1
|
Rathod SS, Agrawal YO. β-Caryophyllene (CB2 agonist) mitigates rotenone-induced neurotoxicity and apoptosis in SH-SY5Y neuroblastoma cells via modulation of GSK-3β/NRF2/HO-1 axis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04281-x. [PMID: 40410551 DOI: 10.1007/s00210-025-04281-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 05/08/2025] [Indexed: 05/25/2025]
Abstract
Rotenone-induced neurotoxicity in SH-SY5Y cells is an essential hallmark of neurodegenerative diseases like Alzheimer's disease (AD) and Parkinson's disease (PD). β-Caryophyllene (BCP), a cannabinoid receptor 2 (CB2) agonist, has anti-inflammatory, antioxidant, and cytoprotective efficacy. The involvement of the GSK-3β/NRF2/HO-1 axis in neuroprotection has garnered attention as a possible mechanism for BCP to exhibit multitargeted neuroprotective effects. Hence, this study investigates the effects of BCP against rotenone-induced neurotoxicity and apoptosis in SH-SY5Y cells, focusing on the involvement of the GSK-3β/NRF2/HO-1 signaling pathway. Initially, we performed the in silico molecular docking of BCP with GSK-3β, NRF2, and HO-1 proteins to ensure the degree of binding affinities. The in vitro MTT assay was performed to evaluate cell viability, followed by the assessment of biomarkers such as LDH leakage, oxidative stress, reactive species, caspase 3 activity, pro-inflammatory markers, and GSK-3β, NRF2, and HO-1 proteins in BCP, as well as specific receptor modulators (chir98023 and quercetin) against the rotenone pre-treated cells. In silico molecular docking studies revealed that BCP exhibits a strong binding affinity for GSK-3β, NRF2, and HO-1 proteins. Also, in vitro studies revealed that BCP (100 µg/ml), as compared to the rotenone-treated group, significantly restored cell viability (72%). Moreover, BCP significantly modulates cell cytotoxicity (LDH leakage), pro-apoptotic, pro-inflammatory, reactive species, and oxidative stress markers. Molecular docking established robust binding affinities of BCP with GSK-3β, NRF2, and HO-1 proteins. Furthermore, protein estimation by ELISA confirmed the BCP-mediated modulation of these pathways. These findings suggest that BCP protects SH-SY5Y cells from rotenone-induced neurotoxicity, offering a potential therapeutic candidate for neurodegenerative diseases like AD.
Collapse
Affiliation(s)
- Sumit S Rathod
- R. C. Patel Institute of Pharmaceutical Education and Research, Dist. Dhule, Shirpur, 425405, Maharashtra, India
| | - Yogeeta O Agrawal
- R. C. Patel Institute of Pharmaceutical Education and Research, Dist. Dhule, Shirpur, 425405, Maharashtra, India.
| |
Collapse
|
2
|
Sheikh MR, Mahmud HH, Hossen MS, Saha D, Uddin ME, Hossain MF, Munshi MK, Sina AAI. Modeling the Interactions Between Chemicals and Proteins to Predict the Health Consequences of Air Pollution. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2025; 22:418. [PMID: 40238577 PMCID: PMC11942511 DOI: 10.3390/ijerph22030418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/04/2025] [Accepted: 03/08/2025] [Indexed: 04/18/2025]
Abstract
The impacts of air pollution on human health have become a major concern, especially with rising greenhouse gas emissions and urban development. This study investigates the molecular mechanisms using the STITCH 4.0 and STRING 9.0 databases to analyze the interaction networks (PCI and PPI) associated with two air pollutants: carbon monoxide and hydrogen sulfide. The functional and pathway analysis related to these pollutants were performed by OmicsBox v.3.0. Additionally, critical proteins and their essential pathways were also identified by the Cytoscape networking tool v.3.10.3. AutoDock vina was employed to hypothetically determine the direct interactions of CO and H2S with the proteins that were found by STITCH. This study revealed that CO and H2S interacted with the different biological processes related to human health, including erythropoiesis, oxidative stress, energy production, amino acids metabolism, and multiple signaling pathways associated with respiratory, cardiovascular, and neurological functions. Six essential proteins were identified based on their degree of centrality, namely, FECH, HMOX1, ALB, CTH, CBS, and CBSL, which regulate various Reactome and KEGG pathways. Molecular docking analysis revealed that CO exhibited a strong interaction with ADI1, demonstrating a binding affinity of -1.9 kcal/mL. Alternately, the binding energy associated with the H2S interaction was notably weak (below -0.9 kcal/mL). This present research highlights the necessity for ongoing investigation into the molecular effects of air pollution to guide public health policies and interventions.
Collapse
Affiliation(s)
- Md. Ramjan Sheikh
- Department of Biochemistry and Molecular Biology, Gono Bishwabidyalay, Dhaka 1344, Bangladesh; (M.R.S.); (M.E.U.); (M.F.H.)
| | - Hasna Heena Mahmud
- Department of Disaster Science and Climate Resilience, University of Dhaka, Dhaka 1000, Bangladesh;
| | - Md. Saikat Hossen
- Department of Soil and Environmental Sciences, University of Barishal, Barishal 8254, Bangladesh;
| | - Disha Saha
- Department of Crop Physiology and Ecology, Hajee Mohammad Danesh Science and Technology University, Dinajpur 5200, Bangladesh;
| | - Md. Ekhlas Uddin
- Department of Biochemistry and Molecular Biology, Gono Bishwabidyalay, Dhaka 1344, Bangladesh; (M.R.S.); (M.E.U.); (M.F.H.)
| | - Md. Fuad Hossain
- Department of Biochemistry and Molecular Biology, Gono Bishwabidyalay, Dhaka 1344, Bangladesh; (M.R.S.); (M.E.U.); (M.F.H.)
| | - Md. Kamruzzaman Munshi
- Institute of Food and Radiation Biology, Bangladesh Atomic Energy Commission, Dhaka 1000, Bangladesh;
| | - Abu Ali Ibn Sina
- Center for Personalized Nanomedicine, Australian Institute for Bioengineering & Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
3
|
Williams KI, Suryadevara P, Zhan CG, Hinds TD, Kipp ZA. Urobilin Derived from Bilirubin Bioconversion Binds Albumin and May Interfere with Bilirubin Interacting with Albumin: Implications for Disease Pathology. Biomedicines 2025; 13:302. [PMID: 40002715 PMCID: PMC11852593 DOI: 10.3390/biomedicines13020302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/19/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Bilirubin is a hydrophobic molecule that binds the carrier protein albumin for transport through systemic circulation. Bilirubin is cleared from the body through the liver and excreted into the intestines, where the microbiota modifies the chemical structure, forming urobilin, which can be reabsorbed into circulation by the hepatic portal vein. Urobilin has no known function. It is also unknown whether urobilin binds albumin for transport in circulation. We hypothesized that because of the likeness of their chemical structures, urobilin would also bind albumin like bilirubin does. Methods: First, we used in silico docking to predict if urobilin would bind to albumin and compared it to the bilirubin binding sites. To test this binding in vitro, we applied bilirubin's fluorescent property, which occurs when it is bound to a protein, including albumin, and exposed to light. We also used this method to determine if urobilin could exhibit autofluorescence when protein bound. Results: We found that bilirubin was predicted to bind albumin at amino acids E208, K212, D237, and K240 through hydrogen bonds. However, urobilin was predicted to bind albumin using different hydrogen bonds at amino acids H67, K240, and E252. We found that urobilin has a fluorescent property that can be quantified when bound to albumin. We performed a concentration response for urobilin-albumin fluorescent binding and observed a direct relationship between the urobilin level and the fluorescence intensity. Conclusions: The in silico docking analysis and autofluorescence results demonstrate that urobilin binds to albumin and might compete with bilirubin. This is the first study to identify a urobilin-binding protein and the important aspects of its physiological function and transport in circulation.
Collapse
Affiliation(s)
- Kevin I. Williams
- Drug & Disease Discovery D3 Research Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40508, USA;
- Department of Biochemistry and Molecular Biology, Centre College, Danville, KY 40422, USA
| | - Priyanka Suryadevara
- Department of Pharmaceutical Sciences and Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY 40508, USA; (P.S.); (C.-G.Z.)
| | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences and Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY 40508, USA; (P.S.); (C.-G.Z.)
| | - Terry D. Hinds
- Drug & Disease Discovery D3 Research Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40508, USA;
- Markey Cancer Center, University of Kentucky, Lexington, KY 40508, USA
- Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY 40508, USA
| | - Zachary A. Kipp
- Drug & Disease Discovery D3 Research Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40508, USA;
| |
Collapse
|
4
|
Kim JW, Byun MS, Yi D, Jung JH, Kong N, Chang YY, Jung G, Ahn H, Lee JY, Kang KM, Sohn CH, Lee YS, Kim YK, Lee DY. Liver function and Alzheimer's brain pathologies: A longitudinal study: Liver and Alzheimer's pathologies. J Prev Alzheimers Dis 2025; 12:100012. [PMID: 39800466 PMCID: PMC12065704 DOI: 10.1016/j.tjpad.2024.100012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
IMPORTANCE The neuropathological links underlying the association between changes in liver function and AD have not yet been clearly elucidated. OBJECTIVE We aimed to examine the relationship between liver function markers and longitudinal changes in Alzheimer's disease (AD) core pathologies. DESIGN Data from the Korean Brain Aging Study for the Early Diagnosis and Prediction of Alzheimer's Disease, a longitudinal cohort study initiated in 2014, were utilized. SETTING Community and memory clinic setting. PARTICIPANTS Three hundred forty-seven older adults. MAIN OUTCOME AND MEASURES Participants underwent baseline and 2-year follow-up evaluations, including liver function assessments and various brain imaging techniques, such as amyloid and tau PET, FDG-PET, and MRI). Liver function indicators [alanine aminotransferase (ALT), aspartate aminotransferase (AST), and total bilirubin] were examined as exposure variables. RESULTS Higher baseline ALT levels were associated with a greater increase in beta-amyloid deposition over 2 years [β = 0.166, Bonferroni-corrected P (PB) = 0.012], while lower total bilirubin levels were associated with a greater increase in tau deposition over the same period (β = -0.570, PB < 0.001). In contrast, AST alone showed no significant association with changes of AD pathologies. CONCLUSIONS AND RELEVANCE The findings suggest a possible link between lower liver function and the accumulation of core AD pathologies in the brain. These results also support the possibility that the liver-brain axis could be a potential target for therapeutic or preventive strategies against AD.
Collapse
Affiliation(s)
- Jee Wook Kim
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, 7 Keunjaebong-gil, Hwaseong, Gyeonggi, 18450, Republic of Korea; Department of Psychiatry, Hallym University College of Medicine, Chuncheon, Gangwon, 24252, Republic of Korea.
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea; Department of Psychiatry, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, 03080, Republic of Korea.
| | - Joon Hyung Jung
- Department of Psychiatry, Chungbuk National University Hospital, Cheongju, 28644, Republic of Korea.
| | - Nayeong Kong
- Department of Psychiatry, Keimyung University Dongsan Hospital, Daegu, 42601, Republic of Korea.
| | - Yoon Young Chang
- Department of Psychiatry, Inje University Sanggye Paik Hospital, Seoul, 01757, Republic of Korea.
| | - Gijung Jung
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea.
| | - Hyejin Ahn
- Interdisciplinary Program of Cognitive Science, Seoul National University College of Humanities, Seoul, 08826, Republic of Korea.
| | - Jun-Young Lee
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, 07061, Republic of Korea.
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, 03080, Republic of Korea.
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, 03080, Republic of Korea.
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, 07061, Republic of Korea.
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea; Department of Psychiatry, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, 03080, Republic of Korea; Interdisciplinary Program of Cognitive Science, Seoul National University College of Humanities, Seoul, 08826, Republic of Korea.
| |
Collapse
|
5
|
Furrow E, Peralta JA, Moore AR, Minor KM, Guerrero C, Hemmila CR, DiCiccio V, Cullen JN, Friedenberg SG, Giger U. Biliverdinuria Caused by Exonic BLVRA Deletions in Two Dogs with Green Urine. Genes (Basel) 2024; 15:1561. [PMID: 39766828 PMCID: PMC11675387 DOI: 10.3390/genes15121561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/11/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: In heme degradation, biliverdin reductase catalyzes the conversion of biliverdin to bilirubin. Defects in the biliverdin reductase A gene (BLVRA) causing biliverdinuria are extraordinarily rare in humans, and this inborn error of metabolism has not been reported in other mammals. The objective of this study was to diagnose biliverdinuria and identify the causal BLVRA variants in two adult mixed-breed dogs with life-long green urine. One of the dogs also had an unexplained regenerative anemia and mild hepatopathy. Methods: Clinicopathological evaluations, urinary mass spectroscopy, and molecular genetic studies were performed. Urine metabolic screening identified increased biliverdin concentrations in both cases relative to control dogs. Results: Whole genome and Sanger sequencing revealed that each case was homozygous for large deletions in BLVRA: UU_Cfam_GSD_1.0/canFam4 chr18:6,532,022-6,551,313 (19,292 bp) in Case 1 and chr18:6,543,863-6,545,908 (2046 bp) in Case 2. These variants were predicted to result in major BLVRA truncations (ENSCAFT00805017018.1 p.[Lys117-Lys296del] and p.[Ala154fs], respectively) and loss of enzyme function. In a genomic variant database, 671 dogs from 63 breeds had coverage over these regions, ruling out homozygosity for the BLVRA deletions. A gene defect for the regenerative anemia in Case 1 was not discovered. Conclusions: While expected to be rare, genotyping for the BLVRA deletions can be used to identify other affected and carrier dogs. This study illustrates the use of targeted metabolic and genomic screening as key diagnostic tools to diagnose a rare metabolic disorder. These are the first confirmed cases of biliverdinuria caused by BLVRA defects in non-human mammals.
Collapse
Affiliation(s)
- Eva Furrow
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA
| | - Jade A. Peralta
- College of Veterinary Medicine & Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - A Russell Moore
- College of Veterinary Medicine & Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Katie M. Minor
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA
| | - Candace Guerrero
- Center for Metabolomics and Proteomics, University of Minnesota, St. Paul, MN 55108, USA
| | - Charlotte R. Hemmila
- Center for Metabolomics and Proteomics, University of Minnesota, St. Paul, MN 55108, USA
| | | | - Jonah N. Cullen
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA
| | | | - Urs Giger
- Clinic for Small Animal Internal Medicine, Vetsuisse Faculty University of Zürich, 8057 Zürich, Switzerland;
| |
Collapse
|
6
|
Chen W, Johansen VBI, Legido-Quigley C. Bridging brain insulin resistance to Alzheimer's pathogenesis. Trends Biochem Sci 2024; 49:939-941. [PMID: 39358050 DOI: 10.1016/j.tibs.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024]
Abstract
Emerging evidence links type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD), with brain insulin resistance (BIR) as a key factor. In a recent study, Lanzillotta et al. reveal that reduced biliverdin reductase-A (BVR-A) impairs glycogen synthase kinase 3β (GSK3β) phosphorylation, causing mitochondrial dysfunction and exacerbating brain insulin resistance in the progression of both T2DM and AD.
Collapse
Affiliation(s)
- Wenqiang Chen
- Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Steno Diabetes Center Copenhagen, Herlev, 2730, Denmark.
| | - Valdemar Brimnes Ingemann Johansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Cristina Legido-Quigley
- Steno Diabetes Center Copenhagen, Herlev, 2730, Denmark; Institute of Pharmaceutical Science, King's College London, London, SE5 9RX, UK
| |
Collapse
|
7
|
Shin JW, Jung KJ, Ryu M, Kim J, Kimm H, Jee SH. Causal association between serum bilirubin and ischemic stroke: multivariable Mendelian randomization. Epidemiol Health 2024; 46:e2024070. [PMID: 39210787 PMCID: PMC11826012 DOI: 10.4178/epih.e2024070] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVES Previous research has predominantly focused on total bilirubin levels without clearly distinguishing between direct and indirect bilirubin. In this study, the differences between these forms were examined, and their potential causal relationships with ischemic stroke were investigated. METHODS Two-sample multivariable Mendelian randomization (MVMR) analysis was employed, extracting summary data on bilirubin from the Korean Cancer Prevention Study-II (n=159,844) and the Korean Genome and Epidemiology Study (n=72,299). Data on ischemic stroke were obtained from BioBank Japan (n=201,800). Colocalization analysis was performed, focusing on the UGT1A1, SLCO1B1, and SLCO1B3 genes, which are the primary loci associated with serum bilirubin levels. RESULTS Crude 2-sample Mendelian randomization analysis revealed a significant negative association between total bilirubin levels and ischemic stroke. However, in MVMR analyses, only indirect bilirubin demonstrated a significant negative association with ischemic stroke (odds ratio, 0.76; 95% confidence interval, 0.59 to 0.98). Colocalization analysis did not identify a shared causal variant between the 3 genetic loci related to indirect bilirubin and the risk of ischemic stroke. CONCLUSIONS Our study establishes a causal association between higher genetically determined levels of serum indirect bilirubin and reduced risk of ischemic stroke in an Asian population. Future research should include more in-depth analysis of shared genetic variants between indirect bilirubin and ischemic stroke.
Collapse
Affiliation(s)
- Jong Won Shin
- Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Ulsan, Korea
| | - Keum Ji Jung
- Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Mikyung Ryu
- Institute on Aging, Ajou University Medical Center, Suwon, Korea
- Basgenbio, Inc., Seoul, Korea
| | | | - Heejin Kimm
- Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Sun Ha Jee
- Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| |
Collapse
|
8
|
Duarte TL, Viveiros N, Godinho C, Duarte D. Heme (dys)homeostasis and liver disease. Front Physiol 2024; 15:1436897. [PMID: 39135705 PMCID: PMC11317413 DOI: 10.3389/fphys.2024.1436897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Heme is essential for a variety of proteins involved in vital physiological functions in the body, such as oxygen transport, drug metabolism, biosynthesis of steroids, signal transduction, antioxidant defense and mitochondrial respiration. However, free heme is potentially cytotoxic due to the capacity of heme iron to promote the oxidation of cellular molecules. The liver plays a central role in heme metabolism by significantly contributing to heme synthesis, heme detoxification, and recycling of heme iron. Conversely, enzymatic defects in the heme biosynthetic pathway originate multisystemic diseases (porphyrias) that are highly associated with liver damage. In addition, there is growing evidence that heme contributes to the outcomes of inflammatory, metabolic and malignant liver diseases. In this review, we summarize the contribution of the liver to heme metabolism and the association of heme dyshomeostasis with liver disease.
Collapse
Affiliation(s)
- Tiago L. Duarte
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Nicole Viveiros
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Catarina Godinho
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| | - Delfim Duarte
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Serviço de Hematologia e Transplantação da Medula Óssea, Instituto Português de Oncologia do Porto Francisco Gentil, E.P.E. (IPO Porto), Porto, Portugal
- Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| |
Collapse
|
9
|
Lizeth ANM, Vanessa BV, María Del Rocio TB, Margarita FC, Damián JM, Alfredo CO, Edgar CE, Placido RF. Hepatoprotective Effect Assessment of C-Phycocyanin on Hepatocellular Carcinoma Rat Model by Using Photoacoustic Spectroscopy. APPLIED SPECTROSCOPY 2024; 78:296-309. [PMID: 38224996 DOI: 10.1177/00037028231222508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary neoplasia of the liver with elevated mortality. Experimental treatment with antioxidants has a beneficial effect on the experimental models of HCC. Arthrospira maxima (spirulina) and its phycocyanin have antitumoral action on different tumoral cells. However, it is unknown whether phycocyanin is the responsible molecule for the antitumoral effect on HCC. Photoacoustic spectroscopy (PAS) stands out among other spectroscopy techniques for its versatility of samples analyzed. This technique makes it possible to obtain the optical absorption spectrum of solid or liquid, dark or transparent samples. Previous studies report that assessing liver damage in rats produced by the modified resistant hepatocyte model (MRHM) is possible by analyzing their blood optical absorption spectrum. This study aimed to investigate, by PAS, the effect of phycocyanin obtained from spirulina on hepatic dysfunction. The optical absorption spectra analysis of the rat blood indicates the damage level induced by the MRHM group, being in concordance with the carried out biological conventional studies results, indicating an increase in the activity of hepatic enzymes, oxidative stress, Bax/Bcl2 ratio, cdk2, and AKT2 expression results, with a reduction in p53 expression. Also, PAS results suggest that phycocyanin decreases induced damage, due to the prevention of the Bax, AKT2, and p53 altered expression and the tumor progression in a HCC rat model.
Collapse
Affiliation(s)
- Alvarado-Noguez Margarita Lizeth
- Departamento de Física, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Col. San Pedro Zacatenco, Ciudad de México, México
| | - Blas-Valdivia Vanessa
- Laboratorio de Neurobiología, Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Alcaldía Gustavo A. Madero, Ciudad de México, México
| | - Thompson-Bonilla María Del Rocio
- Laboratorio de Medicina Genómica, Hospital Regional 1ro de Octubre, ISSSTE, Alcaldía Gustavo A. Madero, Ciudad de México, México
| | - Franco-Colín Margarita
- Laboratorio de Metabolismo I. Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Colonia Unidad Profesional Adolfo López Mateos, Alcaldía Gustavo A. Madero., Ciudad de México, México
| | - Jacinto-Méndez Damián
- Departamento de Física, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Col. San Pedro Zacatenco, Ciudad de México, México
| | - Cruz-Orea Alfredo
- Departamento de Física, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Col. San Pedro Zacatenco, Ciudad de México, México
| | - Cano-Europa Edgar
- Laboratorio de Neurobiología, Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Alcaldía Gustavo A. Madero, Ciudad de México, México
| | - Rojas-Franco Placido
- Laboratorio de Metabolismo I. Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Colonia Unidad Profesional Adolfo López Mateos, Alcaldía Gustavo A. Madero., Ciudad de México, México
| |
Collapse
|
10
|
Paul BD, Pieper AA. Neuroprotective Roles of the Biliverdin Reductase-A/Bilirubin Axis in the Brain. Biomolecules 2024; 14:155. [PMID: 38397392 PMCID: PMC10887292 DOI: 10.3390/biom14020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Biliverdin reductase-A (BVRA) is a multi-functional enzyme with a multitude of important roles in physiologic redox homeostasis. Classically, BVRA is well known for converting the heme metabolite biliverdin to bilirubin, which is a potent antioxidant in both the periphery and the brain. However, BVRA additionally participates in many neuroprotective signaling cascades in the brain that preserve cognition. Here, we review the neuroprotective roles of BVRA and bilirubin in the brain, which together constitute a BVRA/bilirubin axis that influences healthy aging and cognitive function.
Collapse
Affiliation(s)
- Bindu D. Paul
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Lieber Institute for Brain Development, Baltimore, MD 21205, USA
| | - Andrew A. Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106, USA
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
11
|
Mujawar T, Sevelda P, Madea D, Klán P, Švenda J. A Platform for the Synthesis of Oxidation Products of Bilirubin. J Am Chem Soc 2024; 146:1603-1611. [PMID: 38165253 PMCID: PMC10797625 DOI: 10.1021/jacs.3c11778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 01/03/2024]
Abstract
Bilirubin is the principal product of heme catabolism. High concentrations of the pigment are neurotoxic, yet slightly elevated levels are beneficial. Being a potent antioxidant, oxidative transformations of bilirubin occur in vivo and lead to various oxidized fragments. The mechanisms of their formation, intrinsic biological activities, and potential roles in human pathophysiology are poorly understood. Degradation methods have been used to obtain samples of bilirubin oxidation products for research. Here, we report a complementary, fully synthetic method of preparation. Our strategy leverages repeating substitution patterns in the parent tetracyclic pigment. Functionalized ready-to-couple γ-lactone, γ-lactam, and pyrrole monocyclic building blocks were designed and efficiently synthesized. Subsequent modular combinations, supported by metal-catalyzed borylation and cross-coupling chemistries, translated into the concise assembly of the structurally diverse bilirubin oxidation products (BOXes, propentdyopents, and biopyrrins). The discovery of a new photoisomer of biopyrrin A named lumipyrrin is reported. Synthetic bilirubin oxidation products made available in sufficient purity and quantity will support future in vitro and in vivo investigations.
Collapse
Affiliation(s)
- Taufiqueahmed Mujawar
- Department
of Chemistry, Faculty of Science, Masaryk
University, Kamenice 5, Brno 625 00, Czech Republic
| | - Petr Sevelda
- Department
of Chemistry, Faculty of Science, Masaryk
University, Kamenice 5, Brno 625 00, Czech Republic
| | - Dominik Madea
- Department
of Chemistry, Faculty of Science, Masaryk
University, Kamenice 5, Brno 625 00, Czech Republic
- RECETOX,
Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Petr Klán
- Department
of Chemistry, Faculty of Science, Masaryk
University, Kamenice 5, Brno 625 00, Czech Republic
- RECETOX,
Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Jakub Švenda
- Department
of Chemistry, Faculty of Science, Masaryk
University, Kamenice 5, Brno 625 00, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital, Pekařská 53, Brno 656 91, Czech Republic
| |
Collapse
|
12
|
Zhou HL, Grimmett ZW, Venetos NM, Stomberski CT, Qian Z, McLaughlin PJ, Bansal PK, Zhang R, Reynolds JD, Premont RT, Stamler JS. An enzyme that selectively S-nitrosylates proteins to regulate insulin signaling. Cell 2023; 186:5812-5825.e21. [PMID: 38056462 PMCID: PMC10794992 DOI: 10.1016/j.cell.2023.11.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 09/01/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
Acyl-coenzyme A (acyl-CoA) species are cofactors for numerous enzymes that acylate thousands of proteins. Here, we describe an enzyme that uses S-nitroso-CoA (SNO-CoA) as its cofactor to S-nitrosylate multiple proteins (SNO-CoA-assisted nitrosylase, SCAN). Separate domains in SCAN mediate SNO-CoA and substrate binding, allowing SCAN to selectively catalyze SNO transfer from SNO-CoA to SCAN to multiple protein targets, including the insulin receptor (INSR) and insulin receptor substrate 1 (IRS1). Insulin-stimulated S-nitrosylation of INSR/IRS1 by SCAN reduces insulin signaling physiologically, whereas increased SCAN activity in obesity causes INSR/IRS1 hypernitrosylation and insulin resistance. SCAN-deficient mice are thus protected from diabetes. In human skeletal muscle and adipose tissue, SCAN expression increases with body mass index and correlates with INSR S-nitrosylation. S-nitrosylation by SCAN/SNO-CoA thus defines a new enzyme class, a unique mode of receptor tyrosine kinase regulation, and a revised paradigm for NO function in physiology and disease.
Collapse
Affiliation(s)
- Hua-Lin Zhou
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Zachary W Grimmett
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Nicholas M Venetos
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Colin T Stomberski
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Zhaoxia Qian
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Precious J McLaughlin
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Puneet K Bansal
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Rongli Zhang
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - James D Reynolds
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Anesthesiology and Perioperative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Richard T Premont
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Jonathan S Stamler
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.
| |
Collapse
|
13
|
Zeng J, Cai J, Wang D, Liu H, Sun H, Liu J. Heat stress affects dairy cow health status through blood oxygen availability. J Anim Sci Biotechnol 2023; 14:112. [PMID: 37658441 PMCID: PMC10474781 DOI: 10.1186/s40104-023-00915-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 07/06/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND Rises in global warming and extreme weather occurrence make the risk of heat stress (HS) induced by high ambient temperatures more likely in high-yielding dairy cows, resulting in low milk quality and yield. In animals, oxygen is involved in many physiological and metabolic processes, but the effects of HS on oxygen metabolism remain unclear. Thus, the current study aimed to investigate how oxygen metabolism plays a role in health status of dairy cows by measuring the milk yield, milk composition, and blood biochemical variables of cows under different levels of HS: none (No-HS), mild (Mild-HS), and moderate HS (Mod-HS). RESULTS The HS significantly increased rectal temperature (Ptreat < 0.01) and respiration rate (Ptreat < 0.01). Under Mod-HS, greater Na+ (P < 0.05) and lower total CO2, and pH (P < 0.05) were observed relative to those under No-HS and Mild-HS. Oxygen concentrations in both coccygeal artery and mammary vein (Ptreat < 0.01) were lower under Mod-HS than under No-HS. Coccygeal vein concentrations of heat shock protein 90 (HSP90) (P < 0.05) increased during Mod-HS compared with those in cows under No-HS. Malondialdehyde increased during Mod-HS, and glutathione peroxidase (P < 0.01) increased during Mild-HS. Coccygeal vein concentrations of vascular endothelial growth factor (P < 0.01), heme oxygenase-1 (P < 0.01), and hypoxia-inducible factor 1α (P < 0.01) were greater in cows under Mod-HS than those under No-HS. Red blood cell count (P < 0.01) and hemoglobin concentration (P < 0.01) were lower in the coccygeal vein of dairy cows under Mild- and Mod-HS than those of cows under No-HS. CONCLUSIONS Exposure to HS negatively impacts the health status and lactation performance of dairy cows by limiting oxygen metabolism and transportation. However, the specific mechanism by which HS affects mammary function in cows remains unclear and requires further exploration.
Collapse
Affiliation(s)
- Jia Zeng
- Key Laboratory of Dairy Cow Genetic Improvement and Milk Quality Research of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China
- Ministry of Education Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| | - Jie Cai
- Ministry of Education Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| | - Diming Wang
- Ministry of Education Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| | - Hongyun Liu
- Ministry of Education Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| | - Huizeng Sun
- Key Laboratory of Dairy Cow Genetic Improvement and Milk Quality Research of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China
- Ministry of Education Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| | - Jianxin Liu
- Key Laboratory of Dairy Cow Genetic Improvement and Milk Quality Research of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China
- Ministry of Education Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| |
Collapse
|
14
|
Dan Q, Jiang X, Wang R, Dai Z, Sun D. Biogenic Imaging Contrast Agents. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207090. [PMID: 37401173 PMCID: PMC10477908 DOI: 10.1002/advs.202207090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 06/08/2023] [Indexed: 07/05/2023]
Abstract
Imaging contrast agents are widely investigated in preclinical and clinical studies, among which biogenic imaging contrast agents (BICAs) are developing rapidly and playing an increasingly important role in biomedical research ranging from subcellular level to individual level. The unique properties of BICAs, including expression by cells as reporters and specific genetic modification, facilitate various in vitro and in vivo studies, such as quantification of gene expression, observation of protein interactions, visualization of cellular proliferation, monitoring of metabolism, and detection of dysfunctions. Furthermore, in human body, BICAs are remarkably helpful for disease diagnosis when the dysregulation of these agents occurs and can be detected through imaging techniques. There are various BICAs matched with a set of imaging techniques, including fluorescent proteins for fluorescence imaging, gas vesicles for ultrasound imaging, and ferritin for magnetic resonance imaging. In addition, bimodal and multimodal imaging can be realized through combining the functions of different BICAs, which helps overcome the limitations of monomodal imaging. In this review, the focus is on the properties, mechanisms, applications, and future directions of BICAs.
Collapse
Affiliation(s)
- Qing Dan
- Shenzhen Key Laboratory for Drug Addiction and Medication SafetyDepartment of UltrasoundInstitute of Ultrasonic MedicinePeking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhen518036P. R. China
| | - Xinpeng Jiang
- Department of Biomedical EngineeringCollege of Future TechnologyPeking UniversityBeijing100871P. R. China
| | - Run Wang
- Shenzhen Key Laboratory for Drug Addiction and Medication SafetyDepartment of UltrasoundInstitute of Ultrasonic MedicinePeking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhen518036P. R. China
| | - Zhifei Dai
- Department of Biomedical EngineeringCollege of Future TechnologyPeking UniversityBeijing100871P. R. China
| | - Desheng Sun
- Shenzhen Key Laboratory for Drug Addiction and Medication SafetyDepartment of UltrasoundInstitute of Ultrasonic MedicinePeking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhen518036P. R. China
| |
Collapse
|
15
|
Lee WH, Najjar SM, Kahn CR, Hinds TD. Hepatic insulin receptor: new views on the mechanisms of liver disease. Metabolism 2023; 145:155607. [PMID: 37271372 PMCID: PMC10330768 DOI: 10.1016/j.metabol.2023.155607] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/26/2023] [Accepted: 05/28/2023] [Indexed: 06/06/2023]
Abstract
Over 65 % of people with obesity display the metabolic-associated fatty liver disease (MAFLD), which can manifest as steatohepatitis, fibrosis, cirrhosis, or liver cancer. The development and progression of MAFLD involve hepatic insulin resistance and reduced insulin clearance. This review discusses the relationships between altered insulin signaling, hepatic insulin resistance, and reduced insulin clearance in the development of MAFLD and how this provides the impetus for exploring the use of insulin sensitizers to curb this disease. The review also explores the role of the insulin receptor in hepatocytes and hepatic stellate cells and how it signals in metabolic and end-stage liver diseases. Finally, we discuss new research findings that indicate that advanced hepatic diseases may be an insulin-sensitive state in the liver and deliberate whether insulin sensitizers should be used to manage late-stage liver diseases.
Collapse
Affiliation(s)
- Wang-Hsin Lee
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Sonia M Najjar
- Department of Biomedical Sciences and the Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA; Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, KY, USA; Markey Cancer Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
16
|
Jayanti S, Dalla Verde C, Tiribelli C, Gazzin S. Inflammation, Dopaminergic Brain and Bilirubin. Int J Mol Sci 2023; 24:11478. [PMID: 37511235 PMCID: PMC10380707 DOI: 10.3390/ijms241411478] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Dopamine is a well-known neurotransmitter due to its involvement in Parkinson's disease (PD). Dopamine is not only involved in PD but also controls multiple mental and physical activities, such as the pleasure of food, friends and loved ones, music, art, mood, cognition, motivation, fear, affective disorders, addiction, attention deficit disorder, depression, and schizophrenia. Dopaminergic neurons (DOPAn) are susceptible to stressors, and inflammation is a recognized risk for neuronal malfunctioning and cell death in major neurodegenerative diseases. Less is known for non-neurodegenerative conditions. Among the endogenous defenses, bilirubin, a heme metabolite, has been shown to possess important anti-inflammatory activity and, most importantly, to prevent DOPAn demise in an ex vivo model of PD by acting on the tumor necrosis factor-alpha (TNFα). This review summarizes the evidence linking DOPAn, inflammation (when possible, specifically TNFα), and bilirubin as an anti-inflammatory in order to understand what is known, the gaps that need filling, and the hypotheses of anti-inflammatory strategies to preserve dopamine homeostasis with bilirubin included.
Collapse
Affiliation(s)
- Sri Jayanti
- Italian Liver Foundation, Liver Brain Unit “Rita Moretti”, Area Science Park, Bldg. Q, SS 14, Km 163,5, 34149 Trieste, Italy; (S.J.); (C.D.V.); (S.G.)
- Eijkman Research Centre for Molecular Biology, Research Organization for Health, National Research and Innovation Agency, Cibinong 16915, Indonesia
| | - Camilla Dalla Verde
- Italian Liver Foundation, Liver Brain Unit “Rita Moretti”, Area Science Park, Bldg. Q, SS 14, Km 163,5, 34149 Trieste, Italy; (S.J.); (C.D.V.); (S.G.)
| | - Claudio Tiribelli
- Italian Liver Foundation, Liver Brain Unit “Rita Moretti”, Area Science Park, Bldg. Q, SS 14, Km 163,5, 34149 Trieste, Italy; (S.J.); (C.D.V.); (S.G.)
| | - Silvia Gazzin
- Italian Liver Foundation, Liver Brain Unit “Rita Moretti”, Area Science Park, Bldg. Q, SS 14, Km 163,5, 34149 Trieste, Italy; (S.J.); (C.D.V.); (S.G.)
| |
Collapse
|
17
|
Ishikawa K, Xie X, Osaki Y, Miyawaki A, Numata K, Kodama Y. Bilirubin is produced nonenzymatically in plants to maintain chloroplast redox status. SCIENCE ADVANCES 2023; 9:eadh4787. [PMID: 37285441 DOI: 10.1126/sciadv.adh4787] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/01/2023] [Indexed: 06/09/2023]
Abstract
Bilirubin, a potent antioxidant, is a product of heme catabolism in heterotrophs. Heterotrophs mitigate oxidative stress resulting from free heme by catabolism into bilirubin via biliverdin. Although plants also convert heme to biliverdin, they are generally thought to be incapable of producing bilirubin because they lack biliverdin reductase, the enzyme responsible for bilirubin biosynthesis in heterotrophs. Here, we demonstrate that bilirubin is produced in plant chloroplasts. Live-cell imaging using the bilirubin-dependent fluorescent protein UnaG revealed that bilirubin accumulated in chloroplasts. In vitro, bilirubin was produced nonenzymatically through a reaction between biliverdin and reduced form of nicotinamide adenine dinucleotide phosphate at concentrations comparable to those in chloroplasts. In addition, increased bilirubin production led to lower reactive oxygen species levels in chloroplasts. Our data refute the generally accepted pathway of heme degradation in plants and suggest that bilirubin contributes to the maintenance of redox status in chloroplasts.
Collapse
Affiliation(s)
- Kazuya Ishikawa
- Center for Bioscience Research and Education, Utsunomiya University, Tochigi 321-8505, Japan
- Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Xiaonan Xie
- Center for Bioscience Research and Education, Utsunomiya University, Tochigi 321-8505, Japan
| | - Yasuhide Osaki
- Center for Bioscience Research and Education, Utsunomiya University, Tochigi 321-8505, Japan
| | - Atsushi Miyawaki
- Laboratory for Cell Function Dynamics, RIKEN Center for Brain Science, Saitama 351-0198, Japan
- Biotechnological Optics Research Team, RIKEN Center for Advanced Photonics; Saitama, 351-0198, Japan
| | - Keiji Numata
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University; Kyoto, 615-8246, Japan
- Biomacromolecules Research Team, RIKEN Center for Sustainable Resource Science, Saitama 351-0198, Japan
| | - Yutaka Kodama
- Center for Bioscience Research and Education, Utsunomiya University, Tochigi 321-8505, Japan
- Biomacromolecules Research Team, RIKEN Center for Sustainable Resource Science, Saitama 351-0198, Japan
| |
Collapse
|
18
|
Bahou WF, Marchenko N, Nesbitt NM. Metabolic Functions of Biliverdin IXβ Reductase in Redox-Regulated Hematopoietic Cell Fate. Antioxidants (Basel) 2023; 12:antiox12051058. [PMID: 37237924 DOI: 10.3390/antiox12051058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/19/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Cytoprotective heme oxygenases derivatize heme to generate carbon monoxide, ferrous iron, and isomeric biliverdins, followed by rapid NAD(P)H-dependent biliverdin reduction to the antioxidant bilirubin. Recent studies have implicated biliverdin IXβ reductase (BLVRB) in a redox-regulated mechanism of hematopoietic lineage fate restricted to megakaryocyte and erythroid development, a function distinct and non-overlapping from the BLVRA (biliverdin IXα reductase) homologue. In this review, we focus on recent progress in BLVRB biochemistry and genetics, highlighting human, murine, and cell-based studies that position BLVRB-regulated redox function (or ROS accumulation) as a developmentally tuned trigger that governs megakaryocyte/erythroid lineage fate arising from hematopoietic stem cells. BLVRB crystallographic and thermodynamic studies have elucidated critical determinants of substrate utilization, redox coupling and cytoprotection, and have established that inhibitors and substrates bind within the single-Rossmann fold. These advances provide unique opportunities for the development of BLVRB-selective redox inhibitors as novel cellular targets that retain potential for therapeutic applicability in hematopoietic (and other) disorders.
Collapse
Affiliation(s)
- Wadie F Bahou
- Department of Medicine, School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Natalia Marchenko
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Natasha M Nesbitt
- Blood Cell Technologies, 25 Health Sciences Drive, Stony Brook, NY 11790, USA
| |
Collapse
|
19
|
Vitek L, Hinds TD, Stec DE, Tiribelli C. The physiology of bilirubin: health and disease equilibrium. Trends Mol Med 2023; 29:315-328. [PMID: 36828710 PMCID: PMC10023336 DOI: 10.1016/j.molmed.2023.01.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/24/2023]
Abstract
Bilirubin has several physiological functions, both beneficial and harmful. In addition to reactive oxygen species-scavenging activities, bilirubin has potent immunosuppressive effects associated with long-term pathophysiological sequelae. It has been recently recognized as a hormone with endocrine actions and interconnected effects on various cellular signaling pathways. Current studies show that bilirubin also decreases adiposity and prevents metabolic and cardiovascular diseases. All in all, the physiological importance of bilirubin is only now coming to light, and strategies for increasing plasma bilirubin levels to combat chronic diseases are starting to be considered. This review discusses the beneficial effects of increasing plasma bilirubin, incorporates emerging areas of bilirubin biology, and provides key concepts to advance the field.
Collapse
Affiliation(s)
- Libor Vitek
- Fourth Department of Internal Medicine and Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, 120 00 Prague, Czech Republic
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, Barnstable Brown Diabetes Center, Markey Cancer Center, University of Kentucky, Lexington, KY 40508, USA
| | - David E Stec
- Department of Physiology and Biophysics, Cardiorenal, and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | | |
Collapse
|
20
|
Zhang Q, Deng C, Peng M, Li C, Teng Y, Guo S, Wu T, Yi D, Hou Y. Integration of transcriptomic and proteomic analyses reveals protective mechanisms of N-acetylcysteine in indomethacin-stimulated enterocytes. J Nutr Biochem 2023; 112:109231. [PMID: 36435287 DOI: 10.1016/j.jnutbio.2022.109231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 08/07/2022] [Accepted: 10/11/2022] [Indexed: 11/25/2022]
Abstract
Intestinal health is critical for the growth and development of humans and animals. Our previous study has demonstrated that indomethacin (IDMT) could induce intestinal injury in piglets, and N-acetylcysteine (NAC) supplementation contributed to alleviating intestinal injury induced by various stimuli. In this study, we investigated the mechanism of IDMT-induced cell death in IPEC-1 cell lines and explored the role of NAC by using transcriptomic and proteomic analyses. Results showed that cell viability was substantially reduced with the increasing concentrations of IDMT, whereas NAC significantly increased the survival rate of IPEC-1 cells regardless of its addition method. Transcriptomics and proteomics data indicated that terms, such as cell cycle, energy metabolism, and cell proliferation, were significantly enriched by Gene ontology and pathway analyses. Flow cytometer analysis showed that IDMT induced cell cycle arrest at G0/G1 phase. The expression of cell cycle regulatory proteins (CDK1, CCNA2, and CDC45) was decreased by IDMT stimulation. Importantly, NAC treatment repaired IDMT-induced mitochondrial dysfunction by increasing ATP production, decreasing oxygen consumption rate in non-mitochondrial O2 consumption, and increasing the red/green fluorescence ratio. IDMT stimulation significantly increased caspase-3 expression, which was partially reversed by NAC treatment. These results suggest that IDMT-induced cell death may be attributable to disturbance of the cell cycle processes, mitochondria dysfunction and apoptosis, and NAC could confer a protective effect by restoring the mitochondrial function and inhibiting the apoptosis pathway. This study provides a theoretical basis for the pathogenesis of IDMT-induced intestinal injury and guides the clinic application of NAC.
Collapse
Affiliation(s)
- Qian Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Cuifang Deng
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Meng Peng
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Chengcheng Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Yi Teng
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Shuangshuang Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Tao Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Dan Yi
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China.
| | - Yongqing Hou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China.
| |
Collapse
|
21
|
Bates EA, Kipp ZA, Martinez GJ, Badmus OO, Soundarapandian MM, Foster D, Xu M, Creeden JF, Greer JR, Morris AJ, Stec DE, Hinds TD. Suppressing Hepatic UGT1A1 Increases Plasma Bilirubin, Lowers Plasma Urobilin, Reorganizes Kinase Signaling Pathways and Lipid Species and Improves Fatty Liver Disease. Biomolecules 2023; 13:252. [PMID: 36830621 PMCID: PMC9953728 DOI: 10.3390/biom13020252] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Several population studies have observed lower serum bilirubin levels in patients with non-alcoholic fatty liver disease (NAFLD). Yet, treatments to target this metabolic phenotype have not been explored. Therefore, we designed an N-Acetylgalactosamine (GalNAc) labeled RNAi to target the enzyme that clears bilirubin from the blood, the UGT1A1 glucuronyl enzyme (GNUR). In this study, male C57BL/6J mice were fed a high-fat diet (HFD, 60%) for 30 weeks to induce NAFLD and were treated subcutaneously with GNUR or sham (CTRL) once weekly for six weeks while continuing the HFD. The results show that GNUR treatments significantly raised plasma bilirubin levels and reduced plasma levels of the bilirubin catabolized product, urobilin. We show that GNUR decreased liver fat content and ceramide production via lipidomics and lowered fasting blood glucose and insulin levels. We performed extensive kinase activity analyses using our PamGene PamStation kinome technology and found a reorganization of the kinase pathways and a significant decrease in inflammatory mediators with GNUR versus CTRL treatments. These results demonstrate that GNUR increases plasma bilirubin and reduces plasma urobilin, reducing NAFLD and inflammation and improving overall liver health. These data indicate that UGT1A1 antagonism might serve as a treatment for NAFLD and may improve obesity-associated comorbidities.
Collapse
Affiliation(s)
- Evelyn A. Bates
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40508, USA
| | - Zachary A. Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40508, USA
| | - Genesee J. Martinez
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40508, USA
| | - Olufunto O. Badmus
- Department of Physiology & Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | | | | | - Mei Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40508, USA
| | - Justin F. Creeden
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Jennifer R. Greer
- Department of Physiology & Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Andrew J. Morris
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - David E. Stec
- Department of Physiology & Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Terry D. Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40508, USA
- Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY 40508, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40508, USA
| |
Collapse
|
22
|
Flack KD, Vítek L, Fry CS, Stec DE, Hinds TD. Cutting edge concepts: Does bilirubin enhance exercise performance? Front Sports Act Living 2023; 4:1040687. [PMID: 36713945 PMCID: PMC9874874 DOI: 10.3389/fspor.2022.1040687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Exercise performance is dependent on many factors, such as muscular strength and endurance, cardiovascular capacity, liver health, and metabolic flexibility. Recent studies show that plasma levels of bilirubin, which has classically been viewed as a liver dysfunction biomarker, are elevated by exercise training and that elite athletes may have significantly higher levels. Other studies have shown higher plasma bilirubin levels in athletes and active individuals compared to general, sedentary populations. The reason for these adaptions is unclear, but it could be related to bilirubin's antioxidant properties in response to a large number of reactive oxygen species (ROS) that originates from mitochondria during exercise. However, the mechanisms of these are unknown. Current research has re-defined bilirubin as a metabolic hormone that interacts with nuclear receptors to drive gene transcription, which reduces body weight. Bilirubin has been shown to reduce adiposity and improve the cardiovascular system, which might be related to the adaption of bilirubin increasing during exercise. No studies have directly tested if elevating bilirubin levels can influence athletic performance. However, based on the mechanisms proposed in the present review, this seems plausible and an area to consider for future studies. Here, we discuss the importance of bilirubin and exercise and how the combination might improve metabolic health outcomes and possibly athletic performance.
Collapse
Affiliation(s)
- Kyle D. Flack
- Department of Dietetics and Human Nutrition, University of Kentucky, Lexington, KY, United States
| | - Libor Vítek
- 4th Department of Internal Medicine and Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Christopher S. Fry
- Department of Athletic Training and Clinical Nutrition, University of Kentucky College of Medicine, Lexington, KY, United States
- Center for Muscle Biology, University of Kentucky College of Medicine, Lexington, KY, United States
| | - David E. Stec
- Department of Physiology & Biophysics, Cardiorenal, and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS, United States
| | - Terry D. Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
- Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, KY, United States
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
23
|
Kipp ZA, Xu M, Bates EA, Lee WH, Kern PA, Hinds TD. Bilirubin Levels Are Negatively Correlated with Adiposity in Obese Men and Women, and Its Catabolized Product, Urobilin, Is Positively Associated with Insulin Resistance. Antioxidants (Basel) 2023; 12:170. [PMID: 36671031 PMCID: PMC9854555 DOI: 10.3390/antiox12010170] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/03/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023] Open
Abstract
Bilirubin levels in obese humans and rodents have been shown to be lower than in their lean counterparts. Some studies have proposed that the glucuronyl UGT1A1 enzyme that clears bilirubin from the blood increases in the liver with obesity. UGT1A1 clearance of bilirubin allows more conjugated bilirubin to enter the intestine, where it is catabolized into urobilin, which can be then absorbed via the hepatic portal vein. We hypothesized that when bilirubin levels are decreased, the urobilin increases in the plasma of obese humans, as compared to lean humans. To test this, we measured plasma levels of bilirubin and urobilin, body mass index (BMI), adiposity, blood glucose and insulin, and HOMA IR in a small cohort of obese and lean men and women. We found that bilirubin levels negatively correlated with BMI and adiposity in obese men and women, as compared to their lean counterparts. Contrarily, urobilin levels were positively associated with adiposity and BMI. Only obese women were found to be insulin resistant based on significantly higher HOMA IR, as compared to lean women. The urobilin levels were positively associated with HOMA IR in both groups, but women had a stronger linear correlation. These studies indicate that plasma urobilin levels are associated with obesity and its comorbidities, such as insulin resistance.
Collapse
Affiliation(s)
- Zachary A. Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 760 Press Avenue, Healthy Kentucky Research Building, Lexington, KY 40508, USA
| | - Mei Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 760 Press Avenue, Healthy Kentucky Research Building, Lexington, KY 40508, USA
| | - Evelyn A. Bates
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 760 Press Avenue, Healthy Kentucky Research Building, Lexington, KY 40508, USA
| | - Wang-Hsin Lee
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 760 Press Avenue, Healthy Kentucky Research Building, Lexington, KY 40508, USA
| | - Philip A. Kern
- Department of Internal Medicine, Division of Endocrinology, University of Kentucky, Lexington, KY 40508, USA
| | - Terry D. Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 760 Press Avenue, Healthy Kentucky Research Building, Lexington, KY 40508, USA
- Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY 40508, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40508, USA
| |
Collapse
|
24
|
Reiländer S, Schmehl W, Popp K, Nuss K, Kronen P, Verdino D, Wiezorek C, Gutmann M, Hahn L, Däubler C, Meining A, Raschig M, Kaiser F, von Rechenberg B, Scherf-Clavel O, Meinel L. Oral Use of Therapeutic Carbon Monoxide for Anyone, Anywhere, and Anytime. ACS Biomater Sci Eng 2022. [DOI: 10.1021/acsbiomaterials.2c00464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Simon Reiländer
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074Wuerzburg, Germany
| | - Wolfgang Schmehl
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074Wuerzburg, Germany
| | - Kevin Popp
- German Plastics Center (SKZ), Friedrich-Bergius-Ring 22, Wuerzburg97076, Germany
| | - Katja Nuss
- Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
- Competence Center for Applied Biotechnology and Molecular Medicine (CABMM), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
| | - Peter Kronen
- Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
- Competence Center for Applied Biotechnology and Molecular Medicine (CABMM), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
| | - Dagmar Verdino
- Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
- Competence Center for Applied Biotechnology and Molecular Medicine (CABMM), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
| | - Christina Wiezorek
- Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
- Competence Center for Applied Biotechnology and Molecular Medicine (CABMM), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
| | - Marcus Gutmann
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074Wuerzburg, Germany
| | - Lukas Hahn
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074Wuerzburg, Germany
| | - Christof Däubler
- Department of Internal Medicine II, Gastroenterology, University Hospital Wuerzburg, Oberdürrbacherstr. 6, Wuerzburg97080, Germany
| | - Alexander Meining
- Department of Internal Medicine II, Gastroenterology, University Hospital Wuerzburg, Oberdürrbacherstr. 6, Wuerzburg97080, Germany
| | - Martina Raschig
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074Wuerzburg, Germany
| | - Friederike Kaiser
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg, Pleicherwall 2, Würzburg97070, Germany
| | - Brigitte von Rechenberg
- Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
- Competence Center for Applied Biotechnology and Molecular Medicine (CABMM), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
| | - Oliver Scherf-Clavel
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074Wuerzburg, Germany
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074Wuerzburg, Germany
- Helmholtz Institute for RNA-based Infection Biology (HIRI), Würzburg97070, Germany
| |
Collapse
|
25
|
Creeden JF, Kipp ZA, Xu M, Flight RM, Moseley HNB, Martinez GJ, Lee W, Alganem K, Imami AS, McMullen MR, Roychowdhury S, Nawabi AM, Hipp JA, Softic S, Weinman SA, McCullumsmith R, Nagy LE, Hinds TD. Hepatic kinome atlas: An in-depth identification of kinase pathways in liver fibrosis of humans and rodents. Hepatology 2022; 76:1376-1388. [PMID: 35313030 PMCID: PMC9489820 DOI: 10.1002/hep.32467] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/02/2022] [Accepted: 03/12/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Resolution of pathways that converge to induce deleterious effects in hepatic diseases, such as in the later stages, have potential antifibrotic effects that may improve outcomes. We aimed to explore whether humans and rodents display similar fibrotic signaling networks. APPROACH AND RESULTS We assiduously mapped kinase pathways using 340 substrate targets, upstream bioinformatic analysis of kinase pathways, and over 2000 random sampling iterations using the PamGene PamStation kinome microarray chip technology. Using this technology, we characterized a large number of kinases with altered activity in liver fibrosis of both species. Gene expression and immunostaining analyses validated many of these kinases as bona fide signaling events. Surprisingly, the insulin receptor emerged as a considerable protein tyrosine kinase that is hyperactive in fibrotic liver disease in humans and rodents. Discoidin domain receptor tyrosine kinase, activated by collagen that increases during fibrosis, was another hyperactive protein tyrosine kinase in humans and rodents with fibrosis. The serine/threonine kinases found to be the most active in fibrosis were dystrophy type 1 protein kinase and members of the protein kinase family of kinases. We compared the fibrotic events over four models: humans with cirrhosis and three murine models with differing levels of fibrosis, including two models of fatty liver disease with emerging fibrosis. The data demonstrate a high concordance between human and rodent hepatic kinome signaling that focalizes, as shown by our network analysis of detrimental pathways. CONCLUSIONS Our findings establish a comprehensive kinase atlas for liver fibrosis, which identifies analogous signaling events conserved among humans and rodents.
Collapse
Affiliation(s)
- Justin F. Creeden
- Department of NeurosciencesUniversity of Toledo College of Medicine and Life SciencesToledoOhioUSA
| | - Zachary A. Kipp
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Mei Xu
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Robert M. Flight
- Department of Molecular & Cellular BiochemistryUniversity of KentuckyLexingtonKentuckyUSA
- Markey Cancer CenterUniversity of KentuckyLexingtonKentuckyUSA
- Resource Center for Stable Isotope Resolved MetabolomicsUniversity of KentuckyLexingtonKentuckyUSA
| | - Hunter N. B. Moseley
- Department of Molecular & Cellular BiochemistryUniversity of KentuckyLexingtonKentuckyUSA
- Markey Cancer CenterUniversity of KentuckyLexingtonKentuckyUSA
- Resource Center for Stable Isotope Resolved MetabolomicsUniversity of KentuckyLexingtonKentuckyUSA
- Institute for Biomedical InformaticsUniversity of KentuckyLexingtonKentuckyUSA
- Center for Clinical and Translational ScienceUniversity of KentuckyLexingtonKentuckyUSA
| | - Genesee J. Martinez
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Wang‐Hsin Lee
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Khaled Alganem
- Department of NeurosciencesUniversity of Toledo College of Medicine and Life SciencesToledoOhioUSA
| | - Ali S. Imami
- Department of NeurosciencesUniversity of Toledo College of Medicine and Life SciencesToledoOhioUSA
| | - Megan R. McMullen
- Department of Inflammation and ImmunityCleveland ClinicClevelandOhioUSA
| | | | - Atta M. Nawabi
- Division of Transplant and HepatobiliaryDepartment of SurgeryThe University of Kansas Medical CenterKansas CityKansasUSA
| | | | - Samir Softic
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Department of PediatricsUniversity of KentuckyLexingtonKentuckyUSA
| | - Steven A. Weinman
- Department of Internal Medicine and Liver CenterUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Robert McCullumsmith
- Department of NeurosciencesUniversity of Toledo College of Medicine and Life SciencesToledoOhioUSA
- Neurosciences InstituteProMedicaToledoOhioUSA
| | - Laura E. Nagy
- Department of Inflammation and ImmunityCleveland ClinicClevelandOhioUSA
- Department of Gastroenterology and HepatologyCenter for Liver Disease ResearchCleveland ClinicClevelandOhioUSA
- Department of Molecular MedicineCase Western Reserve UniversityClevelandOhioUSA
| | - Terry D. Hinds
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Markey Cancer CenterUniversity of KentuckyLexingtonKentuckyUSA
- Barnstable Brown Diabetes CenterUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| |
Collapse
|
26
|
Molecular mechanisms of metabolic associated fatty liver disease (MAFLD): functional analysis of lipid metabolism pathways. Clin Sci (Lond) 2022; 136:1347-1366. [PMID: 36148775 PMCID: PMC9508552 DOI: 10.1042/cs20220572] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 01/30/2023]
Abstract
The metabolic-associated fatty liver disease (MAFLD) is a condition of fat accumulation in the liver in combination with metabolic dysfunction in the form of overweight or obesity and insulin resistance. It is also associated with an increased cardiovascular disease risk, including hypertension and atherosclerosis. Hepatic lipid metabolism is regulated by a combination of the uptake and export of fatty acids, de novo lipogenesis, and fat utilization by β-oxidation. When the balance between these pathways is altered, hepatic lipid accumulation commences, and long-term activation of inflammatory and fibrotic pathways can progress to worsen the liver disease. This review discusses the details of the molecular mechanisms regulating hepatic lipids and the emerging therapies targeting these pathways as potential future treatments for MAFLD.
Collapse
|
27
|
Cimini FA, Perluigi M, Barchetta I, Cavallo MG, Barone E. Role of Biliverdin Reductase A in the Regulation of Insulin Signaling in Metabolic and Neurodegenerative Diseases: An Update. Int J Mol Sci 2022; 23:5574. [PMID: 35628384 PMCID: PMC9141761 DOI: 10.3390/ijms23105574] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 11/16/2022] Open
Abstract
Insulin signaling is a conserved pathway that orchestrates glucose and lipid metabolism, energy balance, and inflammation, and its dysregulation compromises the homeostasis of multiple systems. Insulin resistance is a shared hallmark of several metabolic diseases, including obesity, metabolic syndrome, and type 2 diabetes, and has been associated with cognitive decline during aging and dementia. Numerous mechanisms promoting the development of peripheral and central insulin resistance have been described, although most of them were not completely clarified. In the last decades, several studies have highlighted that biliverdin reductase-A (BVR-A), over its canonical role in the degradation of heme, acts as a regulator of insulin signaling. Evidence from human and animal studies show that BVR-A alterations are associated with the aberrant activation of insulin signaling, metabolic syndrome, liver steatosis, and visceral adipose tissue inflammation in obese and diabetic individuals. In addition, recent findings demonstrated that reduced BVR-A levels or impaired BVR-A activation contribute to the development of brain insulin resistance and metabolic alterations in Alzheimer's disease. In this narrative review, we will provide an overview on the literature by focusing on the role of BVR-A in the regulation of insulin signaling and how BVR-A alterations impact on cell dysfunctions in both metabolic and neurodegenerative disorders.
Collapse
Affiliation(s)
- Flavia Agata Cimini
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy; (F.A.C.); (I.B.)
| | - Marzia Perluigi
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (M.P.); (E.B.)
| | - Ilaria Barchetta
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy; (F.A.C.); (I.B.)
| | - Maria Gisella Cavallo
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy; (F.A.C.); (I.B.)
| | - Eugenio Barone
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (M.P.); (E.B.)
| |
Collapse
|
28
|
Singh S, Gautam U, Manvi FV. Protective Impact of Edaravone Against ZnO NPs-induced Oxidative Stress in the Human Neuroblastoma SH-SY5Y Cell Line. Cell Mol Neurobiol 2022; 42:1189-1210. [PMID: 33222098 PMCID: PMC11441218 DOI: 10.1007/s10571-020-01011-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 11/13/2020] [Indexed: 10/22/2022]
Abstract
Extensive applications of ZnO NPs (zinc oxide nanoparticles) in daily life have created concern about their biotoxicity. Zinc oxide nanoparticles induce oxidative stress, inflammation, and apoptosis in neurons. Edaravone applies antioxidant agent and anti-inflammatory impacts in the different cells, as evaluated in both in vitro and in vivo experimental models. This study is designed to explore, how edaravone would avert mitochondrial impairment in human neuronal cells against ZnO NPs-induced toxicity. Accordingly, we analyzed here whether a pretreatment (for 24 h) with edaravone (10-100 μM) would enhance mitochondrial protection in the human neuroblastoma cells SH-SY5Y against ZnO NPs-induced toxicity. We found that edaravone at 25 μM averted the ZnO NPs-induced decrease in the amounts of adenosine triphosphate (ATP), just as on the activity of the complexes I and V. Also, edaravone induced an antioxidant activity by diminishing the levels of lipid peroxidation, protein carbonylation, and protein nitration in the mitochondrial membranes. Edaravone blocked the ZnO NPs-induced transcription factor nuclear factor-κB (NF-κB) upregulation. The inhibition of the heme oxygenase-1 (HO-1) enzyme by zinc protoporphyrin IX (ZnPP IX, 10 μM) smothered the preventive impacts brought about by edaravone with respect to mitochondrial function and inflammation. After this examination, it can be concluded that edaravone caused cytoprotective impacts in an HO-1-dependent manner in SH-SY5Y cells against ZnO NPs-induced toxicity.
Collapse
Affiliation(s)
- Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotions Industrial Park (EPIP), Industrial Area, Hajipur, Bihar, India.
- K.L.E. Academy of Higher Education & Research, Belagavi, Karnataka, India.
| | - Upendr Gautam
- Vinayaka Mission's Research Foundation, Ariyanur, Tamil Nadu, India
| | - F V Manvi
- K.L.E. Academy of Higher Education & Research, Belagavi, Karnataka, India
| |
Collapse
|
29
|
Gao Z, Ni X, Zheng B, Sun W, Wan W, Liu H, Ni X, Suo T, Li N, Liu H, Shen S. Biliverdin reductase B impairs cholangiocarcinoma cell motility by inhibiting the Notch/Snail signaling pathway. J Cancer 2022; 13:2159-2170. [PMID: 35517415 PMCID: PMC9066219 DOI: 10.7150/jca.70323] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/22/2022] [Indexed: 12/04/2022] Open
Abstract
Cholangiocarcinoma (CCA) is one of the most lethal types of solid tumors worldwide. Lymph node metastasis is common in the early stage, which is associated with recurrence and reduced survival time after CCA resection. The molecular pathogenesis of CCA is complex and requires extensive investigation. It involves multiple genomic alterations and the dysregulation of signaling pathways. Biliverdin reductase B (BLVRB) is a non-redundant NAD(P)H-dependent biliverdin reductase that regulates cellular redox status by reducing biliverdin to bilirubin. This study aimed at describing the biological functions and molecular mechanisms of BLVRB in human CCA. Prognostic clinical data showed that low expression BLVRB was associated with poor prognosis and lymph node metastasis. BLVRB depletion accelerated epithelial-mesenchymal transition (EMT), cell migration and invasion. In contrast, BLVRB overexpression was associated with reduced EMT and cell migration and invasion in CCA. BLVRB suppression activated Notch signaling, and activated c-Notch enhanced EMT by upregulating Snail expression levels, thereby increasing cell migration and invasion in CCA. Our results identified an unexpected function of BLVRB in CCA migration and invasion through the regulation of Notch/Snail signaling.
Collapse
Affiliation(s)
- Zhihui Gao
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Xiaojian Ni
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Bohao Zheng
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Wentao Sun
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Wenze Wan
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Han Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Xiaoling Ni
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Tao Suo
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Na Li
- Basic Medical Institute; Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Houbao Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
- Department of General Surgery, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai 200031, China
| | - Sheng Shen
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
- Department of General Surgery, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai 200031, China
| |
Collapse
|
30
|
Stec DE, Tiribelli C, Badmus OO, Hinds TD. Novel Function for Bilirubin as a Metabolic Signaling Molecule: Implications for Kidney Diseases. KIDNEY360 2022; 3:945-953. [PMID: 36128497 PMCID: PMC9438427 DOI: 10.34067/kid.0000062022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/24/2022] [Indexed: 01/30/2023]
Abstract
Bilirubin is the end product of the catabolism of heme via the heme oxygenase pathway. Heme oxygenase generates carbon monoxide (CO) and biliverdin from the breakdown of heme, and biliverdin is rapidly reduced to bilirubin by the enzyme biliverdin reductase (BVR). Bilirubin has long been thought of as a toxic product that is only relevant to health when blood levels are severely elevated, such as in clinical jaundice. The physiologic functions of bilirubin correlate with the growing body of evidence demonstrating the protective effects of serum bilirubin against cardiovascular and metabolic diseases. Although the correlative evidence suggests a protective effect of serum bilirubin against many diseases, the mechanism by which bilirubin offers protection against cardiovascular and metabolic diseases remains unanswered. We recently discovered a novel function for bilirubin as a signaling molecule capable of activating the peroxisome proliferator-activated receptor α (PPARα) transcription factor. This review summarizes the new finding of bilirubin as a signaling molecule and proposes several mechanisms by which this novel action of bilirubin may protect against cardiovascular and kidney diseases.
Collapse
Affiliation(s)
- David E. Stec
- Department of Physiology and Biophysics, Cardiorenal, and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | | | - Olufunto O. Badmus
- Department of Physiology and Biophysics, Cardiorenal, and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Terry D. Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky,Barnstable Brown Diabetes Center, University of Kentucky, Lexington, Kentucky,Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
31
|
Reactive Oxygen Species (ROS) and Antioxidants as Immunomodulators in Exercise: Implications for Heme Oxygenase and Bilirubin. Antioxidants (Basel) 2022; 11:antiox11020179. [PMID: 35204062 PMCID: PMC8868548 DOI: 10.3390/antiox11020179] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 02/07/2023] Open
Abstract
Exercise is commonly prescribed as a lifestyle treatment for chronic metabolic diseases as it functions as an insulin sensitizer, cardio-protectant, and essential lifestyle tool for effective weight maintenance. Exercise boosts the production of reactive oxygen species (ROS) and subsequent transient oxidative damage, which also upregulates counterbalancing endogenous antioxidants to protect from ROS-induced damage and inflammation. Exercise elevates heme oxygenase-1 (HO-1) and biliverdin reductase A (BVRA) expression as built-in protective mechanisms, which produce the most potent antioxidant, bilirubin. Together, these mitigate inflammation and adiposity. Moderately raising plasma bilirubin protects in two ways: (1) via its antioxidant capacity to reduce ROS and inflammation, and (2) its newly defined function as a hormone that activates the nuclear receptor transcription factor PPARα. It is now understood that increasing plasma bilirubin can also drive metabolic adaptions, which improve deleterious outcomes of weight gain and obesity, such as inflammation, type II diabetes, and cardiovascular diseases. The main objective of this review is to describe the function of bilirubin as an antioxidant and metabolic hormone and how the HO-1-BVRA-bilirubin-PPARα axis influences inflammation, metabolic function and interacts with exercise to improve outcomes of weight management.
Collapse
|
32
|
Hinds TD, Kipp ZA, Xu M, Yiannikouris FB, Morris AJ, Stec DF, Wahli W, Stec DE. Adipose-Specific PPARα Knockout Mice Have Increased Lipogenesis by PASK-SREBP1 Signaling and a Polarity Shift to Inflammatory Macrophages in White Adipose Tissue. Cells 2021; 11:4. [PMID: 35011564 PMCID: PMC8750478 DOI: 10.3390/cells11010004] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 12/16/2022] Open
Abstract
The nuclear receptor PPARα is associated with reducing adiposity, especially in the liver, where it transactivates genes for β-oxidation. Contrarily, the function of PPARα in extrahepatic tissues is less known. Therefore, we established the first adipose-specific PPARα knockout (PparaFatKO) mice to determine the signaling position of PPARα in adipose tissue expansion that occurs during the development of obesity. To assess the function of PPARα in adiposity, female and male mice were placed on a high-fat diet (HFD) or normal chow for 30 weeks. Only the male PparaFatKO animals had significantly more adiposity in the inguinal white adipose tissue (iWAT) and brown adipose tissue (BAT) with HFD, compared to control littermates. No changes in adiposity were observed in female mice compared to control littermates. In the males, the loss of PPARα signaling in adipocytes caused significantly higher cholesterol esters, activation of the transcription factor sterol regulatory element-binding protein-1 (SREBP-1), and a shift in macrophage polarity from M2 to M1 macrophages. We found that the loss of adipocyte PPARα caused significantly higher expression of the Per-Arnt-Sim kinase (PASK), a kinase that activates SREBP-1. The hyperactivity of the PASK-SREBP-1 axis significantly increased the lipogenesis proteins fatty acid synthase (FAS) and stearoyl-Coenzyme A desaturase 1 (SCD1) and raised the expression of genes for cholesterol metabolism (Scarb1, Abcg1, and Abca1). The loss of adipocyte PPARα increased Nos2 in the males, an M1 macrophage marker indicating that the population of macrophages had changed to proinflammatory. Our results demonstrate the first adipose-specific actions for PPARα in protecting against lipogenesis, inflammation, and cholesterol ester accumulation that leads to adipocyte tissue expansion in obesity.
Collapse
Affiliation(s)
- Terry D. Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40508, USA; (Z.A.K.); (M.X.); (F.B.Y.)
- Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY 40508, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40508, USA
| | - Zachary A. Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40508, USA; (Z.A.K.); (M.X.); (F.B.Y.)
| | - Mei Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40508, USA; (Z.A.K.); (M.X.); (F.B.Y.)
| | - Frederique B. Yiannikouris
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40508, USA; (Z.A.K.); (M.X.); (F.B.Y.)
- Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY 40508, USA
| | - Andrew J. Morris
- Division of Cardiovascular Medicine, College of Medicine, University of Kentucky, Lexington, KY 40508, USA;
- Lexington Veterans Affairs Medical Center, Lexington, KY 40508, USA
| | - Donald F. Stec
- Small Molecule NMR Facility Core, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, USA;
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, Singapore 308232, Singapore;
- Toxalim Research Center in Food Toxicology (UMR 1331), INRAE, ENVT, INP—PURPAN, UPS, Université de Toulouse, F-31300 Toulouse, France
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
| | - David E. Stec
- Department of Physiology & Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
33
|
Bortolussi G, Shi X, ten Bloemendaal L, Banerjee B, De Waart DR, Baj G, Chen W, Oude Elferink RP, Beuers U, Paulusma CC, Stocker R, Muro AF, Bosma PJ. Long-Term Effects of Biliverdin Reductase a Deficiency in Ugt1-/- Mice: Impact on Redox Status and Metabolism. Antioxidants (Basel) 2021; 10:antiox10122029. [PMID: 34943131 PMCID: PMC8698966 DOI: 10.3390/antiox10122029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/07/2021] [Accepted: 12/15/2021] [Indexed: 11/16/2022] Open
Abstract
Accumulation of neurotoxic bilirubin due to a transient neonatal or persistent inherited deficiency of bilirubin glucuronidation activity can cause irreversible brain damage and death. Strategies to inhibit bilirubin production and prevent neurotoxicity in neonatal and adult settings seem promising. We evaluated the impact of Bvra deficiency in neonatal and aged mice, in a background of unconjugated hyperbilirubinemia, by abolishing bilirubin production. We also investigated the disposal of biliverdin during fetal development. In Ugt1−/− mice, Bvra deficiency appeared sufficient to prevent lethality and to normalize bilirubin level in adults. Although biliverdin accumulated in Bvra-deficient fetuses, both Bvra−/− and Bvra−/−Ugt1−/− pups were healthy and reached adulthood having normal liver, brain, and spleen histology, albeit with increased iron levels in the latter. During aging, both Bvra−/− and Bvra−/−Ugt1−/− mice presented normal levels of relevant hematological and metabolic parameters. Interestingly, the oxidative status in erythrocytes from 9-months-old Bvra−/− and Bvra−/−Ugt1−/− mice was significantly reduced. In addition, triglycerides levels in these 9-months-old Bvra−/− mice were significantly higher than WT controls, while Bvra−/−Ugt1−/− tested normal. The normal parameters observed in Bvra−/−Ugt1−/− mice fed chow diet indicate that Bvra inhibition to treat unconjugated hyperbilirubinemia seems safe and effective.
Collapse
Affiliation(s)
- Giulia Bortolussi
- International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy; (G.B.); (B.B.)
| | - Xiaoxia Shi
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (X.S.); (L.t.B.); (D.R.D.W.); (R.P.O.E.); (U.B.); (C.C.P.)
- Key Laboratory of Protein Modification and Disease, School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Lysbeth ten Bloemendaal
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (X.S.); (L.t.B.); (D.R.D.W.); (R.P.O.E.); (U.B.); (C.C.P.)
| | - Bhaswati Banerjee
- International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy; (G.B.); (B.B.)
| | - Dirk R. De Waart
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (X.S.); (L.t.B.); (D.R.D.W.); (R.P.O.E.); (U.B.); (C.C.P.)
| | - Gabriele Baj
- Light Microscopy Imaging Center, Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
| | - Weiyu Chen
- Heart Research Institute, Sydney, NSW 2042, Australia; (W.C.); (R.S.)
| | - Ronald P. Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (X.S.); (L.t.B.); (D.R.D.W.); (R.P.O.E.); (U.B.); (C.C.P.)
| | - Ulrich Beuers
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (X.S.); (L.t.B.); (D.R.D.W.); (R.P.O.E.); (U.B.); (C.C.P.)
| | - Coen C. Paulusma
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (X.S.); (L.t.B.); (D.R.D.W.); (R.P.O.E.); (U.B.); (C.C.P.)
| | - Roland Stocker
- Heart Research Institute, Sydney, NSW 2042, Australia; (W.C.); (R.S.)
| | - Andrés F. Muro
- International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy; (G.B.); (B.B.)
- Correspondence: (A.F.M.); (P.J.B.); Tel.: +39-040-3757369 (A.F.M.); +31-20-566-8850 (P.J.B.)
| | - Piter J. Bosma
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (X.S.); (L.t.B.); (D.R.D.W.); (R.P.O.E.); (U.B.); (C.C.P.)
- Correspondence: (A.F.M.); (P.J.B.); Tel.: +39-040-3757369 (A.F.M.); +31-20-566-8850 (P.J.B.)
| |
Collapse
|
34
|
Cimini FA, Barchetta I, Zuliani I, Pagnotta S, Bertoccini L, Dule S, Zampieri M, Reale A, Baroni MG, Cavallo MG, Barone E. Biliverdin reductase-A protein levels are reduced in type 2 diabetes and are associated with poor glycometabolic control. Life Sci 2021; 284:119913. [PMID: 34453944 DOI: 10.1016/j.lfs.2021.119913] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 12/14/2022]
Abstract
AIM Biliverdin reductase-A (BVR-A) other than its canonical role in the degradation pathway of heme as partner of heme oxygenase-1 (HO1), has recently drawn attention as a protein with pleiotropic functions involved in insulin-glucose homeostasis. However, whether BVR-A expression is altered in type 2 diabetes (T2D) has never been evaluated. MAIN METHODS BVR-A protein levels were evaluated in T2D (n = 44) and non-T2D (n = 29) subjects, who underwent complete clinical workup and routine biochemistry. In parallel, levels HO1, whose expression is regulated by BVR-A as well as levels of tumor necrosis factor α (TNFα), which is a known repressor for BVR-A with pro-inflammatory properties, were also assessed. KEY FINDINGS BVR-A levels were significantly lower in T2D subjects than in non-T2D subjects. Reduced BVR-A levels were associated with greater body mass, systolic blood pressure, fasting blood glucose (FBG), glycated hemoglobin (HbA1c), triglycerides, transaminases and TNFα, and with lower high-density lipoprotein (HDL) levels. Lower BVR-A levels are associated with reduced HO1 protein levels and the multivariate analysis showed that BVR-A represented the main determinant of HO1 levels in T2D after adjustment. In addition, reduced BVR-A levels were able to predict the presence of T2D with AUROC = 0.69. for potential confounders. SIGNIFICANCE Our results demonstrate for the first time that BVR-A protein levels are reduced in T2D individuals, and that this alteration strictly correlates with poor glycometabolic control and a pro-inflammatory state. Hence, these observations reinforce the hypothesis that reduced BVR-A protein levels may represent a key event in the dysregulation of intracellular pathways finally leading to metabolic disorders.
Collapse
Affiliation(s)
| | - Ilaria Barchetta
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Ilaria Zuliani
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Sara Pagnotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Laura Bertoccini
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Sara Dule
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Michele Zampieri
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Anna Reale
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Marco Giorgio Baroni
- Department of Clinical Medicine, Public Health, Life and Environmental Sciences (MeSVA), University of L'Aquila, Italy; Neuroendocrinology and Metabolic Diseases, IRCCS Neuromed, Pozzilli, Is, Italy
| | | | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy.
| |
Collapse
|
35
|
Smedlund KB, Sanchez ER, Hinds TD. FKBP51 and the molecular chaperoning of metabolism. Trends Endocrinol Metab 2021; 32:862-874. [PMID: 34481731 PMCID: PMC8516732 DOI: 10.1016/j.tem.2021.08.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/31/2021] [Accepted: 08/06/2021] [Indexed: 01/30/2023]
Abstract
The molecular chaperone FK506-binding protein 51 (FKBP51) is gaining attention as a meaningful biomarker of metabolic dysfunction. This review examines the emerging contributions of FKBP51 in adipogenesis and lipid metabolism, myogenesis and protein catabolism, and glucocorticoid-induced skin hypoplasia and dermal adipocytes. The FKBP51 signaling mechanisms that may explain these metabolic consequences are discussed. These mechanisms are diverse, with FKBP51 independently and directly regulating phosphorylation cascades and nuclear receptors. We provide a discussion of the newly developed compounds that antagonize FKBP51, which may offer therapeutic advantages for adiposity. These observations suggest we are only beginning to uncover the complex nature of FKBP51 and its molecular chaperoning of metabolism.
Collapse
Affiliation(s)
- Kathryn B Smedlund
- Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Edwin R Sanchez
- Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Terry D Hinds
- Barnstable Brown Diabetes Center, Markey Cancer Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40508, USA.
| |
Collapse
|
36
|
Bilirubin deficiency renders mice susceptible to hepatic steatosis in the absence of insulin resistance. Redox Biol 2021; 47:102152. [PMID: 34610553 PMCID: PMC8498001 DOI: 10.1016/j.redox.2021.102152] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/26/2021] [Accepted: 09/26/2021] [Indexed: 12/31/2022] Open
Abstract
Background & aims Plasma concentrations of bilirubin, a product of heme catabolism formed by biliverdin reductase A (BVRA), inversely associate with the risk of metabolic diseases including hepatic steatosis and diabetes mellitus in humans. Bilirubin has antioxidant and anti-inflammatory activities and may also regulate insulin signaling and peroxisome proliferator-activated receptor alpha (PPARα) activity. However, a causal link between bilirubin and metabolic diseases remains to be established. Here, we used the global Bvra gene knockout (Bvra–/–) mouse as a model of deficiency in bilirubin to assess its role in metabolic diseases. Approach & results We fed mice fat-rich diets to induce hepatic steatosis and insulin resistance. Bile pigments were measured by LC-MS/MS, and hepatic lipids by LC-MS/MS (non-targeted lipidomics), HPLC-UV and Oil-Red-O staining. Oxidative stress was evaluated measuring F2-isoprostanes by GC-MS. Glucose metabolism and insulin sensitivity were verified by glucose and insulin tolerance tests, ex vivo and in vivo glucose uptake, and Western blotting for insulin signaling. Compared with wild type littermates, Bvra–/– mice contained negligible bilirubin in plasma and liver, and they had comparable glucose metabolism and insulin sensitivity. However, Bvra–/– mice exhibited an inflamed and fatty liver phenotype, accompanied by hepatic accumulation of oxidized triacylglycerols and F2-isoprostanes, in association with depletion of α-tocopherol. α-Tocopherol supplementation reversed the hepatic phenotype and observed biochemical changes in Bvra–/– mice. Conclusions Our data suggests that BVRA deficiency renders mice susceptible to oxidative stress-induced hepatic steatosis in the absence of insulin resistance. Low plasma levels of bilirubin associate with increased metabolic disease risk. A direct link between bilirubin and metabolic disease remains to be established. Global BVRA deficiency causes global bilirubin deficiency and a fatty, inflamed liver. This hepatic phenotype is linked to decreased vitamin E and increased lipid oxidation. Vitamin E supplements restore normal liver phenotype in BVRA deficiency.
Collapse
|
37
|
Adin CA. Bilirubin as a Therapeutic Molecule: Challenges and Opportunities. Antioxidants (Basel) 2021; 10:1536. [PMID: 34679671 PMCID: PMC8532879 DOI: 10.3390/antiox10101536] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
There is strong evidence that serum free bilirubin concentration has significant effects on morbidity and mortality in the most significant health conditions of our times, including cardiovascular disease, diabetes, and obesity/metabolic syndrome. Supplementation of bilirubin in animal and experimental models has reproduced these protective effects, but several factors have slowed the application bilirubin as a therapeutic agent in human patients. Bilirubin is poorly soluble in water, and is a complex molecule that is difficult to synthesize. Current sources of this molecule are animal-derived, creating concerns regarding the risk of virus or prion transmission. However, recent developments in nanoparticle drug delivery, biosynthetic strategies, and drug synthesis have opened new avenues for applying bilirubin as a pharmaceutical agent. This article reviews the chemistry and physiology of bilirubin, potential clinical applications and summarizes current strategies for safe and efficient drug delivery.
Collapse
Affiliation(s)
- Christopher A Adin
- Department of Small Animal Clinical Sciences, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
38
|
Abstract
Significance: As the central metabolic organ, the liver is exposed to a variety of potentially cytotoxic, proinflammatory, profibrotic, and carcinogenic stimuli. To protect the organism from these deleterious effects, the liver has evolved a number of defense systems, which include antioxidant substrates and enzymes, anti-inflammatory tools, enzymatic biotransformation systems, and metabolic pathways. Recent Advances: One of the pivotal systems that evolved during phylogenesis was the heme catabolic pathway. Comprising the important enzymes heme oxygenase and biliverdin reductase, this complex pathway has a number of key functions including enzymatic activities, but also cell signaling, and DNA transcription. It further generates two important bile pigments, biliverdin and bilirubin, as well as the gaseous molecule carbon monoxide. These heme degradation products have potent antioxidant, immunosuppressive, and cytoprotective effects. Recent data suggest that the pathway participates in the regulation of metabolic and hormonal processes implicated in the pathogenesis of hepatic and other diseases. Critical Issues: This review discusses the impact of the heme catabolic pathway on major liver diseases, with particular focus on the involvement of cellular targeting and signaling in the pathogenesis of these conditions. Future Directions: To utilize the biological consequences of the heme catabolic pathway, several unique therapeutic strategies have been developed. Research indicates that pharmaceutical, nutraceutical, and lifestyle modifications positively affect the pathway, delivering potentially long-term clinical benefits. However, further well-designed studies are needed to confirm the clinical benefits of these approaches. Antioxid. Redox Signal. 35, 734-752.
Collapse
Affiliation(s)
- Libor Vítek
- Fourth Department of Internal Medicine, and Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
39
|
McClung JA, Levy L, Garcia V, Stec DE, Peterson SJ, Abraham NG. Heme-oxygenase and lipid mediators in obesity and associated cardiometabolic diseases: Therapeutic implications. Pharmacol Ther 2021; 231:107975. [PMID: 34499923 DOI: 10.1016/j.pharmthera.2021.107975] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/08/2021] [Accepted: 07/27/2021] [Indexed: 02/08/2023]
Abstract
Obesity-mediated metabolic syndrome remains the leading cause of death worldwide. Among many potential targets for pharmacological intervention, a promising strategy involves the heme oxygenase (HO) system, specifically its inducible form, HO-1. This review collects and updates much of the current knowledge relevant to pharmacology and clinical medicine concerning HO-1 in metabolic diseases and its effect on lipid metabolism. HO-1 has pleotropic effects that collectively reduce inflammation, while increasing vasodilation and insulin and leptin sensitivity. Recent reports indicate that HO-1 with its antioxidants via the effect of bilirubin increases formation of biologically active lipid metabolites such as epoxyeicosatrienoic acid (EET), omega-3 and other polyunsaturated fatty acids (PUFAs). Similarly, HO-1and bilirubin are potential therapeutic targets in the treatment of fat-induced liver diseases. HO-1-mediated upregulation of EET is capable not only of reversing endothelial dysfunction and hypertension, but also of reversing cardiac remodeling, a hallmark of the metabolic syndrome. This process involves browning of white fat tissue (i.e. formation of healthy adipocytes) and reduced lipotoxicity, which otherwise will be toxic to the heart. More importantly, this review examines the activity of EET in biological systems and a series of pathways that explain its mechanism of action and discusses how these might be exploited for potential therapeutic use. We also discuss the link between cardiac ectopic fat deposition and cardiac function in humans, which is similar to that described in obese mice and is regulated by HO-1-EET-PGC1α signaling, a potent negative regulator of the inflammatory adipokine NOV.
Collapse
Affiliation(s)
- John A McClung
- Department of Medicine, New York Medical College, Valhalla, NY 10595, United States of America
| | - Lior Levy
- Department of Medicine, New York Medical College, Valhalla, NY 10595, United States of America
| | - Victor Garcia
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States of America
| | - David E Stec
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS 39216, United States of America.
| | - Stephen J Peterson
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, United States of America; New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, United States of America
| | - Nader G Abraham
- Department of Medicine, New York Medical College, Valhalla, NY 10595, United States of America; Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States of America.
| |
Collapse
|
40
|
Gordon DM, Hong SH, Kipp ZA, Hinds TD. Identification of Binding Regions of Bilirubin in the Ligand-Binding Pocket of the Peroxisome Proliferator-Activated Receptor-A (PPARalpha). Molecules 2021; 26:molecules26102975. [PMID: 34067839 PMCID: PMC8157031 DOI: 10.3390/molecules26102975] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 11/16/2022] Open
Abstract
Recent work has shown that bilirubin has a hormonal function by binding to the peroxisome proliferator-activated receptor-α (PPARα), a nuclear receptor that drives the transcription of genes to control adiposity. Our previous in silico work predicted three potential amino acids that bilirubin may interact with by hydrogen bonding in the PPARα ligand-binding domain (LBD), which could be responsible for the ligand-induced function. To further reveal the amino acids that bilirubin interacts with in the PPARα LBD, we harnessed bilirubin’s known fluorescent properties when bound to proteins such as albumin. Our work here revealed that bilirubin interacts with threonine 283 (T283) and alanine 333 (A333) for ligand binding. Mutational analysis of T283 and A333 showed significantly reduced bilirubin binding, reductions of 11.4% and 17.0%, respectively. Fenofibrate competitive binding studies for the PPARα LBD showed that bilirubin and fenofibrate possibly interact with different amino acid residues. Furthermore, bilirubin showed no interaction with PPARγ. This is the first study to reveal the amino acids responsible for bilirubin binding in the ligand-binding pocket of PPARα. Our work offers new insight into the mechanistic actions of a well-known molecule, bilirubin, and new fronts into its mechanisms.
Collapse
Affiliation(s)
- Darren M. Gordon
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (D.M.G.); (S.H.H.)
| | - Stephen H. Hong
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (D.M.G.); (S.H.H.)
| | - Zachary A. Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, 760 Press Avenue, Healthy Kentucky Research Building, Lexington, KY 40508, USA;
| | - Terry D. Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, 760 Press Avenue, Healthy Kentucky Research Building, Lexington, KY 40508, USA;
- Correspondence:
| |
Collapse
|
41
|
Widespread severe cerebral elevations of haptoglobin and haemopexin in sporadic Alzheimer's disease: Evidence for a pervasive microvasculopathy. Biochem Biophys Res Commun 2021; 555:89-94. [PMID: 33813281 DOI: 10.1016/j.bbrc.2021.02.107] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 02/22/2021] [Indexed: 12/29/2022]
Abstract
Sporadic Alzheimer's disease (sAD) is the commonest cause of age-related neurodegeneration but there are no available treatments with demonstrated disease-modifying actions. It is therefore relevant to study hitherto-unknown aspects of brain structure and function to seek new disease-related mechanisms that might be targeted by novel disease-modifying interventions. During hypothesis-generating proteomic investigations in a case-control study of sAD, we observed widespread elevations of haptoglobin and haemopexin in all six brain-regions studied, which together represent much of the brain. Measured perturbations were significant, with the posterior probability of upregulation generally >95% and haptoglobin doubling in expression levels on average across deep brain structures (hippocampus, entorhinal cortex and cingulate gyrus) as well as sensory and motor cortices, and cerebellum. Haptoglobin and haemopexin are often regarded as circulating proteins whose main functions are to bind, respectively, the strongly pro-inflammatory extracellular haemoglobin and haeme molecules that form following haemolysis, thereby promoting their clearance and suppressing damage they might otherwise cause, for example, acute kidney injury. To our knowledge, elevations in neither cerebral haptoglobin nor haemopexin have previously been linked to the pathogenesis of sAD. Post-mortem examination of these cases showed no signs of macroscopic cerebral haemorrhage. These findings demonstrate pervasive cerebral elevation of haptoglobin and haemopexin, consistent with low-level intracerebral leakage of haemoglobin and consequent haeme formation throughout sAD brain. They point to a widespread underlying microvasculopathy that facilitates erythrocyte leakage, thereby triggering elevated tissue-free haemoglobin and driving the measured elevations in haptoglobin and haemopexin.
Collapse
|
42
|
Brasil FB, Bertolini Gobbo RC, Souza de Almeida FJ, Luckachaki MD, Dall'Oglio EL, de Oliveira MR. The signaling pathway PI3K/Akt/Nrf2/HO-1 plays a role in the mitochondrial protection promoted by astaxanthin in the SH-SY5Y cells exposed to hydrogen peroxide. Neurochem Int 2021; 146:105024. [PMID: 33775716 DOI: 10.1016/j.neuint.2021.105024] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/26/2021] [Accepted: 03/18/2021] [Indexed: 12/19/2022]
Abstract
The mitochondria are the major source of reactive species in the mammalian cells. Hydrogen peroxide (H2O2) is a potent inducer of redox impairment by a mechanism, at least in part, dependent on its ability to impair mitochondrial function. H2O2 plays an important role in several pathological conditions, including neurodegeneration and cardiovascular diseases. Astaxanthin (AST) is a xanthophyll that may be found in microalgae, crustaceans, and salmon and exhibits antioxidant and anti-inflammatory effects in different cell types. Even though there is evidence pointing to a role for AST as mitochondrial protectant agent, it was not clearly demonstrated how this xanthophyll attenuates mitochondrial stress. Therefore, we investigated here whether and how AST would be able to prevent the H2O2-induced mitochondrial dysfunction in the human neuroblastoma SH-SY5Y cells. We found that AST (20 μM) prevented the H2O2-induced loss of mitochondrial membrane potential (MMP) and decrease in the activity of the Complexes I and V. AST pretreatment blocked the mitochondria-related pro-apoptotic effects elicited by H2O2. AST upregulated the enzyme heme oxygenase-1 (HO-1) and the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) by a mechanism dependent on the phosphoinositide 3-kinase/Akt (PI3K/Akt) signaling pathway. Inhibition of the PI3K/Akt or of the HO-1 enzyme abolished the AST-induced mitochondrial protection in cells challenged with H2O2. Silencing of Nrf2 caused similar effects. Thus, we suggest that AST promotes mitochondrial protection by a mechanism dependent on the PI3K/Akt/Nrf2/HO-1 signaling pathway in SH-SY5Y cells exposed to H2O2.
Collapse
Affiliation(s)
- Flávia Bittencourt Brasil
- Departamento de Ciências da Natureza, Campus Universitário de Rio das Ostras - Universidade Federal Fluminense (UFF), Rio de Janeiro, Brazil
| | - Rênata Cristina Bertolini Gobbo
- Grupo de Estudos em Terapia Mitocondrial, Departamento de Bioquímica "Tuiskon Dick", Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600 (Anexo), CEP 90035-000, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica "Tuiskon Dick", Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Fhelipe Jolner Souza de Almeida
- Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Universidade Federal de Mato Grosso (UFMT), Cuiaba, MT, Brazil; Grupo de Estudos em Neuroquímica e Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, CEP 78060-900, Cuiaba, MT, Brazil
| | - Matheus Dargesso Luckachaki
- Grupo de Estudos em Neuroquímica e Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, CEP 78060-900, Cuiaba, MT, Brazil
| | - Evandro Luiz Dall'Oglio
- Grupo de Estudos em Neuroquímica e Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, CEP 78060-900, Cuiaba, MT, Brazil
| | - Marcos Roberto de Oliveira
- Grupo de Estudos em Terapia Mitocondrial, Departamento de Bioquímica "Tuiskon Dick", Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600 (Anexo), CEP 90035-000, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica "Tuiskon Dick", Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Grupo de Estudos em Neuroquímica e Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, CEP 78060-900, Cuiaba, MT, Brazil.
| |
Collapse
|
43
|
Creeden JF, Gordon DM, Stec DE, Hinds TD. Bilirubin as a metabolic hormone: the physiological relevance of low levels. Am J Physiol Endocrinol Metab 2021; 320:E191-E207. [PMID: 33284088 PMCID: PMC8260361 DOI: 10.1152/ajpendo.00405.2020] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Recent research on bilirubin, a historically well-known waste product of heme catabolism, suggests an entirely new function as a metabolic hormone that drives gene transcription by nuclear receptors. Studies are now revealing that low plasma bilirubin levels, defined as "hypobilirubinemia," are a possible new pathology analogous to the other end of the spectrum of extreme hyperbilirubinemia seen in patients with jaundice and liver dysfunction. Hypobilirubinemia is most commonly seen in patients with metabolic dysfunction, which may lead to cardiovascular complications and possibly stroke. We address the clinical significance of low bilirubin levels. A better understanding of bilirubin's hormonal function may explain why hypobilirubinemia might be deleterious. We present mechanisms by which bilirubin may be protective at mildly elevated levels and research directions that could generate treatment possibilities for patients with hypobilirubinemia, such as targeting of pathways that regulate its production or turnover or the newly designed bilirubin nanoparticles. Our review here calls for a shift in the perspective of an old molecule that could benefit millions of patients with hypobilirubinemia.
Collapse
Affiliation(s)
- Justin F Creeden
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Darren M Gordon
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - David E Stec
- Department of Physiology & Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
44
|
Hinds TD, Creeden JF, Gordon DM, Stec DF, Donald MC, Stec DE. Bilirubin Nanoparticles Reduce Diet-Induced Hepatic Steatosis, Improve Fat Utilization, and Increase Plasma β-Hydroxybutyrate. Front Pharmacol 2020; 11:594574. [PMID: 33390979 PMCID: PMC7775678 DOI: 10.3389/fphar.2020.594574] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/13/2020] [Indexed: 12/18/2022] Open
Abstract
The inverse relationship of plasma bilirubin levels with liver fat accumulation has prompted the possibility of bilirubin as a therapeutic for non-alcoholic fatty liver disease. Here, we used diet-induced obese mice with non-alcoholic fatty liver disease treated with pegylated bilirubin (bilirubin nanoparticles) or vehicle control to determine the impact on hepatic lipid accumulation. The bilirubin nanoparticles significantly reduced hepatic fat, triglyceride accumulation, de novo lipogenesis, and serum levels of liver dysfunction marker aspartate transaminase and ApoB100 containing very-low-density lipoprotein. The bilirubin nanoparticles improved liver function and activated the hepatic β-oxidation pathway by increasing PPARα and acyl-coenzyme A oxidase 1. The bilirubin nanoparticles also significantly elevated plasma levels of the ketone β-hydroxybutyrate and lowered liver fat accumulation. This study demonstrates that bilirubin nanoparticles induce hepatic fat utilization, raise plasma ketones, and reduce hepatic steatosis, opening new therapeutic avenues for NAFLD.
Collapse
Affiliation(s)
- Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Justin F Creeden
- Department of Neurosciences, University of Toledo College of Medicine, Toledo, OH, United States
| | - Darren M Gordon
- Department of Neurosciences, University of Toledo College of Medicine, Toledo, OH, United States
| | - Donald F Stec
- Small Molecule NMR Facility Core, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, United States
| | - Matthew C Donald
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS, United States
| | - David E Stec
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
45
|
Ceccarelli V, Barchetta I, Cimini FA, Bertoccini L, Chiappetta C, Capoccia D, Carletti R, Di Cristofano C, Silecchia G, Fontana M, Leonetti F, Lenzi A, Baroni MG, Barone E, Cavallo MG. Reduced Biliverdin Reductase-A Expression in Visceral Adipose Tissue is Associated with Adipocyte Dysfunction and NAFLD in Human Obesity. Int J Mol Sci 2020; 21:ijms21239091. [PMID: 33260451 PMCID: PMC7730815 DOI: 10.3390/ijms21239091] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
Biliverdin reductase A (BVR-A) is an enzyme involved in the regulation of insulin signalling. Knockout (KO) mice for hepatic BVR-A, on a high-fat diet, develop more severe glucose impairment and hepato-steatosis than the wild type, whereas loss of adipocyte BVR-A is associated with increased visceral adipose tissue (VAT) inflammation and adipocyte size. However, BVR-A expression in human VAT has not been investigated. We evaluated BVR-A mRNA expression levels by real-time PCR in the intra-operative omental biopsy of 38 obese subjects and investigated the association with metabolic impairment, VAT dysfunction, and biopsy-proven non-alcoholic fatty liver disease (NAFLD). Individuals with lower VAT BVR-A mRNA levels had significantly greater VAT IL-8 and Caspase 3 expression than those with higher BVR-A. Lower VAT BVR-A mRNA levels were associated with an increased adipocytes’ size. An association between lower VAT BVR-A expression and higher plasma gamma-glutamyl transpeptidase was also observed. Reduced VAT BVR-A was associated with NAFLD with an odds ratio of 1.38 (95% confidence interval: 1.02–1.9; χ2 test) and with AUROC = 0.89 (p = 0.002, 95% CI = 0.76–1.0). In conclusion, reduced BVR-A expression in omental adipose tissue is associated with VAT dysfunction and NAFLD, suggesting a possible involvement of BVR-A in the regulation of VAT homeostasis in presence of obesity.
Collapse
Affiliation(s)
- Valentina Ceccarelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (I.B.); (F.A.C.); (L.B.); (A.L.)
| | - Ilaria Barchetta
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (I.B.); (F.A.C.); (L.B.); (A.L.)
| | - Flavia Agata Cimini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (I.B.); (F.A.C.); (L.B.); (A.L.)
| | - Laura Bertoccini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (I.B.); (F.A.C.); (L.B.); (A.L.)
| | - Caterina Chiappetta
- Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, 04100 Latina, Italy; (C.C.); (D.C.); (R.C.); (C.D.C.); (G.S.); (F.L.)
| | - Danila Capoccia
- Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, 04100 Latina, Italy; (C.C.); (D.C.); (R.C.); (C.D.C.); (G.S.); (F.L.)
| | - Raffaella Carletti
- Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, 04100 Latina, Italy; (C.C.); (D.C.); (R.C.); (C.D.C.); (G.S.); (F.L.)
| | - Claudio Di Cristofano
- Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, 04100 Latina, Italy; (C.C.); (D.C.); (R.C.); (C.D.C.); (G.S.); (F.L.)
| | - Gianfranco Silecchia
- Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, 04100 Latina, Italy; (C.C.); (D.C.); (R.C.); (C.D.C.); (G.S.); (F.L.)
| | - Mario Fontana
- Department of Biochemical Sciences “A. Rossi-Fanelli” Sapienza University of Rome, 00185 Rome, Italy;
| | - Frida Leonetti
- Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, 04100 Latina, Italy; (C.C.); (D.C.); (R.C.); (C.D.C.); (G.S.); (F.L.)
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (I.B.); (F.A.C.); (L.B.); (A.L.)
| | - Marco Giorgio Baroni
- Department of Clinical Medicine, Public Health, Life and Environmental Sciences (MeSVA), University of L’Aquila, 67100 Coppito, Italy;
- Neuroendocrinology and Metabolic Diseases, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences “A. Rossi-Fanelli” Sapienza University of Rome, 00185 Rome, Italy;
- Correspondence: (E.B.); (M.G.C.); Tel.: +39-(0)6-4997-4692 (M.G.C.)
| | - Maria Gisella Cavallo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (I.B.); (F.A.C.); (L.B.); (A.L.)
- Correspondence: (E.B.); (M.G.C.); Tel.: +39-(0)6-4997-4692 (M.G.C.)
| |
Collapse
|
46
|
Chen K, Yuan T. The role of microbiota in neonatal hyperbilirubinemia. Am J Transl Res 2020; 12:7459-7474. [PMID: 33312382 PMCID: PMC7724329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/24/2020] [Indexed: 06/12/2023]
Abstract
Hyperbilirubinemia accounts for about 60% of full-term and 80% of preterm neonates globally, which is characterized by physiologically elevated unconjugated bilirubin in serum, but abnormally high levels of bilirubin have potential neurotoxic effects. Several factors contribute to the development of neonatal hyperbilirubinemia, including isoimmunization, dysregulated gut flora, genetic alteration and environmental factors. Animal studies have pinpointed the causal roles of several bacteria in bilirubin metabolism. Human studies have revealed microbiota composition in hyperbilirubinemia and found that gut microbiota affect newborns with different severity of hyperbilirubinemia. However, dysbiosis and subsequent changes in microbiota-related metabolic processes are not always considered. This review aims to describe the critical microbiota signatures for neonatal hyperbilirubinemia and focus on the underlying pathogenetic mechanism. These scientific bases give a new and accurate therapeutic strategy for the application of gut microbiota.
Collapse
Affiliation(s)
- Kewei Chen
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health Zhejiang, PR China
| | - Tianming Yuan
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health Zhejiang, PR China
| |
Collapse
|
47
|
Jayanti S, Vítek L, Tiribelli C, Gazzin S. The Role of Bilirubin and the Other "Yellow Players" in Neurodegenerative Diseases. Antioxidants (Basel) 2020; 9:900. [PMID: 32971784 PMCID: PMC7555389 DOI: 10.3390/antiox9090900] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/16/2020] [Accepted: 09/20/2020] [Indexed: 02/07/2023] Open
Abstract
Bilirubin is a yellow endogenous derivate of the heme catabolism. Since the 1980s, it has been recognized as one of the most potent antioxidants in nature, able to counteract 10,000× higher intracellular concentrations of H2O2. In the recent years, not only bilirubin, but also its precursor biliverdin, and the enzymes involved in their productions (namely heme oxygenase and biliverdin reductase; altogether the "yellow players"-YPs) have been recognized playing a protective role in diseases characterized by a chronic prooxidant status. Based on that, there is an ongoing effort in inducing their activity as a therapeutic option. Nevertheless, the understanding of their specific contributions to pathological conditions of the central nervous system (CNS) and their role in these diseases are limited. In this review, we will focus on the most recent evidence linking the role of the YPs specifically to neurodegenerative and neurological conditions. Both the protective, as well as potentially worsening effects of the YP's activity will be discussed.
Collapse
Affiliation(s)
- Sri Jayanti
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy; (S.J.); (C.T.)
- Faculty of Medicine, Universitas Hasanuddin, Makassar 90245, Indonesia
- Molecular Biomedicine Ph.D. Program, University of Trieste, 34127 Trieste, Italy
| | - Libor Vítek
- Institute of Medical Biochemistry and Laboratory Diagnostics, and 4th Department of Internal Medicine, Faculty General Hospital and 1st Faculty of Medicine, Charles University, 12000 Prague, Czech Republic;
| | - Claudio Tiribelli
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy; (S.J.); (C.T.)
| | - Silvia Gazzin
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy; (S.J.); (C.T.)
| |
Collapse
|
48
|
Hinds TD, Creeden JF, Gordon DM, Spegele AC, Britton SL, Koch LG, Stec DE. Rats Genetically Selected for High Aerobic Exercise Capacity Have Elevated Plasma Bilirubin by Upregulation of Hepatic Biliverdin Reductase-A (BVRA) and Suppression of UGT1A1. Antioxidants (Basel) 2020; 9:antiox9090889. [PMID: 32961782 PMCID: PMC7554716 DOI: 10.3390/antiox9090889] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/12/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
Exercise in humans and animals increases plasma bilirubin levels, but the mechanism by which this occurs is unknown. In the present study, we utilized rats genetically selected for high capacity running (HCR) and low capacity running (LCR) to determine pathways in the liver that aerobic exercise modifies to control plasma bilirubin. The HCR rats, compared to the LCR, exhibited significantly higher levels of plasma bilirubin and the hepatic enzyme that produces it, biliverdin reductase-A (BVRA). The HCR also had reduced expression of the glucuronyl hepatic enzyme UGT1A1, which lowers plasma bilirubin. Recently, bilirubin has been shown to activate the peroxisome proliferator-activated receptor-α (PPARα), a ligand-induced transcription factor, and the higher bilirubin HCR rats had significantly increased PPARα-target genes Fgf21, Abcd3, and Gys2. These are known to promote liver function and glycogen storage, which we found by Periodic acid–Schiff (PAS) staining that hepatic glycogen content was higher in the HCR versus the LCR. Our results demonstrate that exercise stimulates pathways that raise plasma bilirubin through alterations in hepatic enzymes involved in bilirubin synthesis and metabolism, improving liver function, and glycogen content. These mechanisms may explain the beneficial effects of exercise on plasma bilirubin levels and health in humans.
Collapse
Affiliation(s)
- Terry D. Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40508, USA
- Correspondence: (T.D.H.J.); (D.E.S.)
| | - Justin F. Creeden
- Department of Neurosciences, University of Toledo College of Medicine, Toledo, OH 43614, USA; (J.F.C.); (D.M.G.)
| | - Darren M. Gordon
- Department of Neurosciences, University of Toledo College of Medicine, Toledo, OH 43614, USA; (J.F.C.); (D.M.G.)
| | - Adam C. Spegele
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA; (A.C.S.); (L.G.K.)
| | - Steven L. Britton
- Department of Anesthesiology, Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Lauren G. Koch
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA; (A.C.S.); (L.G.K.)
| | - David E. Stec
- Center for Excellence in Cardiovascular-Renal Research, Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 North State St, Jackson, MS 392161, USA
- Correspondence: (T.D.H.J.); (D.E.S.)
| |
Collapse
|
49
|
Liu H, Perumal N, Manicam C, Mercieca K, Prokosch V. Proteomics Reveals the Potential Protective Mechanism of Hydrogen Sulfide on Retinal Ganglion Cells in an Ischemia/Reperfusion Injury Animal Model. Pharmaceuticals (Basel) 2020; 13:ph13090213. [PMID: 32867129 PMCID: PMC7557839 DOI: 10.3390/ph13090213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022] Open
Abstract
Glaucoma is the leading cause of irreversible blindness and is characterized by progressive retinal ganglion cell (RGC) degeneration. Hydrogen sulfide (H2S) is a potent neurotransmitter and has been proven to protect RGCs against glaucomatous injury in vitro and in vivo. This study is to provide an overall insight of H2S’s role in glaucoma pathophysiology. Ischemia-reperfusion injury (I/R) was induced in Sprague-Dawley rats (n = 12) by elevating intraocular pressure to 55 mmHg for 60 min. Six of the animals received intravitreal injection of H2S precursor prior to the procedure and the retina was harvested 24 h later. Contralateral eyes were assigned as control. RGCs were quantified and compared within the groups. Retinal proteins were analyzed via label-free mass spectrometry based quantitative proteomics approach. The pathways of the differentially expressed proteins were identified by ingenuity pathway analysis (IPA). H2S significantly improved RGC survival against I/R in vivo (p < 0.001). In total 1115 proteins were identified, 18 key proteins were significantly differentially expressed due to I/R and restored by H2S. Another 11 proteins were differentially expressed following H2S. IPA revealed a significant H2S-mediated activation of pathways related to mitochondrial function, iron homeostasis and vasodilation. This study provides first evidence of the complex role that H2S plays in protecting RGC against I/R.
Collapse
Affiliation(s)
- Hanhan Liu
- Experimental and Translational Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (H.L.); (N.P.); (C.M.)
| | - Natarajan Perumal
- Experimental and Translational Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (H.L.); (N.P.); (C.M.)
| | - Caroline Manicam
- Experimental and Translational Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (H.L.); (N.P.); (C.M.)
| | - Karl Mercieca
- Royal Eye Hospital, School of Medicine, University of Manchester, Manchester M13 9WH, UK;
| | - Verena Prokosch
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
- Correspondence: ; Tel.: +49-1703862250
| |
Collapse
|
50
|
Ayaz-Guner S, Alessio N, Acar MB, Aprile D, Özcan S, Di Bernardo G, Peluso G, Galderisi U. A comparative study on normal and obese mice indicates that the secretome of mesenchymal stromal cells is influenced by tissue environment and physiopathological conditions. Cell Commun Signal 2020; 18:118. [PMID: 32727501 PMCID: PMC7388533 DOI: 10.1186/s12964-020-00614-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The term mesenchymal stromal cells (MSCs) designates an assorted cell population comprised of stem cells, progenitor cells, fibroblasts, and stromal cells. MSCs contribute to the homeostatic maintenance of many organs through paracrine and long-distance signaling. Tissue environment, in both physiological and pathological conditions, may affect the intercellular communication of MSCs. METHODS We performed a secretome analysis of MSCs isolated from subcutaneous adipose tissue (sWAT) and visceral adipose tissue (vWAT), and from bone marrow (BM), of normal and obese mice. RESULTS The MSCs isolated from tissues of healthy mice share a common core of released factors: components of cytoskeletal and extracellular structures; regulators of basic cellular functions, such as protein synthesis and degradation; modulators of endoplasmic reticulum stress; and counteracting oxidative stress. It can be hypothesized that MSC secretome beneficially affects target cells by the horizontal transfer of many released factors. Each type of MSC may exert specific signaling functions, which could be determined by looking at the many factors that are exclusively released from every MSC type. The vWAT-MSCs release factors that play a role in detoxification activity in response to toxic substances and drugs. The sWAT-MSC secretome contains proteins involved in in chondrogenesis, osteogenesis, and angiogenesis. Analysis of BM-MSC secretome revealed that these cells exert a signaling function by remodeling extracellular matrix structures, such as those containing glycosaminoglycans. Obesity status profoundly modified the secretome content of MSCs, impairing the above-described activity and promoting the release of inflammatory factors. CONCLUSION We demonstrated that the content of MSC secretomes depends on tissue microenvironment and that pathological condition may profoundly alter its composition. Video abstract.
Collapse
Affiliation(s)
- Serife Ayaz-Guner
- Department of Molecular Biology and Genetics, Faculty of Life and Natural Science, Abdullah Gül University, Kayseri, Turkey
| | - Nicola Alessio
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | - Mustafa B. Acar
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
- Department of Biology, Faculty of Sciences; Erciyes University, Kayseri, Turkey
| | - Domenico Aprile
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | - Servet Özcan
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
- Department of Biology, Faculty of Sciences; Erciyes University, Kayseri, Turkey
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | | | - Umberto Galderisi
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, 1900 N. 12th St, Philadelphia, PA 19107-6799 USA
| |
Collapse
|