1
|
Zhou Y, Ji P, Sun Q, Gao H, Liu Z. BODIPY-based small molecular probes for fluorescence and photoacoustic dual-modality imaging of superoxide anion in vivo. Talanta 2025; 294:128269. [PMID: 40334509 DOI: 10.1016/j.talanta.2025.128269] [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: 02/13/2025] [Revised: 04/28/2025] [Accepted: 05/03/2025] [Indexed: 05/09/2025]
Abstract
Superoxide anion contributes significantly in the pathological process of acute liver injury. Therefore, real-time in vivo imaging of superoxide anion is of great significance for understanding the pathogenesis. Nevertheless, developing superoxide anion probes that possess high sensitivity and resolution continues to be a challenge. Herein, we report the design of BODIPY-based molecule probes (BDPOS1-2) for fluorescence and photoacoustic dual-modality imaging of superoxide anion. The probes exhibited exceptional selectivity and specificity towards superoxide anion, with a "turn-on" photoacoustic and "turn-off" fluorescence response. They maintained good stability and demonstrated the response behavior to superoxide anion within the pH range of 5-10. BDPOS1-2 can be used for fluorescence imaging endogenous and exogenous superoxide anion in HepG2 cells with detection in the 670-750 nm channel. Notably, galactose-modified BDPOS2 demonstrated selective hepatocyte-targeting capability and achieved dual-modality imaging of superoxide anions during acute liver injury in live mice via capturing photoacoustic signals at 715 nm and fluorescence signals in the 650-690 nm channel. Our findings offer a powerful approach for high-sensitivity and high-resolution in vivo imaging, with considerable potential for early and precise diagnosis of liver injury.
Collapse
Affiliation(s)
- Ying Zhou
- College of Materials Science and Engineering, Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, 210037, China
| | - Peihua Ji
- College of Materials Science and Engineering, Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, 210037, China
| | - Qian Sun
- College of Materials Science and Engineering, Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, 210037, China
| | - Hu Gao
- College of Materials Science and Engineering, Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, 210037, China.
| | - Zhipeng Liu
- College of Materials Science and Engineering, Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, 210037, China.
| |
Collapse
|
2
|
Li Y, Yang X, Li X, Wang S, Chen P, Ma T, Zhang B. Astragaloside IV and cycloastragenol promote liver regeneration through regulation of hepatic oxidative homeostasis and glucose/lipid metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156165. [PMID: 39461202 DOI: 10.1016/j.phymed.2024.156165] [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: 01/26/2024] [Revised: 05/28/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND The regenerative capacity of the liver is pivotal for mitigating various forms of liver injury and requires the rapid proliferation of hepatocytes. Aquaporin-9 (AQP9) provides vital support for hepatocyte proliferation by preserving hydrogen peroxide (H2O2) oxidative balance and glucose/lipid metabolism equilibrium within hepatocytes. Our previous study demonstrated that Radix Astragali (RA) decoction promotes liver regeneration by upregulating hepatic expression of AQP9, possibly via two major active constituents: astragaloside IV (AS-IV) and cycloastragenol (CAG). PURPOSE To verify that upregulated AQP9 expression in hepatocytes maintains liver oxidative balance and glucose/lipid metabolism homeostasis, and is the main pharmacological mechanism by which AS-IV and CAG promote liver regeneration. STUDY DESIGN/METHODS Effects of AS-IV and CAG on liver regeneration were scrutinized using a mouse model of 70 % partial hepatectomy (PHx). AQP9-targeted liver regeneration mediated by AS-IV and CAG was verified using AQP9 gene knockout mice (AQP9-/-). The AQP9 protein expression pattern in hepatocytes was determined using tdTomato-tagged AQP9 transgenic mice (AQP9-RFP). Potential mechanisms of AS-IV and CAG on liver regeneration were studied using real-time quantitative PCR, immunoblotting, staining with hematoxylin and eosin, oil red O, and periodic acid-Schiff, and immunofluorescence, immunohistochemistry, HyPerRed fluorescence, and biochemical analyses. RESULTS AS-IV and CAG promoted substantial liver regeneration and increased hepatic AQP9 expression in wild-type mice (AQP9+/+) following 70 % PHx, but had no discernible benefits in AQP9-/- mice. Both saponin compounds also helped maintain oxidative homeostasis by reducing levels of oxidative stress markers (reactive oxygen species [ROS], H2O2, and malondialdehyde) and elevating levels of ROS scavengers (glutathione and superoxide dismutase) in AQP9+/+ mice post-70 % PHx. This further activated the PI3K-AKT and insulin signaling pathways, thereby fostering liver regeneration. Furthermore, AS-IV and CAG both promoted hepatocyte glycerol uptake, increased gluconeogenesis, facilitated lipolysis, reduced glycolysis, and inhibited glycogen deposition, thus ensuring the energy supply required for liver regeneration. CONCLUSION This research is the first to demonstrate AS-IV and CAG as major active ingredients of RA that promote liver regeneration by upregulating hepatocyte AQP9 expression, improving hepatocyte glucose/lipid metabolism, and reducing oxidative stress damage, constituting a crucial pharmacological mechanism underlying the liver-protective effects of RA. The augmentation of hepatocyte AQP9 expression underscores an important aspect of the Qi-tonifying effect of RA. This study establishes AQP9 as an effective target for regulation of liver regeneration and provides a universal strategy for clinical drug intervention aimed at enhancing liver regeneration.
Collapse
Affiliation(s)
- Yanghao Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023,PR China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Xu Yang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Xiang Li
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Shaodong Wang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Peng Chen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Tonghui Ma
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023,PR China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| | - Bo Zhang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| |
Collapse
|
3
|
Solleiro-Villavicencio H, Méndez-García LA, Ocampo-Aguilera NA, Baltazar-Pérez I, Arreola-Miranda JA, Aguayo-Guerrero JA, Alfaro-Cruz A, González-Chávez A, Fonseca-Sánchez MA, Fragoso JM, Escobedo G. Decreased Hepatic and Serum Levels of IL-10 Concur with Increased Lobular Inflammation in Morbidly Obese Patients. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:862. [PMID: 38929479 PMCID: PMC11205754 DOI: 10.3390/medicina60060862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024]
Abstract
Background and Objectives: Non-alcoholic fatty liver disease (NAFLD) is associated with obesity and ranges from simple steatosis to non-alcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and hepatocellular carcinoma. Accumulating evidence in animal models suggests that loss of interleukin-10 (IL-10) anti-inflammatory actions might contribute to lobular inflammation, considered one of the first steps toward NASH development. However, the role of IL-10 in lobular inflammation remains poorly explored in humans. We examined mRNA and protein levels of IL-10 in liver biopsies and serum samples from morbidly obese patients, investigating the relationship between IL-10 and lobular inflammation degree. Materials and Methods: We prospectively enrolled morbidly obese patients of both sexes, assessing the lobular inflammation grade by the Brunt scoring system to categorize participants into mild (n = 7), moderate (n = 19), or severe (n = 13) lobular inflammation groups. We quantified the hepatic mRNA expression of IL-10 by quantitative polymerase chain reaction and protein IL-10 levels in liver and serum samples by Luminex Assay. We estimated statistical differences by one-way analysis of variance (ANOVA) and Tukey's multiple comparison test. Results: The hepatic expression of IL-10 significantly diminished in patients with severe lobular inflammation compared with the moderate lobular inflammation group (p = 0.01). The hepatic IL-10 protein levels decreased in patients with moderate or severe lobular inflammation compared with the mild lobular inflammation group (p = 0.008 and p = 0.0008, respectively). In circulation, IL-10 also significantly decreased in subjects with moderate or severe lobular inflammation compared with the mild lobular inflammation group (p = 0.005 and p < 0.0001, respectively). Conclusions: In liver biopsies and serum samples of morbidly obese patients, the protein levels of IL-10 progressively decrease as lobular inflammation increases, supporting the hypothesis that lobular inflammation develops because of the loss of the IL-10-mediated anti-inflammatory counterbalance.
Collapse
Affiliation(s)
| | - Lucía Angélica Méndez-García
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| | - Nydia A. Ocampo-Aguilera
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| | - Itzel Baltazar-Pérez
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| | - José A. Arreola-Miranda
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| | - José A. Aguayo-Guerrero
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| | - Ana Alfaro-Cruz
- Pathological Anatomy Department, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico;
| | - Antonio González-Chávez
- Clínica de Atención Integral para Pacientes con Diabetes y Obesidad (CAIDO), General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico;
| | | | - José Manuel Fragoso
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico;
| | - Galileo Escobedo
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| |
Collapse
|
4
|
Yang X, Zhang J, Li Y, Hu H, Li X, Ma T, Zhang B. Si-Ni-San promotes liver regeneration by maintaining hepatic oxidative equilibrium and glucose/lipid metabolism homeostasis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117918. [PMID: 38382654 DOI: 10.1016/j.jep.2024.117918] [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: 11/23/2023] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 02/23/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The efficacy of clinical treatments for various liver diseases is intricately tied to the liver's regenerative capacity. Insufficient or failed liver regeneration is a direct cause of mortality following fulminant hepatic failure and extensive hepatectomy. Si-Ni-San (SNS), a renowned traditional Chinese medicine prescription for harmonizing liver and spleen functions, has shown clinical efficacy in the alleviation of liver injury for thousands of years. However, the precise molecular pharmacological mechanisms underlying its effects remain unclear. AIMS OF THE STUDY This study aimed to investigate the effects of SNS on liver regeneration and elucidate the underlying mechanisms. MATERIALS AND METHODS A mouse model of 70% partial hepatectomy (PHx) was used to analyze the effects of SNS on liver regeneration. Aquaporin-9 knockout mice (AQP9-/-) were used to demonstrate that SNS-mediated enhancement of liver regeneration was AQP9-targeted. A tandem dimer-Tomato-tagged AQP9 transgenic mouse line (AQP9-RFP) was utilized to determine the expression pattern of AQP9 protein in hepatocytes. Immunoblotting, quantitative real-time PCR, staining techniques, and biochemical assays were used to further explore the underlying mechanisms of SNS. RESULTS SNS treatment significantly enhanced liver regeneration and increased AQP9 protein expression in hepatocytes of wild-type mice (AQP9+/+) post 70% PHx, but had no significant effects on AQP9-/- mice. Following 70% PHx, SNS helped maintain hepatic oxidative equilibrium by increasing the levels of reactive oxygen species scavengers glutathione and superoxide dismutase and reducing the levels of oxidative stress molecules H2O2 and malondialdehyde in liver tissues, thereby preserving this crucial process for hepatocyte proliferation. Simultaneously, SNS augmented glycerol uptake by hepatocytes, stimulated gluconeogenesis, and maintained glucose/lipid metabolism homeostasis, ensuring the energy supply required for liver regeneration. CONCLUSIONS This study provides the first evidence that SNS maintains liver oxidative equilibrium and glucose/lipid metabolism homeostasis by upregulating AQP9 expression in hepatocytes, thereby promoting liver regeneration. These findings offer novel insights into the molecular pharmacological mechanisms of SNS in promoting liver regeneration and provide guidance for its clinical application and optimization in liver disease treatment.
Collapse
Affiliation(s)
- Xu Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Junqi Zhang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yanghao Li
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Huiting Hu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiang Li
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tonghui Ma
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Bo Zhang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
5
|
Baradwan S, Gari A, Sabban H, Alshahrani MS, Khadawardi K, Bukhari IA, Alyousef A, Abu-Zaid A. The effect of antioxidant supplementation on dysmenorrhea and endometriosis-associated painful symptoms: a systematic review and meta-analysis of randomized clinical trials. Obstet Gynecol Sci 2024; 67:186-198. [PMID: 38221738 PMCID: PMC10948216 DOI: 10.5468/ogs.23210] [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: 08/25/2023] [Revised: 10/11/2023] [Accepted: 11/09/2023] [Indexed: 01/16/2024] Open
Abstract
This study aimed to review randomized controlled trials (RCTs) investigating the effects of dietary antioxidant supplements on the severity of endometriosis-related pain symptoms. The PubMed/Medline, Scopus, and Web of Science databases were searched until April 2022. Additionally, we manually searched the reference lists. Endpoints were summarized as standardized mean difference (SMD) with 95% confidence intervals (CIs) in a random-effects model. The I2 statistic was used to assess heterogeneity. Ten RCTs were included in this meta-analysis. Overall, 10 studies were related to dysmenorrhea, four to dyspareunia, and four to pelvic pain. Antioxidants significantly reduced dysmenorrhea (SMD, -0.48; 95% CI, -0.82 to -0.13; I2=75.14%). In a subgroup analysis, a significant reduction of dysmenorrhea was observed only in a subset of trials that administered vitamin D (SMD, -0.59; 95% CI, -1.13 to -0.06; I2=69.59%) and melatonin (SMD, -1.40; 95% CI, -2.47 to -0.32; I2=79.15%). Meta-analysis results also suggested that antioxidant supplementation significantly improved pelvic pain (SMD, -1.51; 95% CI, -2.74 to -0.29; I2=93.96%), although they seem not to have a significant beneficial impact on the severity of dyspareunia. Dietary antioxidant supplementation seems to beneficially impact the severity of endometriosis-related dysmenorrhea (with an emphasis on vitamin D and melatonin) and pelvic pain. However, due to the relatively small sample size and high heterogeneity, the findings should be interpreted cautiously, and the importance of further well-designed clinical studies cannot be overstated.
Collapse
Affiliation(s)
- Saeed Baradwan
- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research Center, Jeddah,
Saudi Arabia
| | - Abdulrahim Gari
- Department of Obstetrics and Gynecology, College of Medicine, Umm Al-Qura University, Makkah,
Saudi Arabia
- Department of Obstetrics and Gynecology, Al Salama Hospital, Jeddah,
Saudi Arabia
| | - Hussein Sabban
- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research Center, Jeddah,
Saudi Arabia
- Department of Obstetrics and Gynecology, Faculty of Medicine at Rabigh, King Abdulaziz University, Rabigh,
Saudi Arabia
| | - Majed Saeed Alshahrani
- Department of Obstetrics and Gynecology, Faculty of Medicine, Najran University, Najran,
Saudi Arabia
| | - Khalid Khadawardi
- Department of Obstetrics and Gynecology, College of Medicine, Umm Al-Qura University, Makkah,
Saudi Arabia
| | - Ibtihal Abdulaziz Bukhari
- Department of Obstetrics and Gynecology, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh,
Saudi Arabia
| | - Abdullah Alyousef
- Department of Obstetrics and Gynecology, King Abdullah bin Abdulaziz University Hospital, Riyadh,
Saudi Arabia
| | - Ahmed Abu-Zaid
- Department of Obstetrics and Gynecology, College of Medicine, Alfaisal University, Riyadh,
Saudi Arabia
| |
Collapse
|
6
|
Jia D, Zhao M, Luo J, Li S, Gong J, Cheng M. Dimethyloxalylglycine pretreatment of living donor alleviates both donor and graft liver ischemia-reperfusion injury in rats. Front Pharmacol 2024; 14:1341575. [PMID: 38264531 PMCID: PMC10803591 DOI: 10.3389/fphar.2023.1341575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/22/2023] [Indexed: 01/25/2024] Open
Abstract
Background: Under the circumstance of the increasing waiting list for liver transplantation, living donor liver transplantation (LDLT) can alleviate the shortage of liver donors to some extent. However, how to reduce both donor and graft ischemia-reperfusion injury (IRI) is still an unsolved problem in LDLT. Hypoxia-induced transcription factor 1 (HIF1) activation is considered an important mechanism of cellular adaptation to hypoxia, and early activation of HIF1 may be a new way to alleviate liver IRI. Therefore, we aimed to investigate the impact of the HIF1 stabilizer dimethyloxalylglycine (DMOG) on IRI and the survival rate of donors and recipients of rat LDLT. Methods: Seventy percent partial liver resection and 30% partial liver transplantation were used to simulate donor and recipient of clinical LDLT. Rats were treated with DMOG (40 mg/kg) or with an equivalent amount of saline. The expression of HIF1 and downstream targets was analyzed after 2 h of reperfusion. Liver function and histopathology, apoptosis and oxidative stress levels were detected 6 h after reperfusion. At the same time, the 7-day survival rate of rats was calculated. Results: DMOG pretreatment significantly reduced IR-induced injury in the donor and recipient, which was manifested by reducing liver function damage and promoting tissue recovery. Meanwhile, compared with the untreated group, the oxidative stress level and the cell apoptosis rate were decreased in the group pretreated with DMOG. In addition, the transcription and expression of HIF1 target genes in the DMOG group were significantly enhanced. Remarkably, DMOG also increased the survival rate of the recipient. Conclusion: This study provides the first evidence that DMOG pretreatment of donors significantly alleviates liver IRI in both donors and recipients and increases the survival rate of recipients in LDLT. Therefore, DMOG may be a promising strategy for improving LDLT in the future.
Collapse
Affiliation(s)
| | | | | | | | | | - Mingxiang Cheng
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Terry AQ, Kojima H, Sosa RA, Kaldas FM, Chin JL, Zheng Y, Naini BV, Noguchi D, Nevarez-Mejia J, Jin YP, Busuttil RW, Meyer AS, Gjertson DW, Kupiec-Weglinski JW, Reed EF. Disulfide-HMGB1 signals through TLR4 and TLR9 to induce inflammatory macrophages capable of innate-adaptive crosstalk in human liver transplantation. Am J Transplant 2023; 23:1858-1871. [PMID: 37567451 PMCID: PMC11095628 DOI: 10.1016/j.ajt.2023.08.002] [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: 03/29/2023] [Revised: 06/27/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023]
Abstract
Ischemia-reperfusion injury (IRI) during orthotopic liver transplantation (OLT) contributes to graft rejection and poor clinical outcomes. The disulfide form of high mobility group box 1 (diS-HMGB1), an intracellular protein released during OLT-IRI, induces pro-inflammatory macrophages. How diS-HMGB1 differentiates human monocytes into macrophages capable of activating adaptive immunity remains unknown. We investigated if diS-HMGB1 binds toll-like receptor (TLR) 4 and TLR9 to differentiate monocytes into pro-inflammatory macrophages that activate adaptive immunity and promote graft injury and dysfunction. Assessment of 106 clinical liver tissue and longitudinal blood samples revealed that OLT recipients were more likely to experience IRI and graft dysfunction with increased diS-HMGB1 released during reperfusion. Increased diS-HMGB1 concentration also correlated with TLR4/TLR9 activation, polarization of monocytes into pro-inflammatory macrophages, and production of anti-donor antibodies. In vitro, healthy volunteer monocytes stimulated with purified diS-HMGB1 had increased inflammatory cytokine secretion, antigen presentation machinery, and reactive oxygen species production. TLR4 inhibition primarily impeded cytokine/chemokine and costimulatory molecule programs, whereas TLR9 inhibition decreased HLA-DR and reactive oxygen species production. diS-HMGB1-polarized macrophages also showed increased capacity to present antigens and activate T memory cells. In murine OLT, diS-HMGB1 treatment potentiated ischemia-reperfusion-mediated hepatocellular injury, accompanied by increased serum alanine transaminase levels. This translational study identifies the diS-HMGB1/TLR4/TLR9 axis as potential therapeutic targets in OLT-IRI recipients.
Collapse
Affiliation(s)
- Allyson Q Terry
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Hidenobu Kojima
- Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Rebecca A Sosa
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Fady M Kaldas
- Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jackson L Chin
- Department of Bioengineering, Samueli School of Engineering at UCLA, Los Angeles, California, USA
| | - Ying Zheng
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Bita V Naini
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Daisuke Noguchi
- Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jessica Nevarez-Mejia
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Yi-Ping Jin
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Ronald W Busuttil
- Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Aaron S Meyer
- Department of Bioengineering, Samueli School of Engineering at UCLA, Los Angeles, California, USA
| | - David W Gjertson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA; Department of Biostatistics, Fielding School of Public Health at UCLA, Los Angeles, California, USA
| | - Jerzy W Kupiec-Weglinski
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA; Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Elaine F Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.
| |
Collapse
|
8
|
Kashyap N, Islam M, Kaur H, Tiwari D, Begum A, Bose M, Das CR, Saikia AK, Kalita P, Bose PD, Bose S. Oxidative stress-A key determinant of complications and negative outcome in hepatitis E virus infected pregnancies: A comprehensive account involving cases from northeast India. J Med Virol 2023; 95:e28576. [PMID: 36779361 DOI: 10.1002/jmv.28576] [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: 10/24/2022] [Revised: 01/14/2023] [Accepted: 02/09/2023] [Indexed: 02/14/2023]
Abstract
Regulated oxidative stress (OS) is important during pregnancy. Sporadic studies suggest the significance of deregulated OS in hepatitis E virus (HEV) infected pregnancy, but with limited reactive oxygen species (ROS) or antioxidant markers. The present novel study, therefore, aimed to evaluate the significance of ROS-antioxidant imbalance and resulting altered OS in HEV infected pregnancy complications like preterm delivery (PTD) and outcome. Difference in serum levels of ROS and antioxidant panel of markers were evaluated by ELISA for HEV immunoglobulin M RNA positive genotype 1 cases (including acute [acute viral hepatitis, AVH] and fulminant [fulminant hepatic failure, FHF] cases) and healthy term delivery subjects, and analyzed statistically. Direct ROS marker H2 O2 levels and indirect OS marker for DNA damage 8-hydroxy-2'-deoxyguanosine was significantly increased in HEV-cases compared to controls, and was associated and prognostic factor for PTD and fetal death in HEV cases. A comparatively lower total serum antioxidant capacity was observed in the FHF cases compared to the control subjects and the AVH cases. Glutathione (GSH) levels and superoxide dismutase (SOD) activity were significantly associated with PTD in the FHF sub-cohorts (p = 0.017) and AVH sub-cohorts (p < 0.001), respectively, and was associated with poor prognosis in HEV cases. The serum H2 O2 levels were found to be negatively correlated with SOD activity (p = 0.016) and GSH levels (p = 0.001) in the HEV-AVH cases; and positively correlated with the viral load in HEV cases (p = 0.023). The ROS-antioxidant imbalance resulting OS plays a detrimental associative role in HEV infected pregnancy complications like PTD and adverse pregnancy outcomes; and holds therapeutic significance.
Collapse
Affiliation(s)
- Natasha Kashyap
- Department of Molecular Biology and Biotechnology, Cotton University, Guwahati, Assam, India.,Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | - Mafidul Islam
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | - Harpreet Kaur
- Division of Epidemiology and Communicable Diseases (ECD), Indian Council of Medical Research Headquarters, New Delhi, India
| | - Diptika Tiwari
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | - Anjuma Begum
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | - Moumita Bose
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | - Chandana Ray Das
- Department of Obstetrics and Gynaecology, Gauhati Medical College & Hospital (GMCH), Guwahati, Assam, India
| | - Anjan Kumar Saikia
- Department of Gastroenterology & Hepatology, Guwahati Neurological Research Center (GNRC) Hospitals, Guwahati, Assam, India
| | - Pankaj Kalita
- Department of Ayurveda, SMO (Ayurveda), ESIC Model Hospital, Guwahati, Assam, India
| | - Purabi Deka Bose
- Department of Molecular Biology and Biotechnology, Cotton University, Guwahati, Assam, India
| | - Sujoy Bose
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| |
Collapse
|
9
|
Flores AE, Pascotini ET, Kegler A, Broetto N, Gabbi P, Duarte T, Prado ALC, Duarte MMMF, da Cruz IBM, Dos Santos ARS, Royes LFF, Fighera MR. Worst spasticity in patients post-stroke associated with MNSOD ALA16VAL polymorphism and interleukin-1β. Gene X 2022; 847:146880. [PMID: 36100117 DOI: 10.1016/j.gene.2022.146880] [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: 04/30/2022] [Revised: 08/23/2022] [Accepted: 09/05/2022] [Indexed: 11/04/2022] Open
Abstract
The MnSOD Ala16Val single nucleotide polymorphism (SNP) has shown to be associated to risk factors of several metabolic and vascular diseases. However, little is known about interaction between MnSOD Ala16Val SNP in stroke, a frequent neurologic disease that involves clinic manifestations such as motor deficits and spasticity. In this sense, we decided to investigate the relationship between MnSOD Ala16Val SNP with spasticity in stroke and also its influence on interleukin levels, BDNF, and glycolipid parameters. Eighty post-stroke subjects and 80 healthy controls were investigated. We showed a higher spasticity, levels of total cholesterol, LDL, IL-1β, IL-6, and INF-γ in VV post-stroke group. Interesting, we found a correlation between IL-1β levels and spasticity in VV post-stroke. Triglycerides, glucose levels and caspases (1 and 3) activation were significantly higher, as well as BDNF levels were lower in VV and AV post-stroke. DNA damage was higher in post-stroke group. Thus, we can suggest that the V allele has a worse glycolipid profile, which would facilitate changes in neurovascular homeostasis. These events associated with an increase in inflammatory markers and a reduction in BDNF can contribute with the stroke and a worse clinical evolution in relation to spasticity in patients with VV genotype.
Collapse
Affiliation(s)
- Ariane Ethur Flores
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Eduardo Tanuri Pascotini
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Aline Kegler
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil
| | - Núbia Broetto
- Curso de Fisioterapia, Departamento de Ginástica e Saúde, Universidade Federal de Pelotas, RS, Brazil
| | - Patricia Gabbi
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Thiago Duarte
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | | | - Marta M M F Duarte
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Ivana B M da Cruz
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | | | - Luiz Fernando Freire Royes
- Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil; Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício (BIOEX), Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Michele Rechia Fighera
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil; Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício (BIOEX), Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil.
| |
Collapse
|
10
|
Conde de la Rosa L, Goicoechea L, Torres S, Garcia-Ruiz C, Fernandez-Checa JC. Role of Oxidative Stress in Liver Disorders. LIVERS 2022; 2:283-314. [DOI: 10.3390/livers2040023] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Oxygen is vital for life as it is required for many different enzymatic reactions involved in intermediate metabolism and xenobiotic biotransformation. Moreover, oxygen consumption in the electron transport chain of mitochondria is used to drive the synthesis of ATP to meet the energetic demands of cells. However, toxic free radicals are generated as byproducts of molecular oxygen consumption. Oxidative stress ensues not only when the production of reactive oxygen species (ROS) exceeds the endogenous antioxidant defense mechanism of cells, but it can also occur as a consequence of an unbalance between antioxidant strategies. Given the important role of hepatocytes in the biotransformation and metabolism of xenobiotics, ROS production represents a critical event in liver physiology, and increasing evidence suggests that oxidative stress contributes to the development of many liver diseases. The present review, which is part of the special issue “Oxidant stress in Liver Diseases”, aims to provide an overview of the sources and targets of ROS in different liver diseases and highlights the pivotal role of oxidative stress in cell death. In addition, current antioxidant therapies as treatment options for such disorders and their limitations for future trial design are discussed.
Collapse
Affiliation(s)
- Laura Conde de la Rosa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), 08036 Barcelona, Spain
| | - Leire Goicoechea
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), 08036 Barcelona, Spain
| | - Sandra Torres
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), 08036 Barcelona, Spain
| | - Carmen Garcia-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), 08036 Barcelona, Spain
- Research Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - José C. Fernandez-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), 08036 Barcelona, Spain
- Research Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
11
|
Li X, Zhou Q, Japir AAWMM, Dutta D, Lu N, Ge Z. Protein-Delivering Nanocomplexes with Fenton Reaction-Triggered Cargo Release to Boost Cancer Immunotherapy. ACS NANO 2022; 16:14982-14999. [PMID: 36017992 DOI: 10.1021/acsnano.2c06026] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Immunotherapeutic efficacy of tumors based on immune checkpoint blockade (ICB) therapy is frequently limited by an immunosuppressive tumor microenvironment and cross-reactivity with normal tissues. Herein, we develop reactive oxygen species (ROS)-responsive nanocomplexes with the function of ROS production for delivery and triggered release of anti-mouse programmed death ligand 1 antibody (αPDL1) and glucose oxidase (GOx). GOx and αPDL1 were complexed with oligomerized (-)-epigallocatechin-3-O-gallate (OEGCG), which was followed by chelation with Fe3+ and coverage of the ROS-responsive block copolymer, POEGMA-b-PTKDOPA, consisting of poly(oligo(ethylene glycol)methacrylate) (POEGMA) and the block with thioketal bond-linked dopamine moieties (PTKDOPA) as the side chains. After intravenous injection, the nanocomplexes show prolonged circulation in the bloodstream with a half-life of 8.72 h and efficient tumor accumulation. At the tumor sites, GOx inside the nanocomplexes can produce H2O2 via oxidation of glucose for Fenton reaction to generate hydroxyl radicals (•OH) which further trigger the release of the protein cargos through ROS-responsive cleavage of thioketal bonds. The released GOx improves the production efficiency of •OH to kill cancer cells for release of tumor-associated antigens via chemodynamic therapy (CDT). The enhanced immunogenic cell death (ICD) can activate the immunosuppressive tumor microenvironment and improve the immunotherapy effect of the released αPDL1, which significantly suppresses primary and metastatic tumors. Thus, the nanocomplexes with Fenton reaction-triggered protein release show great potentials to improve the immunotherapeutic efficacy of ICB via combination with CDT.
Collapse
Affiliation(s)
- Xiang Li
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Qinghao Zhou
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Abd Al-Wali Mohammed M Japir
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Debabrata Dutta
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Nannan Lu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Zhishen Ge
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
- School of Chemistry, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| |
Collapse
|
12
|
G Bardallo R, Panisello-Roselló A, Sanchez-Nuno S, Alva N, Roselló-Catafau J, Carbonell T. Nrf2 and oxidative stress in liver ischemia/reperfusion injury. FEBS J 2022; 289:5463-5479. [PMID: 34967991 DOI: 10.1111/febs.16336] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/18/2021] [Accepted: 12/29/2021] [Indexed: 11/28/2022]
Abstract
In response to stress signal, nuclear factor-erythroid 2-related factor 2 (Nrf2) induces the expression of target genes involved in antioxidant defense and detoxification. Nrf2 activity is strictly regulated through a variety of mechanisms, including regulation of Keap1-Nrf2 stability, transcriptional regulation (NF-ĸB, ATF3, ATF4), and post-transcriptional regulation (miRNA), evidencing that transcriptional responses of Nrf2 are critical for the maintenance of homeostasis. Ischemia-reperfusion (IR) injury is a major cause of graft loss and dysfunction in clinical transplantation and organ resection. During the IR process, the generation of reactive oxygen species (ROS) leads to damage from oxidative stress, oxidation of biomolecules, and mitochondrial dysfunction. Oxidative stress can trigger apoptotic and necrotic cell death. Stress factors also result in the assembly of the inflammasome protein complex and the subsequent activation and secretion of proinflammatory cytokines. After Nrf2 activation, the downstream antioxidant upregulation can act as a primary cellular defense against the cytotoxic effects of oxidative stress and help to promote hepatic recovery during IR. The complex crosstalk between Nrf2 and cellular pathways in liver IR injury and the potential therapeutic target of the Nrf2 inducers will be discussed in the present review.
Collapse
Affiliation(s)
- Raquel G Bardallo
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Spain
| | - Arnau Panisello-Roselló
- Experimental Pathology Department, Institute of Biomedical Research of Barcelona (IIBB), CSIC-IDIBAPS, Barcelona, Spain
| | - Sergio Sanchez-Nuno
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Spain
| | - Norma Alva
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Spain
| | - Joan Roselló-Catafau
- Experimental Pathology Department, Institute of Biomedical Research of Barcelona (IIBB), CSIC-IDIBAPS, Barcelona, Spain
| | - Teresa Carbonell
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Spain
| |
Collapse
|
13
|
Krajina I, Stupin A, Šola M, Mihalj M. Oxidative Stress Induced by High Salt Diet—Possible Implications for Development and Clinical Manifestation of Cutaneous Inflammation and Endothelial Dysfunction in Psoriasis vulgaris. Antioxidants (Basel) 2022; 11:antiox11071269. [PMID: 35883760 PMCID: PMC9311978 DOI: 10.3390/antiox11071269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/19/2022] [Accepted: 06/23/2022] [Indexed: 02/07/2023] Open
Abstract
Although oxidative stress is recognized as an important effector mechanism of the immune system, uncontrolled formation of reactive oxygen and nitrogen species promotes excessive tissue damage and leads to disease development. In view of this, increased dietary salt intake has been found to damage redox systems in the vessel wall, resulting in endothelial dysfunction associated with NO uncoupling, inflammation, vascular wall remodeling and, eventually, atherosclerosis. Several studies have reported increased systemic oxidative stress accompanied by reduced antioxidant capacity following a high salt diet. In addition, vigorous ionic effects on the immune mechanisms, such as (trans)differentiation of T lymphocytes are emerging, which together with the evidence of NaCl accumulation in certain tissues warrants a re-examination of the data derived from in vitro research, in which the ionic influence was excluded. Psoriasis vulgaris (PV), as a primarily Th17-driven inflammatory skin disease with proven inflammation-induced accumulation of sodium chloride in the skin, merits our interest in the role of oxidative stress in the pathogenesis of PV, as well as in the possible beneficial effects that could be achieved through modulation of dietary salt intake and antioxidant supplementation.
Collapse
Affiliation(s)
- Ivana Krajina
- Department of Dermatology and Venereology, Osijek University Hospital, J. Huttlera 4, HR-31000 Osijek, Croatia;
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
| | - Ana Stupin
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia;
- Institute and Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
| | - Marija Šola
- Department of Dermatology and Venereology, Osijek University Hospital, J. Huttlera 4, HR-31000 Osijek, Croatia;
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
- Correspondence: (M.Š.); (M.M.); Tel.: +385-31-512-800 (M.M.)
| | - Martina Mihalj
- Department of Dermatology and Venereology, Osijek University Hospital, J. Huttlera 4, HR-31000 Osijek, Croatia;
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia;
- Institute and Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
- Correspondence: (M.Š.); (M.M.); Tel.: +385-31-512-800 (M.M.)
| |
Collapse
|
14
|
Siao YJ, Peng CC, Tung YC, Chen YF. Comparison of Hydrogen Peroxide Secretion From Living Cells Cultured in Different Formats Using Hydrogel-Based LSPR Substrates. Front Bioeng Biotechnol 2022; 10:869184. [PMID: 35464720 PMCID: PMC9031350 DOI: 10.3389/fbioe.2022.869184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/22/2022] [Indexed: 12/04/2022] Open
Abstract
Reactive oxygen species (ROS), a number of reactive molecules and free radicals derived from molecular oxygen, are generated as by-products during mitochondrial electron transport within cells. Physiologically, cells are capable of metabolizing the ROS exploiting specific mechanisms. However, if excessive ROS accumulate inside the cells, it will cause the cells apoptosis or necrosis. Hydrogen peroxide (H2O2) is one of the essential ROS often participating in chemical reactions in organisms and regulating homeostasis in the body. Therefore, rapid and sensitive detection of H2O2 is a significant task in cell biology research. Furthermore, it has been found that cells cultured in different formats can result in different cellular responses and biological activities. In order to investigate the H2O2 secretion from the cells cultured in different formats, a hydrogel-based substrate is exploited to separate relatively large molecular (e.g., proteins) for direct measurement of H2O2 secreted from living cells in complete cell culture medium containing serum. The substrate takes advantage of the localized surface plasmon resonance (LSPR) method based on enzyme immunoprecipitation. In addition, the H2O2 secreted from the cells cultured in different dimensions (suspension of single cells and three-dimensional cell spheroids) treated with identical drugs is measured and compared. The spheroid samples can be prepared with ample amount using a designed microfluidic device with precise control of size. The results show that the H2O2 secretion from the cells are great affected by their culture formats.
Collapse
Affiliation(s)
- Yang-Jyun Siao
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Chien-Chung Peng
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Chung Tung
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
- *Correspondence: Yi-Chung Tung, ; Yih-Fan Chen,
| | - Yih-Fan Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
- *Correspondence: Yi-Chung Tung, ; Yih-Fan Chen,
| |
Collapse
|
15
|
Zhang B, Lv D, Chen Y, Nie W, Jiao Y, Zhang J, Zhou X, Wu X, Chen S, Ma T. Aquaporin-9 facilitates liver regeneration following hepatectomy. Redox Biol 2022; 50:102246. [PMID: 35086002 PMCID: PMC8802049 DOI: 10.1016/j.redox.2022.102246] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/06/2022] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
Aquaporin-9 (AQP9) is an aquaglyceroporin strongly expressed in the basolateral membrane of hepatocytes facing the sinusoids. AQP9 is permeable to hydrogen peroxide (H2O2) and glycerol as well as to water. Here, we report impaired liver regeneration in AQP9−/− mice which involves altered steady-state H2O2 concentration and glucose metabolism in hepatocytes. AQP9−/− mice showed remarkably delayed liver regeneration and increased mortality following 70% or 90% partial hepatectomy. Compared to AQP9+/+ littermates, AQP9−/− mice showed significantly greater hepatic H2O2 concentration and more severe liver injury. Fluorescence measurements indicated impaired H2O2 transport across plasma membrane of primary cultured hepatocytes from AQP9−/− mice, supporting the hypothesis that AQP9 deficiency results in H2O2 accumulation and oxidative injury in regenerating liver because of reduced export of intracellular H2O2 from hepatocytes. The H2O2 overload in AQP9−/− hepatocytes reduced PI3K-Akt and insulin signaling, inhibited autophagy and promoted apoptosis, resulting in impaired proliferation and increased cell death. In addition, hepatocytes from AQP9−/− mice had low liver glycerol and high blood glycerol levels, suggesting decreased glycerol uptake and gluconeogenesis in AQP9−/− hepatocytes. Adeno-associated virus (AAV)-mediated expression of hepatic expression of aquaglyceroporins AQP9 and AQP3 in AQP9−/− mice, but not water-selective channel AQP4, fully rescued the impaired liver regeneration phenotype as well as the oxidative injury and abnormal glucose metabolism. Our data revealed a pivotal role of AQP9 in liver regeneration by regulating hepatocyte H2O2 homeostasis and glucose metabolism, suggesting AQP9 as a novel target to enhance liver regeneration following injury, surgical resection or transplantation. AQP9 mediates H2O2 and glycerol transport across hepatocytes plasma membrane AQP9−/− mice exhibit retained liver regeneration and higher mortality after PH Elevated H2O2 and reduced glucose levels appear in AQP9−/− regenerating liver Replacement of aquaglyceroporin rescued impaired AQP9−/− mouse liver regeneration AQP9 may become a novel target to improve liver regeneration
Collapse
|
16
|
Immune Responses in Leishmaniases: An Overview. Trop Med Infect Dis 2022; 7:tropicalmed7040054. [PMID: 35448829 PMCID: PMC9029249 DOI: 10.3390/tropicalmed7040054] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Leishmaniasis is a parasitic, widespread, and neglected disease that affects more than 90 countries in the world. More than 20 Leishmania species cause different forms of leishmaniasis that range in severity from cutaneous lesions to systemic infection. The diversity of leishmaniasis forms is due to the species of parasite, vector, environmental and social factors, genetic background, nutritional status, as well as immunocompetence of the host. Here, we discuss the role of the immune system, its molecules, and responses in the establishment, development, and outcome of Leishmaniasis, focusing on innate immune cells and Leishmania major interactions.
Collapse
|
17
|
Mohammad Yousof S, Erfan H, Mohamed Hosny M, Shehata SA, El-sayed K. Subacute Toxic Effects of Silver Nanoparticles oral Administration and Withdrawal on the Structure and Function of Adult Albino Rats’ Hepatic Tissue. Saudi J Biol Sci 2022; 29:3890-3898. [PMID: 35844407 PMCID: PMC9280256 DOI: 10.1016/j.sjbs.2022.02.054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022] Open
Abstract
Products containing Silver nanoparticles (Ag NPs) are becoming vastly used in our daily life. The widespread increased introduction of Ag NPs in many aspects of life has raised researchers' concerns regarding their safety and toxicity for biological and environmental life in the past few years. The current study aimed to explore the subsequent effects of Ag NPs withdrawal, following short-term oral administration. Eighteen rats were assigned randomly into three groups (control group "1" and AG NPs treated groups "2" and "3"; 6 animals each). The control group received normal food and tap water while groups 2 & 3 received 0.5 ml of a solution containing 25 ppm Ag NPs for 14 days. Group 2 rats were sacrificed on day 14 whereas group 3 was left for another 14 days of particle cessation followed by euthanasia on day 28. Functional assessment was done by liver enzyme assays, hydrogen peroxide activity, hepatic Bdnf expression, and P53 immunoreactivity. Hepatic tissue structural assessment was done via hematoxylin and eosin, periodic acid-Schiff as well as Masson's trichrome stains. The results revealed a significant elevation of Hydrogen peroxide in group 2 only compared to the control group. Hepatic Bdnf and liver enzymes were both insignificantly affected. Structural abnormalities and enhanced apoptosis in hepatic tissue were found 14 days after ceasing the nanoparticles. In conclusion: Structural and functional insults following Ag NPs oral administration continues after particle withdrawal, and interestingly they do not necessitate apparent reflection on liver enzyme assays.
Collapse
|
18
|
Villa-Jaimes GS, Aguilar-Mora FA, González-Ponce HA, Avelar-González FJ, Martínez Saldaña MC, Buist-Homan M, Moshage H. Biocomponents from Opuntia robusta and Opuntia streptacantha fruits protect against diclofenac-induced acute liver damage in vivo and in vitro. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.104960] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
19
|
Mora FAA, Musheshe N, Arroyave Ospina JC, Geng Y, Soto JM, Rodrigo JA, Alieva T, Buist-Homan M, Lezoualc'h F, Cheng X, Schmidt M, Moshage H. Metformin protects against diclofenac-induced toxicity in primary rat hepatocytes by preserving mitochondrial integrity via a pathway involving EPAC. Biomed Pharmacother 2021; 143:112072. [PMID: 34464747 DOI: 10.1016/j.biopha.2021.112072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/23/2021] [Accepted: 08/17/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE It has been shown that the antidiabetic drug metformin protects hepatocytes against toxicity by various stressors. Chronic or excessive consumption of diclofenac (DF) - a pain-relieving drug, leads to drug-induced liver injury via a mechanism involving mitochondrial damage and ultimately apoptotic death of hepatocytes. However, whether metformin protects against DF-induced toxicity is unknown. Recently, it was also shown that cAMP elevation is protective against DF-induced apoptotic death in hepatocytes, a protective effect primarily involving the downstream cAMP effector EPAC and preservation of mitochondrial function. This study therefore aimed at investigating whether metformin protects against DF-induced toxicity via cAMP-EPACs. EXPERIMENTAL APPROACH Primary rat hepatocytes were exposed to 400 µmol/L DF. CE3F4 or ESI-O5 were used as EPAC-1 or 2 inhibitors respectively. Apoptosis was measured by caspase-3 activity and necrosis by Sytox green staining. Seahorse X96 assay was used to determine mitochondrial function. Mitochondrial reactive oxygen species (ROS) production was measured using MitoSox, mitochondrial MnSOD expression was determined by immunostaining and mitochondrial morphology (fusion and fission ratio) by 3D refractive index imaging. KEY RESULTS Metformin (1 mmol/L) was protective against DF-induced apoptosis in hepatocytes. This protective effect was EPAC-dependent (mainly EPAC-2). Metformin restored mitochondrial morphology in an EPAC-independent manner. DF-induced mitochondrial dysfunction which was demonstrated by decreased oxygen consumption rate, an increased ROS production and a reduced MnSOD level, were all reversed by metformin in an EPAC-dependent manner. CONCLUSION AND IMPLICATIONS Metformin protects hepatocytes against DF-induced toxicity via cAMP-dependent EPAC-2.
Collapse
Affiliation(s)
- Fabio Alejandro Aguilar Mora
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Nshunge Musheshe
- Deptartment Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen University of Groningen, Groningen, The Netherlands.
| | - Johanna C Arroyave Ospina
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Yana Geng
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Juan M Soto
- Department of Optics and Faculty of Physical Sciences, Complutense University of Madrid, Spain.
| | - José A Rodrigo
- Department of Optics and Faculty of Physical Sciences, Complutense University of Madrid, Spain.
| | - Tatiana Alieva
- Department of Optics and Faculty of Physical Sciences, Complutense University of Madrid, Spain.
| | - Manon Buist-Homan
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Frank Lezoualc'h
- Inserm UMR-1048, Institut des Maladies Metaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France.
| | - Xiaodong Cheng
- Department of Integrative Biology & Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Martina Schmidt
- Deptartment Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen University of Groningen, Groningen, The Netherlands.
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
20
|
Wong HY, Langlotz M, Gan-Schreier H, Xu W, Staffer S, Tuma-Kellner S, Liebisch G, Merle U, Chamulitrat W. Constitutive oxidants from hepatocytes of male iPLA2β-null mice increases the externalization of phosphatidylethanolamine on plasma membrane. Free Radic Res 2021; 55:625-633. [PMID: 34696671 DOI: 10.1080/10715762.2021.1987426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
We have found that group VIA calcium-independent phospholipase A2 (iPLA2β) has specificity for hydrolysis of phosphatidylethanolamine (PE) in mouse livers. Phospholipids (PLs) are transported to plasma membrane and some PLs including PE are externalized to maintain membrane PL asymmetry. Here we demonstrated that hepatocytes of iPLA2β-null (KO) mice showed an increase in PE containing palmitate and oleate. We aimed to examine whether externalization of PE on the outer leaflets could be affected by iPLA2β deficiency and its modulation by reactive oxygen species (ROS) or apoptosis. As duramycin has high affinity to PE, we used duramycin conjugated with biotin (DLB) and streptavidin 488 as a probe for detection of externalized PE. Compared to WT, naïve KO hepatocytes showed an increase in both PE externalization and ROS generation. These events were observed in male but not in female KO mice. Hydrogen peroxide or menadione treatment enhanced PE externalization to the same extent for both male/female WT and KO hepatocytes. By indirect immunofluorescence, DLB-streptavidin staining was observed as small punctuated spots on the cell surface of menadione-treated KO hepatocytes. Unlike the reported PS externalization, CD95/FasL treatment did not lead to any increase in PE externalization, and iPLA2β deficiency-dependent PE externalization was also not correlated with apoptosis. Thus, constitutive (but not induced) ROS generation in iPLA2β-deficient hepatocytes leads to PE externalization observed only in male mice. Such PE externalization may imply detrimental effects regarding further oxidation of PE fatty acids and the binding with pathogens on the outer leaflets of hepatocyte plasma membrane.
Collapse
Affiliation(s)
| | - Monika Langlotz
- Center for Molecular Biology, University of Heidelberg, Heidelberg, Germany
| | | | - Weihong Xu
- Heidelberg University Hospital, Heidelberg, Germany
| | | | | | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Regensburg, Regensburg, Germany
| | - Uta Merle
- Heidelberg University Hospital, Heidelberg, Germany
| | | |
Collapse
|
21
|
Wang Y, Liu Y. Neutrophil-Induced Liver Injury and Interactions Between Neutrophils and Liver Sinusoidal Endothelial Cells. Inflammation 2021; 44:1246-1262. [PMID: 33649876 DOI: 10.1007/s10753-021-01442-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 01/29/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022]
Abstract
Neutrophils are the most abundant type of leukocytes with diverse functions in immune defense including production of reactive oxygen species, bacteriocidal proteins, neutrophil extracellular traps, and pro-inflammatory mediators. However, aberrant accumulation of neutrophils in host tissues and excessive release of bacteriocidal compounds can lead to unexpected injury to host organs. Neutrophil-mediated liver injury has been reported in various types of liver diseases including liver ischemia/reperfusion injury, nonalcoholic fatty liver disease, endotoxin-induced liver injury, alcoholic liver disease, and drug-induced liver injury. Yet the mechanisms of neutrophil-induced hepatotoxicity in different liver diseases are complicated. Current knowledge of these mechanisms are summarized in this review. In addition, a substantial body of evidence has emerged showing that liver sinusoidal endothelial cells (LSECs) participate in several key steps of neutrophil-mediated liver injury including neutrophil recruitment, adhesion, transmigration, and activation. This review also highlights the current understanding of the interactions between LSECs and neutrophils in liver injury. The future challenge is to explore new targets for selectively interfering neutrophil-induced liver injury without impairing host defense function against microbial infection. Further understanding the role of LSECs in neutrophil-induced hepatotoxicity would aid in developing more selective therapeutic approaches for liver disease.
Collapse
Affiliation(s)
- Yang Wang
- Department of Gastroenterology, Peking University People's Hospital, No.11, Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Yulan Liu
- Department of Gastroenterology, Peking University People's Hospital, No.11, Xizhimen South Street, Xicheng District, Beijing, 100044, China.
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, China.
| |
Collapse
|
22
|
Liu D, Du J, Sun J, Li M. Parathyroid hormone-related protein inhibits nitrogen-containing bisphosphonate-induced apoptosis of human periodontal ligament fibroblasts by activating MKP1 phosphatase. Bioengineered 2021; 12:1997-2006. [PMID: 34024253 PMCID: PMC8806876 DOI: 10.1080/21655979.2021.1928930] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Massive production of reactive oxygen species (ROS) in human periodontal ligament fibroblasts (HPdLFs) by nitrogen-containing bisphosphonates (BPs) is the main factor causing BP-related osteonecrosis of the jaw. Further, oxidative stress and apoptosis of fibroblasts induced by ROS are closely associated with the activation of MAPK. Parathyroid hormone-related protein (PTHrP) can block the activity of MAPK by regulating the levels of MAPK phosphatase 1 (MKP1). Therefore, it is speculated that PTHrP can inhibit the apoptosis of HPdLFs caused by nitrogen-containing BP via regulating the expression levels of MKP1. Herein, alendronate sodium salt trihydrate (nitrogen-containing BP, FOS) and HPdLFs were co-cultured for 24 h, 48 h, and 72 h, and the levels of ROS and apoptosis were determined, respectively. After 48 h co-culture, FOS significantly increased the levels of ROS and apoptosis, and high phosphorylation levels of p38, ERK1/2 and p66Shc were found in this study. However, the inhibitors of p38 and ERK1/2 significantly reduced the apoptosis of HPdLFs. Interestingly, PTHrP pre-treatment significantly reduced the phosphorylation levels of p38, ERK1/2, and p66Shc. More importantly, MKP1 inhibitor sanguinarine inhibited the dephosphorylation levels of p38, ERK1/2, and p66Shc caused by PTHrP. Altogether, PTHrP can inhibit nitrogen-containing BP-induced apoptosis of HPdLFs by activating MKP1 phosphatase.
Collapse
Affiliation(s)
- Di Liu
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan China.,Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan China
| | - Juan Du
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan China
| | - Jing Sun
- Department of Periodontology, Jinan Stomatological Hospital, Jinan China
| | - Minqi Li
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan China
| |
Collapse
|
23
|
Hu Z, Li L, Lan W, Wei X, Wen X, Wu P, Zhang X, Xi X, Li Y, Wu L, Li W, Liao X. Enrichment of Wee1/CDC2 and NF-κB Signaling Pathway Constituents Mutually Contributes to CDDP Resistance in Human Osteosarcoma. Cancer Res Treat 2021; 54:277-293. [PMID: 33971703 PMCID: PMC8756126 DOI: 10.4143/crt.2021.320] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/08/2021] [Indexed: 02/05/2023] Open
Abstract
Purpose Osteosarcoma (OS) universally exhibits heterogeneity and cisplatin (CDDP) resistance. Although the Wee1/CDC2 and NF-κB pathways were reported to show abnormal activation in some tumor cells with CDDP resistance, whether there is any concrete connection is currently unclear. We explored it in human OS cells. Materials and Methods Multiple OS cell lines were exposed to a Wee1 inhibitor (AZD1775) and CDDP to assess the half-maximal inhibitory concentration values. Western blot, coimmunoprecipitation, confocal immunofluorescence, cell cycle, and CCK-8 assays were performed to explore the connection between the Wee1/CDC2 and NF-κB pathways and their subsequent physiological contribution to CDDP resistance. Finally, CDDP-resistant PDX-OS xenograft models were established to confirm that AZD1775 restores the antitumor effects of CDDP. Results A sensitivity hierarchy of OS cells to CDDP and AZD1775 exists. In the highly CDDP-tolerant cell lines, Wee1 and RelA were physically crosslinked, which resulted in increased abundance of phosphorylated CDC2 (Y15) and RelA (S536) and consequent modulation of cell cycle progression, survival and proliferation. Wee1 inhibition restored the effects of CDDP on these processes in CDDP-resistant OS cells. In addition, animal experiments with CDDP-resistant PDX-OS cells showed that AZD1775 combined with CDDP not only restored CDDP efficacy but also amplified AZD1775 in inhibiting tumor growth and prolonged the median survival of the mice. Conclusion Simultaneous enrichment of molecules in the Wee1/CDC2 and NF-κB pathways and their consequent coactivation is a new molecular mechanism of CDDP resistance in OS cells. OS with this molecular signature may respond well to Wee1 inhibition as an alternative treatment strategy.
Collapse
Affiliation(s)
- Zhengbo Hu
- Derpartment of Orthopedics, Shaoguan First People's Hospital Affiliated to Southern Medical University, Guangdong, China
| | - Lugen Li
- Derpartment of Orthopedics, Shaoguan First People's Hospital Affiliated to Southern Medical University, Guangdong, China
| | - Wenxing Lan
- Derpartment of Orthopedics, Shaoguan First People's Hospital Affiliated to Southern Medical University, Guangdong, China
| | - Xiao Wei
- Derpartment of Orthopedics, Shaoguan First People's Hospital Affiliated to Southern Medical University, Guangdong, China
| | - Xiangyuan Wen
- Derpartment of Orthopedics, Shaoguan First People's Hospital Affiliated to Southern Medical University, Guangdong, China
| | - Penghuan Wu
- Derpartment of Orthopedics, Shaoguan First People's Hospital Affiliated to Southern Medical University, Guangdong, China.,Orthopedics Center, Zhujiang Hospital of Southern Medical Univerty, Guangzhou, China
| | - Xianliao Zhang
- Derpartment of Orthopedics, Shaoguan First People's Hospital Affiliated to Southern Medical University, Guangdong, China.,Orthopedics Center, Zhujiang Hospital of Southern Medical Univerty, Guangzhou, China
| | - Xinhua Xi
- Department of Orthopaedics, the Affiliated Yuebei People's Hospital of Shantou University Medical College, Shaoguan, Guangdong, China
| | - Yufa Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Department of Pathology, Guangdong provincal people's Hospital & Guangdong, Academy of Medical Sciences, Guangzhou, China
| | - Liqi Wu
- Derpartment of Orthopedics, Shaoguan First People's Hospital Affiliated to Southern Medical University, Guangdong, China
| | - Wenhu Li
- Derpartment of Orthopedics, Shaoguan First People's Hospital Affiliated to Southern Medical University, Guangdong, China
| | - Xiaohong Liao
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.,University of Chinese Academy of Social Sciences (Graduate School), Guangzhou, China
| |
Collapse
|
24
|
Aguilar Mora FA, Musheshe N, Oun A, Buist-Homan M, Lezoualc'h F, Cheng X, Schmidt M, Moshage H. Elevated cAMP Protects against Diclofenac-Induced Toxicity in Primary Rat Hepatocytes: A Protective Effect Mediated by the Exchange Protein Directly Activated by cAMP/cAMP-Regulated Guanine Nucleotide Exchange Factors. Mol Pharmacol 2021; 99:294-307. [PMID: 33574047 PMCID: PMC11033960 DOI: 10.1124/molpharm.120.000217] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic consumption of the nonsteroidal anti-inflammatory drug diclofenac may induce drug-induced liver injury (DILI). The mechanism of diclofenac-induced liver injury is partially elucidated and involves mitochondrial damage. Elevated cAMP protects hepatocytes against bile acid-induced injury. However, it is unknown whether cAMP protects against DILI and, if so, which downstream targets of cAMP are implicated in the protective mechanism, including the classic protein kinase A (PKA) pathway or alternative pathways like the exchange protein directly activated by cAMP (EPAC). The aim of this study was to investigate whether cAMP and/or its downstream targets protect against diclofenac-induced injury in hepatocytes. Rat hepatocytes were exposed to 400 µmol/l diclofenac. Apoptosis and necrosis were measured by caspase-3 activity assay and Sytox green staining, respectively. Mitochondrial membrane potential (MMP) was measured by JC-10 staining. mRNA and protein expression were assessed by quantitative polymerase chain reaction (qPCR) and Western blot, respectively. The cAMP-elevating agent 7β-acetoxy-8,13-epoxy-1α,6β,9α-trihydroxylabd-14-en-11-one (forskolin), the pan-phosphodiesterase inhibitor IBMX, and EPAC inhibitors 5,7-dibromo-6-fluoro-3,4-dihydro-2-methyl-1(2H)-quinoline carboxaldehyde (CE3F4) and ESI-O5 were used to assess the role of cAMP and its effectors, PKA or EPAC. Diclofenac exposure induced apoptotic cell death and loss of MMP in hepatocytes. Both forskolin and IBMX prevented diclofenac-induced apoptosis. EPAC inhibition but not PKA inhibition abolished the protective effect of forskolin and IBMX. Forskolin and IBMX preserved the MMP, whereas both EPAC inhibitors diminished this effect. Both EPAC1 and EPAC2 were expressed in hepatocytes and localized in mitochondria. cAMP elevation protects hepatocytes against diclofenac-induced cell death, a process primarily involving EPACs. The cAMP/EPAC pathway may be a novel target for treatment of DILI. SIGNIFICANCE STATEMENT: This study shows two main highlights. First, elevated cAMP levels protect against diclofenac-induced apoptosis in primary hepatocytes via maintenance of mitochondrial integrity. In addition, this study proposes the existence of mitochondrial cAMP-EPAC microdomains in rat hepatocytes, opening new avenues for targeted therapy in drug-induced liver injury (DILI). Both EPAC1 and EPAC2, but not protein kinase A, are responsible for this protective effect. Our findings present cAMP-EPAC as a potential target for the treatment of DILI and liver injury involving mitochondrial dysfunction.
Collapse
Affiliation(s)
- Fabio Alejandro Aguilar Mora
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| | - Nshunge Musheshe
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| | - Asmaa Oun
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| | - Manon Buist-Homan
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| | - Frank Lezoualc'h
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| | - Xiaodong Cheng
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| | - Martina Schmidt
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| | - Han Moshage
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| |
Collapse
|
25
|
González-Ponce HA, Martínez-Saldaña MC, Tepper PG, Quax WJ, Buist-Homan M, Faber KN, Moshage H. Betacyanins, major components in Opuntia red-purple fruits, protect against acetaminophen-induced acute liver failure. Food Res Int 2020; 137:109461. [PMID: 33233135 DOI: 10.1016/j.foodres.2020.109461] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/05/2020] [Accepted: 06/16/2020] [Indexed: 01/06/2023]
Abstract
Acetaminophen (APAP) misuse or overdose is the most important cause of drug-induced acute liver failure. Overdoses of acetaminophen induce oxidative stress and liver injury by the electrophilic metabolite N-acetyl-p-benzoquinone imine (NAPQI). Plant-based medicine has been used for centuries against diseases or intoxications due to their biological activities. The aim of this study was to evaluate the therapeutic value of Opuntia robusta and Opuntia streptacantha fruit extracts against acetaminophen-induced liver damage and to identify the major biocomponents on them. Opuntia fruit extracts were obtained by peeling and squeezing each specie, followed by lyophilization. HPLC was used to characterize the extracts. The effect of the extracts against acetaminophen-induced acute liver injury was evaluated both in vivo and in vitro using biochemical, molecular and histological determinations. The results showed that betacyanins are the main components in the analyzed Opuntia fruit extracts, with betanin as the highest concentration. Therapeutic treatments with Opuntia extracts reduced biochemical, molecular and histological markers of liver (in vivo) and hepatocyte (in vitro) injury. Opuntia extracts reduced the APAP-increased expression of the stress-related gene Gadd45b. Furthermore, Opuntia extracts exerted diverse effects on the antioxidant related genes Sod2, Gclc and Hmox1, independent of their ROS-scavenging ability. Therefore, betacyanins as betanin from Opuntia robusta and Opuntia streptacantha fruits are promising nutraceutical compounds against oxidative liver damage.
Collapse
Affiliation(s)
- Herson Antonio González-Ponce
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, the Netherlands.
| | | | - Pieter G Tepper
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Wim J Quax
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Manon Buist-Homan
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, the Netherlands; Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, the Netherlands.
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, the Netherlands; Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, the Netherlands.
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, the Netherlands; Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, the Netherlands.
| |
Collapse
|
26
|
Lin L, Gong H, Li R, Huang J, Cai M, Lan T, Huang W, Guo Y, Zhou Z, An Y, Chen Z, Liang L, Wang Y, Shuai X, Zhu K. Nanodrug with ROS and pH Dual-Sensitivity Ameliorates Liver Fibrosis via Multicellular Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903138. [PMID: 32274310 PMCID: PMC7140994 DOI: 10.1002/advs.201903138] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/23/2020] [Indexed: 05/09/2023]
Abstract
Liver fibrosis currently represents a global health problem without effective pharmacotherapeutic strategies. The clinical translation of polydatin, a promising natural anti-fibrotic drug candidate with broad anti-inflammatory and antioxidant capabilities, remains a major challenge due to its limited water solubility and tissue absorption. Herein, a polydatin-loaded micelle (PD-MC) based on reactive oxygen species (ROS) and pH dual-sensitive block polymer PEG-P(PBEM-co-DPA) is developed. The micelle exerts great potential in improving the biocompatibility of polydatin and shows highly efficient liver-targeted drug release in response to the fibrotic microenvironment. Both in vitro and in vivo studies demonstrate that PD-MC can significantly suppress inflammatory response and oxidative stress, reduce hepatocyte apoptosis, and avert activation of macrophages and hepatic stellate cells. More excitingly, the blank micelle itself promotes the hepatic ROS consumption at the pathologic site to provide anti-inflammatory benefits. These favorable therapeutic virtues of targeting multiple cell types endow PD-MC with remarkable efficacy with minimal side effects in liver fibrosis treatment. Thus, PD-MC holds great potential to push forward the clinical application of polydatin in pharmacotherapeutic approaches against liver fibrosis.
Collapse
Affiliation(s)
- Liteng Lin
- Laboratory of Interventional RadiologyDepartment of Minimally Invasive Interventional Radiology and Department of RadiologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510260China
| | - Hengye Gong
- PCFM Lab of Ministry of EducationSchool of Material Science and EngineeringSun Yat‐Sen UniversityGuangzhou510275China
| | - Rui Li
- Laboratory of Interventional RadiologyDepartment of Minimally Invasive Interventional Radiology and Department of RadiologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510260China
| | - Jingjun Huang
- Laboratory of Interventional RadiologyDepartment of Minimally Invasive Interventional Radiology and Department of RadiologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510260China
| | - Mingyue Cai
- Laboratory of Interventional RadiologyDepartment of Minimally Invasive Interventional Radiology and Department of RadiologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510260China
| | - Tian Lan
- School of PharmacyGuangdong Pharmaceutical UniversityGuangzhou510006China
| | - Wensou Huang
- Laboratory of Interventional RadiologyDepartment of Minimally Invasive Interventional Radiology and Department of RadiologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510260China
| | - Yongjian Guo
- Laboratory of Interventional RadiologyDepartment of Minimally Invasive Interventional Radiology and Department of RadiologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510260China
| | - Zhimei Zhou
- Laboratory of Interventional RadiologyDepartment of Minimally Invasive Interventional Radiology and Department of RadiologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510260China
| | - Yongcheng An
- Laboratory of Interventional RadiologyDepartment of Minimally Invasive Interventional Radiology and Department of RadiologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510260China
| | - Zhiwei Chen
- Laboratory of Interventional RadiologyDepartment of Minimally Invasive Interventional Radiology and Department of RadiologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510260China
| | - Licong Liang
- Laboratory of Interventional RadiologyDepartment of Minimally Invasive Interventional Radiology and Department of RadiologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510260China
| | - Yong Wang
- College of Chemistry and Materials ScienceJinan UniversityGuangzhou510632China
| | - Xintao Shuai
- PCFM Lab of Ministry of EducationSchool of Material Science and EngineeringSun Yat‐Sen UniversityGuangzhou510275China
| | - Kangshun Zhu
- Laboratory of Interventional RadiologyDepartment of Minimally Invasive Interventional Radiology and Department of RadiologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510260China
| |
Collapse
|
27
|
Ríos-Ocampo WA, Daemen T, Buist-Homan M, Faber KN, Navas MC, Moshage H. Hepatitis C virus core or NS3/4A protein expression preconditions hepatocytes against oxidative stress and endoplasmic reticulum stress. Redox Rep 2020; 24:17-26. [PMID: 30909829 PMCID: PMC6748607 DOI: 10.1080/13510002.2019.1596431] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objectives: The occurrence of oxidative stress and endoplasmic
reticulum (ER) stress in hepatitis C virus (HCV) infection has been demonstrated
and play an important role in liver injury. During viral infection, hepatocytes
must handle not only the replication of the virus, but also inflammatory signals
generating oxidative stress and damage. Although several mechanisms exist to
overcome cellular stress, little attention has been given to the adaptive
response of hepatocytes during exposure to multiple noxious triggers. Methods: In the present study, Huh-7 cells and hepatocytes
expressing HCV Core or NS3/4A proteins, both inducers of oxidative and ER
stress, were additionally challenged with the superoxide anion generator
menadione to mimic external oxidative stress. The production of reactive oxygen
species (ROS) as well as the response to oxidative stress and ER stress were
investigated. Results: We demonstrate that hepatocytes diminish oxidative stress
through a reduction in ROS production, ER-stress markers (HSPA5
[GRP78], sXBP1) and apoptosis (caspase-3 activity) despite
external oxidative stress. Interestingly, the level of the autophagy substrate
protein p62 was downregulated together with HCV Core degradation, suggesting
that hepatocytes can overcome excess oxidative stress through autophagic
degradation of one of the stressors, thereby increasing cell survival. Duscussion: In conclusion, hepatocytes exposed to direct and
indirect oxidative stress inducers are able to cope with cellular stress
associated with viral hepatitis and thus promote cell survival.
Collapse
Affiliation(s)
- W Alfredo Ríos-Ocampo
- a Department of Gastroenterology and Hepatology , University of Groningen, University Medical Center Groningen , Groningen , Netherlands.,b Department Medical Microbiology , University of Groningen, University Medical Center Groningen , Groningen , Netherlands.,c Grupo Gastrohepatología, Facultad de Medicina, Universidad de Antioquia , Medellin , Colombia
| | - Toos Daemen
- b Department Medical Microbiology , University of Groningen, University Medical Center Groningen , Groningen , Netherlands
| | - Manon Buist-Homan
- a Department of Gastroenterology and Hepatology , University of Groningen, University Medical Center Groningen , Groningen , Netherlands.,d Department of Laboratory Medicine , University of Groningen, University Medical Center Groningen , Groningen , Netherlands
| | - Klaas Nico Faber
- a Department of Gastroenterology and Hepatology , University of Groningen, University Medical Center Groningen , Groningen , Netherlands.,d Department of Laboratory Medicine , University of Groningen, University Medical Center Groningen , Groningen , Netherlands
| | - María-Cristina Navas
- c Grupo Gastrohepatología, Facultad de Medicina, Universidad de Antioquia , Medellin , Colombia
| | - Han Moshage
- a Department of Gastroenterology and Hepatology , University of Groningen, University Medical Center Groningen , Groningen , Netherlands.,d Department of Laboratory Medicine , University of Groningen, University Medical Center Groningen , Groningen , Netherlands
| |
Collapse
|
28
|
Cui Y, Zhang M, Leng C, Blokzijl T, Jansen BH, Dijkstra G, Faber KN. Pirfenidone Inhibits Cell Proliferation and Collagen I Production of Primary Human Intestinal Fibroblasts. Cells 2020; 9:cells9030775. [PMID: 32235767 PMCID: PMC7140656 DOI: 10.3390/cells9030775] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/16/2020] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
Intestinal fibrosis is a common complication of inflammatory bowel disease. So far, there is no safe and effective drug for intestinal fibrosis. Pirfenidone is an anti-fibrotic compound available for the treatment of idiopathic pulmonary fibrosis. Here, we explored the anti-proliferative and anti-fibrotic properties of pirfenidone on primary human intestinal fibroblasts (p-hIFs). p-hIFs were cultured in the absence and presence of pirfenidone. Cell proliferation was measured by a real-time cell analyzer (xCELLigence) and BrdU incorporation. Cell motility was monitored by live cell imaging. Cytotoxicity and cell viability were analyzed by Sytox green, Caspase-3 and Water Soluble Tetrazolium Salt-1 (WST-1) assays. Gene expression of fibrosis markers was determined by quantitative reverse transcription PCR (RT-qPCR). The mammalian target of rapamycin (mTOR) signaling was analyzed by Western blotting and type I collagen protein expression additionally by immunofluorescence microscopy. Pirfenidone dose-dependently inhibited p-hIF proliferation and motility, without inducing cell death. Pirfenidone suppressed mRNA levels of genes that contribute to extracellular matrix production, as well as basal and TGF-β1-induced collagen I protein production, which was associated with inhibition of the rapamycin-sensitive mTOR/p70S6K pathway in p-hIFs. Thus, pirfenidone inhibits the proliferation of intestinal fibroblasts and suppresses collagen I production through the TGF-β1/mTOR/p70S6K signaling pathway, which might be a novel and safe anti-fibrotic strategy to treat intestinal fibrosis.
Collapse
Affiliation(s)
- Yingying Cui
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (Y.C.); (M.Z.); (T.B.); (B.H.J.); (G.D.)
| | - Mengfan Zhang
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (Y.C.); (M.Z.); (T.B.); (B.H.J.); (G.D.)
| | - Changsen Leng
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
| | - Tjasso Blokzijl
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (Y.C.); (M.Z.); (T.B.); (B.H.J.); (G.D.)
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Bernadien H. Jansen
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (Y.C.); (M.Z.); (T.B.); (B.H.J.); (G.D.)
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (Y.C.); (M.Z.); (T.B.); (B.H.J.); (G.D.)
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (Y.C.); (M.Z.); (T.B.); (B.H.J.); (G.D.)
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
- Correspondence: ; Tel.: +31-50-3612364
| |
Collapse
|
29
|
Tang Y, Zhang J, Li J, Lei X, Xu D, Wang Y, Li C, Li X, Mao Y. Turnover of bile acids in liver, serum and caecal content by high-fat diet feeding affects hepatic steatosis in rats. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1293-1304. [DOI: 10.1016/j.bbalip.2019.05.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 05/22/2019] [Accepted: 05/30/2019] [Indexed: 12/12/2022]
|
30
|
Strowitzki MJ, Radhakrishnan P, Pavicevic S, Scheer J, Kimmer G, Ritter AS, Tuffs C, Volz C, Vondran F, Harnoss JM, Klose J, Schmidt T, Schneider M. High hepatic expression of PDK4 improves survival upon multimodal treatment of colorectal liver metastases. Br J Cancer 2019; 120:675-688. [PMID: 30808993 PMCID: PMC6461828 DOI: 10.1038/s41416-019-0406-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Patients with borderline resectable colorectal liver metastases (CRLM) frequently receive neoadjuvant chemotherapy (NC) to reduce tumour burden, thus making surgical resection feasible. Even though NC can induce severe liver injury, most studies investigating tissue-based prognostic markers focus on tumour tissue. Here, we assessed the prognostic significance of pyruvate-dehydrogenase-kinase isoenzyme 4 (PDK4) within liver tissue of patients undergoing surgical resection due to CRLM. METHODS Transcript levels of hypoxia-adaptive genes (such as PDK isoenzymes) were assessed in the tissue of healthy liver, corresponding CRLM, healthy colon mucosa and corresponding tumour. Uni- and multivariate analyses were performed. Responses to chemotherapy upon up- or down-regulation of PDK4 were studied in vitro. RESULTS PDK4 expression within healthy liver tissue was associated with increased overall survival and liver function following surgical resection of CRLM. This association was enhanced in patients with NC. PDK4 expression in CRLM tissue did not correlate with overall survival. Up-regulation of PDK4 increased the resistance of hepatocytes and colon cancer cells against chemotherapy-induced toxicity, whereas knockdown of PDK4 enhanced chemotherapy-associated cell damage. CONCLUSION Our findings suggest that up-regulated PDK4 expression reduces hepatic chemotherapy-induced oxidative stress and is associated with improved postoperative liver function in patients undergoing multimodal treatment and resection of CRLM.
Collapse
Affiliation(s)
- Moritz J Strowitzki
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Praveen Radhakrishnan
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Sandra Pavicevic
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Jana Scheer
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Gwendolyn Kimmer
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Alina S Ritter
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Christopher Tuffs
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Claudia Volz
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Florian Vondran
- Regenerative Medicine and Experimental Surgery (ReMediES), Department of General, Visceral and Transplantation Surgery, Hannover Medical School, Hannover, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Hannover, Germany
| | - Jonathan M Harnoss
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Johannes Klose
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Thomas Schmidt
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
31
|
Shen J, Liu M, Xu J, Sun B, Xu H, Zhang W. ARL15 overexpression attenuates high glucose-induced impairment of insulin signaling and oxidative stress in human umbilical vein endothelial cells. Life Sci 2019; 220:127-135. [DOI: 10.1016/j.lfs.2019.01.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/10/2019] [Accepted: 01/18/2019] [Indexed: 02/07/2023]
|
32
|
Liu L, Zhao Y, Lin Y, Zhang R, Luo S, Ye P, Luo M. The antagonistic effect of tamoxifen against d-galactosamine/lipopolysaccharide-induced acute liver failure is associated with reactivation of hepatic nuclear factor-κB. Immunopharmacol Immunotoxicol 2019; 41:192-198. [PMID: 30721100 DOI: 10.1080/08923973.2019.1569044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Context: Tamoxifen (TAM) ameliorates D-galactosamine/lipopolysaccharide (Gal/LPS)-induced acute liver failure (ALF) through its antioxidative effect; thus, this study was designed to determine whether the effectiveness of TAM is related to nuclear factor-κB (NF-κB) reactivation. Materials and methods: Experimental mice were injected with TAM once daily for 3 consecutive days intraperitoneally (i.p). Twelve hours after pretreatment, Gal/LPS was given to mice (i.p) for ALF induction. In the positive control group, N-acetylcysteine (NAC) was administered immediately after ALF establishment. Except for survival observation, other animals were sacrificed 7 h after Gal/LPS treatment. Survival and hepatic failure were evaluated. For the oxidation assessment, the reduced/oxidized glutathione (GSH/GSSG) ratio and hepatic superoxide dismutase (SOD) activity were analyzed using both colorimetry and Western blotting. Lastly, hepatic NF-κB activation was measured through Western blot analysis of p65 and IκBα. Results: The results indicated that pretreatment with TAM dramatically attenuated Gal/LPS-induced ALF, as demonstrated by improved survival (70%), decreased transaminase levels, and reversed histopathological manifestation. In addition, the hepatic GSH/GSSG ratio and SOD activity were decreased in the ALF model. However, to some degree, TAM and NAC effectively prevented this undesirable phenomenon in contrast to the ALF model. Western blotting revealed that compared with mice in the ALF model group, mice treated with TAM or NAC showed reactivation of hepatic NF-κB. Conclusions: Taking the results together with those of other studies, we conclude that TAM may attenuate Gal/LPS-induced ALF by antagonizing oxidative stress through NF-κB reactivation.
Collapse
Affiliation(s)
- Liping Liu
- a The Affiliated Ganzhou Hospital of Nanchang University , Ganzhou , Jiangxi , China
| | - Yongsheng Zhao
- b The People's Hospital of Xinfeng County , Ganzhou , Jiangxi , China
| | - Yan Lin
- a The Affiliated Ganzhou Hospital of Nanchang University , Ganzhou , Jiangxi , China
| | - Rongshan Zhang
- a The Affiliated Ganzhou Hospital of Nanchang University , Ganzhou , Jiangxi , China
| | - Shi Luo
- a The Affiliated Ganzhou Hospital of Nanchang University , Ganzhou , Jiangxi , China
| | - Ping Ye
- a The Affiliated Ganzhou Hospital of Nanchang University , Ganzhou , Jiangxi , China
| | - Mansheng Luo
- a The Affiliated Ganzhou Hospital of Nanchang University , Ganzhou , Jiangxi , China
| |
Collapse
|
33
|
Pascotini MET, Flores DAE, Kegler MA, Konzen MV, Fornari MAL, Arend MJ, Gabbi MP, Gobo MLA, Bochi DGV, Prado DALC, de Carvalho DLM, Duarte DMM, da Cruz DIBM, Moresco DRN, dos Santos DARS, Royes DLFF, Fighera DMR. Brain-Derived Neurotrophic Factor Levels are Lower in Chronic Stroke Patients: A Relation with Manganese-dependent Superoxide Dismutase ALA16VAL Single Nucleotide Polymorphism through Tumor Necrosis Factor-α and Caspases Pathways. J Stroke Cerebrovasc Dis 2018; 27:3020-3029. [DOI: 10.1016/j.jstrokecerebrovasdis.2018.06.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/03/2018] [Accepted: 06/24/2018] [Indexed: 12/19/2022] Open
|
34
|
Su H, Li Y, Hu D, Xie L, Ke H, Zheng X, Chen W. Procyanidin B2 ameliorates free fatty acids-induced hepatic steatosis through regulating TFEB-mediated lysosomal pathway and redox state. Free Radic Biol Med 2018; 126:269-286. [PMID: 30142454 DOI: 10.1016/j.freeradbiomed.2018.08.024] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 08/19/2018] [Accepted: 08/20/2018] [Indexed: 12/11/2022]
Abstract
Procyanidin B2, a naturally occurring phenolic compound, has been reported to exert multiple beneficial functions. However, the effect of procyanidin B2 on free fatty acids (FFAs)-induced hepatic steatosis remains obscure. The present study is therefore aimed to elucidate the protective effect of procyanidin B2 against hepatic steatosis and its underlying mechanism. Herein, we reported that procyanidin B2 attenuated FFAs-induced lipid accumulation and its associated oxidative stress by scavenging excessive ROS and superoxide anion radicals, blocking loss of mitochondrial membrane potential, restoring glutathione content, and increasing activity of antioxidant enzymes (GPx, SOD and CAT) in hepatocytes. Procyanidin B2 mechanistically promoted lipid degradation via modulation of transcription factor EB (TFEB), a master regulator of lysosomal pathway. Molecular docking analysis indicated a possible ligand-binding position of procyanidin B2 with TFEB. In addition, administration of procyanidin B2 resulted in a significant reduction of hepatic fat accumulation in high-fat diet (HFD)-induced obese mice, and also ameliorated HFD-induced metabolic abnormalities, including hyperlipidemia and hyperglycemia. It was confirmed that procyanidin B2 prevented HFD-induced hepatic fat accumulation through down-regulating lipogenesis-related gene expressions (PPARγ, C/EBPα and SREBP-1c), inhibiting pro-inflammatory cytokines production (IL-6 and TNF-α) and increasing antioxidant enzymes activity (GPx, SOD and CAT). Moreover, hepatic fatty acids analysis indicated that procyanidin B2 caused a significant increase in the levels of palmitic acid, oleic acid and linoleic acid. Intriguingly, procyanidin B2 restored the decreased nuclear TFEB expression in HFD-induced liver steatosis and up-regulated its target genes involved in lysosomal pathway (Lamp1, Mcoln, Uvrag), which suggested a previously unrecognized mechanism of procyanidin B2 on ameliorating HFD-induced hepatic steatosis. Taken together, our results demonstrated that procyanidin B2 attenuated FFAs-induced hepatic steatosis through regulating TFEB-mediated lysosomal pathway and redox state, which had important implications that modulation of TFEB might be a potential therapeutic strategy for hepatic steatosis and procyanidin B2 could represent a promising novel agent in the prevention and treatment of non-alcoholic fatty liver disease (NAFLD).
Collapse
Affiliation(s)
- Hongming Su
- Department of Food Science and Nutrition, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China
| | - Yuting Li
- Department of Food Science and Nutrition, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China
| | - Dongwen Hu
- Department of Food Science and Nutrition, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China
| | - Lianghua Xie
- Department of Food Science and Nutrition, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China
| | - Huihui Ke
- Department of Food Science and Nutrition, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China
| | - Xiaodong Zheng
- Department of Food Science and Nutrition, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China
| | - Wei Chen
- Department of Food Science and Nutrition, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
35
|
Wu Z, Wang Y, Meng X, Wang X, Li Z, Qian S, Wei Y, Shu L, Ding Y, Wang P, Peng Y. Total C-21 steroidal glycosides, isolated from the root tuber of Cynanchum auriculatum Royle ex Wight, attenuate hydrogen peroxide-induced oxidative injury and inflammation in L02 cells. Int J Mol Med 2018; 42:3157-3170. [PMID: 30272289 PMCID: PMC6202073 DOI: 10.3892/ijmm.2018.3896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 09/20/2018] [Indexed: 12/22/2022] Open
Abstract
Oxidative stress plays an important role in the pathology of liver disorders. Total C-21 steroidal glycosides (TCSGs), isolated from the root tuber of Cynanchum auriculatum Royle ex Wight, have been reported to exert numerous effects, including liver protective and antioxidant effects. In order to investigate the potential mechanisms underlying the protective effects of TCSGs on liver function, the present study used the human normal liver cell line, L02, to evaluate the effects of TCSGs on hydrogen peroxide (H2O2)-induced oxidative injury and inflammatory responses. The L02 cells were pretreated with various concentrations of TCSGs, followed by exposure to 1.5 mM H2O2. Cell viability was determined by a 3-(4,5-dimethylthiazol-2-yl)-2,5-di-phenyltetrazolium bromide (MTT) assay. The levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), lactate dehydrogenase (LDH) and nitric oxide (NO) were measured using colorimetric assays. The activities of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px) and the production of malondialdehyde (MDA) were also determined. Intracellular reactive oxygen species (ROS) levels were detected using a fluorescent probe. H2O2-induced oxidative toxicity was attenuated following treatment with TCSGs, as indicated by the increase in cell viability, the decreased levels of ALT, AST, LDH, NO, MDA and ROS, and the increased activities of SOD, CAT and GSH-Px. To further explore the possible mechanisms of action of TCSGs, the nuclear factor erythroid 2-related factor 2 (Nrf2) and nuclear factor-κB (NF)-κB pathways were examined. The results revealed that treatment with TCSGs markedly induced Nrf2 nuclear translocation and upregulated the expression of heme oxygenase-1 (HO-1) in the L02 cells damaged by H2O2. In addition, pretreatment with TCSGs inhibited the NF-κB signaling pathway by blocking the degradation of the inhibitor of nuclear factor κBα (IκBα), thereby reducing the expression and nuclear translocation of NF-κB, as well as reducing the expression of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2). On the whole, the findings of this study demonstrate that TCSGs can protect L02 cells against H2O2-induced oxidative toxicity and inflammatory injury by increasing the expression of Nrf2 and HO-1, mediated by the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zhenhui Wu
- Department of Pharmacology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yingyu Wang
- Department of Pharmacology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Xian Meng
- Department of Pharmacology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Xinjie Wang
- State Key Laboratory of Natural Medicines, Research Department of Pharmacognosy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P.R. China
| | - Zhenlin Li
- Department of Pharmacology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Shihui Qian
- Department of Pharmacology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yingjie Wei
- Department of Pharmacology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Luan Shu
- Department of Pharmacology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yongfang Ding
- Department of Pharmacology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Peijuan Wang
- Department of Pharmacology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yunru Peng
- Department of Pharmacology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
36
|
Moirangthem A, Patel T. Mesenchymal stem cell derived extracellular vesicles: a promising new therapeutic approach for hepatic injury. ACTA ACUST UNITED AC 2018; 1. [PMID: 30211398 DOI: 10.21037/biotarget.2017.08.04] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Anuradha Moirangthem
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Tushar Patel
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
37
|
Ismail N, Abdel–Mottaleb Y, Eissa Ahmed AA, El-Maraghy NN. Novel combination of thymoquinone and resveratrol enhances anticancer effect on hepatocellular carcinoma cell line. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2018. [DOI: 10.1016/j.fjps.2017.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
38
|
Han B, Shin HJ, Bak IS, Bak Y, Jeong YL, Kwon T, Park YH, Sun HN, Kim CH, Yu DY. Peroxiredoxin I is important for cancer-cell survival in Ras-induced hepatic tumorigenesis. Oncotarget 2018; 7:68044-68056. [PMID: 27517622 PMCID: PMC5356538 DOI: 10.18632/oncotarget.11172] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 07/27/2016] [Indexed: 02/06/2023] Open
Abstract
Peroxiredoxin I (Prx I), an antioxidant enzyme, has multiple functions in human cancer. However, the role of Prx I in hepatic tumorigenesis has not been characterized. Here we investigated the relevance and underlying mechanism of Prx I in hepatic tumorigenesis. Prx I increased in tumors of hepatocellular carcinoma (HCC) patients that aligned with overexpression of oncogenic H-ras. Prx I also increased in H-rasG12V transfected HCC cells and liver tumors of H-rasG12V transgenic (Tg) mice, indicating that Prx I may be involved in Ras-induced hepatic tumorigenesis. When Prx I was knocked down or deleted in HCC-H-rasG12V cells or H-rasG12V Tg mice, cell colony or tumor formation was significantly reduced that was associated with downregulation of pERK pathway as well as increased intracellular reactive oxygen species (ROS) induced DNA damage and cell death. Overexpressing Prx I markedly increased Ras downstream pERK/FoxM1/Nrf2 signaling pathway and inhibited oxidative damage in HCC cells and H-rasG12V Tg mice. In this study, we found Nrf2 was transcriptionally activated by FoxM1, and Prx I was activated by the H-rasG12V/pERK/FoxM1/Nrf2 pathway and suppressed ROS-induced hepatic cancer-cell death along with formation of a positive feedback loop with Ras/ERK/FoxM1/Nrf2 to promote hepatic tumorigenesis.
Collapse
Affiliation(s)
- Bing Han
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, Korea.,Department of Biology, Chungnam National University, Daejeon, 305-764, Korea
| | - Hye-Jun Shin
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, Korea
| | - In Seon Bak
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, Korea.,Department of Toxicology Evaluation, Graduate School of Preclinical Laboratory Science, Konyang University, Daejeon, 363-700, Korea
| | - Yesol Bak
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, Korea.,Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, 143-701, Korea
| | - Ye-Lin Jeong
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, Korea.,Department of Animal Biosystem Sciences, Chungnam National University, Daejeon, 305-764, Korea
| | - Taeho Kwon
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, Korea
| | - Young-Ho Park
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, Korea
| | - Hu-Nan Sun
- College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, 305-764, Korea
| | - Dae-Yeul Yu
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, Korea
| |
Collapse
|
39
|
González-Ponce HA, Rincón-Sánchez AR, Jaramillo-Juárez F, Moshage H. Natural Dietary Pigments: Potential Mediators against Hepatic Damage Induced by Over-The-Counter Non-Steroidal Anti-Inflammatory and Analgesic Drugs. Nutrients 2018; 10:E117. [PMID: 29364842 PMCID: PMC5852693 DOI: 10.3390/nu10020117] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 12/14/2017] [Accepted: 12/14/2017] [Indexed: 12/19/2022] Open
Abstract
Over-the-counter (OTC) analgesics are among the most widely prescribed and purchased drugs around the world. Most analgesics, including non-steroidal anti-inflammatory drugs (NSAIDs) and acetaminophen, are metabolized in the liver. The hepatocytes are responsible for drug metabolism and detoxification. Cytochrome P450 enzymes are phase I enzymes expressed mainly in hepatocytes and they account for ≈75% of the metabolism of clinically used drugs and other xenobiotics. These metabolic reactions eliminate potentially toxic compounds but, paradoxically, also result in the generation of toxic or carcinogenic metabolites. Cumulative or overdoses of OTC analgesic drugs can induce acute liver failure (ALF) either directly or indirectly after their biotransformation. ALF is the result of massive death of hepatocytes induced by oxidative stress. There is an increased interest in the use of natural dietary products as nutritional supplements and/or medications to prevent or cure many diseases. The therapeutic activity of natural products may be associated with their antioxidant capacity, although additional mechanisms may also play a role (e.g., anti-inflammatory actions). Dietary antioxidants such as flavonoids, betalains and carotenoids play a preventive role against OTC analgesics-induced ALF. In this review, we will summarize the pathobiology of OTC analgesic-induced ALF and the use of natural pigments in its prevention and therapy.
Collapse
Affiliation(s)
- Herson Antonio González-Ponce
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713GZ Groningen, The Netherlands.
| | - Ana Rosa Rincón-Sánchez
- Department of Molecular Biology and Genomics, University Center of Health Sciences, Universidad de Guadalajara, Guadalajara 44340, Mexico.
| | - Fernando Jaramillo-Juárez
- Department of Physiology and Pharmacology, Basic Science Center, Universidad Autónoma de Aguascalientes, Aguascalientes 20131, Mexico.
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713GZ Groningen, The Netherlands.
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9713GZ Groningen, The Netherlands.
| |
Collapse
|
40
|
Zhang H, Zhou X, Wong MHY, Man KY, Pin WK, Yeung JHK, Kwan YW, Leung GPH, Hoi PM, Lee SMY, Chan CO, Mok DKW, Yu PHF, Chan SW. Sichuan pepper attenuates H 2O 2-induced apoptosis via antioxidant activity and up-regulating heme oxygenase-1 gene expression in primary rat hepatocytes. J Food Biochem 2017; 41:e12403. [DOI: 10.1111/jfbc.12403] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Affiliation(s)
- Huan Zhang
- Food Safety and Technology Research Centre, Department of Applied Biology and Chemical Technology; The Hong Kong Polytechnic University; Hong Kong China
| | - Xuelin Zhou
- School of Biomedical Sciences, Faculty of Medicine; The Chinese University of Hong Kong; Hong Kong China
| | - Marcus Ho-Yin Wong
- Food Safety and Technology Research Centre, Department of Applied Biology and Chemical Technology; The Hong Kong Polytechnic University; Hong Kong China
| | - Ka-Yi Man
- Food Safety and Technology Research Centre, Department of Applied Biology and Chemical Technology; The Hong Kong Polytechnic University; Hong Kong China
| | - Wing-Kwan Pin
- Food Safety and Technology Research Centre, Department of Applied Biology and Chemical Technology; The Hong Kong Polytechnic University; Hong Kong China
| | - John Hok-Keung Yeung
- School of Biomedical Sciences, Faculty of Medicine; The Chinese University of Hong Kong; Hong Kong China
| | - Yiu-Wa Kwan
- School of Biomedical Sciences, Faculty of Medicine; The Chinese University of Hong Kong; Hong Kong China
| | - George Pak-Heng Leung
- Department of Pharmacology and Pharmacy, Faculty of Medicine; The University of Hong Kong; Hong Kong China
| | - Pui-Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences; University of Macau; Avenue Padre Tomás Pereira S.J., Taipa Macau China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences; University of Macau; Avenue Padre Tomás Pereira S.J., Taipa Macau China
| | - Chi-On Chan
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Department of Applied Biology and Chemical Technology; The Hong Kong Polytechnic University; Hong Kong China
| | - Daniel Kam-Wah Mok
- Food Safety and Technology Research Centre, Department of Applied Biology and Chemical Technology; The Hong Kong Polytechnic University; Hong Kong China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Department of Applied Biology and Chemical Technology; The Hong Kong Polytechnic University; Hong Kong China
| | - Peter Hoi-Fu Yu
- Department of Food and Health Sciences, Faculty of Science and Technology; Technological and Higher Education Institute of Hong Kong; Hong Kong China
| | - Shun-Wan Chan
- Food Safety and Technology Research Centre, Department of Applied Biology and Chemical Technology; The Hong Kong Polytechnic University; Hong Kong China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Department of Applied Biology and Chemical Technology; The Hong Kong Polytechnic University; Hong Kong China
- Department of Food and Health Sciences, Faculty of Science and Technology; Technological and Higher Education Institute of Hong Kong; Hong Kong China
| |
Collapse
|
41
|
Kučera O, Endlicher R, Rychtrmoc D, Lotková H, Sobotka O, Červinková Z. Acetaminophen toxicity in rat and mouse hepatocytes in vitro. Drug Chem Toxicol 2017; 40:448-456. [PMID: 27960556 DOI: 10.1080/01480545.2016.1255953] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 10/30/2016] [Indexed: 12/17/2022]
Abstract
CONTEXT Acetaminophen (APAP) hepatotoxicity is often studied in primary cultures of hepatocytes of various species, but there are only few works comparing interspecies differences in susceptibility of hepatocytes to APAP in vitro. OBJECTIVES The aim of our work was to compare hepatotoxicity of APAP in rat and mouse hepatocytes in primary cultures. MATERIALS AND METHODS Hepatocytes isolated from male Wistar rats and C57Bl/6J mice were exposed to APAP for up to 24 h. We determined lactate dehydrogenase (LDH) activity in culture medium, activity of cellular dehydrogenases (WST-1) and activity of caspases 3 in cell lysate as markers of cell damage/death. We assessed content of intracellular reduced glutathione, production of reactive oxygen species (ROS) and malondialdehyde (MDA). Respiration of digitonin-permeabilized hepatocytes was measured by high resolution respirometry and mitochondrial membrane potential (MMP) was visualized (JC-1). RESULTS APAP from concentrations of 2.5 and 0.75 mmol/L induced a decrease in viability of rat (p < 0.001) and mouse (p < 0.001) hepatocytes (WST-1), respectively. In contrast to rat hepatocytes, there was no activation of caspase-3 in mouse hepatocytes after APAP treatment. Earlier damage to plasma membrane and faster depletion of reduced glutathione were detected in mouse hepatocytes. Mouse hepatocytes showed increased glutamate + malate-driven respiration in state 4 and higher susceptibility of the outer mitochondrial membrane (OMM) to APAP-induced injury. CONCLUSION APAP displayed dose-dependent toxicity in hepatocytes of both species. Mouse hepatocytes in primary culture however had approximately three-fold higher susceptibility to the toxic effect of APAP when compared to rat hepatocytes.
Collapse
Affiliation(s)
| | - René Endlicher
- b Department of Anatomy , Charles University in Prague, Faculty of Medicine in Hradec Králové , Hradec Králové , Czech Republic
| | | | | | | | | |
Collapse
|
42
|
Xiong W, Zhang W, Yuan W, Du H, Ming K, Yao F, Bai J, Chen Y, Liu J, Wang D, Hu Y, Wu Y. Phosphorylation of Icariin Can Alleviate the Oxidative Stress Caused by the Duck Hepatitis Virus A through Mitogen-Activated Protein Kinases Signaling Pathways. Front Microbiol 2017; 8:1850. [PMID: 29018425 PMCID: PMC5622922 DOI: 10.3389/fmicb.2017.01850] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/11/2017] [Indexed: 12/02/2022] Open
Abstract
The duck virus hepatitis (DVH) caused by the duck hepatitis virus A (DHAV) has produced extensive economic losses to the duck industry. The currently licensed commercial vaccine has shown some defects and does not completely prevent the DVH. Accordingly, a new alternative treatment for this disease is urgently needed. Previous studies have shown that icariin (ICA) and its phosphorylated derivative (pICA) possessed good anti-DHAV effects through direct and indirect antiviral pathways, such as antioxidative stress. But the antioxidant activity showed some differences between ICA and pICA. The aim of this study is to prove that ICA and pICA attenuate oxidative stress caused by DHAV in vitro and in vivo, and to investigate their mechanism of action to explain their differences in antioxidant activities. In vivo, the dynamic deaths, oxidative evaluation indexes and hepatic pathological change scores were detected. When was added the hinokitiol which showed the pro-oxidative effect as an intervention method, pICA still possessed more treatment effect than ICA. The strong correlation between mortality and oxidative stress proves that ICA and pICA alleviate oxidative stress caused by DHAV. This was also demonstrated by the addition of hydrogen peroxide (H2O2) as an intervention method in vitro. pICA can be more effective than ICA to improve duck embryonic hepatocytes (DEHs) viability and reduce the virulence of DHAV. The strong correlation between TCID50 and oxidative stress demonstrates that ICA and pICA can achieve anti-DHAV effects by inhibiting oxidative stress. In addition, the superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) of ICA and pICA showed significant difference. pICA could significantly inhibit the phosphorylation of p38, extra cellular signal regulated Kinase (ERK 1/2) and c-Jun N-terminal kinase (JNK), which were related to mitogen-activated protein kinases (MAPKs) signaling pathways. Ultimately, compared to ICA, pICA exhibited more antioxidant activity that could regulate oxidative stress-related indicators, and inhibited the phosphorylation of MAPKs signaling pathway.
Collapse
Affiliation(s)
- Wen Xiong
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Wei Zhang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Wenjuan Yuan
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hongxu Du
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ke Ming
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Fangke Yao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jingying Bai
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yun Chen
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jiaguo Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yuanliang Hu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yi Wu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
43
|
Nguyen NU, Stamper BD. Polyphenols reported to shift APAP-induced changes in MAPK signaling and toxicity outcomes. Chem Biol Interact 2017; 277:129-136. [PMID: 28918124 DOI: 10.1016/j.cbi.2017.09.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/09/2017] [Accepted: 09/11/2017] [Indexed: 12/20/2022]
Abstract
Due to its widespread availability, acetaminophen (APAP) is the leading cause for drug-induced liver injury in many countries including United States and United Kingdom. When used as recommended, APAP is relatively safe. However, in overdose cases, increased metabolism of APAP to N-acetyl-para-benzoquinoneimine (NAPQI), a reactive metabolite, leads to glutathione (GSH) depletion, oxidative stress, and cellular injury. Throughout this process, a variety of factors play important roles in propagating toxicity, including c-Jun N-terminal kinase (JNK), a member of the mitogen-activated protein kinase (MAPK) family. Because of its involvement in multiple cellular processes, biomarkers associated with MAPK signaling have generated interest as a mechanistic target for protecting against APAP-induced liver injury and hepatocellular injury, in general. This review summarizes mechanistic details by which natural products, specifically those containing polyphenolic moieties, are capable of attenuating APAP-induced toxicity, at least in part through an ability to modulate MAPKs. These compounds include carnosic acid, chlorogenic acid, davallialactone, extracts from Hibiscus sabdariffa, quercetin-based compounds, and resveratrol. Despite variations in the experimental designs across these studies, common pathways and biomarkers were implicated in cytoprotection when polyphenolic compounds were given with APAP, such as enhanced antioxidant gene expression and reversal of APAP-induced changes in oxidative stress markers and MAPK signaling. Overall, an emphasis should be placed on method standardization for future studies if we are to gain a more in-depth understanding of how polyphenolic moieties contribute to cytoprotection during an APAP overdose event.
Collapse
Affiliation(s)
- Ngoc Uy Nguyen
- Pacific University College of Arts & Sciences, 2043 College Way UC #4882, Forest Grove, OR 97116, USA.
| | - Brendan David Stamper
- Pacific University School of Pharmacy, 222 S.E. 8th Avenue #451, Hillsboro, OR 97123, USA.
| |
Collapse
|
44
|
Atanasovska B, Rensen SS, van der Sijde MR, Marsman G, Kumar V, Jonkers I, Withoff S, Shiri-Sverdlov R, Greve JWM, Faber KN, Moshage H, Wijmenga C, van de Sluis B, Hofker MH, Fu J. A liver-specific long noncoding RNA with a role in cell viability is elevated in human nonalcoholic steatohepatitis. Hepatology 2017; 66:794-808. [PMID: 28073183 DOI: 10.1002/hep.29034] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 12/12/2016] [Accepted: 01/06/2017] [Indexed: 12/28/2022]
Abstract
UNLABELLED Hepatocyte apoptosis in nonalcoholic steatohepatitis (NASH) can lead to fibrosis and cirrhosis, which permanently damage the liver. Understanding the regulation of hepatocyte apoptosis is therefore important to identify therapeutic targets that may prevent the progression of NASH to fibrosis. Recently, increasing evidence has shown that long noncoding (lnc) RNAs are involved in various biological processes and that their dysregulation underlies a number of complex human diseases. By performing gene expression profiling of 4,383 lncRNAs in 82 liver samples from individuals with NASH (n = 48), simple steatosis but no NASH (n = 11), and healthy controls (n = 23), we discovered a liver-specific lncRNA (RP11-484N16.1) on chromosome 18 that showed significantly elevated expression in the liver tissue of NASH patients. This lncRNA, which we named lnc18q22.2 based on its chromosomal location, correlated with NASH grade (r = 0.51, P = 8.11 × 10-7 ), lobular inflammation (r = 0.49, P = 2.35 × 10-6 ), and nonalcoholic fatty liver disease activity score (r = 0.48, P = 4.69 × 10-6 ). The association of lnc18q22.2 to liver steatosis and steatohepatitis was replicated in 44 independent liver biopsies (r = 0.47, P = 0.0013). We provided a genetic structure of lnc18q22.2 showing an extended exon 2 in liver. Knockdown of lnc18q22.2 in four different hepatocyte cell lines resulted in severe phenotypes ranging from reduced cell growth to lethality. This observation was consistent with pathway analyses of genes coexpressed with lnc18q22.2 in human liver or affected by lnc18q22.2 knockdown. CONCLUSION We identified an lncRNA that can play an important regulatory role in liver function and provide new insights into the regulation of hepatocyte viability in NASH. (Hepatology 2017;66:794-808).
Collapse
Affiliation(s)
- Biljana Atanasovska
- Department of Pediatrics, Molecular Genetics, University Medical Center Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sander S Rensen
- Department of Surgery, University Hospital Maastricht, and Nutrition and Toxicology Research Institute, University of Maastricht, Maastricht, The Netherlands
| | - Marijke R van der Sijde
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Glenn Marsman
- Department of Pediatrics, Molecular Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - Vinod Kumar
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Iris Jonkers
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sebo Withoff
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ronit Shiri-Sverdlov
- Departments of Molecular Genetics, Molecular Cell Biology, and Population Genetics, Nutrition and Toxicology Research Institute, University of Maastricht, Maastricht, The Netherlands
| | - Jan Willem M Greve
- Department of Surgery, Zuyderland Medical Center, Heerlen, The Netherlands
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University of Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, Center for Liver, Digestive, and Metabolic Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University of Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, Center for Liver, Digestive, and Metabolic Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bart van de Sluis
- Department of Pediatrics, Molecular Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - Marten H Hofker
- Department of Pediatrics, Molecular Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - Jingyuan Fu
- Department of Pediatrics, Molecular Genetics, University Medical Center Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
45
|
Jiang WP, Huang SS, Matsuda Y, Saito H, Uramaru N, Ho HY, Wu JB, Huang GJ. Protective Effects of Tormentic Acid, a Major Component of Suspension Cultures of Eriobotrya japonica Cells, on Acetaminophen-Induced Hepatotoxicity in Mice. Molecules 2017; 22:molecules22050830. [PMID: 28524081 PMCID: PMC6154347 DOI: 10.3390/molecules22050830] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 05/11/2017] [Accepted: 05/15/2017] [Indexed: 12/12/2022] Open
Abstract
An acetaminophen (APAP) overdose can cause hepatotoxicity and lead to fatal liver damage. The hepatoprotective effects of tormentic acid (TA) on acetaminophen (APAP)-induced liver damage were investigated in mice. TA was intraperitoneally (i.p.) administered for six days prior to APAP administration. Pretreatment with TA prevented the elevation of serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), total bilirubin (T-Bil), total cholesterol (TC), triacylglycerol (TG), and liver lipid peroxide levels in APAP-treated mice and markedly reduced APAP-induced histological alterations in liver tissues. Additionally, TA attenuated the APAP-induced production of nitric oxide (NO), reactive oxygen species (ROS), tumor necrosis factor-alpha (TNF-α), interleukin-1beta (IL-1β), and IL-6. Furthermore, the Western blot analysis showed that TA blocked the protein expression of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2), as well as the inhibition of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinases (MAPKs) activation in APAP-injured liver tissues. TA also retained the superoxidase dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT) in the liver. These results suggest that the hepatoprotective effects of TA may be related to its anti-inflammatory effect by decreasing thiobarbituric acid reactive substances (TBARS), iNOS, COX-2, TNF-α, IL-1β, and IL-6, and inhibiting NF-κB and MAPK activation. Antioxidative properties were also observed, as shown by heme oxygenase-1 (HO-1) induction in the liver, and decreases in lipid peroxides and ROS. Therefore, TA may be a potential therapeutic candidate for the prevention of APAP-induced liver injury by inhibiting oxidative stress and inflammation.
Collapse
Affiliation(s)
- Wen-Ping Jiang
- School of Pharmacy, China Medical University, No. 91, Hsueh-Shih R., Taichung 40402, Taiwan.
| | - Shyh-Shyun Huang
- School of Pharmacy, China Medical University, No. 91, Hsueh-Shih R., Taichung 40402, Taiwan.
| | - Yoshikazu Matsuda
- Nihon Pharmaceutical University, 10281, Komuro, Ina-machi, Kitaadachi-gun, Saitama 3620806, Japan.
| | - Hiroshi Saito
- Nihon Pharmaceutical University, 10281, Komuro, Ina-machi, Kitaadachi-gun, Saitama 3620806, Japan.
| | - Naoto Uramaru
- Nihon Pharmaceutical University, 10281, Komuro, Ina-machi, Kitaadachi-gun, Saitama 3620806, Japan.
| | - Hui-Ya Ho
- Jen Li Biotech Company Ltd., Taiping District, Taichung 41143, Taiwan.
| | - Jin-Bin Wu
- School of Pharmacy, China Medical University, No. 91, Hsueh-Shih R., Taichung 40402, Taiwan.
| | - Guan-Jhong Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
46
|
Kwak DH, Kim JS, Chang KT, Choo YK. Aristolochia manshuriensis Kom ethyl acetate extract protects against high-fat diet-induced non-alcoholic steatohepatitis by regulating kinase phosphorylation in mouse. J Vet Sci 2017; 17:279-87. [PMID: 26726030 PMCID: PMC5037294 DOI: 10.4142/jvs.2016.17.3.279] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/11/2015] [Accepted: 12/30/2015] [Indexed: 12/26/2022] Open
Abstract
Aristolochia manshuriensis Kom (AMK) is an herb used as a traditional medicine; however, it causes side effects such as nephrotoxicity and carcinogenicity. Nevertheless, AMK can be applied in specific ways medicinally, including via ingestion of low doses for short periods of time. Non-alcoholic steatohepatitis (NASH) induced the hepatocyte injury and inflammation. The protective effects of AMK against NASH are unclear; therefore, in this study, the protective effects of AMK ethyl acetate extract were investigated in a high-fat diet (HFD)-induced NASH model. We found decreased hepatic steatosis and inflammation, as well as increased levels of lipoproteins during AMK extract treatment. We also observed decreased hepatic lipid peroxidation and triglycerides, as well as suppressed hepatic expression of lipogenic genes in extract-treated livers. Treatment with extract decreased the activation of c-jun N-terminal kinase 1/2 (JNK1/2) and increased the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2). These results demonstrate that the protective effect of the extract against HFD-induced NASH occurred via reductions in reactive oxygen species production, inflammation suppression, and apoptosis related to the suppression of JNK1/2 activation and increased ERK1/2 phosphorylation. Taken together, these results indicate that that ethyl acetate extract of AMK has potential therapeutic effects in the HFD-induced NASH mouse model.
Collapse
Affiliation(s)
- Dong Hoon Kwak
- Institute for Glycoscience College of Natural Sciences, Wonkwang University, Iksan 54538, Korea
| | - Ji-Su Kim
- National Primate Research Center, Korea Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Korea
| | - Kyu-Tae Chang
- National Primate Research Center, Korea Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Korea
| | - Young-Kug Choo
- Institute for Glycoscience College of Natural Sciences, Wonkwang University, Iksan 54538, Korea.,Department of Biological Science, College of Natural Sciences, Wonkwang University, Iksan 54538, Korea
| |
Collapse
|
47
|
Liao CC, Day YJ, Lee HC, Liou JT, Chou AH, Liu FC. ERK Signaling Pathway Plays a Key Role in Baicalin Protection Against Acetaminophen-Induced Liver Injury. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:105-121. [DOI: 10.1142/s0192415x17500082] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Acetaminophen (APAP) overdose causes hepatocytes necrosis and acute liver failure. Baicalin (BA), a major flavonoid of Scutellariae radix, has potent hepatoprotective properties in traditional medicine. In the present study, we investigated the protective effects of BA on a APAP-induced liver injury in a mouse model. The mice received an intraperitoneal hepatotoxic dose of APAP (300[Formula: see text]mg/kg) and after 30[Formula: see text]min, were treated with BA at concentrations of 0, 15, 30, or 60[Formula: see text]mg/kg. After 16[Formula: see text]h of treatment, the mice were sacrificed for further analysis. APAP administration significantly elevated the serum alanine transferase (ALT) enzyme levels and hepatic myeloperoxidase (MPO) activity when compared with control animals. Baicalin treatment significantly attenuated the elevation of liver ALT levels, as well as hepatic MPO activity in a dose- dependent manner (15–60[Formula: see text]mg/kg) in APAP-treated mice. The strongest beneficial effects of BA were seen at a dose of 30[Formula: see text]mg/kg. BA treatment at 30[Formula: see text]mg/kg after APAP overdose reduced elevated hepatic cytokine (TNF-[Formula: see text] and IL-6) levels, and macrophage recruitment around the area of hepatotoxicity in immunohistochemical staining. Significantly, BA treatment can also decrease hepatic phosphorylated extracellular signal-regulated kinase (ERK) expression, which is induced by APAP overdose. Our data suggests that baicalin treatment can effectively attenuate APAP-induced liver injury by down-regulating the ERK signaling pathway and its downstream effectors of inflammatory responses. These results support that baicalin is a potential hepatoprotective agent.
Collapse
Affiliation(s)
- Chia-Chih Liao
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Yuan-Ji Day
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Hung-Chen Lee
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Jiin-Tarng Liou
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - An-Hsun Chou
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Fu-Chao Liu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
48
|
Targeting pathogen metabolism without collateral damage to the host. Sci Rep 2017; 7:40406. [PMID: 28084422 PMCID: PMC5234033 DOI: 10.1038/srep40406] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/06/2016] [Indexed: 12/14/2022] Open
Abstract
The development of drugs that can inactivate disease-causing cells (e.g. cancer cells or parasites) without causing collateral damage to healthy or to host cells is complicated by the fact that many proteins are very similar between organisms. Nevertheless, due to subtle, quantitative differences between the biochemical reaction networks of target cell and host, a drug can limit the flux of the same essential process in one organism more than in another. We identified precise criteria for this ‘network-based’ drug selectivity, which can serve as an alternative or additive to structural differences. We combined computational and experimental approaches to compare energy metabolism in the causative agent of sleeping sickness, Trypanosoma brucei, with that of human erythrocytes, and identified glucose transport and glyceraldehyde-3-phosphate dehydrogenase as the most selective antiparasitic targets. Computational predictions were validated experimentally in a novel parasite-erythrocytes co-culture system. Glucose-transport inhibitors killed trypanosomes without killing erythrocytes, neurons or liver cells.
Collapse
|
49
|
Yan Y, Jiang W, Tan Y, Zou S, Zhang H, Mao F, Gong A, Qian H, Xu W. hucMSC Exosome-Derived GPX1 Is Required for the Recovery of Hepatic Oxidant Injury. Mol Ther 2017; 25:465-479. [PMID: 28089078 DOI: 10.1016/j.ymthe.2016.11.019] [Citation(s) in RCA: 251] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/26/2016] [Accepted: 11/27/2016] [Indexed: 12/13/2022] Open
Abstract
Exosomes are small biological membrane vesicles secreted by various cells, including mesenchymal stem cells (MSCs). We previously reported that MSC-derived exosomes (MSC-Ex) can elicit hepatoprotective effects against toxicant-induced injury. However, the success of MSC-Ex-based therapy for treatment of liver diseases and the underlying mechanisms have not been well characterized. We used human umbilical cord MSC-derived exosome (hucMSC-Ex) administrated by tail vein or oral gavage at different doses and, in engrafted liver mouse models, noted antioxidant and anti-apoptotic effects and rescue from liver failure. A single systemic administration of hucMSC-Ex (16 mg/kg) effectively rescued the recipient mice from carbon tetrachloride (CCl4)-induced liver failure. Moreover, hucMSC-Ex-derived glutathione peroxidase1 (GPX1), which detoxifies CCl4 and H2O2, reduced oxidative stress and apoptosis. Knockdown of GPX1 in hucMSCs abrogated antioxidant and anti-apoptotic abilities of hucMSC-Ex and diminished the hepatoprotective effects of hucMSC-Ex in vitro and in vivo. Thus, hucMSC-Ex promote the recovery of hepatic oxidant injury through the delivery of GPX1.
Collapse
Affiliation(s)
- Yongmin Yan
- Liver Disease and Cancer Institute, School of Medicine, Jiangsu University, 212013 Zhenjiang, Jiangsu, People's Republic of China.
| | - Wenqian Jiang
- Liver Disease and Cancer Institute, School of Medicine, Jiangsu University, 212013 Zhenjiang, Jiangsu, People's Republic of China
| | - Youwen Tan
- The Affiliated Third Hospital of Zhenjiang, Jiangsu University, 212013 Zhenjiang, Jiangsu, People's Republic of China
| | - Shengqiang Zou
- The Affiliated Third Hospital of Zhenjiang, Jiangsu University, 212013 Zhenjiang, Jiangsu, People's Republic of China
| | - Hongguang Zhang
- The Affiliated Third Hospital of Zhenjiang, Jiangsu University, 212013 Zhenjiang, Jiangsu, People's Republic of China
| | - Fei Mao
- Liver Disease and Cancer Institute, School of Medicine, Jiangsu University, 212013 Zhenjiang, Jiangsu, People's Republic of China
| | - Aihua Gong
- Liver Disease and Cancer Institute, School of Medicine, Jiangsu University, 212013 Zhenjiang, Jiangsu, People's Republic of China
| | - Hui Qian
- Liver Disease and Cancer Institute, School of Medicine, Jiangsu University, 212013 Zhenjiang, Jiangsu, People's Republic of China; Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 212013 Zhenjiang, Jiangsu, People's Republic of China
| | - Wenrong Xu
- Liver Disease and Cancer Institute, School of Medicine, Jiangsu University, 212013 Zhenjiang, Jiangsu, People's Republic of China; Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 212013 Zhenjiang, Jiangsu, People's Republic of China.
| |
Collapse
|
50
|
Bai X, Yang P, Zhou Q, Cai B, Buist‐Homan M, Cheng H, Jiang J, Shen D, Li L, Luo X, Faber KN, Moshage H, Shi G. The protective effect of the natural compound hesperetin against fulminant hepatitis in vivo and in vitro. Br J Pharmacol 2017; 174:41-56. [PMID: 27714757 PMCID: PMC5341490 DOI: 10.1111/bph.13645] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 09/21/2016] [Accepted: 09/23/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Liver diseases are mostly accompanied by inflammation and hepatocyte death. Therapeutic approaches targeting both hepatocyte injury and inflammation are not available. Natural compounds are considered as potential treatment for inflammatory liver diseases. Hesperetin, a flavonoid component of citrus fruits, has been reported to have anti-inflammatory properties. The aim of this study was to evaluate the cytoprotective and anti-inflammatory properties of hesperetin both in vitro and in models of fulminant hepatitis. EXPERIMENTAL APPROACH Apoptotic cell death and inflammation were induced in primary cultures of rat hepatocytes by bile acids and cytokine mixture respectively. Apoptosis was quantified by caspase-3 activity and necrosis by LDH release. The concanavalin A (ConA) and D-galactosamine/LPS (D-GalN/LPS) were used as models of fulminant hepatitis. Liver injury was assessed by alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, liver histology and TUNEL assay and inflammation by inducible NOS (iNOS) expression. KEY RESULTS Hesperetin blocked bile acid-induced apoptosis and cytokine-induced inflammation in rat hepatocytes. Moreover, hesperetin improved liver histology and protected against hepatocyte injury in ConA- and D-GalN/LPS-induced fulminant hepatitis, as assessed by TUNEL assay and serum AST and ALT levels. Hesperetin also reduced expression of the inflammatory marker iNOS and the expression and serum levels of TNFα and IFN-γ, the main mediators of cell toxicity in fulminant hepatitis. CONCLUSION AND IMPLICATIONS Hesperetin has anti-inflammatory and cytoprotective actions in models of acute liver toxicity. Hesperetin therefore has therapeutic potential for the treatment of inflammatory liver diseases accompanied by extensive hepatocyte injury, such as fulminant hepatitis.
Collapse
Affiliation(s)
- Xueting Bai
- Department of PharmacologyShantou University Medical CollegeShantouChina
- Department of Gastroenterology and HepatologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Peixuan Yang
- Health Care CenterThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Qiaoling Zhou
- Department of PharmacologyShantou University Medical CollegeShantouChina
| | - Bozhi Cai
- Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Manon Buist‐Homan
- Department of Gastroenterology and HepatologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
- Department of Laboratory MedicineUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - He Cheng
- Department of PharmacologyShantou University Medical CollegeShantouChina
| | - Jiyang Jiang
- Department of PharmacologyShantou University Medical CollegeShantouChina
| | - Daifei Shen
- Department of PharmacologyShantou University Medical CollegeShantouChina
| | - Lijun Li
- Department of PharmacologyShantou University Medical CollegeShantouChina
| | - Xiajiong Luo
- Department of PharmacologyShantou University Medical CollegeShantouChina
| | - Klaas Nico Faber
- Department of Gastroenterology and HepatologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
- Department of Laboratory MedicineUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Han Moshage
- Department of Gastroenterology and HepatologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
- Department of Laboratory MedicineUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Ganggang Shi
- Department of PharmacologyShantou University Medical CollegeShantouChina
| |
Collapse
|