes to egress, regulate, and eventually escape from the host immune system, revealing another key function of exosomes apart from their recognised role in intercellular communication. This review will discuss how the hepatitis viruses exploit exosome biogenesis and transport capacity to establish successful infection and spread. Then, we will outline the contribution of exosomes in viral persistence and liver disease progression.
Collapse
|
23
|
Malacarne IT, Takeshita WM, de Souza DV, Dos Anjos Rosario B, de Barros Viana M, Renno ACM, Salvadori DMF, Ribeiro DA. Is micronucleus assay in oral exfoliated cells a useful biomarker for biomonitoring populations exposed to pesticides? A systematic review with meta-analysis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:64392-64403. [PMID: 35854069 DOI: 10.1007/s11356-022-22015-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/11/2022] [Indexed: 06/15/2023]
Abstract
The systematic review (SR) with meta-analysis aimed to infer if micronucleus assay using oral mucosal cells a useful biomarker for biomonitoring populations continuously exposed to pesticides (EP). The SR has been made in accordance with the PRISMA-P guidelines. The PICOS strategy has focused to answer the following question: "Does exposure to pesticides cause genetic damage in oral cells?" The literature search was made in the following scientific databases: Web of Science, PubMed/Medline, and Scopus. The approach was defined as follows: standardized mean difference (SMD) and 95% confidence intervals (CI). The quality assessment of manuscripts was obtained by the EPHPP (Effective Public Health Practice Project). The GRADE tool was chosen for assessing the quality of evidence. A total of 108 articles were selected in this setting. After screening abstracts and titles, 23 manuscripts were evaluated for eligibility. After reviewing the studies, two were considered weak and 22 were classified as moderate or strong. The meta-analysis data pointed out statistically significant differences in volunteers exposed to EP (SMD = 1.23, 95% CI, 0.69 to 1.77, p < 0.001), with a Tau2 = 1.44; Chi2 = 566.38, and p < 0.001, so that the selected manuscripts were considered heterogeneous and the I2 of 97% indicated high heterogeneity. Taken together, this review was able to validate the micronucleus assay in oral exfoliated cells as a useful biomarker in individuals continuously exposed to EP because the studies categorized as moderate and strong have demonstrated positive response related to mutagenesis.
Collapse
Affiliation(s)
- Ingra Tais Malacarne
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, UNIFESP, Rua Silva Jardim, 136, Room 332, Vila Mathias, Santos, SP, 11050-020, Brazil
| | | | - Daniel Vitor de Souza
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, UNIFESP, Rua Silva Jardim, 136, Room 332, Vila Mathias, Santos, SP, 11050-020, Brazil
| | - Barbara Dos Anjos Rosario
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, UNIFESP, Rua Silva Jardim, 136, Room 332, Vila Mathias, Santos, SP, 11050-020, Brazil
| | - Milena de Barros Viana
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, UNIFESP, Rua Silva Jardim, 136, Room 332, Vila Mathias, Santos, SP, 11050-020, Brazil
| | - Ana Claudia Muniz Renno
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, UNIFESP, Rua Silva Jardim, 136, Room 332, Vila Mathias, Santos, SP, 11050-020, Brazil
| | | | - Daniel Araki Ribeiro
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, UNIFESP, Rua Silva Jardim, 136, Room 332, Vila Mathias, Santos, SP, 11050-020, Brazil.
| |
Collapse
|
24
|
Microcystin Contamination and Toxicity: Implications for Agriculture and Public Health. Toxins (Basel) 2022; 14:toxins14050350. [PMID: 35622596 PMCID: PMC9145844 DOI: 10.3390/toxins14050350] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/02/2022] [Accepted: 05/12/2022] [Indexed: 01/02/2023] Open
Abstract
Microcystins are natural hepatotoxic metabolites secreted by cyanobacteria in aquatic ecosystems. When present at elevated concentrations, microcystins can affect water quality aesthetics; contaminate drinking water reservoirs and recreational waters; disrupt normal ecosystem functioning; and cause health hazards to animals, plants, and humans. Animal and human exposures to microcystins generally result from ingesting contaminated drinking water or physically contacting tainted water. Much research has identified a multitude of liver problems from oral exposure to microcystins, varying from hepatocellular damage to primary liver cancer. Provisional guidelines for microcystins in drinking and recreational water have been established to prevent toxic exposures and protect public health. With increasing occurrences of eutrophication in freshwater systems, microcystin contamination in groundwater and surface waters is growing, posing threats to aquatic and terrestrial plants and agricultural soils used for crop production. These microcystins are often transferred to crops via irrigation with local sources of water, such as bloom-forming lakes and ponds. Microcystins can survive in high quantities in various parts of plants (roots, stems, and leaves) due to their high chemical stability and low molecular weight, increasing health risks for consumers of agricultural products. Studies have indicated potential health risks associated with contaminated fruits and vegetables sourced from irrigated water containing microcystins. This review considers the exposure risk to humans, plants, and the environment due to the presence of microcystins in local water reservoirs used for drinking and irrigation. Additional studies are needed to understand the specific health impacts associated with the consumption of microcystin-contaminated agricultural plants.
Collapse
|
25
|
Li K, Tharwat M, Larson EL, Felgendreff P, Hosseiniasl SM, Rmilah AA, Safwat K, Ross JJ, Nyberg SL. Re-Endothelialization of Decellularized Liver Scaffolds: A Step for Bioengineered Liver Transplantation. Front Bioeng Biotechnol 2022; 10:833163. [PMID: 35360393 PMCID: PMC8960611 DOI: 10.3389/fbioe.2022.833163] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/11/2022] [Indexed: 12/12/2022] Open
Abstract
Bioengineered livers (BELs) are an attractive therapeutic alternative to address the donor organ shortage for liver transplantation. The goal of BELs technology aims at replacement or regeneration of the native human liver. A variety of approaches have been proposed for tissue engineering of transplantable livers; the current review will highlight the decellularization-recellularization approach to BELs. For example, vascular patency and appropriate cell distribution and expansion are critical components in the production of successful BELs. Proper solutions to these components of BELs have challenged its development. Several strategies, such as heparin immobilization, heparin-gelatin, REDV peptide, and anti-CD31 aptamer have been developed to extend the vascular patency of revascularized bioengineered livers (rBELs). Other novel methods have been developed to enhance cell seeding of parenchymal cells and to increase graft functionality during both bench and in vivo perfusion. These enhanced methods have been associated with up to 15 days of survival in large animal (porcine) models of heterotopic transplantation but have not yet permitted extended survival after implantation of BELs in the orthotopic position. This review will highlight both the remaining challenges and the potential for clinical application of functional bioengineered grafts.
Collapse
Affiliation(s)
- Kewei Li
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- Department of Pediatric Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Mohammad Tharwat
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- General Surgery Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ellen L. Larson
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Philipp Felgendreff
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- Department for General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| | | | - Anan Abu Rmilah
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Khaled Safwat
- General Surgery Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | | - Scott L. Nyberg
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
- *Correspondence: Scott L. Nyberg,
| |
Collapse
|