1
|
Joubert MBV, Ingaramo PI, Collins P, D'Alessandro ME. Astaxanthin improves lipotoxicity, lipid peroxidation and oxidative stress in kidney of sucrose-rich diet-fed rats. J Nutr Biochem 2025; 135:109779. [PMID: 39374743 DOI: 10.1016/j.jnutbio.2024.109779] [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: 05/10/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/09/2024]
Abstract
Metabolic Syndrome (MS) is a cluster of metabolic risk factors, characterized by abdominal obesity, dyslipidemia, hypertension, insulin resistance, among others. The purpose of the study was to evaluate the astaxanthin (AXT) effects extracted from freshwater crab (Dilocarcinus pagei) at the Paraná Basin on lipotoxicity, lipid peroxidation and oxidative stress in the kidney of rats fed with a sucrose-rich diet (SRD). We hypothesized that daily administration of AXT prevents kidney damage by reducing lipotoxicity, lipid peroxidation, and reactive oxygen species (ROS), and by improving antioxidant enzyme defenses and crosstalk between NrF2 and NF-ĸB transcription factors. Male Wistar rats were fed a reference diet (RD), RD+AXT, SRD and SRD+AXT (AXT daily oral dose: [10 mg/kg body weight]) for 90 days. Systolic and diastolic blood pressure, biochemical assays in serum and urine were evaluated. Renal cortex samples were taken for histological analysis, determination of triglyceride content, ROS, thiobarbituric acid reactive substances (TBARS), catalase (CAT), glutathione peroxidase (GPx) and glutathione reductase (GR) enzyme activities and glutathione content (GSH). 4-HNE, NrF2, and NF-ĸB p65 expression were analyzed by immunohistochemistry. We demonstrated that daily oral supplementation of AXT to animals fed a SRD reduced systolic and diastolic blood pressure, histological renal damage, lipid accumulation, ROS and lipid peroxidation, and increased CAT and GPx activities. NrF2 protein expression in renal cortex was increased, whilst NF-ĸB p65 was reduced. AXT extracted from freshwater crabs (Dilocarcinus pagei) may be promising nutritional strategy for the prevention of renal alterations present in this model.
Collapse
Affiliation(s)
- Michelle Berenice Vega Joubert
- Laboratorio de Estudio de Enfermedades Metabólicas relacionadas con la Nutrición, Departamento de Ciencias Biológicas, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Paola Inés Ingaramo
- Departamento de Fisiopatología Ambiental, Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Cs. Biológicas. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Pablo Collins
- Departamento de Acuicultura, Instituto Nacional de Limnología (INALI), Universidad Nacional del Litoral- Consejo Nacional de Investigaciones Cientificas y Técnicas (CONICET), Santa Fe, Argentina
| | - María Eugenia D'Alessandro
- Laboratorio de Estudio de Enfermedades Metabólicas relacionadas con la Nutrición, Departamento de Ciencias Biológicas, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina.
| |
Collapse
|
2
|
Heine N, Weber A, Pautsch A, Gottschling D, Uphues I, Bauer M, Ebenhoch R, Magarkar A, Nosse B, Kley JT. Discovery of BI-9787, a potent zwitterionic ketohexokinase inhibitor with oral bioavailability. Bioorg Med Chem Lett 2024; 112:129930. [PMID: 39179180 DOI: 10.1016/j.bmcl.2024.129930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/01/2024] [Accepted: 08/15/2024] [Indexed: 08/26/2024]
Abstract
Fructose metabolism by ketohexokinase (KHK) is implicated in a variety of metabolic disorders. KHK inhibition is a potential therapeutic strategy for the treatment of diseases including diabetes, non-alcoholic fatty liver disease, and non-alcoholic steatohepatitis. The first small-molecule KHK-inhibitors have entered clinical trials, but it remains unclear if systemic inhibition of KHK by small-molecules will eventually benefit patients. Here we report the discovery of BI-9787, a potent, zwitterionic KHK inhibitor characterized by high permeability and favorable oral rat pharmacokinetics. BI-9787 was identified by optimizing chemical starting points generated via a ligand-based virtual screening of Boehringer's virtual library of synthetically accessible compounds (BICLAIM). It serves as a high-quality in vitro and in vivo tool compound for investigating the role of fructose metabolism in disease.
Collapse
Affiliation(s)
- Niklas Heine
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Alexander Weber
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Alexander Pautsch
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Dirk Gottschling
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Ingo Uphues
- Boehringer Ingelheim Pharma GmbH & Co. KG, Cardiometabolic Diseases Research, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Margit Bauer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Rebecca Ebenhoch
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Aniket Magarkar
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Bernd Nosse
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany; Boehringer Ingelheim International GmbH, Business Development & Licensing, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Jörg Thomas Kley
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany.
| |
Collapse
|
3
|
Jin S, Wang YS, Huang JC, Wang TT, Li BY, Guo B, Yue ZP. Osthole exhibits the remedial potential for polycystic ovary syndrome mice through Nrf2-Foxo1-GSH-NF-κB pathway. Cell Biol Int 2024; 48:1111-1123. [PMID: 38741282 DOI: 10.1002/cbin.12170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/03/2024] [Accepted: 04/26/2024] [Indexed: 05/16/2024]
Abstract
Polycystic ovary syndrome (PCOS) is the primary cause of female infertility with a lack of universal therapeutic regimen. Although osthole exhibits numerous pharmacological activities in treating various diseases, its therapeutic effect on PCOS is undiscovered. The present study found that application of osthole improved the symptoms of PCOS mice through preventing ovarian granulosa cells (GCs) production of more estrogen and alleviating the liberation of pro-inflammatory cytokine interleukin (IL)-1β, IL-6, and tumor necrosis factor alpha. Meanwhile, osthole enhanced ovarian antioxidant capacity and alleviated intracellular reactive oxygen species (ROS) accumulation with a concurrent attenuation for oxidative stress, while intervention of antioxidant enzymic activity and glutathione (GSH) synthesis neutralized the salvation of osthole on GCs secretory disorder and chronic inflammation. Further analysis revealed that osthole restored the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and forkhead box O 1 (Foxo1) whose repression antagonized the amelioration of osthole on the insufficiency of antioxidant capacity and accumulation of ROS. Moreover, Nrf2 served as an intermedium to mediate the regulation of osthole on Foxo1. Additionally, osthole restricted the phosphorylation of IκBα and nuclear factor kappa B (NF-κB) subunit p65 by DHEA and weakened the transcriptional activity of NF-κB, but this effectiveness was abrogated by the obstruction of Nrf2 and Foxo1, whereas adjunction of GSH renewed the redemptive effect of osthole on NF-κB whose activation caused an invalidation of osthole in rescuing the aberration of GCs secretory function and inflammation response. Collectively, osthole might relieve the symptoms of PCOS mice via Nrf2-Foxo1-GSH-NF-κB pathway.
Collapse
Affiliation(s)
- Shan Jin
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
- Reproductive Medical Center, The Second Hospital of Jilin University, Changchun, P.R. China
| | - Yu-Si Wang
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Ji-Cheng Huang
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Ting-Ting Wang
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Bai-Yu Li
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Bin Guo
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Zhan-Peng Yue
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| |
Collapse
|
4
|
Mohamed B, Ghareib SA, Alsemeh AE, El-Sayed SS. Telmisartan ameliorates nephropathy and restores the hippo pathway in rats with metabolic syndrome. Eur J Pharmacol 2024; 973:176605. [PMID: 38653362 DOI: 10.1016/j.ejphar.2024.176605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
The main objective of this study was to determine if the telmisartan-ameliorative effects of metabolic syndrome (MetS)-evoked nephropathy are attributed to the Hippo pathway. A secondary objective was to investigate the potential of vitamin D3 to enhance telmisartan-favourable effects. A diet composed of 24% fat and 3% salt, along with drinking water containing 10% fructose, was administered for 12 weeks to induce MetS. MetS-rats were given telmisartan (5 mg/kg/day), vitamin D3 (10 μg/kg/day) or both by gavage, starting in the sixth week of experimental diet administration. Assessments performed at closure included renal function, histological examination, catalase, malondialdehyde (MDA), nuclear factor kappa-B (NF-κB), interleukin-6 (IL-6), peroxisome proliferator-activated receptor-γ (PPAR-γ), phosphatase and tensin homolog (PTEN), and transforming growth factor-β (TGF-β). Matrix metalloproteinase-9 (MMP-9) immunostaining was conducted. The expression of the Hippo pathway components, as well as that of angiotensin II type 1 and type 2 (AT1 and AT2), receptors was evaluated. Telmisartan attenuated MetS-evoked nephropathy, as demonstrated by improvement of renal function and histological features, enhancement of catalase, reduction of MDA, inflammation (NF-κB, IL-6), and renal fibrosis (increased PPAR-γ and PTEN and reduced MMP-9 and TGF-β). Telmisartan downregulated AT1-receptor, upregulated AT2-receptor and restored the Hippo pathway. Vitamin D3 replicated most of the telmisartan-elicited effects and enhanced the antifibrotic actions of telmisartan. The alleviative effects of telmisartan on MetS-evoked nephropathy may be related to the restoration of the Hippo pathway. The combination of vitamin D3 and telmisartan exerted more favourable effects on metabolic and nephropathic biomarkers compared with either one administered alone.
Collapse
Affiliation(s)
- Badria Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Salah A Ghareib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Amira Ebrahim Alsemeh
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt.
| | - Shaimaa S El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
5
|
Arellano Buendia AS, Juárez Rojas JG, García-Arroyo F, Aparicio Trejo OE, Sánchez-Muñoz F, Argüello-García R, Sánchez-Lozada LG, Bojalil R, Osorio-Alonso H. Antioxidant and anti-inflammatory effects of allicin in the kidney of an experimental model of metabolic syndrome. PeerJ 2023; 11:e16132. [PMID: 37786577 PMCID: PMC10541809 DOI: 10.7717/peerj.16132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 08/28/2023] [Indexed: 10/04/2023] Open
Abstract
Background Recent studies have suggested that metabolic syndrome (MS) encompasses a group of risk factors for developing chronic kidney disease (CKD). This work aimed to evaluate the antioxidant and anti-inflammatory effects of allicin in the kidney from an experimental model of MS. Methods Male Wistar rats (220-250 g) were used, and three experimental groups (n = 6) were formed: control (C), metabolic syndrome (MS), and MS treated with allicin (16 mg/Kg/day, gastric gavage) (MS+A). MS was considered when an increase of 20% in at least three parameters (body weight, systolic blood pressure (SBP), fasting blood glucose (FBG), or dyslipidemia) was observed compared to the C group. After the MS diagnosis, allicin was administered for 30 days. Results Before the treatment with allicin, the MS group showed more significant body weight gain, increased SBP, and FBG, glucose intolerance, and dyslipidemia. In addition, increased markers of kidney damage in urine and blood. Moreover, the MS increased oxidative stress and inflammation in the kidney compared to group C. The allicin treatment prevented further weight gain, reduced SBP, FBG, glucose intolerance, and dyslipidemia. Also, markers of kidney damage in urine and blood were decreased. Further, the oxidative stress and inflammation were decreased in the renal cortex of the MS+A compared to the MS group. Conclusion Allicin exerts its beneficial effects on the metabolic syndrome by considerably reducing systemic and renal inflammation as well as the oxidative stress. These effects were mediated through the Nrf2 pathway. The results suggest allicin may be a therapeutic alternative for treating kidney injury induced by the metabolic syndrome risk factors.
Collapse
Affiliation(s)
- Abraham Said Arellano Buendia
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Mexico, Xochimilco, Mexico
- Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico, Tlalpan, México
| | | | - Fernando García-Arroyo
- Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico, Tlalpan, México
| | | | - Fausto Sánchez-Muñoz
- Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico, Tlalpan, México
| | - Raúl Argüello-García
- Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del IPN, México, Gustavo A. Madero, México
| | | | - Rafael Bojalil
- Atención a la Salud, Universidad Autónoma Metropolitana, Mexico, Xochimilco, México
| | - Horacio Osorio-Alonso
- Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico, Tlalpan, México
| |
Collapse
|
6
|
Johnson RJ, Lanaspa MA, Sanchez-Lozada LG, Tolan D, Nakagawa T, Ishimoto T, Andres-Hernando A, Rodriguez-Iturbe B, Stenvinkel P. The fructose survival hypothesis for obesity. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220230. [PMID: 37482773 PMCID: PMC10363705 DOI: 10.1098/rstb.2022.0230] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 05/04/2023] [Indexed: 07/25/2023] Open
Abstract
The fructose survival hypothesis proposes that obesity and metabolic disorders may have developed from over-stimulation of an evolutionary-based biologic response (survival switch) that aims to protect animals in advance of crisis. The response is characterized by hunger, thirst, foraging, weight gain, fat accumulation, insulin resistance, systemic inflammation and increased blood pressure. The process is initiated by the ingestion of fructose or by stimulating endogenous fructose production via the polyol pathway. Unlike other nutrients, fructose reduces the active energy (adenosine triphosphate) in the cell, while blocking its regeneration from fat stores. This is mediated by intracellular uric acid, mitochondrial oxidative stress, the inhibition of AMP kinase and stimulation of vasopressin. Mitochondrial oxidative phosphorylation is suppressed, and glycolysis stimulated. While this response is aimed to be modest and short-lived, the response in humans is exaggerated due to gain of 'thrifty genes' coupled with a western diet rich in foods that contain or generate fructose. We propose excessive fructose metabolism not only explains obesity but the epidemics of diabetes, hypertension, non-alcoholic fatty liver disease, obesity-associated cancers, vascular and Alzheimer's dementia, and even ageing. Moreover, the hypothesis unites current hypotheses on obesity. Reducing activation and/or blocking this pathway and stimulating mitochondrial regeneration may benefit health-span. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part I)'.
Collapse
Affiliation(s)
- Richard J. Johnson
- Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO 80016, USA
| | - Miguel A. Lanaspa
- Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO 80016, USA
| | - L. Gabriela Sanchez-Lozada
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología ‘Ignacio Chavez’, Mexico City 14080, Mexico
| | - Dean Tolan
- Biology Department, Boston University, Boston, MA 02215, USA
| | - Takahiko Nakagawa
- Department of Nephrology, Rakuwakai-Otowa Hospital, Kyoto 607-8062, Japan
| | - Takuji Ishimoto
- Department of Nephrology and Rheumatology, Aichi Medical University, Aichi 480-1103, Japan
| | - Ana Andres-Hernando
- Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO 80016, USA
| | - Bernardo Rodriguez-Iturbe
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición ‘Salvador Zubirán’, Mexico City 14080, Mexico
| | - Peter Stenvinkel
- Department of Renal Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
| |
Collapse
|
7
|
Cuevas-López B, Romero-Ramirez EI, García-Arroyo FE, Tapia E, León-Contreras JC, Silva-Palacios A, Roldán FJ, Campos ONM, Hernandez-Esquivel L, Marín-Hernández A, Gonzaga-Sánchez JG, Hernández-Pando R, Pedraza-Chaverri J, Sánchez-Lozada LG, Aparicio-Trejo OE. NAC Pre-Administration Prevents Cardiac Mitochondrial Bioenergetics, Dynamics, Biogenesis, and Redox Alteration in Folic Acid-AKI-Induced Cardio-Renal Syndrome Type 3. Antioxidants (Basel) 2023; 12:1592. [PMID: 37627587 PMCID: PMC10451243 DOI: 10.3390/antiox12081592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
The incidence of kidney disease is increasing worldwide. Acute kidney injury (AKI) can strongly favor cardio-renal syndrome (CRS) type 3 development. However, the mechanism involved in CRS development is not entirely understood. In this sense, mitochondrial impairment in both organs has become a central axis in CRS physiopathology. This study aimed to elucidate the molecular mechanisms associated with cardiac mitochondrial impairment and its role in CRS development in the folic acid-induced AKI (FA-AKI) model. Our results showed that 48 h after FA-AKI, the administration of N-acetyl-cysteine (NAC), a mitochondrial glutathione regulator, prevented the early increase in inflammatory and cell death markers and oxidative stress in the heart. This was associated with the ability of NAC to protect heart mitochondrial bioenergetics, principally oxidative phosphorylation (OXPHOS) and membrane potential, through complex I activity and the preservation of glutathione balance, thus preventing mitochondrial dynamics shifting to fission and the decreases in mitochondrial biogenesis and mass. Our data show, for the first time, that mitochondrial bioenergetics impairment plays a critical role in the mechanism that leads to heart damage. Furthermore, NAC heart mitochondrial preservation during an AKI event can be a valuable strategy to prevent CRS type 3 development.
Collapse
Affiliation(s)
- Belén Cuevas-López
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Edgar Ignacio Romero-Ramirez
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Fernando E. García-Arroyo
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Edilia Tapia
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Juan Carlos León-Contreras
- Experimental Pathology Section, National Institute of Medical Sciences and Nutrition “Salvador Zubirán”, Mexico City 14000, Mexico; (J.C.L.-C.); (R.H.-P.)
| | - Alejandro Silva-Palacios
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico;
| | - Francisco-Javier Roldán
- Outpatient Department, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico;
| | - Omar Noel Medina Campos
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City 04510, Mexico; (O.N.M.C.); (J.P.-C.)
| | - Luz Hernandez-Esquivel
- Department of Biochemistry, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (L.H.-E.); (A.M.-H.)
| | - Alvaro Marín-Hernández
- Department of Biochemistry, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (L.H.-E.); (A.M.-H.)
| | - José Guillermo Gonzaga-Sánchez
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Rogelio Hernández-Pando
- Experimental Pathology Section, National Institute of Medical Sciences and Nutrition “Salvador Zubirán”, Mexico City 14000, Mexico; (J.C.L.-C.); (R.H.-P.)
| | - José Pedraza-Chaverri
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City 04510, Mexico; (O.N.M.C.); (J.P.-C.)
| | - Laura Gabriela Sánchez-Lozada
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Omar Emiliano Aparicio-Trejo
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| |
Collapse
|
8
|
García-Arroyo FE, Gonzaga-Sánchez G, Silva-Palacios A, Roldán FJ, Loredo-Mendoza ML, Alvarez-Alvarez YQ, de los Santos Coyotl JA, Vélez Orozco KA, Tapia E, Osorio-Alonso H, Arellano-Buendía AS, Sánchez-Gloria JL, Lanaspa MA, Johnson RJ, Sánchez-Lozada LG. Osthole Prevents Heart Damage Induced by Diet-Induced Metabolic Syndrome: Role of Fructokinase (KHK). Antioxidants (Basel) 2023; 12:1023. [PMID: 37237888 PMCID: PMC10215822 DOI: 10.3390/antiox12051023] [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: 03/16/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
There is increasing evidence that either ingested or produced fructose may have a role in metabolic syndrome. While not commonly considered a criterion for metabolic syndrome, cardiac hypertrophy is often associated with metabolic syndrome, and its presence carries increased cardiovascular risk. Recently it has been shown that fructose and fructokinase C (KHK) can be induced in cardiac tissue. Here we tested whether diet-induced metabolic syndrome causes heart disease associated with increased fructose content and metabolism and whether it can be prevented with a fructokinase inhibitor (osthole). Male Wistar rats were provided a control diet (C) or high fat/sugar diet for 30 days (MS), with half of the latter group receiving osthol (MS+OT, 40 mg/kg/d). The Western diet increased fructose, uric acid, and triglyceride concentrations in cardiac tissue associated with cardiac hypertrophy, local hypoxia, oxidative stress, and increased activity and expression of KHK in cardiac tissue. Osthole reversed these effects. We conclude that the cardiac changes in metabolic syndrome involve increased fructose content and its metabolism and that blocking fructokinase can provide cardiac benefit through the inhibition of KHK with modulation of hypoxia, oxidative stress, hypertrophy, and fibrosis.
Collapse
Affiliation(s)
- Fernando E. García-Arroyo
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Guillermo Gonzaga-Sánchez
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Alejandro Silva-Palacios
- Department of Cardiovascular Biomedicine, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Francisco Javier Roldán
- Department of External Consultation, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - María L. Loredo-Mendoza
- Department of Pathology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | | | - Jesus A. de los Santos Coyotl
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Kevin A. Vélez Orozco
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Edilia Tapia
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Horacio Osorio-Alonso
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Abraham S. Arellano-Buendía
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - José L. Sánchez-Gloria
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Miguel A. Lanaspa
- Renal Diseases and Hypertension, University of Colorado, Aurora, CO 80045, USA
| | - Richard J. Johnson
- Renal Diseases and Hypertension, University of Colorado, Aurora, CO 80045, USA
| | - Laura Gabriela Sánchez-Lozada
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| |
Collapse
|
9
|
Di Stasi LC. Natural Coumarin Derivatives Activating Nrf2 Signaling Pathway as Lead Compounds for the Design and Synthesis of Intestinal Anti-Inflammatory Drugs. Pharmaceuticals (Basel) 2023; 16:ph16040511. [PMID: 37111267 PMCID: PMC10142712 DOI: 10.3390/ph16040511] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Nrf2 (nuclear factor erythroid 2-related factor 2) is a transcription factor related to stress response and cellular homeostasis that plays a key role in maintaining the redox system. The imbalance of the redox system is a triggering factor for the initiation and progression of non-communicable diseases (NCDs), including Inflammatory Bowel Disease (IBD). Nrf2 and its inhibitor Kelch-like ECH-associated protein 1 (Keap1) are the main regulators of oxidative stress and their activation has been recognized as a promising strategy for the treatment or prevention of several acute and chronic diseases. Moreover, activation of Nrf2/keap signaling pathway promotes inhibition of NF-κB, a transcriptional factor related to pro-inflammatory cytokines expression, synchronically promoting an anti-inflammatory response. Several natural coumarins have been reported as potent antioxidant and intestinal anti-inflammatory compounds, acting by different mechanisms, mainly as a modulator of Nrf2/keap signaling pathway. Based on in vivo and in vitro studies, this review focuses on the natural coumarins obtained from both plant products and fermentative processes of food plants by gut microbiota, which activate Nrf2/keap signaling pathway and produce intestinal anti-inflammatory activity. Although gut metabolites urolithin A and urolithin B as well as other plant-derived coumarins display intestinal anti-inflammatory activity modulating Nrf2 signaling pathway, in vitro and in vivo studies are necessary for better pharmacological characterization and evaluation of their potential as lead compounds. Esculetin, 4-methylesculetin, daphnetin, osthole, and imperatorin are the most promising coumarin derivatives as lead compounds for the design and synthesis of Nrf2 activators with intestinal anti-inflammatory activity. However, further structure-activity relationships studies with coumarin derivatives in experimental models of intestinal inflammation and subsequent clinical trials in health and disease volunteers are essential to determine the efficacy and safety in IBD patients.
Collapse
Affiliation(s)
- Luiz C Di Stasi
- Laboratory of Phytomedicines, Pharmacology and Biotechnology (PhytoPharmaTech), Department of Biophysics and Pharmacology, São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil
| |
Collapse
|
10
|
Osthole Inhibits M1 Macrophage Polarization and Attenuates Osteolysis in a Mouse Skull Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:2975193. [PMID: 36686380 PMCID: PMC9851800 DOI: 10.1155/2023/2975193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 01/15/2023]
Abstract
Excessive bone resorption due to increased inflammatory factors is a common feature of inflammatory lytic bone diseases. This group of diseases is effectively treated with drugs. In recent years, many studies have reported that traditional Chinese medicine herbs have substantial effects on inflammation, osteoclast differentiation and maturation, and bone destruction. Herein, we investigated the effects of osthole (OST) on lipopolysaccharide- (LPS-) induced macrophage polarization, inflammatory responses, and osteolysis. In vitro, we used immunofluorescence and quantitative real-time polymerase chain reaction assays to confirm whether bone marrow-derived macrophages showed an increased expression of inflammatory factors, such as interleukin-6, iNOS, CCR7, and CD86, in the presence of LPS. However, we found that such expression was suppressed and that the M2 macrophage expression increased in the presence of OST. OST reduced LPS- and RANKL-induced intracellular reactive oxygen species production in the bone marrow-derived macrophages. Further, it potently suppressed osteoclast differentiation and osteoclast-specific gene expression by suppressing the P38/MAPK and NF-κB pathways. Consistent with the in vitro observations, OST greatly ameliorated LPS-induced bone resorption and modulated the ratio of macrophages at the site of osteolysis. Taken together, OST has great potential for use in the management of osteolytic diseases.
Collapse
|
11
|
Monkfish ( Lophius litulon) Peptides Ameliorate High-Fat-Diet-Induced Nephrotoxicity by Reducing Oxidative Stress and Inflammation via Regulation of Intestinal Flora. Molecules 2022; 28:molecules28010245. [PMID: 36615439 PMCID: PMC9822466 DOI: 10.3390/molecules28010245] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/15/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Renal damage and intestinal flora imbalance due to lipotoxicity are particularly significant in terms of oxidative stress and inflammation, which can be alleviated with bioactive peptides. The monkfish (Lophius litulon) is rich in proteins, which can be used as a source of quality bioactive peptides. This study aimed to examine the protective effect of monkfish peptides on renal injury and their potential role in regulating gut microbiota. METHODS Monkfish meat was hydrolyzed using neutral protease and filtered, and the component with the highest elimination rate of 2,2-diphenyl-1-picrylhydrazyl was named lophius litulon peptides (LPs). Lipid nephrotoxicity was induced via high-fat diet (HFD) feeding for 8 weeks and then treated with LPs. Oxidative stress, inflammatory factors, and intestinal flora were evaluated. RESULTS LP (200 mg/kg) therapy reduced serum creatinine, uric acid, and blood urea nitrogen levels by 49.5%, 31.6%, and 31.6%, respectively. Renal vesicles and tubules were considerably improved with this treatment. Moreover, the activities of superoxide dismutase, glutathione peroxidase, and total antioxidant capacity increased significantly by 198.7%, 167.9%, 61.5%, and 89.4%, respectively. LPs attenuated the upregulation of HFD-induced Toll-like receptor 4 and phospho-nuclear factor-kappa B and increased the protein levels of heme oxygenase 1, nicotinamide quinone oxidoreductase 1, and nuclear factor erythroid 2-related factor 2. The dysbiosis of intestinal microbiota improved after LP treatment. CONCLUSIONS LPs significantly improve antioxidant activity, reduce inflammatory cytokine levels, and regulate intestinal dysbiosis. Thus, LPs are potential compounds that can alleviate HFD-induced renal lipotoxicity.
Collapse
|
12
|
Khalil HE, Abdelwahab MF, Ibrahim HIM, AlYahya KA, Altaweel AA, Alasoom AJ, Burshed HA, Alshawush MM, Waz S. Cichoriin, a Biocoumarin, Mitigates Oxidative Stress and Associated Adverse Dysfunctions on High-Fat Diet-Induced Obesity in Rats. Life (Basel) 2022; 12:1731. [PMID: 36362887 PMCID: PMC9694194 DOI: 10.3390/life12111731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 08/27/2023] Open
Abstract
Metabolic dysfunctions linked to obesity carry the risk of co-morbidities such as diabetes, hepatorenal, and cardiovascular diseases. Coumarins are believed to display several biological effects on diverse adverse health conditions. This study was conducted to uncover the impact of cichoriin on high-fat diet (HFD)-induced obese rats. Methods: Obesity was induced in twenty rats by exposure to an HFD for six weeks. The rats were randomly divided into five groups; group I comprised five healthy rats and was considered the control one. On the other hand, the HFD-induced rats were divided into the following (five per each group): group II (the HFD group), groups III (cichoriin 50 mg/kg) and IV (cichoriin 100 mg/kg) as the treatment groups, and group V received atorvastatin (10 mg/kg) (as a standard). Triglycerides (TG), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), alanine transaminase (ALT), aspartate transaminase (AST), creatine kinase MB (CK-MB), urea, creatinine, the hepatic and renal malondialdehyde (MDA) as well as reduced glutathione (GSH) levels were assessed. Histopathological analysis of the heart, kidney, and liver tissues was investigated. mRNA and protein expressions of the peroxisome proliferator-activated receptor gamma (PPAR-γ) were estimated. Results: The administration of cichoriin alleviated HFD-induced metabolic dysfunctions and improved the histopathological characteristics of the heart, kidney, and liver. Additionally, the treatment improved the lipid profile and hepatic and renal functions, as well as the oxidative balance state. Cichoriin demonstrated an upregulation of the mRNA and protein expressions of PPAR-γ. Taken together, these findings are the first report on the beneficial role of cichoriin in alleviating adverse metabolic effects in HFD-induced obesity and adapting it into an innovative obesity management strategy.
Collapse
Affiliation(s)
- Hany Ezzat Khalil
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Miada F. Abdelwahab
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Hairul-Islam Mohamed Ibrahim
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Pondicherry Centre for Biological Science and Educational Trust, Puducherry 605004, India
| | - Khalid A. AlYahya
- Department of Surgery, College of Medicine, King Faisal University, Al-Ahsa 36363, Saudi Arabia
| | - Abdullah Abdulhamid Altaweel
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Abdullah Jalal Alasoom
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Hussein Ali Burshed
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Marwan Mohamed Alshawush
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Shaimaa Waz
- Department of Biochemistry, Faculty of Pharmacy, Minia University, El-Minia 61511, Egypt
| |
Collapse
|
13
|
Lin L, Tan W, Pan X, Tian E, Wu Z, Yang J. Metabolic Syndrome-Related Kidney Injury: A Review and Update. Front Endocrinol (Lausanne) 2022; 13:904001. [PMID: 35813613 PMCID: PMC9261267 DOI: 10.3389/fendo.2022.904001] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/09/2022] [Indexed: 11/15/2022] Open
Abstract
Metabolic syndrome (MetS) includes visceral obesity, hyperglycemia, dyslipidemia, and hypertension. The prevalence of MetS is 20-25%, which is an important risk factor for chronic kidney disease (CKD). MetS causes effects on renal pathophysiology, including glomerular hyperfiltration, RAAS, microalbuminuria, profibrotic factors and podocyte injury. This review compares several criteria of MetS and analyzes their differences. MetS and the pathogenesis of CKD includes insulin resistance, obesity, dyslipidemia, inflammation, oxidative stress, and endothelial dysfunction. The intervention of MetS-related renal damage is the focus of this article and includes controlling body weight, hypertension, hyperglycemia, and hyperlipidemia, requiring all components to meet the criteria. In addition, interventions such as endoplasmic reticulum stress, oxidative stress, gut microbiota, body metabolism, appetite inhibition, podocyte apoptosis, and mesenchymal stem cells are reviewed.
Collapse
Affiliation(s)
- Lirong Lin
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Wei Tan
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Xianfeng Pan
- Department of Nephrology, Chongqing Kaizhou District People’s Hospital of Chongqing, Chongqing, China
| | - En Tian
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Zhifeng Wu
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Jurong Yang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| |
Collapse
|
14
|
Shiels PG, Painer J, Natterson-Horowitz B, Johnson RJ, Miranda JJ, Stenvinkel P. Manipulating the exposome to enable better ageing. Biochem J 2021; 478:2889-2898. [PMID: 34319404 PMCID: PMC8331090 DOI: 10.1042/bcj20200958] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/03/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023]
Abstract
The sum total of life course exposures creates an exposome that has a significant impact on age-related health. Understanding the interplay between exposome factors and the (epi) genome, offers pertinent insights into the ageing process and its relationship with the accumulation of allostatic load. We propose to exploit this to develop a biomimetic approach that will provide insight into how evolution through natural selection in other species has solved many age related human health issues. In particular, we will emphasise the need to reconnect a more mechanistic approach to medical science with a broader natural sciences approach, using biomimetics to mitigate the global burden of age related ill health. In particular, we will discuss how such an approach indicates leverage of the activities of the Nrf 2 gene to enhance health span via reintroduction of the classical 'Food as Medicine' concept, including modulation of the microbiome and the creation of more salutogenic and biophilic environments. Additionally, we will discuss how this approach integrates with novel and developing senotherapies.
Collapse
Affiliation(s)
- Paul G. Shiels
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, U.K
| | - Johanna Painer
- Research Institute of Wildlife Ecology, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine, Savoyenstreet 1, 1160 Vienna, Austria
| | - Barbara Natterson-Horowitz
- Department of Human Evolutionary Biology, UCLA Division of Cardiology, Co-Director, Evolutionary Medicine Program at UCLA, Harvard University, California, U.S.A
| | - Richard J. Johnson
- Division of Renal Diseases, University of Colorado Anschutz Medical Campus, Aurora, Colorado, U.S.A
| | - Jaime J. Miranda
- CRONICAS Centre of Excellence in Chronic Diseases, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| |
Collapse
|