1
|
Zhou J, Tang J, Zhang C, Li G, Lin X, Liao S, Luo J, Yu G, Zheng F, Guo Z, Shao W, Hu H, Xu L, Wu S, Li H. ALKBH5 targets ACSL4 mRNA stability to modulate ferroptosis in hyperbilirubinemia-induced brain damage. Free Radic Biol Med 2024; 220:271-287. [PMID: 38734267 DOI: 10.1016/j.freeradbiomed.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/18/2024] [Accepted: 05/08/2024] [Indexed: 05/13/2024]
Abstract
Bilirubin-induced brain damage is a serious clinical consequence of hyperbilirubinemia, yet the underlying molecular mechanisms remain largely unknown. Ferroptosis, an iron-dependent cell death, is characterized by iron overload and lipid peroxidation. Here, we report a novel regulatory mechanism of demethylase AlkB homolog 5 (ALKBH5) in acyl-coenzyme A synthetase long-chain family member 4 (ACSL4)-mediated ferroptosis in hyperbilirubinemia. Hyperdifferential PC12 cells and newborn Sprague-Dawley rats were used to establish in vitro and in vivo hyperbilirubinemia models, respectively. Proteomics, coupled with bioinformatics analysis, first suggested the important role of ferroptosis in hyperbilirubinemia-induced brain damage. In vitro experiments showed that ferroptosis is activated in hyperbilirubinemia, and ferroptosis inhibitors (desferrioxamine and ferrostatin-1) treatment effectively alleviates hyperbilirubinemia-induced oxidative damage. Notably, we observed that the ferroptosis in hyperbilirubinemia was regulated by m6A modification through the downregulation of ALKBH5 expression. MeRIP-seq and RIP-seq showed that ALKBH5 may trigger hyperbilirubinemia ferroptosis by stabilizing ACSL4 mRNA via m6A modification. Further, hyperbilirubinemia-induced oxidative damage was alleviated through ACSL4 genetic knockdown or rosiglitazone-mediated chemical repression but was exacerbated by ACSL4 overexpression. Mechanistically, ALKBH5 promotes ACSL4 mRNA stability and ferroptosis by combining the 669 and 2015 m6A modified sites within 3' UTR of ACSL4 mRNA. Our findings unveil a novel molecular mechanism of ferroptosis and suggest that m6A-dependent ferroptosis could be an underlying clinical target for the therapy of hyperbilirubinemia.
Collapse
Affiliation(s)
- Jinfu Zhou
- Medical Genetic Diagnosis and Therapy Center, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, Fujian Province, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Jianping Tang
- Medical Genetic Diagnosis and Therapy Center, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Chenran Zhang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Guilin Li
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Xinpei Lin
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Sining Liao
- Center for Disease Control and Prevention of Shantou, Shantou, Guangdong, 515000, China
| | - Jinying Luo
- Obstetrics and Gynecology Department, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Guangxia Yu
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Fuli Zheng
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Zhenkun Guo
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Wenya Shao
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Hong Hu
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Liangpu Xu
- Medical Genetic Diagnosis and Therapy Center, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, Fujian Province, China.
| | - Siying Wu
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China.
| | - Huangyuan Li
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China.
| |
Collapse
|
2
|
Liu HW, Gong LN, Lai K, Yu XF, Liu ZQ, Li MX, Yin XL, Liang M, Shi HS, Jiang LH, Yang W, Shi HB, Wang LY, Yin SK. Bilirubin gates the TRPM2 channel as a direct agonist to exacerbate ischemic brain damage. Neuron 2023; 111:1609-1625.e6. [PMID: 36921602 DOI: 10.1016/j.neuron.2023.02.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 10/18/2022] [Accepted: 02/13/2023] [Indexed: 03/16/2023]
Abstract
Stroke prognosis is negatively associated with an elevation of serum bilirubin, but how bilirubin worsens outcomes remains mysterious. We report that post-, but not pre-, stroke bilirubin levels among inpatients scale with infarct volume. In mouse models, bilirubin increases neuronal excitability and ischemic infarct, whereas ischemic insults induce the release of endogenous bilirubin, all of which are attenuated by knockout of the TRPM2 channel or its antagonist A23. Independent of canonical TRPM2 intracellular agonists, bilirubin and its metabolic derivatives gate the channel opening, whereas A23 antagonizes it by binding to the same cavity. Knocking in a loss of binding point mutation for bilirubin, TRPM2-D1066A, effectively antagonizes ischemic neurotoxicity in mice. These findings suggest a vicious cycle of stroke injury in which initial ischemic insults trigger the release of endogenous bilirubin from injured cells, which potentially acts as a volume neurotransmitter to activate TRPM2 channels, aggravating Ca2+-dependent brain injury.
Collapse
Affiliation(s)
- Han-Wei Liu
- Department of Otorhinolaryngology Head & Neck Surgery, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Li-Na Gong
- Department of Otorhinolaryngology Head & Neck Surgery, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ke Lai
- Department of Otorhinolaryngology Head & Neck Surgery, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Xia-Fei Yu
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhen-Qi Liu
- Department of Otorhinolaryngology Head & Neck Surgery, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ming-Xian Li
- Department of Otorhinolaryngology Head & Neck Surgery, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xin-Lu Yin
- Department of Otorhinolaryngology Head & Neck Surgery, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Department of Head & Neck Surgery, Renji Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Min Liang
- Department of Otorhinolaryngology Head & Neck Surgery, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Department of Otorhinolaryngology Head & Neck Surgery, Xinhua Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Hao-Song Shi
- Department of Otorhinolaryngology Head & Neck Surgery, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Lin-Hua Jiang
- Department of Physiology and Pathophysiology, School of Basic Sciences, Xinxiang Medical University, Xinxiang, Henan Province 453003, China; School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Wei Yang
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hai-Bo Shi
- Department of Otorhinolaryngology Head & Neck Surgery, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Lu-Yang Wang
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Shan-Kai Yin
- Department of Otorhinolaryngology Head & Neck Surgery, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
3
|
Pranty AI, Shumka S, Adjaye J. Bilirubin-Induced Neurological Damage: Current and Emerging iPSC-Derived Brain Organoid Models. Cells 2022; 11:2647. [PMID: 36078055 PMCID: PMC9454749 DOI: 10.3390/cells11172647] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/04/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Bilirubin-induced neurological damage (BIND) has been a subject of studies for decades, yet the molecular mechanisms at the core of this damage remain largely unknown. Throughout the years, many in vivo chronic bilirubin encephalopathy models, such as the Gunn rat and transgenic mice, have further elucidated the molecular basis of bilirubin neurotoxicity as well as the correlations between high levels of unconjugated bilirubin (UCB) and brain damage. Regardless of being invaluable, these models cannot accurately recapitulate the human brain and liver system; therefore, establishing a physiologically recapitulating in vitro model has become a prerequisite to unveil the breadth of complexities that accompany the detrimental effects of UCB on the liver and developing human brain. Stem-cell-derived 3D brain organoid models offer a promising platform as they bear more resemblance to the human brain system compared to existing models. This review provides an explicit picture of the current state of the art, advancements, and challenges faced by the various models as well as the possibilities of using stem-cell-derived 3D organoids as an efficient tool to be included in research, drug screening, and therapeutic strategies for future clinical applications.
Collapse
Affiliation(s)
| | | | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Faculty of Medicine, Heinrich-Heine University, Moorenstrasse 5, 40225 Dusseldorf, Germany
| |
Collapse
|
4
|
Shahrokhi SZ, Karami Tehrani FS, Salami S. Induction of cell apoptosis by biliverdin reductase inhibitor in MCF-7 and MDA-MB-468 breast cancer cell lines: Experimental and in silico studies. EXCLI JOURNAL 2021; 20:1502-1516. [PMID: 34924900 PMCID: PMC8678058 DOI: 10.17179/excli2021-4069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/15/2021] [Indexed: 01/03/2023]
Abstract
Biliverdin reductase, biliverdin and bilirubin are known as important components of cellular signaling pathways that play major roles in cell proliferation and apoptosis, although their physiological relevance is still under evaluation. This study was designed to investigate the expression and activity of BVR-A and its apoptotic effect in the breast cancer cell lines, MCF-7 and MDA-MB-468. The expression of BVR-A was examined by real-time PCR and western blot analysis. Bilirubin concentration was measured by HPLC and molecular docking was performed to identify an appropriate inhibitor for BVR-A. To detect cell apoptosis, annexin V-PI staining, caspase-3, -8, and -9 activities were evaluated. Cell viability was reduced by biliverdin, in a dose-dependent manner, and an intrinsic apoptotic response occurred which was evidenced by caspase-3 and -9 activities. The intra- and extracellular concentrations of bilirubin were higher in MCF-7 cells than those of MDA-MB-468 cells. The expression of BVR-A, at mRNA and protein levels, in MCF-7 was also higher than that of MDA-MB-468 cells. Treatment of both cell lines with biliverdin plus DTNB, a BVR-A inhibitor, increased the cell death significantly when compared with biliverdin alone. Using annexin V-PI staining and assessment of caspase-3 activity, it was confirmed that biliverdin together with DTNB increases apoptosis in breast cancer cells. In conclusion, biliverdin has an important role in cell apoptosis and inhibition of biliverdin reductase increases the apoptotic effect of biliverdin.
Collapse
Affiliation(s)
- Seyedeh Zahra Shahrokhi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Siamak Salami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Hu JH, Fan P, Zhang LR, Chen CY, Xu J, Huang J, Lu WT, Zhu SJ, Qiu GP, Xu SY, Ran JH, Gan SW. Neuroglobin expression and function in the temporal cortex of bilirubin encephalopathy rats. Anat Rec (Hoboken) 2021; 305:254-264. [PMID: 34358403 DOI: 10.1002/ar.24734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 06/10/2021] [Accepted: 06/24/2021] [Indexed: 11/09/2022]
Affiliation(s)
- Jia-Heng Hu
- Institute of Neuroscience, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Ping Fan
- Department of Gynecology and Obstetrics, The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Li-Rong Zhang
- Institute of Neuroscience, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Chun-Yan Chen
- Institute of Neuroscience, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Jin Xu
- Institute of Neuroscience, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Juan Huang
- Institute of Neuroscience, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Wei-Tian Lu
- Institute of Neuroscience, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Shu-Juan Zhu
- Institute of Neuroscience, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Guo-Ping Qiu
- Institute of Neuroscience, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Shi-Ye Xu
- Institute of Neuroscience, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Jian-Hua Ran
- Institute of Neuroscience, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Sheng-Wei Gan
- Institute of Neuroscience, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Bilirubin disrupts calcium homeostasis in neonatal hippocampal neurons: a new pathway of neurotoxicity. Arch Toxicol 2020; 94:845-855. [DOI: 10.1007/s00204-020-02659-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/03/2020] [Indexed: 12/22/2022]
|
7
|
Canesin G, Hejazi SM, Swanson KD, Wegiel B. Heme-Derived Metabolic Signals Dictate Immune Responses. Front Immunol 2020; 11:66. [PMID: 32082323 PMCID: PMC7005208 DOI: 10.3389/fimmu.2020.00066] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/10/2020] [Indexed: 12/21/2022] Open
Abstract
Heme is one of the most abundant molecules in the body acting as the functional core of hemoglobin/myoglobin involved in the O2/CO2 carrying in the blood and tissues, redox enzymes and cytochromes in mitochondria. However, free heme is toxic and therefore its removal is a significant priority for the host. Heme is a well-established danger-associated molecular pattern (DAMP), which binds to toll-like receptor 4 (TLR4) to induce immune responses. Heme-derived metabolites including the bile pigments, biliverdin (BV) and bilirubin (BR), were first identified as toxic drivers of neonatal jaundice in 1800 but have only recently been appreciated as endogenous drivers of multiple signaling pathways involved in protection from oxidative stress and regulators of immune responses. The tissue concentration of heme, BV and BR is tightly controlled. Heme oxygenase-1 (HO-1, encoded by HMOX1) produces BV by heme degradation, while biliverdin reductase-A (BLVR-A) generates BR by the subsequent conversion of BV. BLVR-A is a fascinating protein that possesses a classical protein kinase domain, which is activated in response to BV binding to its enzymatic site and initiates the downstream mitogen-activated protein kinases (MAPK) and phosphatidylinositol 3-kinase (PI3K) pathways. This links BLVR-A activity to cell growth and survival pathways. BLVR-A also contains a bZip DNA binding domain and a nuclear export sequence (NES) and acts as a transcription factor to regulate the expression of immune modulatory genes. Here we will discuss the role of heme-related immune response and the potential for targeting the heme system for therapies directed toward hepatitis and cancer.
Collapse
Affiliation(s)
- Giacomo Canesin
- Department of Surgery, Cancer Research Institute and Transplant Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Seyed M. Hejazi
- Department of Surgery, Cancer Research Institute and Transplant Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Kenneth D. Swanson
- Brain Tumor Center and Neuro-Oncology Unit, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Barbara Wegiel
- Department of Surgery, Cancer Research Institute and Transplant Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
8
|
Abstract
Schizophrenia is a complex syndrome of unknown etiology and difficult to manage. Unconjugated bilirubin has been researched as a potential biological marker of this syndrome. The objective of this review article was to gather the studies published to date on the relationship between this molecule and schizophrenia. Broad inclusion criteria have been used (PRISMA) to include as many relevant studies as possible. Fourteen studies were selected: 3 analyzed the effects of unconjugated hyperbilirubinemia in animal models; 6 demonstrated an increased incidence of schizophrenia in patients with increased unconjugated bilirubin; 2 reported an increased incidence of the disease in patients with decreased unconjugated bilirubin; and 3 linked an increased incidence of schizophrenia with an increased excretion of the oxidative product of bilirubin, the so-called biopyrrins. Because of apparently contradictory reported results, the hypothesis that the relationship between schizophrenia and unconjugated bilirubin was not linear and that there was an inflammatory dysfunction explaining this was considered. The 2 most accepted models for the pathophysiology of schizophrenia are described, and the possible role of the molecule in each is clarified. The bilirubin buffer system and its role in antioxidant defense was explored. The average levels of unconjugated bilirubin in patients with schizophrenia, schizoaffective disorder, and bipolar disorder were also compared, having been hypothesized that these diseases could be different points of a same pathological spectrum. Finally, it was concluded that unconjugated bilirubin is a promising molecule that could be used as a possible biological marker for schizophrenia, and the necessity of subsequent efforts for its research was considered.
Collapse
|
9
|
Ruiz-Gaspà S, Guañabens N, Jurado S, Dubreuil M, Combalia A, Peris P, Monegal A, Parés A. Bile acids and bilirubin effects on osteoblastic gene profile. Implications in the pathogenesis of osteoporosis in liver diseases. Gene 2019; 725:144167. [PMID: 31639434 DOI: 10.1016/j.gene.2019.144167] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 12/16/2022]
Abstract
Osteoporosis in advanced cholestatic and end-stage liver disease is related to low bone formation. Previous studies have demonstrated the deleterious consequences of lithocholic acid (LCA) and bilirubin on osteoblastic cells. These effects are partially or completely neutralized by ursodeoxycholic acid (UDCA). We have assessed the differential gene expression of osteoblastic cells under different culture conditions. The experiments were performed in human osteosarcoma cells (Saos-2) cultured with LCA (10 μM), bilirubin (50 μM) or UDCA (10 and 100 μM) at 2 and 24 h. Expression of 87 genes related to bone metabolism and other signalling pathways were assessed by TaqMan micro fluidic cards. Several genes were up-regulated by LCA, most of them pro-apoptotic (BAX, BCL10, BCL2L13, BCL2L14), but also MGP (matrix Gla protein), BGLAP (osteocalcin), SPP1 (osteopontin) and CYP24A1, and down-regulated bone morphogenic protein genes (BMP3 and BMP4) and DKK1 (Dickkopf-related protein 1). Parallel effects were observed with bilirubin, which up-regulated apoptotic genes and CSF2 (colony-stimulating factor 2) and down-regulated antiapoptotic genes (BCL2 and BCL2L1), BMP3, BMP4 and RUNX2. UDCA 100 μM had specific consequences since differential expression was observed, up-regulating BMP2, BMP4, BMP7, CALCR (calcitonin receptor), SPOCK3 (osteonectin), BGLAP (osteocalcin) and SPP1 (osteopontin), and down-regulating pro-apoptotic genes. Furthermore, most of the differential expression changes induced by both LCA and bilirubin were partially or completely neutralized by UDCA. Conclusion: Our observations reveal novel target genes, whose regulation by retained substances of cholestasis may provide additional insights into the pathogenesis of osteoporosis in cholestatic and end-stage liver diseases.
Collapse
Affiliation(s)
- Silvia Ruiz-Gaspà
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Spain
| | - Nuria Guañabens
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Spain; Metabolic Bone Diseases Unit, Department of Rheumatology, Hospital Clínic, IDIBAPS, University of Barcelona, Spain.
| | - Susana Jurado
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Spain
| | - Marta Dubreuil
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Spain
| | - Andres Combalia
- Metabolic Bone Diseases Unit, Department of Rheumatology, Hospital Clínic, IDIBAPS, University of Barcelona, Spain
| | - Pilar Peris
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Spain; Metabolic Bone Diseases Unit, Department of Rheumatology, Hospital Clínic, IDIBAPS, University of Barcelona, Spain
| | - Ana Monegal
- Metabolic Bone Diseases Unit, Department of Rheumatology, Hospital Clínic, IDIBAPS, University of Barcelona, Spain
| | - Albert Parés
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Spain; Liver Unit, Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona, Spain
| |
Collapse
|
10
|
Nandi S, Biswas S. A recyclable post-synthetically modified Al(iii) based metal–organic framework for fast and selective fluorogenic recognition of bilirubin in human biofluids. Dalton Trans 2019; 48:9266-9275. [DOI: 10.1039/c9dt01180c] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The ultra-fast, highly sensitive and selective sensing features of bilirubin in human biofluids by a post-synthetically modified Al(iii) MOF are presented.
Collapse
Affiliation(s)
- Soutick Nandi
- Department of Chemistry
- Indian Institute of Technology Guwahati
- India
| | - Shyam Biswas
- Department of Chemistry
- Indian Institute of Technology Guwahati
- India
| |
Collapse
|
11
|
Bilirubin-Induced Oxidative Stress Leads to DNA Damage in the Cerebellum of Hyperbilirubinemic Neonatal Mice and Activates DNA Double-Strand Break Repair Pathways in Human Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1801243. [PMID: 30598724 PMCID: PMC6287157 DOI: 10.1155/2018/1801243] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/19/2018] [Accepted: 10/02/2018] [Indexed: 12/27/2022]
Abstract
Unconjugated bilirubin is considered a potent antioxidant when present at moderate levels. However, at high concentrations, it produces severe neurological damage and death associated with kernicterus due to oxidative stress and other mechanisms. While it is widely recognized that oxidative stress by different toxic insults results in severe damage to cellular macromolecules, especially to DNA, no data are available either on DNA damage in the brain triggered by hyperbilirubinemia during the neonatal period or on the activation of DNA repair mechanisms. Here, using a mouse model of neonatal hyperbilirubinemia, we demonstrated that DNA damage occurs in vivo in the cerebellum, the brain region most affected by bilirubin toxicity. We studied the mechanisms associated with potential toxic action of bilirubin on DNA in in vitro models, which showed significant increases in DNA damage when neuronal and nonneuronal cells were treated with 140 nM of free bilirubin (Bf), as determined by γH2AX Western blot and immunofluorescence analyses. Cotreatment of cells with N-acetyl-cysteine, a potent oxidative-stress inhibitor, prevented DNA damage by bilirubin, supporting the concept that DNA damage was caused by bilirubin-induced oxidative stress. Bilirubin treatment also activated the main DNA repair pathways through homologous recombination (HR) and nonhomologous end joining (NHEJ), which may be adaptive responses to repair bilirubin-induced DNA damage. Since DNA damage may be another important factor contributing to neuronal death and bilirubin encephalopathy, these results contribute to the understanding of the mechanisms associated with bilirubin toxicity and may be of relevance in neonates affected with severe hyperbilirubinemia.
Collapse
|
12
|
Deliktaş M, Ergin H, Demiray A, Akça H, Özdemir ÖMA, Özdemir MB. Caffeine prevents bilirubin-induced cytotoxicity in cultured newborn rat astrocytes. J Matern Fetal Neonatal Med 2018; 32:1813-1819. [PMID: 29295636 DOI: 10.1080/14767058.2017.1419175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Unconjugated bilirubin (UCB) may cause neurotoxicity in preterm neonates due to immaturity of UGT1A1 leading to bilirubin accumulation in the brain. Caffeine used in the treatment of apnea of prematurity was reported to decrease mechanical ventilation requirement, the frequencies of bronchopulmonary dysplasia, patent ductus arteriosus, cerebral palsy and neurodevelopmental disorders in very low birth weight infants. However, the effect of caffeine on hyperbilirubinemia was not yet clarified. METHODS We used astrocyte cell cultures obtained from 2-day-old Wistar albino rats via modified Cole and de Vellis method. UCB concentration toxic to 50% of astrocytes, and caffeine concentration increasing cell viability 100% were used in experiments. While no medication was applied to the control group, UCB (50 μM) and caffeine (100 μM) were applied to the bilirubin and caffeine groups for 24 h. Prophylactic and therapeutic caffeine groups were treated with caffeine 4 h before and after UCB exposure. The effects of caffeine were investigated in rat astrocytes exposed to UCB in terms of cell viability, apoptosis, antioxidant defense, proinflammatory cytokines, and Toll-like receptor (TLR)s. RESULTS Compared to the control group, UCB increased apoptosis, malondialdehyde (MDA), tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, total nitrate/nitrite, and TLR4 levels, and decreased cell viability, catalase (CAT), glutathione peroxidase (GPx), superoxide dismutase (SOD) activities, glutathione, and TLR9 levels (for all p < .001). Conversely, prophylactic and therapeutic caffeine improved the detrimental effects of UCB. CONCLUSIONS Caffeine seems encouraging for the prevention and treatment of bilirubin neurotoxicity in rats by means of its antiapoptotic, antioxidant, anti-inflammatory, anti-nitrosative, and anti-TLR-4 properties.
Collapse
Affiliation(s)
- Mehmet Deliktaş
- a Department of Pediatrics, Division of Neonatology, Faculty of Medicine , Pamukkale University , Denizli , Turkey
| | - Hacer Ergin
- a Department of Pediatrics, Division of Neonatology, Faculty of Medicine , Pamukkale University , Denizli , Turkey
| | - Aydın Demiray
- b Department of Medical Biology, Division of Neonatology, Faculty of Medicine , Pamukkale University , Denizli , Turkey
| | - Hakan Akça
- b Department of Medical Biology, Division of Neonatology, Faculty of Medicine , Pamukkale University , Denizli , Turkey
| | - Özmert M A Özdemir
- a Department of Pediatrics, Division of Neonatology, Faculty of Medicine , Pamukkale University , Denizli , Turkey
| | - Mehmet Bülent Özdemir
- c Department of Anatomy, Division of Neonatology, Faculty of Medicine , Pamukkale University , Denizli , Turkey
| |
Collapse
|
13
|
The activation of autophagy protects neurons and astrocytes against bilirubin-induced cytotoxicity. Neurosci Lett 2017; 661:96-103. [PMID: 28965934 DOI: 10.1016/j.neulet.2017.09.056] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/27/2017] [Accepted: 09/27/2017] [Indexed: 12/21/2022]
Abstract
Unconjugated bilirubin (UCB) neurotoxicity involves oxidative stress, calcium signaling and ER-stress. The same insults can also induce autophagy, a process of "self-eating", with both a pro-survival or a pro-apoptotic role. Our aim was to study the outcome of autophagy activation by UCB in the highly sensitive neuronal SH-SY5Y cells and in the resistant astrocytoma U87 cells. Upon treatment with a toxic dose of UCB, the conversion of LC3-I to LC3-II was detected in both cell lines. Inhibition of autophagy by E64d before UCB treatment increased SH-SY5Y cell mortality and made U87 cells sensitive to UCB. In SH-SY5Y autophagy related genes ATG8 (5 folds), ATG18 (5 folds), p62 (3 folds) and FAM 129A (4.5 folds) were induced 8h after UCB treatment while DDIT4 upregulation (13 folds) started at 4h. mTORC1 inactivation by UCB was confirmed by phosphorylation of 4EBP1. UCB induced LC3-II conversion was completely prevented by pretreating cells with the calcium chelator BAPTA and reduced by 65% using the ER-stress inhibitor 4-PBA. Pretreatment with the PKC inhibitor reduced LC3 mRNA by 70% as compared to cells exposed to UCB alone. Finally, autophagy induction by Trifluoroperazine (TFP) increased the cell viability of rat hippocampal primary neurons upon UCB treatment from 60% to 80%. In SH-SY5Y cells, TFP pretreatment blocked the UCB-induced cleaved caspase-3 protein expression, decreased LDH release from 50% to 23%, reduced the UCB-induction of HO1, CHOP and IL-8 mRNAs by 85%, 70% and 97%. Collectively these data indicate that the activation of autophagy protects neuronal cells from UCB cytotoxicity. The mechanisms of autophagy activation by UCB involves mTOR/ER-stress/PKC/calcium signaling.
Collapse
|
14
|
Figueira I, Garcia G, Pimpão RC, Terrasso AP, Costa I, Almeida AF, Tavares L, Pais TF, Pinto P, Ventura MR, Filipe A, McDougall GJ, Stewart D, Kim KS, Palmela I, Brites D, Brito MA, Brito C, Santos CN. Polyphenols journey through blood-brain barrier towards neuronal protection. Sci Rep 2017; 7:11456. [PMID: 28904352 PMCID: PMC5597593 DOI: 10.1038/s41598-017-11512-6] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 08/08/2017] [Indexed: 01/03/2023] Open
Abstract
Age-related complications such as neurodegenerative disorders are increasing and remain cureless. The possibility of altering the progression or the development of these multifactorial diseases through diet is an emerging and attractive approach with increasing experimental support. We examined the potential of known bioavailable phenolic sulfates, arising from colonic metabolism of berries, to influence hallmarks of neurodegenerative processes. In silico predictions and in vitro transport studies across blood-brain barrier (BBB) endothelial cells, at circulating concentrations, provided evidence for differential transport, likely related to chemical structure. Moreover, endothelial metabolism of these phenolic sulfates produced a plethora of novel chemical entities with further potential bioactivies. Pre-conditioning with phenolic sulfates improved cellular responses to oxidative, excitotoxicity and inflammatory injuries and this attenuation of neuroinflammation was achieved via modulation of NF-κB pathway. Our results support the hypothesis that these small molecules, derived from dietary (poly)phenols may cross the BBB, reach brain cells, modulate microglia-mediated inflammation and exert neuroprotective effects, with potential for alleviation of neurodegenerative diseases.
Collapse
Affiliation(s)
- I Figueira
- Instituto de Tecnologia Quı́mica e Biológica - António Xavier, Universidade Nova de Lisboa, Av. da República, EAN, 2781-901, Oeiras, Portugal.,Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal
| | - G Garcia
- Instituto de Tecnologia Quı́mica e Biológica - António Xavier, Universidade Nova de Lisboa, Av. da República, EAN, 2781-901, Oeiras, Portugal.,Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal
| | - R C Pimpão
- Instituto de Tecnologia Quı́mica e Biológica - António Xavier, Universidade Nova de Lisboa, Av. da República, EAN, 2781-901, Oeiras, Portugal.,Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal
| | - A P Terrasso
- Instituto de Tecnologia Quı́mica e Biológica - António Xavier, Universidade Nova de Lisboa, Av. da República, EAN, 2781-901, Oeiras, Portugal.,Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal
| | - I Costa
- Instituto de Tecnologia Quı́mica e Biológica - António Xavier, Universidade Nova de Lisboa, Av. da República, EAN, 2781-901, Oeiras, Portugal.,Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal
| | - A F Almeida
- Instituto de Tecnologia Quı́mica e Biológica - António Xavier, Universidade Nova de Lisboa, Av. da República, EAN, 2781-901, Oeiras, Portugal.,Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal
| | - L Tavares
- Instituto de Tecnologia Quı́mica e Biológica - António Xavier, Universidade Nova de Lisboa, Av. da República, EAN, 2781-901, Oeiras, Portugal.,Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal
| | - T F Pais
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, 2780-156, Oeiras, Portugal
| | - P Pinto
- Instituto de Tecnologia Quı́mica e Biológica - António Xavier, Universidade Nova de Lisboa, Av. da República, EAN, 2781-901, Oeiras, Portugal.,Escola Superior Agrária, Instituto Politécnico de Santarém, Qta do Galinheiro, Santarém, Portugal
| | - M R Ventura
- Instituto de Tecnologia Quı́mica e Biológica - António Xavier, Universidade Nova de Lisboa, Av. da República, EAN, 2781-901, Oeiras, Portugal
| | - A Filipe
- Medical Department, Grupo Tecnimede, 2710-089, Sintra, Portugal
| | - G J McDougall
- The James Hutton Institute, Invergowrie, Dundee, DD2 5DA, Scotland, United Kingdom
| | - D Stewart
- The James Hutton Institute, Invergowrie, Dundee, DD2 5DA, Scotland, United Kingdom.,Engineering and Physical Sciences, Heriot Watt University, Edinburgh, EH14 4AS, Scotland, United Kingdom.,NIBIO, Norwegian Institute of Bioeconomy Research, Pb 115, NO-1431, Ås, Norway
| | - K S Kim
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, 600 North Wolfe Street Park 256, Baltimore, MD21287, USA
| | - I Palmela
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - D Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - M A Brito
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - C Brito
- Instituto de Tecnologia Quı́mica e Biológica - António Xavier, Universidade Nova de Lisboa, Av. da República, EAN, 2781-901, Oeiras, Portugal.,Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal
| | - C N Santos
- Instituto de Tecnologia Quı́mica e Biológica - António Xavier, Universidade Nova de Lisboa, Av. da República, EAN, 2781-901, Oeiras, Portugal. .,Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal.
| |
Collapse
|
15
|
Gou P, Qi X, Yuan R, Li H, Gao X, Wang J, Zhang B. Tet1-mediated DNA demethylation involves in neuron damage induced by bilirubin in vitro. Toxicol Mech Methods 2017; 28:55-61. [DOI: 10.1080/15376516.2017.1357775] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Panhong Gou
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Xiaoling Qi
- Institute of Occupational Health and Environment Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Rui Yuan
- Institute of Occupational Health and Environment Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Haojie Li
- Institute of Occupational Health and Environment Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Xiaoling Gao
- Clinical Research and Translational Medicine Institute, Gansu Provincial People’s Hospital, Lanzhou, China
| | - Junling Wang
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Benzhong Zhang
- Institute of Occupational Health and Environment Health, School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
16
|
Gleixner AM, Hutchison DF, Sannino S, Bhatia TN, Leak LC, Flaherty PT, Wipf P, Brodsky JL, Leak RK. N-Acetyl-l-Cysteine Protects Astrocytes against Proteotoxicity without Recourse to Glutathione. Mol Pharmacol 2017; 92:564-575. [PMID: 28830914 DOI: 10.1124/mol.117.109926] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/17/2017] [Indexed: 02/06/2023] Open
Abstract
N-acetyl-l-cysteine (NAC) exhibits protective properties in brain injury models and has undergone a number of clinical trials. Most studies of NAC have focused on neurons. However, neuroprotection may be complemented by the protection of astrocytes because healthier astrocytes can better support the viability of neurons. Here, we show that NAC can protect astrocytes against protein misfolding stress (proteotoxicity), the hallmark of neurodegenerative disorders. Although NAC is thought to be a glutathione precursor, NAC protected primary astrocytes from the toxicity of the proteasome inhibitor MG132 without eliciting any increase in glutathione. Furthermore, glutathione depletion failed to attenuate the protective effects of NAC. MG132 elicited a robust increase in the folding chaperone heat shock protein 70 (Hsp70), and NAC mitigated this effect. Nevertheless, three independent inhibitors of Hsp70 function ablated the protective effects of NAC, suggesting that NAC may help preserve Hsp70 chaperone activity and improve protein quality control without need for Hsp70 induction. Consistent with this view, NAC abolished an increase in ubiquitinated proteins in MG132-treated astrocytes. However, NAC did not affect the loss of proteasome activity in response to MG132, demonstrating that it boosted protein homeostasis and cell viability without directly interfering with the efficacy of this proteasome inhibitor. The thiol-containing molecules l-cysteine and d-cysteine both mimicked the protective effects of NAC, whereas the thiol-lacking molecule N-acetyl-S-methyl-l-cysteine failed to exert protection or blunt the rise in ubiquitinated proteins. Collectively, these findings suggest that the thiol group in NAC is required for its effects on glial viability and protein quality control.
Collapse
Affiliation(s)
- Amanda M Gleixner
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Daniel F Hutchison
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Sara Sannino
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Tarun N Bhatia
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Lillian C Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Patrick T Flaherty
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Peter Wipf
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Jeffrey L Brodsky
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| |
Collapse
|
17
|
Zhou YF, Zhang C, Yang G, Qian ZM, Zhang MW, Ma J, Zhang FL, Ke Y. Hepcidin Protects Neuron from Hemin-Mediated Injury by Reducing Iron. Front Physiol 2017; 8:332. [PMID: 28588503 PMCID: PMC5440571 DOI: 10.3389/fphys.2017.00332] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 05/08/2017] [Indexed: 01/05/2023] Open
Abstract
Hemin plays a key role in mediating secondary neuronal injury after intracerebral hemorrhage (ICH) and the cell toxicity of hemin is thought to be due to iron that is liberated when hemin is degraded. In a recent study, we demonstrated the iron regulatory hormone hepcidin reduces brain iron in iron-overloaded rats. Therefore, we hypothesized that hepcidin might be able to reduce iron and then protect neurons from hemin or iron-mediated neurotoxicity in hemin-treated neuronal cells. Here, we tested the hypothesis and demonstrated that ad-hepcidin and hepcidin peptide both have the ability to suppress the hemin-induced increase in LDH release and apoptotic cell numbers, to reduce cell iron and ferritin contents, and to inhibit expression of transferrin receptor 1, divalent metal transporter 1, and ferroportin 1 in hemin-treated neurons. We conclude that hepcidin protects neuron from hemin-mediated injury by reducing iron via inhibition of expression of iron transport proteins.
Collapse
Affiliation(s)
- Yu-Fu Zhou
- Laboratory of Neuropharmacology, School of Pharmacy, Fudan UniversityShanghai, China
| | - Chao Zhang
- Laboratory of Neuropharmacology, School of Pharmacy, Fudan UniversityShanghai, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong KongShatin, Hong Kong
| | - Guang Yang
- Laboratory of Neuropharmacology, School of Pharmacy, Fudan UniversityShanghai, China
| | - Zhong-Ming Qian
- Laboratory of Neuropharmacology, School of Pharmacy, Fudan UniversityShanghai, China
| | - Meng-Wan Zhang
- Laboratory of Neuropharmacology, School of Pharmacy, Fudan UniversityShanghai, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong KongShatin, Hong Kong
| | - Juan Ma
- Laboratory of Neuropharmacology, School of Pharmacy, Fudan UniversityShanghai, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong KongShatin, Hong Kong
| | - Fa-Li Zhang
- Laboratory of Neuropharmacology, School of Pharmacy, Fudan UniversityShanghai, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong KongShatin, Hong Kong
| | - Ya Ke
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong KongShatin, Hong Kong
| |
Collapse
|
18
|
Vodret S, Bortolussi G, Jašprová J, Vitek L, Muro AF. Inflammatory signature of cerebellar neurodegeneration during neonatal hyperbilirubinemia in Ugt1 -/- mouse model. J Neuroinflammation 2017; 14:64. [PMID: 28340583 PMCID: PMC5366125 DOI: 10.1186/s12974-017-0838-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 03/12/2017] [Indexed: 12/14/2022] Open
Abstract
Background Severe hyperbilirubinemia is toxic during central nervous system development. Prolonged and uncontrolled high levels of unconjugated bilirubin lead to bilirubin-induced neurological damage and eventually death by kernicterus. Bilirubin neurotoxicity is characterized by a wide array of neurological deficits, including irreversible abnormalities in motor, sensitive and cognitive functions, due to bilirubin accumulation in the brain. Despite the abundant literature documenting the in vitro and in vivo toxic effects of bilirubin, it is unclear which molecular and cellular events actually characterize bilirubin-induced neurodegeneration in vivo. Methods We used a mouse model of neonatal hyperbilirubinemia to temporally and spatially define the response of the developing cerebellum to the bilirubin insult. Results We showed that the exposure of developing cerebellum to sustained bilirubin levels induces the activation of oxidative stress, ER stress and inflammatory markers at the early stages of the disease onset. In particular, we identified TNFα and NFKβ as key mediators of bilirubin-induced inflammatory response. Moreover, we reported that M1 type microglia is increasingly activated during disease progression. Failure to counteract this overwhelming stress condition resulted in the induction of the apoptotic pathway and the generation of the glial scar. Finally, bilirubin induced the autophagy pathway in the stages preceding death of the animals. Conclusions This study demonstrates that inflammation is a key contributor to bilirubin damage that cooperates with ER stress in the onset of neurotoxicity. Pharmacological modulation of the inflammatory pathway may be a potential intervention target to ameliorate neonatal lethality in Ugt1-/- mice. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0838-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Simone Vodret
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 99, 34149, Trieste, Italy
| | - Giulia Bortolussi
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 99, 34149, Trieste, Italy.
| | - Jana Jašprová
- Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University, 120 00, Prague, Czech Republic
| | - Libor Vitek
- Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University, 120 00, Prague, Czech Republic.,Fourth Department of Internal Medicine, First Faculty of Medicine, Charles University, 120 00, Prague, Czech Republic
| | - Andrés F Muro
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 99, 34149, Trieste, Italy.
| |
Collapse
|
19
|
Qaisiya M, Brischetto C, Jašprová J, Vitek L, Tiribelli C, Bellarosa C. Bilirubin-induced ER stress contributes to the inflammatory response and apoptosis in neuronal cells. Arch Toxicol 2016; 91:1847-1858. [PMID: 27578021 DOI: 10.1007/s00204-016-1835-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 08/24/2016] [Indexed: 02/06/2023]
Abstract
Unconjugated bilirubin (UCB) in newborns may lead to bilirubin neurotoxicity. Few studies investigated the activation of endoplasmic reticulum stress (ER stress) by UCB. We performed an in vitro comparative study using undifferentiated SH-SY5Y, differentiated GI-ME-N neuronal cells and human U87 astrocytoma cells. ER stress and its contribution to inflammation and apoptosis induced by UCB were analyzed. Cytotoxicity, ER stress and inflammation were observed only in neuronal cells, despite intracellular UCB accumulation in all three cell types. UCB toxicity was enhanced in undifferentiated SH-SY5Y cells and correlated with a higher mRNA expression of pro-apoptotic CHOP. Mouse embryonic fibroblast knockout for CHOP and CHOP siRNA-silenced SH-SY5Y increased cells viability upon UCB exposure. In SH-SY5Y, ER stress inhibition by 4-phenylbutyric acid reduced UCB-induced apoptosis and decreased the cleaved forms of caspase-3 and PARP proteins. Reporter gene assay and PERK siRNA showed that IL-8 induction by UCB is transcriptionally regulated by NFкB and PERK signaling. These data suggest that ER stress has an important role in the UCB-induced inflammation and apoptosis, and that targeting ER stress may represent a potential therapeutic approach to decrease UCB-induced neurotoxicity.
Collapse
Affiliation(s)
- Mohammed Qaisiya
- Fondazione Italiana Fegato ONLUS, Italian Liver Foundation ONLUS, AREA Science Park Basovizza Bldg Q, 34149, Trieste, Italy.
| | - Cristina Brischetto
- Fondazione Italiana Fegato ONLUS, Italian Liver Foundation ONLUS, AREA Science Park Basovizza Bldg Q, 34149, Trieste, Italy
| | - Jana Jašprová
- Institute of Medical Biochemistry and Laboratory Medicine, 1st Faculty of Medicine, Charles University in Prague, 12000, Prague, Czech Republic
| | - Libor Vitek
- Institute of Medical Biochemistry and Laboratory Medicine, 1st Faculty of Medicine, Charles University in Prague, 12000, Prague, Czech Republic.,4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University in Prague, 12000, Prague, Czech Republic
| | - Claudio Tiribelli
- Fondazione Italiana Fegato ONLUS, Italian Liver Foundation ONLUS, AREA Science Park Basovizza Bldg Q, 34149, Trieste, Italy.,Department of Medical Sciences, University of Trieste, 34149, Trieste, Italy
| | - Cristina Bellarosa
- Fondazione Italiana Fegato ONLUS, Italian Liver Foundation ONLUS, AREA Science Park Basovizza Bldg Q, 34149, Trieste, Italy
| |
Collapse
|
20
|
Tomasini MC, Borelli AC, Beggiato S, Tanganelli S, Loche A, Cacciaglia R, Ferraro L, Antonelli T. GET73 Prevents Ethanol-Induced Neurotoxicity in Primary Cultures of Rat Hippocampal Neurons. Alcohol Alcohol 2015; 51:128-35. [PMID: 26271115 DOI: 10.1093/alcalc/agv094] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 07/23/2015] [Indexed: 12/29/2022] Open
Abstract
AIMS N-[(4-trifluoromethyl) benzyl] 4-methoxybutyramide (GET73) may be considered a promising therapeutic agent for the treatment of alcohol use disorders. The compound displayed anti-alcohol and anxiolytic properties in rat. In the present study, an in vitro experimental model of chronic ethanol treatment was used to investigate the ability of the compound to counteract the ethanol-induced neurotoxicity. METHODS Primary cultures of rat hippocampal neurons were exposed to ethanol (75 mM; 4 days) and the neuroprotective effects of GET73 were assessed by evaluating cell viability, cell morphology, glutamate levels and reactive oxygen species production. RESULTS The exposure to ethanol induced a reduction of cell viability, an alteration of cytoskeleton, a decrease in extracellular glutamate levels and an increase of reactive oxygen species production. The addiction of GET73 (1 and 10 µM) 1 h before and during chronic ethanol exposure prevented all the above ethanol-induced effects. Based on the proposed GET73 mechanism of action, the effects of mGlu5 receptor negative allosteric modulator, 2-methyl-6-(phenylethynyl)-pyridine (MPEP), on ethanol-induced reduction of cell viability were also assessed. The results indicated that the addiction of MPEP (100 µM) 1 h before and during chronic ethanol exposure prevented the ethanol-induced cell viability reduction. CONCLUSION The present findings provide the first evidence that GET73 shows a neuroprotective role against ethanol-induced neurotoxicity in primary cultures of rat hippocampal neurons. Together with previous findings, these results suggest that GET73 possesses multifaceted properties thus lending further support to the significance of developing GET73 as a therapeutic tool for use in the treatment of alcohol use disorders.
Collapse
Affiliation(s)
- Maria C Tomasini
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy IRET Foundation, Ozzano Emilia, Bologna, Italy
| | - Andrea C Borelli
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Sarah Beggiato
- IRET Foundation, Ozzano Emilia, Bologna, Italy Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Sergio Tanganelli
- IRET Foundation, Ozzano Emilia, Bologna, Italy Department of Medical Sciences, University of Ferrara, Ferrara, Italy LTTA Centre, University of Ferrara, Ferrara, Italy
| | | | | | - Luca Ferraro
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy IRET Foundation, Ozzano Emilia, Bologna, Italy LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Tiziana Antonelli
- IRET Foundation, Ozzano Emilia, Bologna, Italy Department of Medical Sciences, University of Ferrara, Ferrara, Italy LTTA Centre, University of Ferrara, Ferrara, Italy
| |
Collapse
|
21
|
Impairment of enzymatic antioxidant defenses is associated with bilirubin-induced neuronal cell death in the cerebellum of Ugt1 KO mice. Cell Death Dis 2015; 6:e1739. [PMID: 25950469 PMCID: PMC4669693 DOI: 10.1038/cddis.2015.113] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 03/16/2015] [Accepted: 03/18/2015] [Indexed: 01/14/2023]
Abstract
Severe hyperbilirubinemia is toxic during central nervous system development. Prolonged and uncontrolled high levels of unconjugated bilirubin lead to bilirubin-induced encephalopathy and eventually death by kernicterus. Despite extensive studies, the molecular and cellular mechanisms of bilirubin toxicity are still poorly defined. To fill this gap, we investigated the molecular processes underlying neuronal injury in a mouse model of severe neonatal jaundice, which develops hyperbilirubinemia as a consequence of a null mutation in the Ugt1 gene. These mutant mice show cerebellar abnormalities and hypoplasia, neuronal cell death and die shortly after birth because of bilirubin neurotoxicity. To identify protein changes associated with bilirubin-induced cell death, we performed proteomic analysis of cerebella from Ugt1 mutant and wild-type mice. Proteomic data pointed-out to oxidoreductase activities or antioxidant processes as important intracellular mechanisms altered during bilirubin-induced neurotoxicity. In particular, they revealed that down-representation of DJ-1, superoxide dismutase, peroxiredoxins 2 and 6 was associated with hyperbilirubinemia in the cerebellum of mutant mice. Interestingly, the reduction in protein levels seems to result from post-translational mechanisms because we did not detect significant quantitative differences in the corresponding mRNAs. We also observed an increase in neuro-specific enolase 2 both in the cerebellum and in the serum of mutant mice, supporting its potential use as a biomarker of bilirubin-induced neurological damage. In conclusion, our data show that different protective mechanisms fail to contrast oxidative burst in bilirubin-affected brain regions, ultimately leading to neurodegeneration.
Collapse
|
22
|
Cross-talk between neurons and astrocytes in response to bilirubin: adverse secondary impacts. Neurotox Res 2015; 26:1-15. [PMID: 24122290 DOI: 10.1007/s12640-013-9427-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 09/23/2013] [Accepted: 09/24/2013] [Indexed: 12/25/2022]
Abstract
Previous studies using monotypic nerve cell cultures have shown that bilirubin-induced neurological dysfunction (BIND) involves apoptosis and necrosis-like cell death, following neuritic atrophy and astrocyte activation,and that glycoursodeoxycholic acid (GUDCA) has therapeutic efficacy against BIND. Cross-talk between neurons and astrocytes may protect or aggravate neurotoxicity by unconjugated bilirubin (UCB). In a previous work we have shown that bidirectional signaling during astrocyte-neuron recognition attenuates neuronal damage by UCB. Here, we investigated whether the establishment of neuron-astrocyte homeostasis prior to cell exposure to UCB was instead associated with a lower resistance of neurons to UCB toxicity, and if the pro-survival properties of GUDCA were replicated in that experimental model. We have introduced a 24 h adaptation period for neuron-glia communication prior to the 48 h treatment with UCB. In such conditions, UCB induced glial activation, which aggravated neuronal damage, comprising increased apoptosis,cell demise and neuritic atrophy, which were completely prevented in the presence of GUDCA. Neuronal multidrug resistance-associated protein 1 expression and tumor necrosis factor-a secretion, although unchanged by UCB, increased in the presence of astrocytes. The rise in S100B and nitric oxide in the co-cultures medium may have contributed to UCB neurotoxicity. Since the levels of these diffusible molecules did not change by GUDCA we may assume that they are not directly involved in its beneficial effects. Data indicate that astrocytes, in an indirect neuron-astrocyte co-culture model and after homeostatic setting regulation of the system, are critically influencing neurodegeneration by UCB, and support GUDCA for the prevention of BIND.
Collapse
|
23
|
Hu W, Cheng X, Ye X, Zhao L, Huang Y, Zhu H, Yan Z, Wang X, Wang X, Bai G, Gao H. Ex vivo (1)H nuclear magnetic resonance spectroscopy reveals systematic alterations in cerebral metabolites as the key pathogenetic mechanism of bilirubin encephalopathy. Mol Brain 2014; 7:87. [PMID: 25424547 PMCID: PMC4252999 DOI: 10.1186/s13041-014-0087-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 11/13/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Bilirubin encephalopathy (BE) is a severe neurologic sequelae induced by hyperbilirubinemia in newborns. However, the pathogenetic mechanisms underlying the clinical syndromes of BE remain ambiguous. Ex vivo (1)H nuclear magnetic resonance (NMR) spectroscopy was used to measure changes in the concentrations of cerebral metabolites in various brain areas of newborn 9-day-old rats subjected to bilirubin to explore the related mechanisms of BE. RESULTS When measured 0.5 hr after injection of bilirubin, levels of the amino acid neurotransmitters glutamate (Glu), glutamine (Gln), and γ-aminobutyric acid (GABA) in hippocampus and occipital cortex significantly decreased, by contrast, levels of aspartate (Asp) considerably increased. In the cerebellum, Glu and Gln levels significantly decreased, while GABA, and Asp levels showed no significant differences. In BE 24 hr rats, all of the metabolic changes observed returned to normal in the hippocampus and occipital cortex; however, levels of Glu, Gln, GABA, and glycine significantly increased in the cerebellum. CONCLUSIONS These metabolic changes for the neurotransmitters are mostly likely the result of a shift in the steady-state equilibrium of the Gln-Glu-GABA metabolic cycle between astrocytes and neurons, in a region-specific manner. Changes in energy metabolism and the tricarboxylic acid cycle may also be involved in the pathogenesis of BE.
Collapse
Affiliation(s)
- Wenyi Hu
- Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Xiaojie Cheng
- Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Xinjian Ye
- Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Liangcai Zhao
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Yanan Huang
- Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Huanle Zhu
- Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Zhihan Yan
- Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Xuebao Wang
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Xiaojie Wang
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Guanghui Bai
- Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Hongchang Gao
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
24
|
Tidmarsh GF, Wong RJ, Stevenson DK. End-tidal carbon monoxide and hemolysis. J Perinatol 2014; 34:577-81. [PMID: 24743136 DOI: 10.1038/jp.2014.66] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 02/21/2014] [Accepted: 03/13/2014] [Indexed: 12/13/2022]
Abstract
Hemolytic disease in newborns can result from a number of conditions, which can place such infants at an increased risk for the development of severe hyperbilirubinemia. Because the catabolism of heme produces equimolar amounts of carbon monoxide (CO) and bilirubin, measurements of end-tidal breath CO (corrected for ambient CO) or ETCOc can serve as an index of hemolysis as well as of bilirubin production from any cause. Elevated levels of ETCOc have been correlated with blood carboxyhemoglobin levels and thus hemolysis. However, the detection of hemolysis can be a clinically challenging problem in newborns. Here, we describe the importance of determining ETCOc levels and their application in identifying infants at risk for developing hyperbilirubinemia associated with hemolysis and other causes of increased bilirubin production.
Collapse
Affiliation(s)
- G F Tidmarsh
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - R J Wong
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - D K Stevenson
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
25
|
Qaisiya M, Coda Zabetta CD, Bellarosa C, Tiribelli C. Bilirubin mediated oxidative stress involves antioxidant response activation via Nrf2 pathway. Cell Signal 2013; 26:512-20. [PMID: 24308969 DOI: 10.1016/j.cellsig.2013.11.029] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 11/21/2013] [Accepted: 11/26/2013] [Indexed: 12/22/2022]
Abstract
Unconjugated bilirubin (UCB) is responsible for neonatal jaundice and high level of free bilirubin (Bf) can lead to kernicterus. Previous studies suggest that oxidative stress is a critical component of UCB-induced neurotoxicity. The Nrf2 pathway is a powerful sensor for cellular redox state and is activated directly by oxidative stress and/or indirectly by stress response protein kinases. Activated Nrf2 translocates to nucleus, binds to Antioxidant Response Element (ARE), and enhances the up-regulation of cytoprotective genes that mediate cell survival. The aim of the present study was to investigate the role of Nrf2 pathway in cell response to bilirubin mediated oxidative stress in the neuroblastoma SH-SY5Y cell line. Cells exposed to a toxic concentration of UCB (140 nM Bf) showed an increased intracellular ROS levels and enhanced nuclear accumulation of Nrf2 protein. UCB stimulated transcriptional induction of ARE-GFP reporter gene and induced mRNA expression of multiple antioxidant response genes as: xCT, Gly1, γGCL-m, γGCL-c, HO-1, NQO1, FTH, ME1, and ATF3. Nrf2 siRNA decreased UCB induced mRNA expression of HO1 (75%), NQO1 (54%), and FTH (40%). The Nrf2-related HO-1 induction was reduced to 60% in cells pre-treated with antioxidant (NAC) or specific signaling pathway inhibitors for PKC, P38α and MEK1/2 (80, 40 and 25%, respectively). In conclusion, we demonstrated that SH-SY5Y cells undergo an adaptive response against UCB-mediated oxidative stress by activation of multiple antioxidant response, in part through Nrf2 pathway.
Collapse
Affiliation(s)
- Mohammed Qaisiya
- Fondazione Italiana Fegato ONLUS, Italian Liver Foundation ONLUS, Bldg Q AREA Science Park - Basovizza Campus, 34149 Trieste, Italy.
| | - Carlos Daniel Coda Zabetta
- Fondazione Italiana Fegato ONLUS, Italian Liver Foundation ONLUS, Bldg Q AREA Science Park - Basovizza Campus, 34149 Trieste, Italy.
| | - Cristina Bellarosa
- Fondazione Italiana Fegato ONLUS, Italian Liver Foundation ONLUS, Bldg Q AREA Science Park - Basovizza Campus, 34149 Trieste, Italy.
| | - Claudio Tiribelli
- Fondazione Italiana Fegato ONLUS, Italian Liver Foundation ONLUS, Bldg Q AREA Science Park - Basovizza Campus, 34149 Trieste, Italy; Department of Medical Sciences, University of Trieste, 34100 Trieste, Italy.
| |
Collapse
|
26
|
Cytochrome P450 2A5 and bilirubin: Mechanisms of gene regulation and cytoprotection. Toxicol Appl Pharmacol 2013; 270:129-38. [DOI: 10.1016/j.taap.2013.04.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 04/16/2013] [Accepted: 04/18/2013] [Indexed: 12/19/2022]
|
27
|
Lidong Z, Xiaoquan W, Tao C, Weiwei G, Chang L, Shiming Y. Hyperbilirubinemia and Auditory Neuropathy. J Otol 2013. [DOI: 10.1016/s1672-2930(13)50001-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
28
|
Erdinc K, Sarici SU, Akgul EO, Agilli M, Ozcan O. Relationship between neonatal adrenomedullin and bilirubin levels. J Matern Fetal Neonatal Med 2013; 27:30-5. [DOI: 10.3109/14767058.2013.799655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
29
|
Cross-Talk Between Neurons and Astrocytes in Response to Bilirubin: Early Beneficial Effects. Neurochem Res 2013; 38:644-59. [DOI: 10.1007/s11064-012-0963-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 12/17/2012] [Accepted: 12/21/2012] [Indexed: 12/31/2022]
|
30
|
Tricellulin expression in brain endothelial and neural cells. Cell Tissue Res 2012; 351:397-407. [DOI: 10.1007/s00441-012-1529-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 11/08/2012] [Indexed: 10/27/2022]
|
31
|
Babu R, Bagley JH, Di C, Friedman AH, Adamson C. Thrombin and hemin as central factors in the mechanisms of intracerebral hemorrhage-induced secondary brain injury and as potential targets for intervention. Neurosurg Focus 2012; 32:E8. [PMID: 22463118 DOI: 10.3171/2012.1.focus11366] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stoke that may cause significant morbidity and mortality. Brain injury due to ICH initially occurs within the first few hours as a result of mass effect due to hematoma formation. However, there is increasing interest in the mechanisms of secondary brain injury as many patients continue to deteriorate clinically despite no signs of rehemorrhage or hematoma expansion. This continued insult after primary hemorrhage is believed to be mediated by the cytotoxic, excitotoxic, oxidative, and inflammatory effects of intraparenchymal blood. The main factors responsible for this injury are thrombin and erythrocyte contents such as hemoglobin. Therapies including thrombin inhibitors, N-methyl-D-aspartate antagonists, chelators to bind free iron, and antiinflammatory drugs are currently under investigation for reducing this secondary brain injury. This review will discuss the molecular mechanisms of brain injury as a result of intraparenchymal blood, potential targets for therapeutic intervention, and treatment strategies currently in development.
Collapse
Affiliation(s)
- Ranjith Babu
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | |
Collapse
|
32
|
Lipid peroxidation is not the primary mechanism of bilirubin-induced neurologic dysfunction in jaundiced Gunn rat pups. Pediatr Res 2012; 72:455-9. [PMID: 22902434 DOI: 10.1038/pr.2012.111] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Hazardous levels of bilirubin produce oxidative stress in vitro and may play a role in the genesis of bilirubin-induced neurologic dysfunction (BIND). We hypothesized that the antioxidants taurourosdeoxycholic acid (TUDCA), 12S-hydroxy-1,12-pyrazolinominocycline (PMIN), and minocycline (MNC) inhibit oxidative stress and block BIND in hyperbilirubinemic j/j Gunn rat pups that were given sulfadimethoxine to induce bilirubin encephalopathy. METHODS At peak postnatal hyperbilirubinemia, j/j Gunn rat pups were dosed with sulfadimethoxine to induce bilirubin encephalopathy. Pups were given TUDCA, PMIN, MNC, or vehicle pretreatment (15 min before sulfadimethoxine). After 24 h, BIND was scored by using a rating scale of neurobehavior and cerebellar tissue 4-hydroxynonenal and protein carbonyl dinitrophenyl content were determined. Nonjaundiced heterozygous N/j pups served as controls. RESULTS Administration of sulfadimethoxine induced BIND and lipid peroxidation but not protein oxidation in hyperbilirubinemic j/j pups. TUDCA, PMIN, and MNC each reduced lipid peroxidation to basal levels observed in nonjaundiced N/j controls, but only MNC prevented BIND. CONCLUSION These findings show that lipid peroxidation inhibition alone is not sufficient to prevent BIND. We speculate that the neuroprotective efficacy of MNC against BIND involves action(s) independent of, or in addition to, its antioxidant effects.
Collapse
|
33
|
Khan NM, Poduval TB. Bilirubin augments radiation injury and leads to increased infection and mortality in mice: molecular mechanisms. Free Radic Biol Med 2012; 53:1152-69. [PMID: 22819982 DOI: 10.1016/j.freeradbiomed.2012.07.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 07/08/2012] [Indexed: 01/15/2023]
Abstract
Our earlier results demonstrated that clinically relevant concentrations of unconjugated bilirubin (UCB) possessed immunotoxic effects. Whole-body irradiation (WBI) with 1 to 6 Gy leads to acute radiation syndrome, immunosuppression, and makes the host susceptible to infection. Since hyperbilirubinemia has been shown to be associated with several types of cancer, the present studies were undertaken to evaluate the radiomodifying effects of UCB in radiation-exposed mice having elevated levels of UCB. Pretreatment of splenic lymphocytes with UCB (1-50 μM at UCB/BSA ratio <1) augmented radiation-induced DNA strand breaks, MMP loss, calcium release, and apoptosis. Combination treatment of mice with UCB (50mg/kg bw) followed by WBI (2 Gy) 0.5h later, resulted in significantly increased splenic atrophy, bone marrow aplasia, decreased counts of peritoneal exudate cells, and different splenocyte subsets such as CD3+ T, CD4+ T, CD8+ T, CD19+ B, and CD14+ macrophages as compared to either UCB or WBI treatment. Hematological studies showed that WBI-induced lymphopenia, thrombocytopenia, and neutropenia were further aggravated in the combination treatment group. UCB pretreatment of mice potentiated WBI-induced apoptosis and decreased WBI-induced loss of functional response of various immune cells leading to augmentation of immunosuppression and infection susceptibility caused by WBI. In an acute bacterial peritonitis model, UCB pretreatment of mice significantly increased WBI-induced proinflammatory cytokines, nitric oxide, and peritoneal bacterial load resulting in increased infection and death. Studies using the pharmacological inhibitor of p38MAPK demonstrated the involvement of p38MAPK activation in the inflammatory cascade of peritonitis. These findings should prove useful in understanding the potential risk to hyperbilirubinemic patients during radiotherapy and victims of acute radiation exposure in the course of radiation accidents.
Collapse
Affiliation(s)
- Nazir M Khan
- Immunology and Hyperthermia Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India.
| | | |
Collapse
|
34
|
ER Stress, Mitochondrial Dysfunction and Calpain/JNK Activation are Involved in Oligodendrocyte Precursor Cell Death by Unconjugated Bilirubin. Neuromolecular Med 2012; 14:285-302. [DOI: 10.1007/s12017-012-8187-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 06/01/2012] [Indexed: 12/24/2022]
|
35
|
Brites D. The evolving landscape of neurotoxicity by unconjugated bilirubin: role of glial cells and inflammation. Front Pharmacol 2012; 3:88. [PMID: 22661946 PMCID: PMC3361682 DOI: 10.3389/fphar.2012.00088] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 04/23/2012] [Indexed: 12/13/2022] Open
Abstract
Unconjugated hyperbilirubinemia is a common condition in the first week of postnatal life. Although generally harmless, some neonates may develop very high levels of unconjugated bilirubin (UCB), which may surpass the protective mechanisms of the brain in preventing UCB accumulation. In this case, both short-term and long-term neurodevelopmental disabilities, such as acute and chronic UCB encephalopathy, known as kernicterus, or more subtle alterations defined as bilirubin-induced neurological dysfunction (BIND) may be produced. There is a tremendous variability in babies' vulnerability toward UCB for reasons not yet explained, but preterm birth, sepsis, hypoxia, and hemolytic disease are comprised as risk factors. Therefore, UCB levels and neurological abnormalities are not strictly correlated. Even nowadays, the mechanisms of UCB neurotoxicity are still unclear, as are specific biomarkers, and little is known about lasting sequelae attributable to hyperbilirubinemia. On autopsy, UCB was shown to be within neurons, neuronal processes, and microglia, and to produce loss of neurons, demyelination, and gliosis. In isolated cell cultures, UCB was shown to impair neuronal arborization and to induce the release of pro-inflammatory cytokines from microglia and astrocytes. However, cell dependent sensitivity to UCB toxicity and the role of each nerve cell type remains not fully understood. This review provides a comprehensive insight into cell susceptibilities and molecular targets of UCB in neurons, astrocytes, and oligodendrocytes, and on phenotypic and functional responses of microglia to UCB. Interplay among glia elements and cross-talk with neurons, with a special emphasis in the UCB-induced immunostimulation, and the role of sepsis in BIND pathogenesis are highlighted. New and interesting data on the anti-inflammatory and antioxidant activities of different pharmacological agents are also presented, as novel and promising additional therapeutic approaches to BIND.
Collapse
Affiliation(s)
- Dora Brites
- Neuron Glia Biology in Health and Disease Unit, Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon Lisbon, Portugal
| |
Collapse
|
36
|
Gazzin S, Strazielle N, Tiribelli C, Ghersi-Egea JF. Transport and metabolism at blood-brain interfaces and in neural cells: relevance to bilirubin-induced encephalopathy. Front Pharmacol 2012; 3:89. [PMID: 22629246 PMCID: PMC3355510 DOI: 10.3389/fphar.2012.00089] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 04/25/2012] [Indexed: 01/16/2023] Open
Abstract
Bilirubin, the end-product of heme catabolism, circulates in non-pathological plasma mostly as a protein-bound species. When bilirubin concentration builds up, the free fraction of the molecule increases. Unbound bilirubin then diffuses across blood-brain interfaces (BBIs) into the brain, where it accumulates and exerts neurotoxic effects. In this classical view of bilirubin neurotoxicity, BBIs act merely as structural barriers impeding the penetration of the pigment-bound carrier protein, and neural cells are considered as passive targets of its toxicity. Yet, the role of BBIs in the occurrence of bilirubin encephalopathy appears more complex than being simple barriers to the diffusion of bilirubin, and neural cells such as astrocytes and neurons can play an active role in controlling the balance between the neuroprotective and neurotoxic effects of bilirubin. This article reviews the emerging in vivo and in vitro data showing that transport and metabolic detoxification mechanisms at the blood-brain and blood-cerebrospinal fluid barriers may modulate bilirubin flux across both cellular interfaces, and that these protective functions can be affected in chronic unconjugated hyperbilirubinemia. Then the in vivo and in vitro arguments in favor of the physiological antioxidant function of intracerebral bilirubin are presented, as well as the potential role of transporters such as ABCC1 and metabolizing enzymes such as cytochromes P-450 in setting the cerebral cell- and structure-specific toxicity of bilirubin following hyperbilirubinemia. The relevance of these data to the pathophysiology of bilirubin-induced neurological diseases is discussed.
Collapse
Affiliation(s)
- Silvia Gazzin
- Italian Liver Foundation, AREA Science Park Basovizza Trieste, Italy
| | | | | | | |
Collapse
|
37
|
Becerir C, Kılıç İ, Şahin Ö, Özdemir Ö, Tokgün O, Özdemir B, Akca H. The protective effect of docosahexaenoic acid on the bilirubin neurotoxicity. J Enzyme Inhib Med Chem 2012; 28:801-7. [DOI: 10.3109/14756366.2012.684053] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | | | | | | | - Onur Tokgün
- Department of Medical Biology,
Denizli, Turkey
| | - Bülent Özdemir
- Department Anatomy, Pamukkale University Faculty of Medicine,
Denizli, Turkey
| | - Hakan Akca
- Department of Medical Biology,
Denizli, Turkey
| |
Collapse
|
38
|
Cardoso FL, Kittel Á, Veszelka S, Palmela I, Tóth A, Brites D, Deli MA, Brito MA. Exposure to lipopolysaccharide and/or unconjugated bilirubin impair the integrity and function of brain microvascular endothelial cells. PLoS One 2012; 7:e35919. [PMID: 22586454 PMCID: PMC3346740 DOI: 10.1371/journal.pone.0035919] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 03/27/2012] [Indexed: 11/21/2022] Open
Abstract
Background Sepsis and jaundice are common conditions in newborns that can lead to brain damage. Though lipopolysaccharide (LPS) is known to alter the integrity of the blood-brain barrier (BBB), little is known on the effects of unconjugated bilirubin (UCB) and even less on the joint effects of UCB and LPS on brain microvascular endothelial cells (BMEC). Methodology/Principal Findings Monolayers of primary rat BMEC were treated with 1 µg/ml LPS and/or 50 µM UCB, in the presence of 100 µM human serum albumin, for 4 or 24 h. Co-cultures of BMEC with astroglial cells, a more complex BBB model, were used in selected experiments. LPS led to apoptosis and UCB induced both apoptotic and necrotic-like cell death. LPS and UCB led to inhibition of P-glycoprotein and activation of matrix metalloproteinases-2 and -9 in mono-cultures. Transmission electron microscopy evidenced apoptotic bodies, as well as damaged mitochondria and rough endoplasmic reticulum in BMEC by either insult. Shorter cell contacts and increased caveolae-like invaginations were noticeable in LPS-treated cells and loss of intercellular junctions was observed upon treatment with UCB. Both compounds triggered impairment of endothelial permeability and transendothelial electrical resistance both in mono- and co-cultures. The functional changes were confirmed by alterations in immunostaining for junctional proteins β-catenin, ZO-1 and claudin-5. Enlargement of intercellular spaces, and redistribution of junctional proteins were found in BMEC after exposure to LPS and UCB. Conclusions LPS and/or UCB exert direct toxic effects on BMEC, with distinct temporal profiles and mechanisms of action. Therefore, the impairment of brain endothelial integrity upon exposure to these neurotoxins may favor their access to the brain, thus increasing the risk of injury and requiring adequate clinical management of sepsis and jaundice in the neonatal period.
Collapse
Affiliation(s)
- Filipa L. Cardoso
- Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - Ágnes Kittel
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Szilvia Veszelka
- Laboratory of Molecular Neurobiology, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Inês Palmela
- Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - Andrea Tóth
- Laboratory of Molecular Neurobiology, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Dora Brites
- Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - Mária A. Deli
- Laboratory of Molecular Neurobiology, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Maria A. Brito
- Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
- * E-mail:
| |
Collapse
|
39
|
Brito MA, Zurolo E, Pereira P, Barroso C, Aronica E, Brites D. Cerebellar axon/myelin loss, angiogenic sprouting, and neuronal increase of vascular endothelial growth factor in a preterm infant with kernicterus. J Child Neurol 2012; 27:615-24. [PMID: 22190497 DOI: 10.1177/0883073811423975] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We performed histologic and immunohistochemical analysis of cerebellar sections from a preterm infant (32 weeks 5 days) dead on the 4th day of life with the diagnosis of kernicterus and compared the results with 1 age-matched nonicteric patient. Poorer Luxol fast blue-periodic acid Schiff and Bodian-Luxol fast blue stainings as well as neurofilament expression were observed in the kernicterus case, indicating loss of axon neurites and myelin fibers. Elevated claudin-5 and cluster of differentiation 34 expression associated with increased blood vessel density suggests bilirubin-induced angiogenic sprouting. Upregulation of vascular endothelial growth factor and its receptor 2 was observed in nucleus dentatus and Purkinje neurons. Although upregulation of multidrug resistance-associated protein 1 was increased in cerebellar neurons, it was not able to prevent bilirubin-induced neurotoxicity. These data add new insights into the pathophysiology of kernicterus, revealing vascular endothelial growth factor and its receptor 2, as well as angiogenic sprouting, as new players in neurologic damage by unconjugated bilirubin.
Collapse
Affiliation(s)
- Maria A Brito
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL) and Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal.
| | | | | | | | | | | |
Collapse
|
40
|
Khan NM, Poduval TB. Immunomodulatory and immunotoxic effects of bilirubin: molecular mechanisms. J Leukoc Biol 2011; 90:997-1015. [PMID: 21807743 DOI: 10.1189/jlb.0211070] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The immunomodulatory and immunotoxic effects of purified UCB have not been evaluated previously at clinically relevant UCB concentrations and UCB:BSA ratios. To delineate the molecular mechanism of UCB-induced immunomodulation, immune cells were exposed to clinically relevant concentrations of UCB. It inhibited LPS-induced B cell proliferation and cytokine production from splenic macrophages. UCB (≥25 μM) was toxic to unfractionated splenocytes, splenic T cells, B cells, macrophages, LPS-stimulated CD19(+) B cells, human PBMCs, and RBCs. Purified UCB also was found to be toxic to splenocytes and human PBMCs. UCB induced necrosis and apoptosis in splenocytes. UCB activated the extrinsic and intrinsic pathways of apoptosis, as reflected by the markers, such as CD95, caspase-8, Bax, MMP, cytoplasmic Ca(+2), caspase-3, and DNA fragmentation. UCB depleted GSH and activated p38MAPK. NAC, caspase inhibitors, and p38MAPK inhibitor attenuated the UCB-induced apoptosis. In vivo administration of ≥25 mg/kbw UCB induced atrophy of spleen, depletion of bone marrow cells, and leukopenia and decreased lymphocyte count and the T and B cell response to mitogens. UCB administration to mice led to induction of oxidative stress, activation of p38MAPK, and cell death in splenocytes. These parameters were attenuated by the injection of NAC and the p38MAPK inhibitor. Our results demonstrate for the first time that clinically relevant concentrations of UCB induce apoptosis and necrosis in immune cells by depleting cellular GSH. These findings should prove useful in understanding the immunosuppression associated with hyperbilirubinemia.
Collapse
Affiliation(s)
- Nazir M Khan
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai, India
| | | |
Collapse
|
41
|
Vaz AR, Silva SL, Barateiro A, Falcão AS, Fernandes A, Brito MA, Brites D. Selective vulnerability of rat brain regions to unconjugated bilirubin. Mol Cell Neurosci 2011; 48:82-93. [PMID: 21708263 DOI: 10.1016/j.mcn.2011.06.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2010] [Revised: 06/06/2011] [Accepted: 06/10/2011] [Indexed: 12/22/2022] Open
Abstract
Hippocampus is one of the brain regions most vulnerable to unconjugated bilirubin (UCB) encephalopathy, although cerebellum also shows selective yellow staining in kernicterus. We previously demonstrated that UCB induces oxidative stress in cortical neurons, disruption of neuronal network dynamics, either in developing cortical or hippocampal neurons, and that immature cortical neurons are more prone to UCB-induced injury. Here, we studied if immature rat neurons isolated from cortex, cerebellum and hippocampus present distinct features of oxidative stress and cell dysfunction upon UCB exposure. We also explored whether oxidative damage and its regulation contribute to neuronal dysfunction induced by hyperbilirubinemia, considering neurite extension and ramification, as well as cell death. Our results show that UCB induces nitric oxide synthase expression, as well as production of nitrites and cyclic guanosine monophosphate in immature neurons, mainly in those from hippocampus. After exposure to UCB, hippocampal neurons presented the highest content of reactive oxygen species, disruption of glutathione redox status and cell death, when compared to neurons from cortex or cerebellum. In particular, the results indicate that cells exposed to UCB undertake an adaptive response that involves DJ-1, a multifunctional neuroprotective protein implicated in the maintenance of cellular oxidation status. However, longer neuronal exposure to UCB caused down-regulation of DJ-1 expression, especially in hippocampal neurons. In addition, a greater impairment in neurite outgrowth and branching following UCB treatment was also noticed in immature neurons from hippocampus. Interestingly, pre-incubation with N-acetylcysteine, a precursor of glutathione synthesis, protected neurons from UCB-induced oxidative stress and necrotic cell death, preventing DJ-1 down-regulation and neuritic impairment. Taken together, these data point to oxidative injury and disruption of neuritic network as hallmarks in hippocampal susceptibility to UCB. Most importantly, they also suggest that local differences in glutathione content may account to the different susceptibility between brain regions exposed to UCB.
Collapse
Affiliation(s)
- Ana Rita Vaz
- Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | | | | | | | | | | | | |
Collapse
|
42
|
Dang TN, Bishop GM, Dringen R, Robinson SR. The metabolism and toxicity of hemin in astrocytes. Glia 2011; 59:1540-50. [PMID: 21681819 DOI: 10.1002/glia.21198] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2010] [Accepted: 05/18/2011] [Indexed: 01/09/2023]
Abstract
Hemin is cytotoxic, and contributes to the brain damage that accompanies hemorrhagic stroke. In order to better understand the basis of hemin toxicity in astrocytes, the present study quantified hemin metabolism and compared it to the pattern of cell death. Heme oxygenase-1 (HO-1) expression was first evident after 2 h incubation with hemin, with maximal expression being observed by 24 h. Despite the induction of HO-1, it was found that the proportion of hemin metabolized by astrocytes remained fairly constant throughout the 24 h period, with 70-80% of intracellular hemin remaining intact. A period of cell loss began after 2 h exposure to hemin, which gradually increased in severity to reach a maximum by 24 h. This cell loss could not be attenuated by the iron chelator, 1,10-phenanthroline, or by several antioxidant compounds (Trolox, N-acetyl-L-cysteine and N-tert-butyl-α-phenylnitrone), indicating that the mechanism of hemin toxicity does not involve iron. While these results make it unlikely that hemin toxicity is due to interactions with endogenous H(2)O(2), hemin toxicity was increased in the presence of supraphysiological levels of H(2)O(2) and this increase was ameliorated by PHEN, indicating that the iron released from hemin can be toxic under some pathological conditions. However, when H(2)O(2) is present at physiological levels, the toxicity of hemin appears to be caused by other mechanisms that may involve bilirubin and carbon monoxide in this model system.
Collapse
Affiliation(s)
- Theresa N Dang
- Blood-Brain Interactions Group, School of Psychology and Psychiatry, Monash University, Clayton, VIC, Australia.
| | | | | | | |
Collapse
|
43
|
Vaz AR, Silva SL, Barateiro A, Fernandes A, Falcão AS, Brito MA, Brites D. Pro-inflammatory cytokines intensify the activation of NO/NOS, JNK1/2 and caspase cascades in immature neurons exposed to elevated levels of unconjugated bilirubin. Exp Neurol 2011; 229:381-90. [DOI: 10.1016/j.expneurol.2011.03.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 03/07/2011] [Indexed: 01/30/2023]
|
44
|
Abstract
Encephalopathy by hyperbilirubinemia in infants has been described for decades, but neither the underlying cellular and molecular mechanisms nor the selective pattern of bilirubin deposition in the brain is well understood. The brain is composed of highly specialized and diverse populations of cells, represented by neurons and glia that comprise astrocytes, oligodendrocytes, and microglia. Although microscopic evaluation of icteric brain sections revealed bilirubin within neurons, neuronal processes, and microglia, cell dependent-sensitivity to bilirubin toxicity and the role of each nerve cell type are poorly understood. Even less considered are glial and neuronal pathologic alterations as integrated phenomena. The available knowledge on reactivity of glial cells to bilirubin and on the impairment to neuronal network dynamics that it causes, here summarized, suggests that a better comprehension of the interplay between neurons and glia is essential to understand bilirubin neurotoxicity and highlight potential molecular targets that may lead to disease-modifying therapeutic approaches.
Collapse
Affiliation(s)
- Dora Brites
- Research Institute for Medicines and Pharmaceutical Sciences (iMedUL), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal.
| |
Collapse
|
45
|
Gazzin S, Berengeno AL, Strazielle N, Fazzari F, Raseni A, Ostrow JD, Wennberg R, Ghersi-Egea JF, Tiribelli C. Modulation of Mrp1 (ABCc1) and Pgp (ABCb1) by bilirubin at the blood-CSF and blood-brain barriers in the Gunn rat. PLoS One 2011; 6:e16165. [PMID: 21297965 PMCID: PMC3031532 DOI: 10.1371/journal.pone.0016165] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 12/07/2010] [Indexed: 01/03/2023] Open
Abstract
Accumulation of unconjugated bilirubin (UCB) in the brain causes bilirubin encephalopathy. Pgp (ABCb1) and Mrp1 (ABCc1), highly expressed in the blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB) respectively, may modulate the accumulation of UCB in brain. We examined the effect of prolonged exposure to elevated concentrations of UCB on expression of the two transporters in homozygous, jaundiced (jj) Gunn rats compared to heterozygous, not jaundiced (Jj) littermates at different developmental stages (2, 9, 17 and 60 days after birth). BBB Pgp protein expression was low in both jj and Jj pups at 9 days (about 16-27% of adult values), despite the up-regulation in jj animals (2 and 1.3 fold higher than age matched Jj animals at P9 and P17-P60, respectively); Mrp1 protein expression was barely detectable. Conversely, at the BCSFB Mrp1 protein expression was rather high (60-70% of the adult values) in both jj and Jj at P2, but was markedly (50%) down-regulated in jj pups starting at P9, particularly in the 4(th) ventricle choroid plexuses: Pgp was almost undetectable. The Mrp1 protein down regulation was accompanied by a modest up-regulation of mRNA, suggesting a translational rather than a transcriptional inhibition. In vitro exposure of choroid plexus epithelial cells obtained from normal rats to UCB, also resulted in a down-regulation of Mrp1 protein. These data suggest that down-regulation of Mrp1 protein at the BSCFB, resulting from a direct effect of UCB on epithelial cells, may impact the Mrp1-mediated neuroprotective functions of the blood-cerebrospinal fluid barrier and actually potentiate UCB neurotoxicity.
Collapse
|
46
|
Fernandes A, Barateiro A, Falcão AS, Silva SLA, Vaz AR, Brito MA, Silva RFM, Brites D. Astrocyte reactivity to unconjugated bilirubin requires TNF-α and IL-1β receptor signaling pathways. Glia 2010; 59:14-25. [PMID: 20967881 DOI: 10.1002/glia.21072] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Revised: 07/30/2010] [Accepted: 08/10/2010] [Indexed: 12/31/2022]
Abstract
Jaundice and sepsis are common neonatal conditions that can lead to neurodevelopment sequelae, namely if present at the same time. We have reported that tumor necrosis factor (TNF)-α and interleukin (IL)-1β are produced by cultured neurons and mainly by glial cells exposed to unconjugated bilirubin (UCB). The effects of these cytokines are mediated by cell surface receptors through a nuclear factor (NF)-κB-dependent pathway that we have showed to be activated by UCB. The present study was designed to evaluate the role of TNF-α and IL-1β signaling on astrocyte reactivity to UCB in rat cortical astrocytes. Exposure of astrocytes to UCB increased the expression of both TNF-α receptor (TNFR)1 and IL-1β receptor (IL-1R)1, but not TNFR2, as well as their activation, observed by augmented binding of receptors' molecular adaptors, TRAF2 and TRAF6, respectively. Silencing of TNFR1, using siRNA technology, or blockade of IL-1β cascade, using its endogenous antagonist, IL-1 receptor antagonist (IL-1ra), prevented UCB-induced cytokine release and NF-κB activation. Interestingly, lack of TNF-α signal transduction reduced UCB-induced cell death for short periods of incubation, although an increase was observed after extended exposure; in contrast, inhibition of IL-1β cascade produced a sustained blockade of astrocyte injury by UCB. Together, our data show that inflammatory pathways are activated during in vitro exposure of rat cortical astrocytes to UCB and that this activation is prolonged in time. This supports the concept that inflammatory pathways play a role in brain damage by UCB, and that they may represent important pharmacological targets.
Collapse
Affiliation(s)
- Adelaide Fernandes
- Research Institute for Medicines and Pharmaceutical Sciences (IMedUL), Faculdade de Farmácia, University of Lisbon, Lisbon, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Oakes GH, Bend JR. Global changes in gene regulation demonstrate that unconjugated bilirubin is able to upregulate and activate select components of the endoplasmic reticulum stress response pathway. J Biochem Mol Toxicol 2010; 24:73-88. [PMID: 20196124 DOI: 10.1002/jbt.20313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Elevated concentrations of unconjugated bilirubin (UCB) are responsible for neonatal jaundice and can eventually lead to kernicterus or death. The molecular mechanism of UCB toxicity is incompletely elucidated. The purpose of this study was to analyze changes in gene regulation mediated by UCB to determine novel pathways that contribute to UCB-mediated toxicity. We employed microarray analysis to determine changes in gene regulation mediated by UCB at both pro- (50 microM) and antioxidant (70 nM) concentrations in Hepa 1c1c7 cells at 1 and 6 h. The changes observed in select genes were validated with qPCR. Using immunoblot analysis, we validated these changes at the protein level for select genes and documented the activation of two proteins involved in the endoplasmic reticulum (ER) stress pathway, eIF2 alpha and PERK. Following treatment with 50 microM UCB, microarray analysis revealed the upregulation of many genes involved in ER stress (ATF3, BiP, CHOP, Dnajb1, and Herp). We demonstrate that upregulation of the proapoptotic transcription factor CHOP results in increased intracellular protein content. It was determined that activation of proteins involved in ER stress was an early event in UCB toxicity as eIF2 alpha and PERK were both phosphorylated and activated by 1 h posttreatment. We also demonstrate that procaspase-12 content, a proposed initiator caspase in ER stress-mediated apoptosis, is decreased by 4 h posttreatment. In conclusion, this study demonstrates that elevated concentrations of UCB (50 microM) are able to activate select components of the ER stress pathway in Hepa 1c1c7 cells, which may contribute to UCB-mediated apoptosis.
Collapse
Affiliation(s)
- Garth H Oakes
- Department of Physiology & Pharmacology, Siebens-Drake Medical Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, 1400 Western Road, London, Ontario, Canada
| | | |
Collapse
|
48
|
Brito MA, Vaz AR, Silva SL, Falcão AS, Fernandes A, Silva RFM, Brites D. N-methyl-aspartate receptor and neuronal nitric oxide synthase activation mediate bilirubin-induced neurotoxicity. Mol Med 2010; 16:372-80. [PMID: 20593111 PMCID: PMC2935951 DOI: 10.2119/molmed.2009.00152] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Accepted: 06/28/2010] [Indexed: 01/02/2023] Open
Abstract
Hyperbilirubinemia may lead to neurotoxicity and neuronal death. Although the mechanisms of nerve cell damage by unconjugated bilirubin (UCB) appear to involve a disruption of the redox status and excitotoxicity, the contribution of nitric oxide (NO·) and of N-methyl-D-aspartate (NMDA) glutamate receptors is unclear. We investigated the role of NO· and NMDA glutamate receptors in the pathways of nerve cell demise by UCB. Neurons were incubated with 100 micromol/L UCB, in the presence of 100 micromol/L human serum albumin for 4 h at 37ºC, alone or in combination with N-ω-nitro-L-arginine methyl ester (L-NAME) (an inhibitor of neuronal nitric oxide synthase [nNOS]), hemoglobin (an NO· scavenger) or (+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate (MK-801) (an NMDA-receptor antagonist). Exposure to UCB led to increased expression of nNOS and production of both NO· and cyclic guanosine 3',5'-monophosphate (cGMP), along with protein oxidation and depletion of glutathione. These events concurred for cell dysfunction and death and were counteracted by L-NAME. Moreover, the UCB-induced loss of neuronal viability was abolished by hemoglobin, whereas the activation of nNOS and production of both NO· and cGMP were counteracted by MK-801, resulting in significant protection from cell dysfunction and death. These results reinforce the involvement of oxidative stress by showing that nerve cell damage by UCB is mediated by NO· and therefore is counteracted by NO· inhibitors or scavengers. Our findings strongly suggest that the activation of nNOS and neurotoxicity occur through the engagement of NMDA receptors. These data reveal a role for overstimulation of glutamate receptors in mediating oxidative damage by UCB.
Collapse
Affiliation(s)
- Maria A Brito
- Research Institute for Medicines and Pharmaceutical Sciences, Faculdade de Farmácia, University of Lisbon, Lisbon, Portugal.
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Despite a century of research, several clinically relevant areas of bilirubin biochemistry remain controversial, poorly understood, or unrecognized. These include: (i) The structure and molecularity of bilirubin under physiological environments such as membranes, brain tissue and when bound to proteins. Related to this is the large number of structurally different bilirubin species that may occur in blood under pathological conditions and their potential effects on measurements of bilirubin and free bilirubin. (ii) The mechanism of phototherapy, the neurotoxicity of the photoisomers produced and their influence on measurements of bilirubin and free bilirubin. (iii) The role of membrane transporters in the passage of unconjugated bilirubin across the placenta, intestine, vascular epithelium, blood-brain barrier, and into the liver. (iv) Biochemical mechanisms of bilirubin toxicity, pharmacologic prevention of kernicterus, the contribution of bilirubin to antioxidant defenses, and the practical value of free bilirubin measurements for identifying infants at most risk of kernicterus.
Collapse
|
50
|
Deganuto M, Cesaratto L, Bellarosa C, Calligaris R, Vilotti S, Renzone G, Foti R, Scaloni A, Gustincich S, Quadrifoglio F, Tiribelli C, Tell G. A proteomic approach to the bilirubin-induced toxicity in neuronal cells reveals a protective function of DJ-1 protein. Proteomics 2010; 10:1645-57. [DOI: 10.1002/pmic.200900579] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|