1
|
El-Mahrouk SR, El-Ghiaty MA, El-Kadi AOS. The role of nuclear factor erythroid 2-related factor 2 (NRF2) in arsenic toxicity. J Environ Sci (China) 2025; 150:632-644. [PMID: 39306435 DOI: 10.1016/j.jes.2024.02.027] [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: 11/09/2023] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 09/25/2024]
Abstract
Arsenic, a naturally occurring toxic element, manifests in various chemical forms and is widespread in the environment. Exposure to arsenic is a well-established risk factor for an elevated incidence of various cancers and chronic diseases. The crux of arsenic-mediated toxicity lies in its ability to induce oxidative stress, characterized by an unsettling imbalance between oxidants and antioxidants, accompanied by the rampant generation of reactive oxygen species and free radicals. In response to this oxidative turmoil, cells deploy their defense mechanisms, prominently featuring the redox-sensitive transcription factor known as nuclear factor erythroid 2-related factor 2 (NRF2). NRF2 stands as a primary guardian against the oxidative harm wrought by arsenic. When oxidative stress activates NRF2, it orchestrates a symphony of downstream antioxidant genes, leading to the activation of pivotal antioxidant enzymes like glutathione-S-transferase, heme oxygenase-1, and NAD(P)H: quinone oxidoreductase 1. This comprehensive review embarks on the intricate and diverse ways by which various arsenicals influence the NRF2 antioxidant pathway and its downstream targets, shedding light on their roles in defending against arsenic exposure toxic effects. It offers valuable insights into targeting NRF2 as a strategy for safeguarding against or treating the harmful and carcinogenic consequences of arsenic exposure.
Collapse
Affiliation(s)
- Sara R El-Mahrouk
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; Faculty of Pharmacy, Tanta University, Tanta, Gharbia, Egypt
| | - Mahmoud A El-Ghiaty
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2E1, Canada.
| |
Collapse
|
2
|
Tramer F, Sist P, Cardenas-Perez R, Urbani R, Bortolussi G, Passamonti S. Combined fluorometric analysis of biliverdin and bilirubin by the recombinant protein HUG. MethodsX 2024; 13:102979. [PMID: 39430781 PMCID: PMC11490793 DOI: 10.1016/j.mex.2024.102979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/24/2024] [Indexed: 10/22/2024] Open
Abstract
Biliverdin is a secondary metabolite of heme catabolism. It is formed by the reaction catalyzed by heme oxygenase, which converts the heme group contained in proteins such as hemoglobin, myoglobin, cytochromes, and catalase into biliverdin, iron (II) and CO in equimolar amounts, consuming NADPH. Biliverdin is then reduced to bilirubin by biliverdin reductase. Biliverdin and bilirubin form a redox couple and are important for the redox homeostasis of cells. Heme oxygenase-1 is an inducible enzyme that is induced by hypoxic conditions, increased availability of heme or proinflammatory mechanisms such as LPS, UV radiation, etc. In addition, both heme oxygenase-1 and biliverdin reductase play roles other than catalysis by modulating specific metabolic pathways at the transcriptional level. There is a need for affordable assays to analyze these bile pigments in biological and clinical samples. Here we present a method for the combined determination of biliverdin and bilirubin that utilizes the specific binding of bilirubin to the fluorescent recombinant fusion protein HUG and the enzymatic conversion of biliverdin to bilirubin.•This method enables the combined measurement of bilirubin and biliverdin in the nM range.•The method does not require solvent extraction or protein precipitation of the samples.
Collapse
Affiliation(s)
- Federica Tramer
- Department of Life Sciences, University of Trieste, via Giorgieri 1, I‐34127, Trieste, Italy
| | - Paola Sist
- Department of Life Sciences, University of Trieste, via Giorgieri 1, I‐34127, Trieste, Italy
| | - Rocio Cardenas-Perez
- Department of Life Sciences, University of Trieste, via Giorgieri 1, I‐34127, Trieste, Italy
- Group of Bioengineering in Regeneration and Cancer, Biogipuzkoa Health Research Institute, San Sebastián, Spain
| | - Ranieri Urbani
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, via Giorgieri 1, I‐34127, Trieste, Italy
| | - Giulia Bortolussi
- International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy
| | - Sabina Passamonti
- Department of Life Sciences, University of Trieste, via Giorgieri 1, I‐34127, Trieste, Italy
| |
Collapse
|
3
|
Rallabandi S, Amin SB. Bilirubinemia and retinopathy of prematurity in infants ≤ 29 weeks' gestational age. Pediatr Res 2024:10.1038/s41390-024-03479-8. [PMID: 39191947 DOI: 10.1038/s41390-024-03479-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/16/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Bilirubin is a known antioxidant. We hypothesized that the degree of bilirubinemia is inversely associated with oxidative stress-mediated retinopathy of prematurity (ROP). METHODS A prospective study was performed to determine if bilirubinemia is associated with ROP in infants ≤ 29 wks' gestational age and birth weights ≤ 1000 g. Total serum bilirubin (TSB) levels were measured at least twice daily during the first week and at least daily during the second week to determine peak and mean TSB levels. The outcomes were the incidence and severity of ROP. RESULTS Of 193 infants studied, 110 developed ROP: 53 had mild (stage 1 in zone 2 or 3); 31 had moderate (stage 2 in zone 2 or 3); and 26 had severe ROP ( ≥ stage 3 in any zone or any stage in zone 1). Mean TSB levels were associated with the severity (adjusted odds ratio [AOR] 0.68, 95% confidence interval [CI]:0.47-0.98, p = 0.04), but not with the incidence of ROP (AOR 0.81, 95% CI:0.52-1.27, p = 0.3). Peak TSB levels were also associated with the severity (AOR 0.71, 95% CI:0.52-0.96, p = 0.02), but not with the incidence of ROP (AOR 0.71, 95% CI:0.48-1.01, p = 0.07). CONCLUSIONS Bilirubinemia may be protective against severity of ROP. IMPACT Retinopathy of prematurity (ROP) is a disease mediated by oxidative stress. Increasing antioxidant status may decrease the incidence and severity of ROP. Bilirubin is a known antioxidant; however, findings from observational studies evaluating the role of bilirubinemia against the incidence and/or severity of ROP in premature infants are conflicting. Our findings from this prospective study suggests that bilirubinemia may be protective against the severity of ROP.
Collapse
Affiliation(s)
- Srujana Rallabandi
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Sanjiv B Amin
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Michigan, Central Michigan University, Detroit, MI, USA.
- Department of Pediatrics, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
4
|
Salloom RJ, Ahmad IM, Sahtout DZ, Baine MJ, Abdalla MY. Heme Oxygenase-1 and Prostate Cancer: Function, Regulation, and Implication in Cancer Therapy. Int J Mol Sci 2024; 25:9195. [PMID: 39273143 PMCID: PMC11394971 DOI: 10.3390/ijms25179195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Prostate cancer (PC) is a significant cause of mortality in men worldwide, hence the need for a comprehensive understanding of the molecular mechanisms underlying its progression and resistance to treatment. Heme oxygenase-1 (HO-1), an inducible enzyme involved in heme catabolism, has emerged as a critical player in cancer biology, including PC. This review explores the multifaceted role of HO-1 in PC, encompassing its function, regulation, and implications in cancer therapy. HO-1 influences cell proliferation, anti-apoptotic pathways, angiogenesis, and the tumor microenvironment, thereby influencing tumor growth and metastasis. HO-1 has also been associated with therapy resistance, affecting response to standard treatments. Moreover, HO-1 plays a significant role in immune modulation, affecting the tumor immune microenvironment and potentially influencing therapy outcomes. Understanding the intricate balance of HO-1 in PC is vital for developing effective therapeutic strategies. This review further explores the potential of targeting HO-1 as a therapeutic approach, highlighting challenges and opportunities. Additionally, clinical implications are discussed, focusing on the prognostic value of HO-1 expression and the development of novel combined therapies to augment PC sensitivity to standard treatment strategies. Ultimately, unraveling the complexities of HO-1 in PC biology will provide critical insights into personalized treatment approaches for PC patients.
Collapse
Affiliation(s)
- Ramia J. Salloom
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.J.S.); (D.Z.S.)
| | - Iman M. Ahmad
- Department of Clinical, Diagnostic, and Therapeutic Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Dania Z. Sahtout
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.J.S.); (D.Z.S.)
| | - Michael J. Baine
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Maher Y. Abdalla
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.J.S.); (D.Z.S.)
| |
Collapse
|
5
|
Kciuk M, Garg N, Dhankhar S, Saini M, Mujwar S, Devi S, Chauhan S, Singh TG, Singh R, Marciniak B, Gielecińska A, Kontek R. Exploring the Comprehensive Neuroprotective and Anticancer Potential of Afzelin. Pharmaceuticals (Basel) 2024; 17:701. [PMID: 38931368 PMCID: PMC11206995 DOI: 10.3390/ph17060701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, Huntington's disease, and others) and cancer, seemingly disparate in their etiology and manifestation, exhibit intriguing associations in certain cellular and molecular processes. Both cancer and neurodegenerative diseases involve the deregulation of cellular processes such as apoptosis, proliferation, and DNA repair and pose a significant global health challenge. Afzelin (kaempferol 3-O-rhamnoside) is a flavonoid compound abundant in various plant sources. Afzelin exhibits a diverse range of biological activities, offering promising prospects for the treatment of diseases hallmarked by oxidative stress and deregulation of cell death pathways. Its protective potential against oxidative stress is also promising for alleviating the side effects of chemotherapy. This review explores the potential therapeutic implications of afzelin, including its capacity to mitigate oxidative stress, modulate inflammation, and promote cellular regeneration in neurodegenerative and cancer diseases.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90-237 Lodz, Poland; (M.K.); (B.M.); (A.G.); (R.K.)
| | - Nitika Garg
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; (N.G.); (S.D.); (S.D.); (T.G.S.)
| | - Sanchit Dhankhar
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; (N.G.); (S.D.); (S.D.); (T.G.S.)
| | - Monika Saini
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be) University, Mullana, Ambala 133207, Haryana, India;
- Swami Vivekanand College of Pharmacy, Ramnagar, Banur 140601, Punjab, India
| | - Somdutt Mujwar
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; (N.G.); (S.D.); (S.D.); (T.G.S.)
| | - Sushma Devi
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; (N.G.); (S.D.); (S.D.); (T.G.S.)
| | - Samrat Chauhan
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; (N.G.); (S.D.); (S.D.); (T.G.S.)
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; (N.G.); (S.D.); (S.D.); (T.G.S.)
| | - Randhir Singh
- Department of Pharmacology, Central University of Punjab, Bathinda 151401, Punjab, India;
| | - Beata Marciniak
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90-237 Lodz, Poland; (M.K.); (B.M.); (A.G.); (R.K.)
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90-237 Lodz, Poland; (M.K.); (B.M.); (A.G.); (R.K.)
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90-237 Lodz, Poland; (M.K.); (B.M.); (A.G.); (R.K.)
| |
Collapse
|
6
|
Lee S, Lee SA, Shinn J, Lee Y. Hyaluronic Acid-Bilirubin Nanoparticles as a Tumor Microenvironment Reactive Oxygen Species-Responsive Nanomedicine for Targeted Cancer Therapy. Int J Nanomedicine 2024; 19:4893-4906. [PMID: 38828202 PMCID: PMC11141580 DOI: 10.2147/ijn.s460468] [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] [Received: 03/01/2024] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
Introduction The tumor microenvironment (TME) has attracted considerable attention as a potential therapeutic target for cancer. High levels of reactive oxygen species (ROS) in the TME may act as a stimulus for drug release. In this study, we have developed ROS-responsive hyaluronic acid-bilirubin nanoparticles (HABN) loaded with doxorubicin (DOX@HABN) for the specific delivery and release of DOX in tumor tissue. The hyaluronic acid shell of the nanoparticles acts as an active targeting ligand that can specifically bind to CD44-overexpressing tumors. The bilirubin core has intrinsic anti-cancer activity and ROS-responsive solubility change properties. Methods & Results DOX@HABN showed the HA shell-mediated targeting ability, ROS-responsive disruption leading to ROS-mediated drug release, and synergistic anti-cancer activity against ROS-overproducing CD44-overexpressing HeLa cells. Additionally, intravenously administered HABN-Cy5.5 showed remarkable tumor-targeting ability in HeLa tumor-bearing mice with limited distribution in major organs. Finally, intravenous injection of DOX@HABN into HeLa tumor-bearing mice showed synergistic anti-tumor efficacy without noticeable side effects. Conclusion These findings suggest that DOX@HABN has significant potential as a cancer-targeting and TME ROS-responsive nanomedicine for targeted cancer treatment.
Collapse
Affiliation(s)
- Seonju Lee
- College of Pharmacy, Ewha Womans University, Seoul, 03760, South Korea
| | - Seon Ah Lee
- College of Pharmacy, Ewha Womans University, Seoul, 03760, South Korea
| | - Jongyoon Shinn
- College of Pharmacy, Ewha Womans University, Seoul, 03760, South Korea
| | - Yonghyun Lee
- College of Pharmacy, Ewha Womans University, Seoul, 03760, South Korea
| |
Collapse
|
7
|
Herrald AL, Ambrogi EK, Mirica KA. Electrochemical Detection of Gasotransmitters: Status and Roadmap. ACS Sens 2024; 9:1682-1705. [PMID: 38593007 PMCID: PMC11196117 DOI: 10.1021/acssensors.3c02529] [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] [Indexed: 04/11/2024]
Abstract
Gasotransmitters, including nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), are a class of gaseous, endogenous signaling molecules that interact with one another in the regulation of critical cardiovascular, immune, and neurological processes. The development of analytical sensing mechanisms for gasotransmitters, especially multianalyte mechanisms, holds vast importance and constitutes a growing area of study. This review provides an overview of electrochemical sensing mechanisms with an emphasis on opportunities in multianalyte sensing. Electrochemical methods demonstrate good sensitivity, adequate selectivity, and the most well-developed potential for the multianalyte detection of gasotransmitters. Future research will likely address challenges with sensor stability and biocompatibility (i.e., sensor lifetime and cytotoxicity), sensor miniaturization, and multianalyte detection in biological settings.
Collapse
Affiliation(s)
- Audrey L Herrald
- Department of Chemistry, Burke Laboratory, Dartmouth College, 41 College Street, Hanover, New Hampshire 03755, United States
| | - Emma K Ambrogi
- Department of Chemistry, Burke Laboratory, Dartmouth College, 41 College Street, Hanover, New Hampshire 03755, United States
| | - Katherine A Mirica
- Department of Chemistry, Burke Laboratory, Dartmouth College, 41 College Street, Hanover, New Hampshire 03755, United States
| |
Collapse
|
8
|
Panda S, Phan H, Karlin KD. Heme-copper and Heme O 2-derived synthetic (bioinorganic) chemistry toward an understanding of cytochrome c oxidase dioxygen chemistry. J Inorg Biochem 2023; 249:112367. [PMID: 37742491 PMCID: PMC10615892 DOI: 10.1016/j.jinorgbio.2023.112367] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/22/2023] [Accepted: 09/07/2023] [Indexed: 09/26/2023]
Abstract
Cytochrome c oxidase (CcO), also widely known as mitochondrial electron-transport-chain complex IV, is a multi-subunit transmembrane protein responsible for catalyzing the last step of the electron transport chain, dioxygen reduction to water, which is essential to the establishment and maintenance of the membrane proton gradient that drives ATP synthesis. Although many intermediates in the CcO catalytic cycle have been spectroscopically and/or computationally authenticated, the specifics regarding the IP intermediate, hypothesized to be a heme-Cu (hydro)peroxo species whose O-O bond homolysis is supported by a hydrogen-bonding network of water molecules, are largely obscured by the fast kinetics of the A (FeIII-O2•-/CuI/Tyr) → PM (FeIV=O/CuII-OH/Tyr•) step. In this review, we have focused on the recent advancements in the design, development, and characterization of synthetic heme-peroxo‑copper model complexes, which can circumvent the abovementioned limitation, for the investigation of the formation of IP and its O-O cleavage chemistry. Novel findings regarding (a) proton and electron transfer (PT/ET) processes, together with their contributions to exogenous phenol induced O-O cleavage, (b) the stereo-electronic tunability of the secondary coordination sphere (especially hydrogen-bonding) on the geometric and spin state alteration of the heme-peroxo‑copper unit, and (c) a plausible mechanism for the Tyr-His cofactor biogenesis, are discussed in great detail. Additionally, since the ferric-superoxide and the ferryl-oxo (Compound II) species are critically involved in the CcO catalytic cycle, this review also highlights a few fundamental aspects of these heme-only (i.e., without copper) species, including the structural and reactivity influences of electron-donating trans-axial ligands and Lewis acid-promoted H-bonding.
Collapse
Affiliation(s)
- Sanjib Panda
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Hai Phan
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kenneth D Karlin
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
9
|
Martínez A, López-Rull I, Fargallo JA. To Prevent Oxidative Stress, What about Protoporphyrin IX, Biliverdin, and Bilirubin? Antioxidants (Basel) 2023; 12:1662. [PMID: 37759965 PMCID: PMC10525153 DOI: 10.3390/antiox12091662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
The pigments responsible for eggshell color and patterning in birds are protoporphyrin IX (PP) and biliverdin (BV). Both are involved in the catalytic degradation of the hemo group. Bilirubin (BR), another pigment, is produced when BV is broken down. PP, BV, and BR are free radical scavengers. In this study, we theoretically investigated the antioxidant capacities of these three biological meaningful molecules using Density Functional Theory calculations. First, two antioxidant mechanisms were analyzed for PP, BV, and BR: electron transfer and Hydrogen Atom Transfer. Second, since PP and BV interact with the calcium carbonate matrix of the eggshell, we analyzed the interaction of these pigments with Ca2+ and investigated their chelate compounds. Third, we explored the pro-oxidant properties of PP and BV, which have been proposed for PP when photoactivated to the triplet state, but not for BV. Our results show that PP, BV, and BR are just as good antiradical as other important natural pigments (carotenoids). Neither the antiradical properties of PP and BV nor the UV-visible spectra change due to the presence of calcium, suggesting that the signaling function of these pigments is not affected by the link with Ca2+. Finally, we found that both PP and BV (alone and when linked to Ca2+) can transfer energy from its triplet state to molecular-oxygen-producing singlet oxygen, indicating their pro-oxidant capacity. This investigation answers important questions about the function of these pigments, which may help to understand their influence on the reproductive success of birds.
Collapse
Affiliation(s)
- Ana Martínez
- Departamento de Materiales de Baja Dimensionalidad, Instituto de Investigaciones en Materiales, Universidad Nacional Autónoma de México, Circuito Exterior S. N., Ciudad Universitaria, Ciudad de México 04510, Mexico
| | - Isabel López-Rull
- Departamento Biología y Geología, Física y Química Inorgánica, Área de Biodiversidad y Conservación, Universidad Rey Juan Carlos, C/Tulipán s/n., 28933 Madrid, Spain;
| | - Juan A. Fargallo
- Departamento de Ecología Evolutiva, Museo Nacional de Ciencias Naturales-CSIC, C/José Gutiérrez Abascal 2, 28006 Madrid, Spain;
| |
Collapse
|
10
|
Sami DH, Soliman AS, Khowailed AA, Alruhaimi RS, Hassanein EHM, Kamel EM, Mahmoud AM. The protective effect of 7-hydroxycoumarin against cisplatin-induced liver injury is mediated via attenuation of oxidative stress and inflammation and upregulation of Nrf2/HO-1 pathway. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:80181-80191. [PMID: 37291353 DOI: 10.1007/s11356-023-27879-1] [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: 05/03/2023] [Accepted: 05/19/2023] [Indexed: 06/10/2023]
Abstract
Cisplatin (CIS) is an effective chemotherapy against different solid cancers. However, the adverse effects, including hepatotoxicity, limit its clinical use. 7-hydroxycoumarin (7-HC) possesses antioxidant and hepatoprotective activities, but its protective effect against CIS hepatotoxicity has not been investigated. This study evaluated the effect of 7-HC on liver injury, oxidative stress (OS), and inflammation provoked by CIS. Rats received 7-HC (25, 50, and 100 mg/kg) orally for 2 weeks followed by intraperitoneal injection of CIS (7 mg/kg) at day 15. CIS increased serum transaminases, alkaline phosphatase (ALP), and bilirubin and provoked tissue injury accompanied by elevated reactive oxygen species (ROS), malondialdehyde (MDA), and nitric oxide (NO). Liver nuclear factor (NF)-κB p65, inducible NO synthase (iNOS), pro-inflammatory cytokines, Bax, and caspase-3 were upregulated, and antioxidant defenses and Bcl-2 were decreased in CIS-treated rats, while 7-HC prevented liver injury and ameliorated OS, inflammatory and apoptosis markers. In addition, 7-HC enhanced nuclear factor erythroid 2-related factor 2 (Nrf2), and heme oxygenase (HO)-1 in CIS-administered rats and in silico studies revealed its binding affinity toward HO-1. In conclusion, 7-HC protected against CIS hepatotoxicity by mitigating OS and inflammatory response and modulating Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Demiana H Sami
- Physiology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Ayman S Soliman
- Physiology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Akef A Khowailed
- Physiology Department, Faculty of Medicine, Cairo University, Giza, 12613, Egypt
| | - Reem S Alruhaimi
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, 11671, Saudi Arabia
| | - Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University-Assiut Branch, Assiut, 71524, Egypt
| | - Emadeldin M Kamel
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Ayman M Mahmoud
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt.
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, M1 5GD, UK.
| |
Collapse
|
11
|
Minic S, Annighöfer B, Milcic M, Maignen F, Brûlet A, Combet S. The effects of biliverdin on pressure-induced unfolding of apomyoglobin: The specific role of Zn 2+ ions. Int J Biol Macromol 2023:125549. [PMID: 37356686 DOI: 10.1016/j.ijbiomac.2023.125549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/05/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
Apomyoglobin (apoMb), a model protein in biochemistry, exhibits a strong propensity to bind various ligands, which makes it a good candidate as a carrier of bioactive hydrophobic drugs. The stability of its hydrophobic pocket determines its potential as a carrier of bioactive compounds. High pressure (HP) is a potent tool for studying protein stability, revealing the specific role of hydrophobic cavities in unfolding. We probed the effects of biliverdin (BV) binding and its complex with Zn2+ ions on the structure and HP stability of apoMb. CD spectroscopy and SAXS measurements revealed that BV and BV-Zn2+ complexes make the apoMb structure more compact with higher α-helical content. We performed in-situ HP measurements of apoMb intrinsic fluorescence to demonstrate the ability of BV to stabilise apoMb structure at HP conditions. Furthermore, the presence of Zn2+ within the apoMb-BV complex significantly enhances the BV stabilisation effect. In-situ visible absorption study of BV chromophore confirmed the ability of Zn2+ to increase the stability of apoMb-BV complex under HP: the onset of complex dissociation is shifted by ~100 MPa in the presence of Zn2+. By combining HP-fluorescence and HP-visible absorption spectroscopy, our strategy highlights the crucial role of tetrapyrrole-metal complexes in stabilising apoMb hydrophobic pocket.
Collapse
Affiliation(s)
- Simeon Minic
- Laboratoire Léon-Brillouin (LLB), UMR12 CEA CNRS, Université Paris-Saclay, F-91191 Gif-sur-Yvette CEDEX, France; Center of Excellence for Molecular Food Sciences, Department of Biochemistry, University of Belgrade - Faculty of Chemistry, Belgrade, Serbia.
| | - Burkhard Annighöfer
- Laboratoire Léon-Brillouin (LLB), UMR12 CEA CNRS, Université Paris-Saclay, F-91191 Gif-sur-Yvette CEDEX, France
| | - Milos Milcic
- Department of General and Inorganic Chemistry, Faculty of Chemistry, University of Belgrade, Belgrade, Serbia
| | - François Maignen
- Laboratoire Léon-Brillouin (LLB), UMR12 CEA CNRS, Université Paris-Saclay, F-91191 Gif-sur-Yvette CEDEX, France
| | - Annie Brûlet
- Laboratoire Léon-Brillouin (LLB), UMR12 CEA CNRS, Université Paris-Saclay, F-91191 Gif-sur-Yvette CEDEX, France
| | - Sophie Combet
- Laboratoire Léon-Brillouin (LLB), UMR12 CEA CNRS, Université Paris-Saclay, F-91191 Gif-sur-Yvette CEDEX, France.
| |
Collapse
|
12
|
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
|
13
|
Cao S, Yang Y, He L, Hang Y, Yan X, Shi H, Wu J, Ouyang Z. Cryo-EM structures of mitochondrial ABC transporter ABCB10 in apo and biliverdin-bound form. Nat Commun 2023; 14:2030. [PMID: 37041204 PMCID: PMC10090120 DOI: 10.1038/s41467-023-37851-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 04/03/2023] [Indexed: 04/13/2023] Open
Abstract
ABCB10, a member of ABC transporter superfamily that locates in the inner membrane of mitochondria, plays crucial roles in hemoglobin synthesis, antioxidative stress and stabilization of the iron transporter mitoferrin-1. Recently, it was found that ABCB10 is a mitochondrial biliverdin exporter. However, the molecular mechanism of biliverdin export by ABCB10 remains elusive. Here we report the cryo-EM structures of ABCB10 in apo (ABCB10-apo) and biliverdin-bound form (ABCB10-BV) at 3.67 Å and 2.85 Å resolution, respectively. ABCB10-apo adopts a wide-open conformation and may thus represent the apo form structure. ABCB10-BV forms a closed conformation and biliverdin situates in a hydrophobic pocket in one protomer and bridges the interaction through hydrogen bonds with the opposing one. We also identify cholesterols sandwiched by BVs and discuss the export dynamics based on these structural and biochemical observations.
Collapse
Affiliation(s)
- Sheng Cao
- Wuxi Biortus Biosciences Co. Ltd., 6 Dongsheng Western Road, 214437, Jiangyin, Jiangsu, China
| | - Yihu Yang
- Wuxi Biortus Biosciences Co. Ltd., 6 Dongsheng Western Road, 214437, Jiangyin, Jiangsu, China
| | - Lili He
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, 430030, Wuhan, Hubei Province, China
| | - Yumo Hang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, 430030, Wuhan, Hubei Province, China
| | - Xiaodong Yan
- Wuxi Biortus Biosciences Co. Ltd., 6 Dongsheng Western Road, 214437, Jiangyin, Jiangsu, China
| | - Hui Shi
- Wuxi Biortus Biosciences Co. Ltd., 6 Dongsheng Western Road, 214437, Jiangyin, Jiangsu, China
| | - Jiaquan Wu
- Wuxi Biortus Biosciences Co. Ltd., 6 Dongsheng Western Road, 214437, Jiangyin, Jiangsu, China
| | - Zhuqing Ouyang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, 430030, Wuhan, Hubei Province, China.
| |
Collapse
|
14
|
Perez-Ortiz G, Sidda JD, Peate J, Ciccarelli D, Ding Y, Barry SM. Production of copropophyrin III, biliverdin and bilirubin by the rufomycin producer, Streptomyces atratus. Front Microbiol 2023; 14:1092166. [PMID: 37007481 PMCID: PMC10060970 DOI: 10.3389/fmicb.2023.1092166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/31/2023] [Indexed: 03/18/2023] Open
Abstract
Heme is best known for its role as a versatile prosthetic group in prokaryotic and eukaryotic proteins with diverse biological functions including gas and electron transport, as well as a wide array of redox chemistry. However, free heme and related tetrapyrroles also have important roles in the cell. In several bacterial strains, heme biosynthetic precursors and degradation products have been proposed to function as signaling molecules, ion chelators, antioxidants and photoprotectants. While the uptake and degradation of heme by bacterial pathogens is well studied, less is understood about the physiological role of these processes and their products in non-pathogenic bacteria. Streptomyces are slow growing soil bacteria known for their extraordinary capacity to produce complex secondary metabolites, particularly many clinically used antibiotics. Here we report the unambiguous identification of three tetrapyrrole metabolites from heme metabolism, coproporphyrin III, biliverdin and bilirubin, in culture extracts of the rufomycin antibiotic producing Streptomyces atratus DSM41673. We propose that biliverdin and bilirubin may combat oxidative stress induced by nitric oxide production during rufomycin biosynthesis, and indicate the genes involved in their production. This is, to our knowledge, the first report of the production of all three of these tetrapyrroles by a Streptomycete.
Collapse
Affiliation(s)
| | | | | | | | | | - Sarah M. Barry
- Department of Chemistry, Faculty of Natural & Mathematical Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
15
|
Chen MS, Wang JX, Zhang H, Cui JG, Zhao Y, Li JL. Novel Role of Hemeoxygenase-1 in Phthalate-Induced Renal Proximal Tubule Cell Ferroptosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:2579-2589. [PMID: 36696656 DOI: 10.1021/acs.jafc.2c07762] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Phthalates are widely used to improve the flexibility of poly(vinyl chloride) (PVC) polymer agriculture products. Di(2-ethylhexyl) phthalate (DEHP) is a type of addition to plastic and can lead to many health problems. Hemeoxygenase-1 (HO-1) is an extremely important molecule that releases enzymatic products to promote ferroptosis. This research aimed to explore the function of HO-1 in DEHP-induced renal proximal tubule cell ferroptosis. In the experiment, ICR male mice are exposed to (0, 50, 200, and 500 mg/kg BW/day) DEHP for 28 days. Here, we observed that DEHP induced glomeruli atrophy and the tubules swell. Furthermore, DEHP exposure could increase ferrous iron content and decrease antioxidant activity. We also found that DEHP exposure increased the expression of nuclear factor-erythroid 2 p45-related factor 2 (NFE2L2) in the nucleus. In particular, the expression of (HO-1) is significantly increased both in protein and mRNA levels. Glutathione peroxidase 4 (GPX4) as an endogenous control of ferroptosis was downregulated, which proved the occurrence of ferroptosis. In the study, exposure to DEHP activated the NFE2L2/HO-1 signaling pathway and resulted in ferroptosis of the proximal tubule. This research connects ferroptosis with HO-1, providing new insights into the potential roles of phthalates in nephrotoxicity.
Collapse
|
16
|
Brasil FB, de Almeida FJS, Luckachaki MD, Dall'Oglio EL, de Oliveira MR. The isothiocyanate sulforaphane prevents mitochondrial impairment and neuroinflammation in the human dopaminergic SH-SY5Y and in the mouse microglial BV2 cells: role for heme oxygenase-1. Metab Brain Dis 2023; 38:419-435. [PMID: 35469083 DOI: 10.1007/s11011-022-00990-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/18/2022] [Indexed: 01/25/2023]
Abstract
Sulforaphane (SFN) promotes protective effects in different cell types. Nonetheless, it remains to be clarified by which mechanism SFN exerts benefits in mammalian cells. Mitochondria are a major source of adenosine triphosphate (ATP) and reactive species in nucleated cells. Mitochondrial impairment result in cellular redox biology disruption, bioenergetic status collapse, and inflammation. Evidence suggest that mitochondrial dysfunction plays a role in neurological disorders. Since a cure was not discovered yet to some of these diseases, investigating strategies to promote mitochondrial protection is pharmacologically relevant and may improve life quality of patients suffering from these maladies. Natural molecules, such as SFN, are potent inducers of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) and, consequently, stimulate the expression of genes whose products, such as heme oxygenase-1 (HO-1), induce cytoprotective actions in mammalian tissues. In this work, we investigated whether SFN (5 µM) would be capable to prevent the dysfunctions caused by chlorpyrifos (CPF) on the human dopaminergic SH-SY5Y cells. Moreover, we examined the effects of a pretreatment with SFN at the same concentration on the mouse microglial BV2 cells stimulated by lipopolysaccharide (LPS) in an experimental model of neuroinflammation. SFN prevented the mitochondrial impairment and the neuroinflammation caused by the chemical stressors in both cell types. Inhibition of heme oxygenase-1 (HO-1) suppressed the mitochondrial protection and anti-inflammatory action afforded by SFN in this experimental model. Overall, SFN promoted cytoprotection by a mechanism dependent on the HO-1 enzyme in the SH-SY5Y and BV2 cells.
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
| | - 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, Cuiaba, MT, CEP 78060-900, 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, Cuiaba, MT, CEP 78060-900, 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, Cuiaba, MT, CEP 78060-900, Brazil
| | - Marcos Roberto de Oliveira
- 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, Cuiaba, MT, CEP 78060-900, Brazil.
| |
Collapse
|
17
|
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: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [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
|
18
|
Jung W, Lee DY, Moon E, Jon S. Nanoparticles derived from naturally occurring metal chelators for theranostic applications. Adv Drug Deliv Rev 2022; 191:114620. [PMID: 36379406 DOI: 10.1016/j.addr.2022.114620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 11/15/2022]
Abstract
Metals are indispensable for the activities of all living things, from single-celled organisms to higher organisms, including humans. Beyond their intrinsic quality as metal ions, metals help creatures to maintain requisite biological processes by forming coordination complexes with endogenous ligands that are broadly distributed in nature. These types of naturally occurring chelating reactions are found through the kingdoms of life, including bacteria, plants and animals. Mimicking these naturally occurring coordination complexes with intrinsic biocompatibility may offer an opportunity to develop nanomedicine toward clinical applications. Herein, we introduce representative examples of naturally occurring coordination complexes in a selection of model organisms and highlight such bio-inspired metal-chelating nanomaterials for theranostic applications.
Collapse
Affiliation(s)
- Wonsik Jung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea
| | - Dong Yun Lee
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Seoul 05505, Republic of Korea; Translational Biomedical Research Group, Biomedical Research Center, Asan Institute for Life Science, Asan Medical Center, 88 Olympic-ro 43-gil, Seoul 05505, Republic of Korea.
| | - Eugene Moon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea
| | - Sangyong Jon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea.
| |
Collapse
|
19
|
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
|
20
|
DPP9 as a Potential Novel Mediator in Gastrointestinal Virus Infection. Antioxidants (Basel) 2022; 11:antiox11112177. [DOI: 10.3390/antiox11112177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Dipeptidyl peptidase 9 (DPP9) is a member of the dipeptidyl peptidase IV family. Inhibition of DPP9 has recently been shown to activate the nucleotide-binding domain leucine-rich repeat 1 (NLRP1) inflammasome. NLRP1 is known to bind nucleic acids with high affinity and directly interact with double stranded RNA, which plays a key role in viral replication. DPP9 has also recently emerged as a key gene related to lung-inflammation in critical SARS-CoV-2 infection. Importantly, DPP9 activity is strongly dependent on the oxidative status. Here, we explored the potential role of DPP9 in the gastrointestinal tract. We performed transcriptomics analyses of colon (microarray, n = 37) and jejunal (RNA sequencing, n = 31) biopsies from two independent cohorts as well as plasma metabolomics analyses in two independent cohorts (n = 37 and n = 795). The expression of DPP9 in the jejunum, colon, and blood was significantly associated with circulating biomarkers of oxidative stress (uric acid, bilirubin). It was also associated positively with the expression of transcription factors (NRF-2) and genes (SOD, CAT, GPX) encoding for antioxidant enzymes, but negatively with that of genes (XDH, NOX) and transcription factors (NF-KB) involved in ROS-generating enzymes. Gene co-expression patterns associated with DPP9 identified several genes participating in antiviral pathways in both tissues. Notably, DPP9 expression in the colon and plasma was strongly positively associated with several circulating nucleotide catabolites (hypoxanthine, uric acid, 3-ureidopropionic acid) with important roles in the generation of ROS and viral infection, as well as other metabolites related to oxidative stress (Resolvin D1, glutamate-containing dipeptides). Gene-drug enrichment analyses identified artenimol, puromycin, anisomycin, 3-phenyllactic acid, and linezolid as the most promising drugs targeting these DPP9-associated genes. We have identified a novel potential pathogenic mechanism of viral infection in the digestive tract and promising existing drugs that can be repositioned against viral infection.
Collapse
|
21
|
Hormesis and Oxidative Distress: Pathophysiology of Reactive Oxygen Species and the Open Question of Antioxidant Modulation and Supplementation. Antioxidants (Basel) 2022; 11:antiox11081613. [PMID: 36009331 PMCID: PMC9405171 DOI: 10.3390/antiox11081613] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022] Open
Abstract
Alterations of redox homeostasis leads to a condition of resilience known as hormesis that is due to the activation of redox-sensitive pathways stimulating cell proliferation, growth, differentiation, and angiogenesis. Instead, supraphysiological production of reactive oxygen species (ROS) exceeds antioxidant defence and leads to oxidative distress. This condition induces damage to biomolecules and is responsible or co-responsible for the onset of several chronic pathologies. Thus, a dietary antioxidant supplementation has been proposed in order to prevent aging, cardiovascular and degenerative diseases as well as carcinogenesis. However, this approach has failed to demonstrate efficacy, often leading to harmful side effects, in particular in patients affected by cancer. In this latter case, an approach based on endogenous antioxidant depletion, leading to ROS overproduction, has shown an interesting potential for enhancing susceptibility of patients to anticancer therapies. Therefore, a deep investigation of molecular pathways involved in redox balance is crucial in order to identify new molecular targets useful for the development of more effective therapeutic approaches. The review herein provides an overview of the pathophysiological role of ROS and focuses the attention on positive and negative aspects of antioxidant modulation with the intent to find new insights for a successful clinical application.
Collapse
|
22
|
Role of transcranial Doppler in assessment of cerebral blood flow in full term neonates with extreme unconjugated hyperbilirubinemia. J Ultrasound 2022; 26:175-184. [PMID: 35969370 PMCID: PMC10063702 DOI: 10.1007/s40477-022-00704-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/24/2022] [Indexed: 10/15/2022] Open
Abstract
PURPOSE To evaluate the difference in cerebral blood flow in neonates with and without extreme unconjugated hyperbilirubinemia. METHODS Transcranial Doppler parameters of 26 full term newborns with extreme unconjugated hyperbilirubinemia (UCH) were compared to 13 postnatal age and sex matched normal healthy neonates serving as controls. Resistance index (RI), pulsatility index (PI) and peak systolic velocity (PSV) were measured in the middle cerebral, internal carotid and posterior cerebral arteries on both sides by transcranial color Doppler ultrasound. RESULTS An increase in cerebral blood flow (decreased RI, PI and increased PSV) was observed in the extreme unconjugated hyperbilirubinemia (UCH) group. There was positive correlation between total serum bilirubin level and peak systolic velocity and vice versa with resistivity and pulsatility indices. Eight neonates developed clinical features of acute bilirubin encephalopathy and showed significantly increased peak systolic velocity in the right middle cerebral artery compared to those with normal outcome. Resistivity index and pulsatility index were lower in patients managed by exchange transfusion compared to those managed with phototherapy. CONCLUSION An increase in cerebral blood flow was observed in neonates with UCH compared to those without hyperbilirubinemia. By assessing the cerebral blood flow velocity, resistivity index (RI), and pulsatility index (PI) of particular intracranial arteries, the transcranial Doppler can identify the at-risk neonates, for development of neurological affliction in extreme unconjugated hyperbilirubinemia.
Collapse
|
23
|
Highly sensitive and selective detection of free bilirubin using blue emitting graphene quantum dots (GQDs). J CHEM SCI 2022. [DOI: 10.1007/s12039-022-02079-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
24
|
Abstract
Sunlight drives phototrophic metabolism, which affects redox conditions and produces substrates for nonphototrophs. These environmental parameters fluctuate daily due to Earth’s rotation, and nonphototrophic organisms can therefore benefit from the ability to respond to, or even anticipate, such changes. Circadian rhythms, such as daily changes in body temperature, in host organisms can also affect local conditions for colonizing bacteria. Here, we investigated the effects of light/dark and temperature cycling on biofilms of the opportunistic pathogen Pseudomonas aeruginosa PA14. We grew biofilms in the presence of a respiratory indicator dye and found that enhanced dye reduction occurred in biofilm zones that formed during dark intervals and at lower temperatures. This pattern formation occurred with cycling of blue, red, or far-red light, and a screen of mutants representing potential sensory proteins identified two with defects in pattern formation, specifically under red light cycling. We also found that the physiological states of biofilm subzones formed under specific light and temperature conditions were retained during subsequent condition cycling. Light/dark and temperature cycling affected expression of genes involved in primary metabolic pathways and redox homeostasis, including those encoding electron transport chain components. Consistent with this, we found that cbb3-type oxidases contribute to dye reduction under light/dark cycling conditions. Together, our results indicate that cyclic changes in light exposure and temperature have lasting effects on redox metabolism in biofilms formed by a nonphototrophic, pathogenic bacterium.
Collapse
|
25
|
El-Ghiaty MA, Alqahtani MA, El-Kadi AOS. Down-regulation of hepatic cytochromes P450 1A1 and 1A2 by arsenic trioxide (ATO) in vivo and in vitro: A role of heme oxygenase 1. Chem Biol Interact 2022; 364:110049. [PMID: 35872050 DOI: 10.1016/j.cbi.2022.110049] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 07/03/2022] [Accepted: 07/13/2022] [Indexed: 11/03/2022]
Abstract
Arsenic trioxide (ATO) has evolved from an environmental threat to a successful therapy for acute promyelocytic leukemia (APL) and probably for solid tumors in the future. However, its efficacy comes at a cost of multi-organ toxicity whose mechanism remains unresolved. Arsenicals have been reported to modulate cytochrome P450 1A (CYP1A) enzymes, thus modifying activation/detoxification of drugs/procarcinogens. Therefore, this study aimed to investigate the possible effects of ATO on CYP1A1 and CYP1A2, in absence and presence of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) using in vivo and in vitro models. For this purpose, C57BL/6 mice were intraperitoneally injected with 8 mg/kg ATO with or without 15 μg/kg TCDD for 6 and 24 h. Furthermore, HepG2 cells were treated with ATO (1, 5, and 10 μM) with or without 1 nM TCDD for 6 and 24 h. ATO significantly inhibited TCDD-mediated induction of CYP1A1/1A2 mRNA, protein, and activity in both models. ATO differentially modulated CYP1A1/1A2 basal levels in vivo. We also demonstrated that ATO downregulates CYP1A through inhibiting the transcriptional activation of its regulatory element at both basal and inducible levels. Additionally, ATO significantly induced mRNA and protein of heme oxygenase 1 (HMOX1) in vivo and in vitro. In HepG2 cells, inhibition of HMOX1 by tin (IV) mesoporphyrin (IX) (SnMP) resulted in a partial restoration of the TCDD-mediated induction of CYP1A1 activity that was inhibited by ATO co-exposure. Our findings show that ATO alters both constitutive and inducible CYP1A1/1A2 expressions through transcriptional and HMOX1-mediated post-translational mechanisms. This implies the possible involvement of ATO in clearance-related consequences for the substrates of these enzymes such as drug-drug interactions or suboptimal toxicant elimination.
Collapse
Affiliation(s)
- Mahmoud A El-Ghiaty
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Mohammed A Alqahtani
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
26
|
Lin YY, Takemoto JY, Chang CWT, Peng CA. Mesobiliverdin IXα ameliorates osteoporosis via promoting osteogenic differentiation of mesenchymal stem cells. Biochem Biophys Res Commun 2022; 619:56-61. [PMID: 35738065 DOI: 10.1016/j.bbrc.2022.06.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022]
Abstract
Heme oxygenase-1 (HO-1) expression promotes osteogenesis, but the mechanisms remain unclear and therapeutic strategies using it to target bone disorders such as osteoporosis have not progressed. Mesobiliverdin IXα is a naturally occurring bilin analog of HO-1 catalytic product biliverdin IXα. Inclusion of mesobiliverdin IXα in the feed diet of ovariectomized osteoporotic mice was observed to increase femur bone volume, trabecular thickness and osteogenesis serum markers osteoprotegrin and osteocalcin and to decrease bone resorption serum markers cross-linked N-teleopeptide and tartrate-resistant acid phosphatase 5b. Moreover, in vitro exposure of human bone marrow mesenchymal stem cells to mesobiliverdin IXα enhanced osteogenic differentiation efficiency by two-fold over non-exposed controls. Our results imply that mesobiliverdin IXα promotes osteogenesis in ways that reflect the potential therapeutic effects of induced HO-1 expression in alleviating osteoporosis.
Collapse
Affiliation(s)
- Yuan-Yu Lin
- Department of Animal Science and Technology, National Taiwan University, Taipei City, Taiwan
| | - Jon Y Takemoto
- Department of Biology, Utah State University, Logan, UT, United States
| | - Cheng-Wei T Chang
- Department of Chemistry and Biochemistry, Utah State University, Logan, UT, United States
| | - Ching-An Peng
- Department of Chemical and Biological Engineering, University of Idaho, Moscow, ID, United States.
| |
Collapse
|
27
|
Brasil FB, de Almeida FJS, Luckachaki MD, Dall'Oglio EL, de Oliveira MR. A Pretreatment with Isoorientin Attenuates Redox Disruption, Mitochondrial Impairment, and Inflammation Caused by Chlorpyrifos in a Dopaminergic Cell Line: Involvement of the Nrf2/HO-1 Axis. Neurotox Res 2022; 40:1043-1056. [PMID: 35583593 DOI: 10.1007/s12640-022-00517-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
Abstract
The C-glucosyl flavone isoorientin (ISO) is obtained by humans from the diet and exhibits several cytoprotective effects, as demonstrated in different experimental models. However, it was not previously shown whether ISO would be able to prevent mitochondrial impairment in cells exposed to a chemical stressor. Thus, we treated the human neuroblastoma SH-SY5Y cells with ISO (0.5-20 µM) for 18 h before a challenge with chlorpyrifos (CPF) at 100 µM for additional 24 h. We observed that ISO prevented the CPF-induced lipid peroxidation and protein carbonylation and nitration in the membranes of mitochondria extracted from CPF-treated cells. ISO also attenuated the CPF-elicited increase in the production of reactive species in this experimental model. Moreover, ISO prevented the CPF-induced disruption in the activity of components of the oxidative phosphorylation (OXPHOS) system in the SH-SY5Y cells. ISO also promoted an anti-inflammatory action in the cells exposed to CPF. CPF caused a decrease in the activity of the enzyme heme oxygenase-1 (HO-1), a cytoprotective agent. On the other hand, ISO upregulated HO-1 activity in SH-SY5Y cells. Inhibition of HO-1 by zinc protoporphyrin-IX (ZnPP-IX) suppressed the cytoprotection induced by ISO in the CPF-treated cells. Besides, silencing of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) abolished the ISO-induced HO-1 upregulation and mitochondrial benefits induced by this flavone on the CPF-challenged cells. Thus, ISO protected mitochondria of the CPF-treated cells by an Nrf2/HO-1-dependent fashion in the SH-SY5Y cells.
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
| | - Fhelipe Jolner Souza de Almeida
- Programa de Pós-Graduação Em Ciências da Saúde (PPGCS), Universidade Federal de Mato Grosso (UFMT), Cuiaba, Mato Grosso, 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, Cuiaba, Mato Grosso, CEP 78060-900, 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, Cuiaba, Mato Grosso, CEP 78060-900, 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, Cuiaba, Mato Grosso, CEP 78060-900, Brazil
| | - Marcos Roberto de Oliveira
- 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, Cuiaba, Mato Grosso, CEP 78060-900, Brazil.
| |
Collapse
|
28
|
de Oliveira J, Denadai MB, Costa DL. Crosstalk between Heme Oxygenase-1 and Iron Metabolism in Macrophages: Implications for the Modulation of Inflammation and Immunity. Antioxidants (Basel) 2022; 11:861. [PMID: 35624725 PMCID: PMC9137896 DOI: 10.3390/antiox11050861] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 12/16/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is an enzyme that catalyzes the degradation of heme, releasing equimolar amounts of carbon monoxide (CO), biliverdin (BV), and iron. The anti-inflammatory and antioxidant properties of HO-1 activity are conferred in part by the release of CO and BV and are extensively characterized. However, iron constitutes an important product of HO-1 activity involved in the regulation of several cellular biological processes. The macrophage-mediated recycling of heme molecules, in particular those contained in hemoglobin, constitutes the major mechanism through which living organisms acquire iron. This process is finely regulated by the activities of HO-1 and of the iron exporter protein ferroportin. The expression of both proteins can be induced or suppressed in response to pro- and anti-inflammatory stimuli in macrophages from different tissues, which alters the intracellular iron concentrations of these cells. As we discuss in this review article, changes in intracellular iron levels play important roles in the regulation of cellular oxidation reactions as well as in the transcriptional and translational regulation of the expression of proteins related to inflammation and immune responses, and therefore, iron metabolism represents a potential target for the development of novel therapeutic strategies focused on the modulation of immunity and inflammation.
Collapse
Affiliation(s)
- Joseana de Oliveira
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirao Preto 14049-900, Brazil; (J.d.O.); (M.B.D.)
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirao Preto 14049-900, Brazil
| | - Marina B. Denadai
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirao Preto 14049-900, Brazil; (J.d.O.); (M.B.D.)
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirao Preto 14049-900, Brazil
| | - Diego L. Costa
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirao Preto 14049-900, Brazil; (J.d.O.); (M.B.D.)
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirao Preto 14049-900, Brazil
| |
Collapse
|
29
|
Keum H, Yoo D, Jon S. Photomedicine based on heme-derived compounds. Adv Drug Deliv Rev 2022; 182:114134. [PMID: 35122881 DOI: 10.1016/j.addr.2022.114134] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/16/2021] [Accepted: 01/30/2022] [Indexed: 02/08/2023]
Abstract
Photoimaging and phototherapy have become major platforms for the diagnosis and treatment of various health complications. These applications require a photosensitizer (PS) that is capable of absorbing light from a source and converting it into other energy forms for detection and therapy. While synthetic inorganic materials such as quantum dots and gold nanorods have been widely explored for their medical diagnosis and photodynamic (PDT) and photothermal (PTT) therapy capabilities, translation of these technologies has lagged, primarily owing to potential cytotoxicity and immunogenicity issues. Of the various photoreactive molecules, the naturally occurring endogenous compound heme, a constituent of red blood cells, and its derivatives, porphyrin, biliverdin and bilirubin, have shown immense potential as noteworthy candidates for clinically translatable photoreactive agents, as evidenced by previous reports. While porphyrin-based photomedicines have attracted significant attention and are well documented, research on photomedicines based on two other heme-derived compounds, biliverdin and bilirubin, has been relatively lacking. In this review, we summarize the unique photoproperties of heme-derived compounds and outline recent efforts to use them in biomedical imaging and phototherapy applications.
Collapse
|
30
|
Yang S, Ouyang J, Lu Y, Harypursat V, Chen Y. A Dual Role of Heme Oxygenase-1 in Tuberculosis. Front Immunol 2022; 13:842858. [PMID: 35281042 PMCID: PMC8913507 DOI: 10.3389/fimmu.2022.842858] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/08/2022] [Indexed: 12/19/2022] Open
Abstract
Iron metabolism is vital for the survival of both humans and microorganisms. Heme oxygenase-1 (HO-1) is an essential stress-response enzyme highly expressed in the lungs, and catabolizes heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV)/bilirubin (BR), especially in pathological conditions which cause oxidative stress and inflammation. Ferrous iron (Fe2+) is an important raw material for the synthesis of hemoglobin in red blood cells, and patients with iron deficiency are often associated with decreased cellular immunity. CO and BR can inhibit oxidative stress and inflammation. Thus, HO-1 is regarded as a cytoprotective molecule during the infection process. However, recent study has unveiled new information regarding HO-1. Being a highly infectious pathogenic bacterium, Mycobacterium tuberculosis (MTB) infection causes acute oxidative stress, and increases the expression of HO-1, which may in turn facilitate MTB survival and growth due to increased iron availability. Moreover, in severe cases of MTB infection, excessive reactive oxygen species (ROS) and free iron (Fe2+) due to high levels of HO-1 can lead to lipid peroxidation and ferroptosis, which may promote further MTB dissemination from cells undergoing ferroptosis. Therefore, it is important to understand and illustrate the dual role of HO-1 in tuberculosis. Herein, we critically review the interplay among HO-1, tuberculosis, and the host, thus paving the way for development of potential strategies for modulating HO-1 and iron metabolism.
Collapse
|
31
|
Guo Y, Zhao H, Lin Z, Ye T, Xu D, Zeng Q. Heme in Cardiovascular Diseases: A Ubiquitous Dangerous Molecule Worthy of Vigilance. Front Cell Dev Biol 2022; 9:781839. [PMID: 35127704 PMCID: PMC8807526 DOI: 10.3389/fcell.2021.781839] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/13/2021] [Indexed: 12/20/2022] Open
Abstract
Heme, the protoporphyrin IX iron complex is widely present in the human body and it is involved in oxygen storage, electron transfer, and enzymatic reactions. However, free heme can be toxic as it catalyzes the production of reactive oxygen species, oxidizes lipids and proteins, and causes DNA damage, thereby inducing a pro-inflammatory environment. The generation, metabolism, and degradation of heme in the human body are regulated by precise mechanisms to ensure that heme remains non-toxic. However, in several types of cardiovascular diseases, impaired metabolism and exposure to heme may occur in pathological processes, including neovascularization, internal hemorrhage, ischemia, and reperfusion. Based on years of research, in this review, we aimed to summarize the underlying mechanisms by which heme contributes to the development of cardiovascular diseases through oxidative stress, relative pathway gene expression regulation and phenotypic changes in cells. Excess heme plays a detrimental role in atherosclerosis, heart failure, myocardial ischemia-reperfusion injury, degenerative aortic valve stenosis, cardiac iron overload. Recent researches revealed that in some cases heme involved in cardiac damage though ferroptosis. Thus, heme concentrations beyond normal levels are dangerous. Further research on the role of heme in cardiovascular diseases is needed.
Collapse
Affiliation(s)
- Yuyang Guo
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Hengli Zhao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Zhibin Lin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Taochun Ye
- Department of Cardiopulmonary Rehabilitation, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
- *Correspondence: Qingchun Zeng, ; Dingli Xu,
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
- *Correspondence: Qingchun Zeng, ; Dingli Xu,
| |
Collapse
|
32
|
Almahi WA, Yu KN, Mohammed F, Kong P, Han W. Hemin enhances radiosensitivity of lung cancer cells through ferroptosis. Exp Cell Res 2022; 410:112946. [PMID: 34826424 DOI: 10.1016/j.yexcr.2021.112946] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 11/16/2021] [Accepted: 11/21/2021] [Indexed: 01/08/2023]
Abstract
The principle underlying radiotherapy is to kill cancer cells while minimizing the harmful effects on non-cancer cells, which has still remained as a major challenge. In relation, ferroptosis has recently been proposed as a novel mechanism of radiation-induced cell death. In this study, we investigated and demonstrated the role of Hemin as an iron overloading agent in the generation of reactive oxygen species (ROS) induced by ionizing radiation in lung cancer and non-cancer cells. It was found that the presence of Hemin in irradiated lung cancer cells enhanced the productivity of initial ROS, resulting in lipid peroxidation and subsequent ferroptosis. We observed that application of Hemin as a co-treatment increased the activity of GPx4 degradation in both cancer and normal lung cells. Furthermore, Hemin protected normal lung cells against radiation-induced cell death, in that it suppressed ROS after radiation, and boosted the production of bilirubin which was a lipophilic ROS antioxidant. In addition, we demonstrated significant FTH1 expression in normal lung cells when compared to lung cancer cells, which prevented iron from playing a role in increasing IR-induced cell death. Our findings demonstrated that Hemin had a dual function in enhancing the radiosensitivity of ferroptosis in lung cancer cells while promoting cell survival in normal lung cells.
Collapse
Affiliation(s)
- Waleed Abdelbagi Almahi
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, 230026, China; Sudan Atomic Energy Commission, Nuclear Applications in Biological Sciences Institute, Radiobiology and Cancer Researches Department, Khartoum 11111, P.O Box 3001, Sudan.
| | - K N Yu
- Department of Physics, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, 999077, Hong Kong, People's Republic of China; State Key Laboratory in Marine Pollution, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, 999077, Hong Kong, People's Republic of China.
| | - Fathelrahman Mohammed
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, Anhui, China.
| | - Peizhong Kong
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
| | - Wei Han
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
| |
Collapse
|
33
|
Jayakumar D, S Narasimhan KK, Periandavan K. Triad role of hepcidin, ferroportin, and Nrf2 in cardiac iron metabolism: From health to disease. J Trace Elem Med Biol 2022; 69:126882. [PMID: 34710708 DOI: 10.1016/j.jtemb.2021.126882] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/29/2021] [Accepted: 10/19/2021] [Indexed: 11/28/2022]
Abstract
Iron is an essential trace element required for several vital physiological and developmental processes, including erythropoiesis, bone, and neuronal development. Iron metabolism and oxygen homeostasis are interlinked to perform a vital role in the functionality of the heart. The metabolic machinery of the heart utilizes almost 90 % of oxygen through the electron transport chain. To handle this tremendous level of oxygen, the iron metabolism in the heart is utmost crucial. Iron availability to the heart is therefore tightly regulated by (i) the hepcidin/ferroportin axis, which controls dietary iron absorption, storage, and recycling, and (ii) iron regulatory proteins 1 and 2 (IRP1/2) via hypoxia inducible factor 1 (HIF1) pathway. Despite iron being vital to the heart, recent investigations have demonstrated that iron imbalance is a common manifestation in conditions of heart failure (HF), since free iron readily transforms between Fe2+ and Fe3+via the Fenton reaction, leading to reactive oxygen species (ROS) production and oxidative damage. Therefore, to combat iron-mediated oxidative stress, targeting Nrf2/ARE antioxidant signaling is rational. The involvement of Nrf2 in regulating several genes engaged in heme synthesis, iron storage, and iron export is beginning to be uncovered. Consequently, it is possible that Nrf2/hepcidin/ferroportin might act as an epicenter connecting iron metabolism to redox alterations. However, the mechanism bridging the two remains obscure. In this review, we tried to summarize the contemporary insight of how cardiomyocytes regulate intracellular iron levels and discussed the mechanisms linking cardiac dysfunction with iron imbalance. Further, we emphasized the impact of Nrf2 on the interplay between systemic/cardiac iron control in the context of heart disease, particularly in myocardial ischemia and HF.
Collapse
Affiliation(s)
- Deepthy Jayakumar
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute for Basic Medical Sciences, University of Madras, Chennai, 600113, Tamil Nadu, India
| | - Kishore Kumar S Narasimhan
- Department of Pharmacology and Neurosciences, Creighton University, 2500 California Plaza, Omaha, NE, USA
| | - Kalaiselvi Periandavan
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute for Basic Medical Sciences, University of Madras, Chennai, 600113, Tamil Nadu, India.
| |
Collapse
|
34
|
Brasil FB, de Almeida FJS, Luckachaki MD, Dall'Oglio EL, de Oliveira MR. Pinocembrin pretreatment counteracts the chlorpyrifos-induced HO-1 downregulation, mitochondrial dysfunction, and inflammation in the SH-SY5Y cells. Metab Brain Dis 2021; 36:2377-2391. [PMID: 34338973 DOI: 10.1007/s11011-021-00803-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 07/22/2021] [Indexed: 12/14/2022]
Abstract
Chlorpyrifos (CPF), an insecticide, induces pro-oxidant, pro-inflammatory, and pro-apoptotic effects in animal cells. Contamination with CPF occurs not only in farms, since CPF is found in the food consumed in homes. Recently, it was demonstrated that CPF affects the mitochondria, inhibiting components of the electron transfer chain (ETC), causing loss of mitochondrial membrane potential (MMP), and reducing the synthesis of adenosine triphosphate (ATP) by the Complex V. Pinocembrin (PB) is found in propolis and exhibits antioxidant, anti-inflammatory, and anti-apoptotic effects in mammalian cells. PB is a potent inducer of the nuclear factor erythroid 2-related factor 2 (Nrf2), which is a major transcription factor controlling the expression of heme oxygease-1 (HO-1), among others. In the present work, we investigated whether PB would be able to prevent the mitochondrial and immune dysfunctions in the human neuroblastoma SH-SY5Y cells exposed to CPF. PB was tested at 1-25 µM for 4 h before the administration of CPF at 100 µM for additional 24 h. We found that PB prevented the CPF-induced inhibition of ETC, loss of MMP, and decline in the ATP synthesis. PB also promoted anti-inflammatory actions in this experimental model. Silencing of Nrf2 or inhibition of HO-1 suppressed the PB-induced effects in the CPF-challenged cells. Thus, PB promoted beneficial effects by a mechanism dependent on the Nrf2/HO-1/CO + BR axis in the CPF-treated cells.
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
| | - 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, Cuiaba, MT, CEP 78060-900, 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, Cuiaba, MT, CEP 78060-900, 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, Cuiaba, MT, CEP 78060-900, Brazil
| | - Marcos Roberto de Oliveira
- 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, Cuiaba, MT, CEP 78060-900, Brazil.
| |
Collapse
|
35
|
Wu T, Pang Y, Ai HW. Circularly Permuted Far-Red Fluorescent Proteins. BIOSENSORS 2021; 11:438. [PMID: 34821654 PMCID: PMC8615523 DOI: 10.3390/bios11110438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/22/2022]
Abstract
The color palette of genetically encoded fluorescent protein indicators (GEFPIs) has expanded rapidly in recent years. GEFPIs with excitation and emission within the "optical window" above 600 nm are expected to be superior in many aspects, such as enhanced tissue penetration, reduced autofluorescence and scattering, and lower phototoxicity. Circular permutation of fluorescent proteins (FPs) is often the first step in the process of developing single-FP-based GEFPIs. This study explored the tolerance of two far-red FPs, mMaroon1 and mCarmine, towards circular permutation. Several initial constructs were built according to previously reported circularly permuted topologies for other FP analogs. Mutagenesis was then performed on these constructs and screened for fluorescent variants. As a result, five circularly permuted far-red FPs (cpFrFPs) with excitation and emission maxima longer than 600 nm were identified. Some displayed appreciable brightness and efficient chromophore maturation. These cpFrFPs variants could be intriguing starting points to further engineer far-red GEFPIs for in vivo tissue imaging.
Collapse
Affiliation(s)
- Tianchen Wu
- Department of Molecular Physiology and Biological Physics, and Center for Membrane and Cell Physiology, University of Virginia School of Medicine, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA; (T.W.); (Y.P.)
| | - Yu Pang
- Department of Molecular Physiology and Biological Physics, and Center for Membrane and Cell Physiology, University of Virginia School of Medicine, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA; (T.W.); (Y.P.)
- Department of Chemistry, University of Virginia, Charlottesville, VA 22908, USA
| | - Hui-wang Ai
- Department of Molecular Physiology and Biological Physics, and Center for Membrane and Cell Physiology, University of Virginia School of Medicine, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA; (T.W.); (Y.P.)
- Department of Chemistry, University of Virginia, Charlottesville, VA 22908, USA
- The UVA Cancer Center, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
36
|
Nunes B, Pinheiro D, Gomes A. Effect of sublethal concentrations of the antiparasitic ivermectin on the polychaeta species Hediste diversicolor: biochemical and behavioral responses. ECOTOXICOLOGY (LONDON, ENGLAND) 2021; 30:1841-1853. [PMID: 34224071 DOI: 10.1007/s10646-021-02444-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 06/13/2023]
Abstract
Pharmaceutical drugs have emerged as major micropollutants in aquatic ecosystems. Their presence has been systematically reported in monitoring surveys, and their wide distribution and constant presence in the wild is a direct consequence of their massive use, in both human and veterinary therapeutics. Drugs used to treat parasitic infections in livestock are major contaminants, given the amounts in which they are administered, and reach the aquatic compartment in high amounts, where they may affect non target species. Some of these drugs are prone to find their final deposit in sediments of estuarine areas, exerting their toxic effects preferentially at these locations. Sediment dwelling organisms of coastal areas, such as polychaetas, are especially prone to have their major physiological functions compromised after being exposed to pharmaceutical drugs. Ivermectin is one of the most used antiparasitic drugs, and its effects are not limited to biochemical traits, but also behavioral features may be compromised considering their neurotoxic actions. Despite these putative effects, little is known about their toxicity on polychaetas. The present study aimed to characterize the toxicity of realistic levels of ivermectin on the polychaeta Hediste diversicolor, in biochemical and behavioral terms. The obtained results showed that low levels of ivermectin are capable of causing significant disturbances in mobility and burrowing activity of exposed worms, as well as alterations of metabolic and anti-oxidant defense efficacy of exposed animals, suggesting that its environmental presence may mean a major environmental concern.
Collapse
Affiliation(s)
- B Nunes
- Departamento de Biologia da Universidade de Aveiro, Campus de Santiago, 3810-193, Aveiro, Portugal.
- Centro de Estudos do Ambiente e do Mar (CESAM), Campus de Santiago, Universidade de Aveiro, 3810-193, Aveiro, Portugal.
| | - D Pinheiro
- Departamento de Física da Universidade de Aveiro, Campus de Santiago, 3810-193, Aveiro, Portugal
| | - A Gomes
- Departamento de Biologia da Universidade de Aveiro, Campus de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
37
|
Piotrowski ER, Tift MS, Crocker DE, Pearson AB, Vázquez-Medina JP, Keith AD, Khudyakov JI. Ontogeny of Carbon Monoxide-Related Gene Expression in a Deep-Diving Marine Mammal. Front Physiol 2021; 12:762102. [PMID: 34744798 PMCID: PMC8567018 DOI: 10.3389/fphys.2021.762102] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Marine mammals such as northern elephant seals (NES) routinely experience hypoxemia and ischemia-reperfusion events to many tissues during deep dives with no apparent adverse effects. Adaptations to diving include increased antioxidants and elevated oxygen storage capacity associated with high hemoprotein content in blood and muscle. The natural turnover of heme by heme oxygenase enzymes (encoded by HMOX1 and HMOX2) produces endogenous carbon monoxide (CO), which is present at high levels in NES blood and has been shown to have cytoprotective effects in laboratory systems exposed to hypoxia. To understand how pathways associated with endogenous CO production and signaling change across ontogeny in diving mammals, we measured muscle CO and baseline expression of 17 CO-related genes in skeletal muscle and whole blood of three age classes of NES. Muscle CO levels approached those of animals exposed to high exogenous CO, increased with age, and were significantly correlated with gene expression levels. Muscle expression of genes associated with CO production and antioxidant defenses (HMOX1, BVR, GPX3, PRDX1) increased with age and was highest in adult females, while that of genes associated with protection from lipid peroxidation (GPX4, PRDX6, PRDX1, SIRT1) was highest in adult males. In contrast, muscle expression of mitochondrial biogenesis regulators (PGC1A, ESRRA, ESRRG) was highest in pups, while genes associated with inflammation (HMOX2, NRF2, IL1B) did not vary with age or sex. Blood expression of genes involved in regulation of inflammation (IL1B, NRF2, BVR, IL10) was highest in pups, while HMOX1, HMOX2 and pro-inflammatory markers (TLR4, CCL4, PRDX1, TNFA) did not vary with age. We propose that ontogenetic upregulation of baseline HMOX1 expression in skeletal muscle of NES may, in part, underlie increases in CO levels and expression of genes encoding antioxidant enzymes. HMOX2, in turn, may play a role in regulating inflammation related to ischemia and reperfusion in muscle and circulating immune cells. Our data suggest putative ontogenetic mechanisms that may enable phocid pups to transition to a deep-diving lifestyle, including high baseline expression of genes associated with mitochondrial biogenesis and immune system activation during postnatal development and increased expression of genes associated with protection from lipid peroxidation in adulthood.
Collapse
Affiliation(s)
| | - Michael S. Tift
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, NC, United States
| | - Daniel E. Crocker
- Biology Department, Sonoma State University, Rohnert Park, CA, United States
| | - Anna B. Pearson
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, NC, United States
| | - José P. Vázquez-Medina
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Anna D. Keith
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| | - Jane I. Khudyakov
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| |
Collapse
|
38
|
Chu ECP, Wong AYL, Sim P, Krüger F. Exploring scraping therapy: Contemporary views on an ancient healing - A review. J Family Med Prim Care 2021; 10:2757-2762. [PMID: 34660401 PMCID: PMC8483130 DOI: 10.4103/jfmpc.jfmpc_360_21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/27/2021] [Accepted: 05/06/2021] [Indexed: 11/17/2022] Open
Abstract
Gua sha is a traditional healing technique that aims to create petechiae on the skin for a believed therapeutic benefit. Natural healings are mostly based on repeated observations and anecdotal information. Hypothetical model for healing does not always fit the modern understanding. Yet, the mechanisms underlying Gua Sha have not been empirically established. Contemporary scientific research can now explain some events of traditional therapies that were once a mystery. It is assumed that Gua Sha therapy can serve as a mechanical signal to enhance the immune surveillance function of the skin during the natural resolving of the petechiae, through which scraping may result in therapeutic benefits. The current review, without judging the past hypothetical model, attempts to interpret the experience of the ancient healings in terms of contemporary views and concepts.
Collapse
Affiliation(s)
- Eric Chun Pu Chu
- New York Chiropractic and Physiotherapy Centre, New York Medical Group, Hong Kong SAR, China
| | - Arnold Yu Lok Wong
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Patrick Sim
- Australian Chiropractic College, Adelaide, South Australia, Australia
| | - Friso Krüger
- Chiropraktische Familienpraxis, Lüneburg, Germany
| |
Collapse
|
39
|
Basiglio CL, Crocenzi FA, Sánchez Pozzi EJ, Roma MG. Oxidative Stress and Localization Status of Hepatocellular Transporters: Impact on Bile Secretion and Role of Signaling Pathways. Antioxid Redox Signal 2021; 35:808-831. [PMID: 34293961 DOI: 10.1089/ars.2021.0021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Significance: Most hepatopathies are primarily or secondarily cholestatic in nature. Oxidative stress (OS) is a frequent trait among them, and impairs the machinery to generate bile by triggering endocytic internalization of hepatocellular transporters, thus causing cholestasis. This is critical, since it leads to accelerated transporter degradation, which could explain the common post-transcriptional downregulation of transporter expression in human cholestatic diseases. Recent Advances: The mechanisms involved in OS-induced hepatocellular transporter internalization are being revealed. Filamentous actin (F-actin) cytoskeleton disorganization and/or detachment of crosslinking actin proteins that afford transporter stability have been characterized as causal factors. Activation of redox-sensitive signaling pathways leading to changes in phosphorylation status of these structures is involved, including Ca2+-mediated activation of "classical" and "novel" protein kinase C (PKC) isoforms or redox-signaling cascades downstream of NADPH oxidase. Critical Issues: Despite the well-known occurrence of hepatocellular transporter internalization in human hepatopathies, the cholestatic implications of this phenomenon have been overlooked. Accordingly, no specific treatment has been established in the clinical practice for its prevention/reversion. Future Directions: We need to improve our knowledge on the pro-oxidant triggering factors and the multiple signaling pathways that mediate this oxidative injury in each cholestatic hepatopathy, so as to envisage tailor-made therapeutic strategies for each case. Meanwhile, administration of antioxidants or heme oxygenase-1 induction to elevate the hepatocellular levels of the endogenous scavenger bilirubin are promising alternatives that need to be re-evaluated and implemented. They may complement current treatments in cholestasis aimed to enhance transcriptional carrier expression, by providing membrane stability to the newly synthesized carriers. Antioxid. Redox Signal. 35, 808-831.
Collapse
Affiliation(s)
- Cecilia L Basiglio
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| | - Fernando A Crocenzi
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| | - Enrique J Sánchez Pozzi
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| | - Marcelo G Roma
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| |
Collapse
|
40
|
Erem AS, Razzaque MS. Vitamin D-independent benefits of safe sunlight exposure. J Steroid Biochem Mol Biol 2021; 213:105957. [PMID: 34329737 DOI: 10.1016/j.jsbmb.2021.105957] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/15/2021] [Accepted: 07/25/2021] [Indexed: 01/10/2023]
Abstract
This review examines the beneficial effects of ultraviolet radiation on systemic autoimmune diseases, including multiple sclerosis and type I diabetes, where the epidemiological evidence for the vitamin D-independent effects of sunlight is most apparent. Ultraviolet radiation, in addition to its role in the synthesis of vitamin D, stimulates anti-inflammatory pathways, alters the composition of dendritic cells, T cells, and T regulatory cells, and induces nitric oxide synthase and heme oxygenase metabolic pathways, which may directly or indirectly mitigate disease progression and susceptibility. Recent work has also explored how the immune-modulating functions of ultraviolet radiation affect type II diabetes, cancer, and the current global pandemic caused by SARS-CoV-2. These diseases are particularly important amidst global changes in lifestyle that result in unhealthy eating, increased sedentary habits, and alcohol and tobacco consumption. Compelling epidemiological data shows increased ultraviolet radiation associated with reduced rates of certain cancers, such as colorectal cancer, breast cancer, non-Hodgkins lymphoma, and ultraviolet radiation exposure correlated with susceptibility and mortality rates of COVID-19. Therefore, understanding the effects of ultraviolet radiation on both vitamin D-dependent and -independent pathways is necessary to understand how they influence the course of many human diseases.
Collapse
Affiliation(s)
- Anna S Erem
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mohammed S Razzaque
- Department of Pathology, Lake Erie College of Osteopathic Medicine, Erie, PA, USA.
| |
Collapse
|
41
|
The Role of HO-1 and Its Crosstalk with Oxidative Stress in Cancer Cell Survival. Cells 2021; 10:cells10092401. [PMID: 34572050 PMCID: PMC8471703 DOI: 10.3390/cells10092401] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 12/19/2022] Open
Abstract
Heme oxygenases (HOs) act on heme degradation to produce carbon monoxide (CO), free iron, ferritin, and biliverdin. Upregulation of cellular HO-1 levels is signature of oxidative stress for its downstream effects particularly under pro-oxidative status. Subcellular traffics of HO-1 to different organelles constitute a network of interactions compromising a variety of effectors such as pro-oxidants, ROS, mitochondrial enzymes, and nucleic transcription factors. Some of the compartmentalized HO-1 have been demonstrated as functioning in the progression of cancer. Emerging data show the multiple roles of HO-1 in tumorigenesis from pathogenesis to the progression to malignancy, metastasis, and even resistance to therapy. However, the role of HO-1 in tumorigenesis has not been systematically addressed. This review describes the crosstalk between HO-1 and oxidative stress, and following redox regulation in the tumorigenesis. HO-1-regulated signaling pathways are also summarized. This review aims to integrate basic information and current progress of HO-1 in cancer research in order to enhance the understandings and facilitate following studies.
Collapse
|
42
|
Žiberna L, Jenko-Pražnikar Z, Petelin A. Serum Bilirubin Levels in Overweight and Obese Individuals: The Importance of Anti-Inflammatory and Antioxidant Responses. Antioxidants (Basel) 2021; 10:antiox10091352. [PMID: 34572984 PMCID: PMC8472302 DOI: 10.3390/antiox10091352] [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: 07/23/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 12/14/2022] Open
Abstract
Obesity is a chronic condition involving low-grade inflammation and increased oxidative stress; thus, obese and overweight people have lower values of serum bilirubin. Essentially, bilirubin is a potent endogenous antioxidant molecule with anti-inflammatory, immunomodulatory, antithrombotic, and endocrine properties. This review paper presents the interplay between obesity-related pathological processes and bilirubin, with a focus on adipose tissue and adipokines. We discuss potential strategies to mildly increase serum bilirubin levels in obese patients as an adjunctive therapeutic approach.
Collapse
Affiliation(s)
- Lovro Žiberna
- Institute of Pharmacology and Experimental Toxicology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia;
| | | | - Ana Petelin
- Faculty of Health Sciences, University of Primorska, SI-6310 Izola, Slovenia;
- Correspondence: ; Tel.: +386-5-66-2469
| |
Collapse
|
43
|
Fu K, Garvan CS, Heaton SC, Nagaraja N, Doré S. Association of Serum Bilirubin with the Severity and Outcomes of Intracerebral Hemorrhages. Antioxidants (Basel) 2021; 10:antiox10091346. [PMID: 34572977 PMCID: PMC8465680 DOI: 10.3390/antiox10091346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 11/22/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is the second most common subtype of stroke, and it is often associated with a high mortality rate and significant morbidity among survivors. Recent studies have shown that bilirubin, a product of heme metabolism, can exhibit cytoprotective, antioxidant and, anti-inflammatory properties. However, little is known about the role of bilirubin in combating several pathophysiological pathways caused by intracerebral bleeding in patients with ICH. In this study, data were collected retrospectively on 276 patients with ICH who were admitted to a university hospital between 5 January 2014 and 31 December 2017. We assessed the relationship between levels of total, direct, and indirect serum bilirubin and assessments of initial stroke severity and clinical outcomes by using Spearman’s rank correlation and Kruskal-Wallis H tests. A secondary examination of the carrier protein albumin was also undertaken. Our study found that higher levels of direct bilirubin were correlated with worse admission Glasgow Coma Scales (GCS) (rs = −0.17, p = 0.011), worse admission ICH Scores (rs = 0.19, p = 0.008), and worse discharge modified Rankin Scales (mRS) (rs = 0.15, p = 0.045). Direct bilirubin was still significantly correlated with discharge mRS after adjusting for temperature at admission (rs = 0.16, p = 0.047), oxygen saturation at admission (rs = 0.15, p = 0.048), white blood cell count (rs = 0.18, p = 0.023), or Troponin T (rs = 0.25, p = 0.001) using partial Spearman’s correlation. No statistical significance was found between levels of total or indirect bilirubin and assessments of stroke severity and outcomes. In contrast, higher levels of albumin were correlated with better admission GCS (rs = 0.13, p = 0.027), discharge GCS (rs = 0.15, p = 0.013), and discharge mRS (rs = −0.16, p = 0.023). We found that levels of total bilirubin, direct bilirubin, and albumin were all significantly related to discharge outcomes classified by discharge destinations (p = 0.036, p = 0.014, p = 0.016, respectively; Kruskal-Wallis H tests). In conclusion, higher direct bilirubin levels were associated with greater stroke severity at presentation and worse outcomes at discharge among patients with ICH. Higher levels of albumin were associated with lower stroke severity and better clinical outcomes. Future prospective studies on the free bioactive bilirubin are needed to better understand the intricate relationships between bilirubin and ICH.
Collapse
Affiliation(s)
- Kai Fu
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL 32610, USA; (K.F.); (C.S.G.)
| | - Cynthia S. Garvan
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL 32610, USA; (K.F.); (C.S.G.)
| | - Shelley C. Heaton
- Department of Clinical & Health Psychology, University of Florida College of Public Health and Health Professions, Gainesville, FL 32610, USA;
| | - Nandakumar Nagaraja
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL 32610, USA;
| | - Sylvain Doré
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL 32610, USA; (K.F.); (C.S.G.)
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL 32610, USA;
- Center for Translational Research in Neurodegenerative Disease, Departments of Psychiatry, Pharmaceutics, and Neuroscience, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL 32610, USA
- Correspondence: ; Tel.: +1-352-273-9663
| |
Collapse
|
44
|
Hyperbilirubinemia Maintained by Chronic Supplementation of Unconjugated Bilirubin Improves the Clinical Course of Experimental Autoimmune Arthritis. Int J Mol Sci 2021; 22:ijms22168662. [PMID: 34445367 PMCID: PMC8395408 DOI: 10.3390/ijms22168662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/06/2021] [Accepted: 08/06/2021] [Indexed: 11/23/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic multisystem disease, therapy of which remains a challenge for basic research. The present work examined the effect of unconjugated bilirubin (UCB) administration in adjuvant-induced arthritis (AIA)—an experimental model, in which oxidative stress (OS), inflammation and inadequate immune response are often similar to RA. Male Lewis rats were randomized into groups: CO—control, AIA—untreated adjuvant-induced arthritis, AIA-BIL—adjuvant-induced arthritis administrated UCB, CO-BIL—control with administrated UCB. UCB was administered intraperitoneally 200 mg/kg of body weight daily from 14th day of the experiment, when clinical signs of the disease are fully manifested, to 28th day, the end of the experiment. AIA was induced by a single intradermal immunization at the base of the tail with suspension of Mycobacterium butyricum in incomplete Freund’s adjuvant. Clinical, hematologic, biochemical and histologic examinations were performed. UCB administration to animals with AIA lead to a significant decrease in hind paws volume, plasma levels of C-reactive protein (CRP) and ceruloplasmin, drop of leukocytes, lymphocytes, erythrocytes, hemoglobin and an increase in platelet count. UCB administration caused significantly lowered oxidative damage to DNA in arthritic animals, whereas in healthy controls it induced considerable oxidative damage to DNA. UCB administration also induced atrophy of the spleen and thymus in AIA and CO animals comparing to untreated animals. Histological signs of joint damage assessed by neutrophils infiltration and deposition of fibrin were significantly reduced by UCB administration. The effects of exogenously administered UCB to the animals with adjuvant-induced arthritis might be identified as therapeutic, in contrast to the effects of UCB administration in healthy animals rather classified as toxic.
Collapse
|
45
|
Brasil FB, de Almeida FJS, Luckachaki MD, Dall'Oglio EL, de Oliveira MR. Suppression of Mitochondria-Related Bioenergetics Collapse and Redox Impairment by Tanshinone I, a Diterpenoid Found in Salvia miltiorrhiza Bunge (Danshen), in the Human Dopaminergic SH-SY5Y Cell Line Exposed to Chlorpyrifos. Neurotox Res 2021; 39:1495-1510. [PMID: 34351569 DOI: 10.1007/s12640-021-00400-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/24/2021] [Accepted: 07/27/2021] [Indexed: 01/01/2023]
Abstract
Tanshinone I (T-I, C18H12O3) is a diterpene found in Salvia miltiorrhiza Bunge (Danshen) and promotes cytoprotection in several experimental models. Chlorpyrifos (CPF) is an agrochemical that causes bioenergetics failure, redox impairment, inflammation, and cell death in animal tissues. Here, we investigated whether T-I would be able to prevent the consequences resulting from the exposure of the human dopaminergic SH-SY5Y cells to CPF. We found that a pretreatment with T-I at 2.5 µM for 2 h suppressed lipid peroxidation and protein carbonylation and nitration on the membranes of mitochondria extracted from the CPF-treated cells. Also, T-I reduced the production of radical superoxide (O2-•) by the mitochondria of the CPF-challenged cells. The production of nitric oxide (NO•) and hydrogen peroxide (H2O2) was also decreased by T-I in the cells exposed to CPF. The CPF-induced decrease in the activity of the complexes I-III, II-III, and V was abolished by a pretreatment with T-I. Loss of mitochondrial membrane potential (ΔΨm) and reduction in the production of adenosine triphosphate (ATP) were also prevented by T-I in the CPF-treated cells. T-I also induced anti-inflammatory effects in the CPF-treated cells by decreasing the levels of interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) and the activity of the nuclear factor-κB (NF-κB). Inhibition of heme oxygenase-1 (HO-1) or silencing of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) blocked the T-I-promoted mitochondrial protection and anti-inflammatory action. Overall, T-I depended on the Nrf2/HO-1 axis to prevent the deleterious effects caused by CPF in this experimental model.
Collapse
Affiliation(s)
- Flávia Bittencourt Brasil
- Department of Natural Sciences, Rio das Ostras Universitary Campus - Fluminense Federal University (UFF), Rio de Janeiro, Brazil
| | - Fhelipe Jolner Souza de Almeida
- Graduate Program in Health Sciences (PPGCS), Federal University of Mato Grosso (UFMT), Cuiaba, MT, Brazil
- Research Group in Neurochemistry and Neurobiology of Bioactive Molecules, Federal University of Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, Cuiaba, MT, 2367, 78060-900, Brazil
| | - Matheus Dargesso Luckachaki
- Research Group in Neurochemistry and Neurobiology of Bioactive Molecules, Federal University of Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, Cuiaba, MT, 2367, 78060-900, Brazil
| | - Evandro Luiz Dall'Oglio
- Research Group in Neurochemistry and Neurobiology of Bioactive Molecules, Federal University of Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, Cuiaba, MT, 2367, 78060-900, Brazil
| | - Marcos Roberto de Oliveira
- Research Group in Neurochemistry and Neurobiology of Bioactive Molecules, Federal University of Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, Cuiaba, MT, 2367, 78060-900, Brazil.
| |
Collapse
|
46
|
Zahra KF, Lefter R, Ali A, Abdellah EC, Trus C, Ciobica A, Timofte D. The Involvement of the Oxidative Stress Status in Cancer Pathology: A Double View on the Role of the Antioxidants. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9965916. [PMID: 34394838 PMCID: PMC8360750 DOI: 10.1155/2021/9965916] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/19/2021] [Indexed: 12/24/2022]
Abstract
Oxygen-free radicals, reactive oxygen species (ROS) or reactive nitrogen species (RNS), are known by their "double-sided" nature in biological systems. The beneficial effects of ROS involve physiological roles as weapons in the arsenal of the immune system (destroying bacteria within phagocytic cells) and role in programmed cell death (apoptosis). On the other hand, the redox imbalance in favor of the prooxidants results in an overproduction of the ROS/RNS leading to oxidative stress. This imbalance can, therefore, be related to oncogenic stimulation. High levels of ROS disrupt cellular processes by nonspecifically attacking proteins, lipids, and DNA. It appears that DNA damage is the key player in cancer initiation and the formation of 8-OH-G, a potential biomarker for carcinogenesis. The harmful effect of ROS is neutralized by an antioxidant protection treatment as they convert ROS into less reactive species. However, contradictory epidemiological results show that supplementation above physiological doses recommended for antioxidants and taken over a long period can lead to harmful effects and even increase the risk of cancer. Thus, we are describing here some of the latest updates on the involvement of oxidative stress in cancer pathology and a double view on the role of the antioxidants in this context and how this could be relevant in the management and pathology of cancer.
Collapse
Affiliation(s)
- Kamal Fatima Zahra
- Faculty of Sciences and Techniques, Laboratory of Physical Chemistry of Processes and Materials/Agri-Food and Health, Hassan First University, B.P. 539, 26000 Settat, Morocco
| | - Radu Lefter
- Center of Biomedical Research, Romanian Academy, 8th Carol I Avenue, 700506 Iasi, Romania
| | - Ahmad Ali
- Department of Life Sciences, University of Mumbai, Vidyanagari, Santacruz (East), Mumbai 400098, India
| | - Ech-Chahad Abdellah
- Faculty of Sciences and Techniques, Laboratory of Physical Chemistry of Processes and Materials, Hassan First University, B.P. 539, 26000 Settat, Morocco
| | - Constantin Trus
- Department of Morphological and Functional Sciences, Faculty of Medicine, Dunarea de Jos University, 800008 Galati, Romania
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University, 11th Carol I Avenue, 700506 Iasi, Romania
| | - Daniel Timofte
- Faculty of Medicine, “Grigore T. Popa”, University of Medicine and Pharmacy, Strada Universitatii 16, 700115 Iasi, Romania
| |
Collapse
|
47
|
Huan S, Jin S, Liu H, Xia W, Liang G, Xu S, Fang X, Li C, Wang Q, Sun X, Li Y. Fine particulate matter exposure and perturbation of serum metabolome: A longitudinal study in Baoding, China. CHEMOSPHERE 2021; 276:130102. [PMID: 33684857 DOI: 10.1016/j.chemosphere.2021.130102] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/11/2021] [Accepted: 02/22/2021] [Indexed: 06/12/2023]
Abstract
Metabolomics represents a powerful tool for measuring environmental exposures and biological responses to unveil potential mechanisms. Few studies have investigated the effects of exposure to fine particulate matter (PM2.5) longitudinally on serum metabolomics in regions with high-level PM2.5. Therefore, we examined the changes of serum metabolomics corresponding to individual PM2.5 exposure levels in spring and autumn among 63 healthy college students in Baoding city, Hebei, China. The metabolic profiling was determined by ultra-performance liquid chromatography coupled to quadrupole time-of-flight mass spectrometry. The average level of individual PM2.5 in the spring was 1.82-fold higher than in the autumn (240 μg/m3 vs 132 μg/m3). Males were exposed to a higher level of PM2.5 than females in the spring. Metabolic profiling was clearly separated by orthogonal partial least square-discriminant analysis in males but not in females. In the analysis of the associations between the metabolome and PM2.5 of the two seasons, the changes of 14 serum metabolites were significantly associated with PM2.5 in males. The metabolites related to heme metabolism (bilirubin, biliverdin), energy metabolism and oxidative stress (2-Octenoylcarnitine, N-Heptanoylglycine, and acetylcysteine), phospholipid metabolism (lysophosphatidic acid, phospholipid acid, and lysophosphatidylethanolamine), and tryptophan metabolism (N-Acetylserotonin, indolepyruvate, and melatonin) were decreased in the range of 2.16%-6.80% for each 10 μg/m3 increase of PM2.5, while thyrotropin-releasing hormone, glutathione, and phosphatidylethanolamine related to energy metabolism and oxidative stress, and phospholipid metabolism were increased in the range of 2.95%-4.90% for each 10 μg/m3 increase of PM2.5. This longitudinal study suggests that higher PM2.5 exposure may induce perturbations in serum metabolic signaling related to oxidative stress and inflammation, and males may be more prone to these metabolic perturbations.
Collapse
Affiliation(s)
- Shu Huan
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China; State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China
| | - Shuna Jin
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China; State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China
| | - Hongxiu Liu
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China; State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China; New York University, New York, 10016, United States
| | - Wei Xia
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China; State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China.
| | - Gaodao Liang
- Institute of Environmental Health, Wuhan Centers for Disease Prevention & Control, Wuhan, Hubei, 430024, PR China.
| | - Shunqing Xu
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China; State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China
| | - Xingjie Fang
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China; State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China
| | - Chunhui Li
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China; State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China
| | - Qianqian Wang
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China; State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China
| | - Xiaojie Sun
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China; State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China
| | - Yuanyuan Li
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China; State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, PR China
| |
Collapse
|
48
|
Sterling JP, Lombardi VC. Decreasing the Likelihood of Multiple Organ Dysfunction Syndrome in Burn Injury with Early Antioxidant Treatment. Antioxidants (Basel) 2021; 10:antiox10081192. [PMID: 34439439 PMCID: PMC8389021 DOI: 10.3390/antiox10081192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 01/21/2023] Open
Abstract
Major burn trauma initiates a cascade of physiological events that cause profound stress on the body, resulting in significant complications which often lead to death. An understanding of these events may afford earlier and more precise interventions which, in turn, may reduce these complications, thus, improving patient outcomes. Burn trauma is associated with numerous inflammatory events that result in the release of free radicals, which promote oxidative stress and subsequent tissue damage. These mass-inflammatory events affect the body systemically, leading to several detrimental responses including complement activation, excessive histamine release, decrease in blood pressure, release of reactive oxygen species, and ultimately multiple organ dysfunction syndrome (MODS). However, recent studies conducted on the use of antioxidants as a part of a burn treatment protocol have shown promising results. In this review, we will discuss the current research and advancements in the treatment of burn trauma with the use of antioxidants, and how the early administration of antioxidant can possibly reduce the risk of developing MODS.
Collapse
Affiliation(s)
| | - Vincent C. Lombardi
- Department of Microbiology and Immunology, University of Nevada, Reno, School of Medicine, Reno, NV 89557, USA
- Correspondence: ; Tel.: +1-775-682-8278
| |
Collapse
|
49
|
Rashdan NA, Zhai B, Lovern PC. Fluid shear stress regulates placental growth factor expression via heme oxygenase 1 and iron. Sci Rep 2021; 11:14912. [PMID: 34290391 PMCID: PMC8295300 DOI: 10.1038/s41598-021-94559-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/13/2021] [Indexed: 12/29/2022] Open
Abstract
Increased fluid shear stress (FSS) is a key initiating stimulus for arteriogenesis, the outward remodeling of collateral arterioles in response to upstream occlusion. Placental growth factor (PLGF) is an important arteriogenic mediator. We previously showed that elevated FSS increases PLGF in a reactive oxygen species (ROS)-dependent fashion both in vitro and ex vivo. Heme oxygenase 1 (HO-1) is a cytoprotective enzyme that is upregulated by stress and has arteriogenic effects. In the current study, we used isolated murine mesentery arterioles and co-cultures of human coronary artery endothelial cells (EC) and smooth muscle cells (SMC) to test the hypothesis that HO-1 mediates the effects of FSS on PLGF. HO-1 mRNA was increased by conditions of increased flow and shear stress in both co-cultures and vessels. Both inhibition of HO-1 with zinc protoporphyrin and HO-1 knockdown abolished the effect of FSS on PLGF. Conversely, induction of HO-1 activity increased PLGF. To determine which HO-1 product upregulates PLGF, co-cultures were treated with a CO donor (CORM-A1), biliverdin, ferric ammonium citrate (FAC), or iron-nitrilotriacetic acid (iron-NTA). Of these FAC and iron-NTA induced an increase PLGF expression. This study demonstrates that FSS acts through iron to induce pro-arteriogenic PLGF, suggesting iron supplementation as a novel potential treatment for revascularization.
Collapse
Affiliation(s)
- Nabil A Rashdan
- Department of Molecular and Cellular Physiology, Louisiana State University, Shreveport, LA, USA
| | - Bo Zhai
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Pamela C Lovern
- Department of Physiological Sciences, Oklahoma State University, 264 McElroy Hall, Stillwater, OK, 74078, USA.
| |
Collapse
|
50
|
Suihara S, Ishisaka A, Murakami A. (-)-Epigallocatechin-3-O-gallate at a high concentration may induce lipolysis via ATP consumption by activation of stress defense mechanisms. Biosci Biotechnol Biochem 2021; 85:411-420. [PMID: 33604623 DOI: 10.1093/bbb/zbaa056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/30/2020] [Indexed: 01/02/2023]
Abstract
Green tea catechins have thus far been demonstrated to have antiobesity effects in a variety of experimental models. However, upstream molecular events triggering those phenomena remain to be identified. In this study, we found that (-)-epigallocatechin-3-O-gallate (EGCG) promoted lipolysis in lipid-loaded Huh7 human hepatoma cells. Notably, EGCG at a high concentration induced both oxidative stress and protein stress (proteo-stress), leading to activation of stress defense mechanisms, such as mRNA expressions of antioxidant and phase-2 detoxifying enzymes, and heat shock proteins. Furthermore, EGCG decreased the level of intracellular ATP, while glucose uptake from culture media was promoted possibly for energy homeostasis. EGCG also upregulated the expression of adipose triglyceride lipase, and activated AMP-activated protein kinase. Collectively, these results suggest that EGCG induces lipolysis to compensate for ATP reduction derived from activation of stress defense systems against its oxidative and proteo-stress properties.
Collapse
Affiliation(s)
- Satoki Suihara
- Department of Food Science and Nutrition, School of Human Science and Environment, University of Hyogo, Himeji, Hyogo, Japan
| | - Akari Ishisaka
- Department of Food Science and Nutrition, School of Human Science and Environment, University of Hyogo, Himeji, Hyogo, Japan.,Research Institute for Food and Nutritional Sciences, University of Hyogo, Himeji, Hyogo, Japan
| | - Akira Murakami
- Department of Food Science and Nutrition, School of Human Science and Environment, University of Hyogo, Himeji, Hyogo, Japan.,Research Institute for Food and Nutritional Sciences, University of Hyogo, Himeji, Hyogo, Japan
| |
Collapse
|