1
|
Davenport BN, Williams A, Regnault TRH, Jones HN, Wilson RL. Placenta hIGF1 nanoparticle treatment in guinea pigs mitigates FGR-associated fetal sex-dependent effects on liver metabolism-related signaling pathways. Am J Physiol Endocrinol Metab 2025; 328:E395-E409. [PMID: 39907801 DOI: 10.1152/ajpendo.00440.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/03/2024] [Accepted: 01/19/2025] [Indexed: 02/06/2025]
Abstract
Fetal development in an adverse in utero environment significantly increases the risk of developing metabolic diseases in later life, including dyslipidemia, nonalcoholic fatty liver diseases, and diabetes. The aim of this study was to determine whether improving the in utero fetal growth environment with a placental nanoparticle gene therapy would ameliorate fetal growth restriction (FGR)-associated dysregulation of fetal hepatic lipid and glucose metabolism-related signaling pathways. Using the guinea pig maternal nutrient restriction (MNR) model of placental insufficiency and FGR, placenta efficiency and fetal weight were significantly improved following three administrations of a nonviral polymer-based nanoparticle gene therapy to the placenta from mid-pregnancy (gestational day 35) until gestational day 52. The nanoparticle gene therapy transiently increased expression of human insulin-like growth factor 1 (hIGF1) in placenta trophoblast. Fetal liver tissue was collected near-term at gestational day 60. Fetal sex-specific differences in liver gene and protein expression of profibrosis and glucose metabolism-related factors were demonstrated in sham-treated FGR fetuses but not observed in FGR fetuses who received placental hIGF1 nanoparticle treatment. Increased plasma bilirubin, an indirect measure of hepatic activity, was also demonstrated with placental hIGF1 nanoparticle treatment. We speculate that the changes in liver gene and protein expression and increased liver activity that result in similar expression profiles to appropriately growing control fetuses might confer protection against increased susceptibility to aberrant liver physiology in later life. Overall, this work opens avenues for future research assessing the translational prospect of mitigating FGR-induced metabolic derangements.NEW & NOTEWORTHY A placenta-specific nonviral polymer-based nanoparticle gene therapy that improves placenta nutrient transport and near-term fetal weight ameliorates growth restriction-associated changes to fetal liver activity, and cholesterol and glucose/nutrient homeostasis genes/proteins that might confer protection against increased susceptibility to aberrant liver physiology in later life. This knowledge may have implications toward removing predispositions that increase the risk of metabolic diseases, including diabetes, dyslipidemia, and nonalcoholic fatty liver disease in later life.
Collapse
Affiliation(s)
- Baylea N Davenport
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, Florida, United States
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| | - Alyssa Williams
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, Florida, United States
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| | - Timothy R H Regnault
- Departments of Obstetrics and Gynaecology, Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Helen N Jones
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, Florida, United States
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| | - Rebecca L Wilson
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, Florida, United States
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
2
|
Ma F, Longo M, Meroni M, Bhattacharya D, Paolini E, Mughal S, Hussain S, Anand SK, Gupta N, Zhu Y, Navarro-Corcuera A, Li K, Prakash S, Cogliati B, Wang S, Huang X, Wang X, Yurdagul A, Rom O, Wang L, Fried SK, Dongiovanni P, Friedman SL, Cai B. EHBP1 suppresses liver fibrosis in metabolic dysfunction-associated steatohepatitis. Cell Metab 2025:S1550-4131(25)00019-1. [PMID: 40015280 DOI: 10.1016/j.cmet.2025.01.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 11/27/2024] [Accepted: 01/22/2025] [Indexed: 03/01/2025]
Abstract
Excess cholesterol accumulation contributes to fibrogenesis in metabolic dysfunction-associated steatohepatitis (MASH), but how hepatic cholesterol metabolism becomes dysregulated in MASH is not completely understood. We show that human fibrotic MASH livers have decreased EH-domain-binding protein 1 (EHBP1), a genome-wide association study (GWAS) locus associated with low-density lipoprotein (LDL) cholesterol, and that EHBP1 loss- and gain-of-function increase and decrease MASH fibrosis in mice, respectively. Mechanistic studies reveal that EHBP1 promotes sortilin-mediated PCSK9 secretion, leading to LDL receptor (LDLR) degradation, decreased LDL uptake, and reduced TAZ, a fibrogenic effector. At a cellular level, EHBP1 deficiency affects the intracellular localization of retromer, a protein complex required for sortilin stabilization. Our therapeutic approach to stabilizing retromer is effective in mitigating MASH fibrosis. Moreover, we show that the tumor necrosis factor alpha (TNF-α)/peroxisome proliferator-activated receptor alpha (PPARα) pathway suppresses EHBP1 in MASH. These data not only provide mechanistic insights into the role of EHBP1 in cholesterol metabolism and MASH fibrosis but also elucidate an interplay between inflammation and EHBP1-mediated cholesterol metabolism.
Collapse
Affiliation(s)
- Fanglin Ma
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Miriam Longo
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Marica Meroni
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Dipankar Bhattacharya
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Erika Paolini
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Shama Mughal
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Syed Hussain
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sumit Kumar Anand
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Neha Gupta
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yiwei Zhu
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Amaia Navarro-Corcuera
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kenneth Li
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Satya Prakash
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bruno Cogliati
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shuang Wang
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xin Huang
- Columbia Center for Human Development, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Liheng Wang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Susan K Fried
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Scott L Friedman
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
3
|
Wishna-Kadawarage RN, Połtowicz K, Hickey RM, Siwek M. Modulation of gene expression in immune-related organs by in ovo stimulation with probiotics and prophybiotics in broiler chickens. J Appl Genet 2025; 66:195-205. [PMID: 38987456 PMCID: PMC11761985 DOI: 10.1007/s13353-024-00891-y] [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: 05/17/2024] [Revised: 06/20/2024] [Accepted: 06/28/2024] [Indexed: 07/12/2024]
Abstract
In ovo stimulation has been studied intensively as an alternative to antibiotic use in poultry production. We investigated the potential use of a probiotic in combination with a phytobiotic as a prophybiotic for in ovo stimulation and reported its beneficial effects on the gut microbiome of broiler chickens. The current study further investigates the gene expression in the immune-related organs of these chickens to understand the tissue-specific immunomodulatory effects of the treatments. The selected prophybiotic (Leuconostoc mesenteroides with garlic aqueous extract) and its probiotic component alone were injected into ROSS308 chicken eggs on the 12th day of incubation, and gene expression in cecal tonsils, spleen, and liver at 35 days of age was determined using qPCR method. The relative expression of each treatment was compared to the positive control, chickens injected with physiological saline in ovo. The results displayed a downregulation of pro- and anti-inflammatory cytokines in the cecal tonsils of the probiotic group and the liver of the prophybiotic group. The spleen displayed upregulated AVBD1 in both groups and upregulated IL1-β in the probiotic group. The probiotic group displayed increased expression of genes related to metabolism of energy (COX16), protein (mTOR), and lipids (CYP46A1) whereas the prophybiotic group displayed reduced expression of genes related to cholesterol synthesis (SREBP1) and glucose transportation (SLC2A2) in the liver. In conclusion, Leuconostoc mesenteroides differentially modulated gene expression in chickens when administered in ovo in combination with garlic aqueous extract. Further in ovo studies with different prophybiotic combinations are required to optimize the benefits in broiler chickens.
Collapse
Affiliation(s)
- Ramesha N Wishna-Kadawarage
- Department of Animal Biotechnology and Genetics, Faculty of Animal Breeding and Biology, Bydgoszcz University of Science and Technology, Mazowiecka 28, 85-084, Bydgoszcz, Poland.
| | - Katarzyna Połtowicz
- Department of Poultry Breeding, National Research Institute of Animal Production, Krakowska 1, 32-083, Balice, Poland
| | - Rita M Hickey
- Teagasc Food Research Centre, Moorepark, P61 C996, Fermoy, Co. Cork, Ireland
| | - Maria Siwek
- Department of Animal Biotechnology and Genetics, Faculty of Animal Breeding and Biology, Bydgoszcz University of Science and Technology, Mazowiecka 28, 85-084, Bydgoszcz, Poland
| |
Collapse
|
4
|
Kar P, Çiftci G, Çiftci A. Potential benefit of Lactobacillus acidophilus supplementation to rats fed with a high-fat diet on serum lipid profile, kidney amyloid protein and tumor necrosis factor-alpha level. Prostaglandins Other Lipid Mediat 2024; 175:106911. [PMID: 39321864 DOI: 10.1016/j.prostaglandins.2024.106911] [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: 03/01/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
AIM It was aimed to determine the potential effect of Lactobacillus acidophilus supplementation on rats exposed to an experimental high-fat diet on serum lipid profile and kidney total beta amyloid protein (TBAP) and Tumor Necrosis Factor-alpha (TNF-α) levels. METHODS 24 male Sprague-Dawley rats were used in the study to establish 4 groups. Standard rat food (SD) was provided to Group 1 as the control; Group 2 was fed a high-fat diet (HFD); Group 3 consumed SD and received L. acidophilus probiotics; Group 4 was fed HFD and received L. acidophilus probiotics. Body weights were determined weekly during the 12-week trial period. At the end of the experiment, TBP and TNF-α levels in the serum and kidney tissue of the rats were measured by ELISA method. Serum total cholesterol (TC), triglyceride (TG), HDL, LDL, urea and creatinine levels and paraoxanase, amylase and lipase activities were determined by spectrophotometric method on the analyzer device. RESULTS When the control (Group 1) group and Group 2 were compared at the end of the experiment, it was found that Group 2 had gained the most weight and that both the blood and kidney tissue levels of TNF-α and TBAP, as well as the quantities of TG, TK, LDL, and urea, were significantly greater (P<0.05). Serum HDL, PON and amylase levels were found to be significantly low (P>0.05). TG, TK, LDL, urea, and the levels of TNF-α and TBAP in serum and renal tissue were shown to be lower in the groups who received L. acidophilus probiotics (Groups 3, 4) when compared to Group 2 (P>0.05). It was observed that HDL, PON and amylase levels increased and approached the control group (P<0.05). CONCLUSION The study's findings showed that probiotic supplementation improved blood levels of TG, TC, HDL, LDL, urea, PON, and amylase as well as serum and kidney tissue levels of TNF-α and TBAP in obese rats fed a high-fat diet.
Collapse
Affiliation(s)
- Pınar Kar
- Department of Veterinary Biochemistry, Faculty of Veterinary Medicine, University of Ondokuz Mayıs, Samsun, Turkey
| | - Gülay Çiftci
- Department of Veterinary Biochemistry, Faculty of Veterinary Medicine, University of Ondokuz Mayıs, Samsun, Turkey.
| | - Alper Çiftci
- Department of Veterinary Microbiology, Faculty of Veterinary Medicine, University of Ondokuz Mayıs, Samsun, Turkey
| |
Collapse
|
5
|
Antelo-Cea DA, Martínez-Rojas L, Cabrerizo-Ibáñez I, Roudi Rashtabady A, Hernández-Alvarez MI. Regulation of Mitochondrial and Peroxisomal Metabolism in Female Obesity and Type 2 Diabetes. Int J Mol Sci 2024; 25:11237. [PMID: 39457018 PMCID: PMC11508381 DOI: 10.3390/ijms252011237] [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: 10/03/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Obesity and type 2 diabetes (T2D) are widespread metabolic disorders that significantly impact global health today, affecting approximately 17% of adults worldwide with obesity and 9.3% with T2D. Both conditions are closely linked to disruptions in lipid metabolism, where peroxisomes play a pivotal role. Mitochondria and peroxisomes are vital organelles responsible for lipid and energy regulation, including the β-oxidation and oxidation of very long-chain fatty acids (VLCFAs), cholesterol biosynthesis, and bile acid metabolism. These processes are significantly influenced by estrogens, highlighting the interplay between these organelles' function and hormonal regulation in the development and progression of metabolic diseases, such as obesity, metabolic dysfunction-associated fatty liver disease (MAFLD), and T2D. Estrogens modulate lipid metabolism through interactions with nuclear receptors, like peroxisome proliferator-activated receptors (PPARs), which are crucial for maintaining metabolic balance. Estrogen deficiency, such as in postmenopausal women, impairs PPAR regulation, leading to lipid accumulation and increased risk of metabolic disorders. The disruption of peroxisomal-mitochondrial function and estrogen regulation exacerbates lipid imbalances, contributing to insulin resistance and ROS accumulation. This review emphasizes the critical role of these organelles and estrogens in lipid metabolism and their implications for metabolic health, suggesting that therapeutic strategies, including hormone replacement therapy, may offer potential benefits in treating and preventing metabolic diseases.
Collapse
Affiliation(s)
- Damián A. Antelo-Cea
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
| | - Laura Martínez-Rojas
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
| | - Izan Cabrerizo-Ibáñez
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
| | - Ayda Roudi Rashtabady
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
| | - María Isabel Hernández-Alvarez
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
6
|
Seidel F, Vreeken D, Custers E, Wiesmann M, Özsezen S, van Duyvenvoorde W, Caspers M, Menke A, Morrison MC, Verschuren L, Duering M, Hazebroek EJ, Kiliaan AJ, Kleemann R. Metabolic dysfunction-associated steatotic liver disease is associated with effects on cerebral perfusion and white matter integrity. Heliyon 2024; 10:e38516. [PMID: 39391513 PMCID: PMC11466594 DOI: 10.1016/j.heliyon.2024.e38516] [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: 04/04/2024] [Revised: 08/29/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
It is unclear whether early metabolic and inflammatory aberrations in the liver are associated with detrimental changes in brain structure and cognitive function. This cross-sectional study examines putative associations between metabolic dysfunction-associated steatotic liver disease (MASLD) and brain health in 36-55 year-old participants with obesity (n = 70) from the BARICO study (BAriatric surgery Rijnstate and Radboudumc neuroImaging and Cognition in Obesity). The participants underwent brain magnetic resonance imaging to study brain volumes and cortical thickness (3T MRI including T1-weighted magnetization-prepared rapid gradient-echo sequence), cerebral blood perfusion (arterial spin labeling) and white matter integrity (diffusion weighted imaging to assess mean-skeletonized mean diffusivity and fluid-attenuated inversion recovery to detect the presence of white matter hyperintensities (WMH)). The participants additionally performed neuropsychological tests to assess global cognition, working and episodic memory, verbal fluency and the ability to shift attention. Liver biopsies were collected and liver dysfunction was examined with histopathological, biochemical, and gene expression analyses. Linear regression analyses were performed between liver and brain parameters and the influence of body-mass index, diabetes and hypertension was explored. Early stages of liver disease were not associated with cognitive status but with cerebrovascular changes independently of age, sex, BMI, diabetes and hypertension: hepatic fibrosis development was associated with higher spatial coefficient of variation (sCoV) in the nucleus accumbens (NAcc), reflecting greater variations in cerebral perfusion and reduced vascular efficiency. Elevated hepatic levels of free cholesterol and cholesteryl esters were associated with increased WMH, indicating cerebral small vessel disease. RNA-seq and pathway analyses identified associations between sCoV in NAcc and WMH and the expression of hepatic genes involved in inflammation and cellular stress. Additionally, sCoV in NAcc correlated with plasma IL-6 levels suggesting that systemic-low grade inflammation may, at least partly, mediate this relationship. In conclusion, this study demonstrates that specific features of liver dysfunction (e.g. free cholesterol, onset of fibrosis) are associated with subtle cerebrovascular impairments, when changes in cognitive performance are not yet noticeable. These findings highlight the need for future research on therapeutic strategies that normalize metabolic-inflammatory aberrations in the liver to reduce the risk of cognitive decline.
Collapse
Affiliation(s)
- Florine Seidel
- Department Medical Imaging, Anatomy, Radboud Alzheimer Center, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Geert Grooteplein 21N, 6525 EZ Nijmegen, the Netherlands
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| | - Debby Vreeken
- Department Medical Imaging, Anatomy, Radboud Alzheimer Center, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Geert Grooteplein 21N, 6525 EZ Nijmegen, the Netherlands
- Department of Bariatric Surgery, Vitalys, part of Rijnstate hospital, Postbus 9555, 6800 TA Arnhem, the Netherlands
| | - Emma Custers
- Department Medical Imaging, Anatomy, Radboud Alzheimer Center, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Geert Grooteplein 21N, 6525 EZ Nijmegen, the Netherlands
- Department of Bariatric Surgery, Vitalys, part of Rijnstate hospital, Postbus 9555, 6800 TA Arnhem, the Netherlands
| | - Maximilian Wiesmann
- Department Medical Imaging, Anatomy, Radboud Alzheimer Center, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Geert Grooteplein 21N, 6525 EZ Nijmegen, the Netherlands
| | - Serdar Özsezen
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| | - Wim van Duyvenvoorde
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| | - Martien Caspers
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| | - Aswin Menke
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| | - Martine C. Morrison
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| | - Marco Duering
- Institute for Stroke and Dementia Research (ISD), University Hospital LMU Munich, Feodor-Lynen-Straße 17, 81377 Munich, Germany
- Medical Imaging Analysis Center (MIAC) and Department of Biomedical Engineering, University of Basel, Marktgasse 8, CH-4051 Basel, Switzerland
| | - Eric J. Hazebroek
- Department of Bariatric Surgery, Vitalys, part of Rijnstate hospital, Postbus 9555, 6800 TA Arnhem, the Netherlands
- Division of Human Nutrition and Health, Wageningen University, Postbus 17 6700 AA Wageningen Wageningen, the Netherlands
| | - Amanda J. Kiliaan
- Department Medical Imaging, Anatomy, Radboud Alzheimer Center, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Geert Grooteplein 21N, 6525 EZ Nijmegen, the Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| |
Collapse
|
7
|
Yadav KK, Boley PA, Khatiwada S, Lee CM, Bhandari M, Kenney SP. Development of fatty liver disease model using high cholesterol and low choline diet in white leghorn chickens. Vet Res Commun 2024; 48:2489-2497. [PMID: 38861204 PMCID: PMC11315703 DOI: 10.1007/s11259-024-10420-1] [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: 02/21/2024] [Accepted: 05/17/2024] [Indexed: 06/12/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD), which shows similar symptoms as fatty liver hemorrhage syndrome (FLHS) in chickens, is the most common cause of chronic liver disease and cancer in humans. NAFLD patients and FLHS in chickens have demonstrated severe liver disorders when infected by emerging strains of human hepatitis E virus (HEV) and avian HEV, respectively. We sought to develop a fatty liver disease chicken model by altering the diet of 3-week-old white leghorn chickens. The high cholesterol, and low choline (HCLC) diet included 7.6% fat with additional 2% cholesterol and 800 mg/kg choline in comparison to 5.3% fat, and 1,300 mg/kg choline in the regular diet. Our diet induced fatty liver avian model successfully recapitulates the clinical features seen during NAFLD in humans and FLHS in chickens, including hyperlipidemia and hepatic steatosis, as indicated by significantly higher serum triglycerides, serum cholesterol, liver triglycerides, cholesterol, and fatty acids. By developing this chicken model, we expect to provide a platform to explore the role of lipids in the liver pathology linked with viral infections and contribute to the development of prophylactic interventions.
Collapse
Affiliation(s)
- Kush Kumar Yadav
- Center for Food Animal Health (CFAH), Department of Animal Sciences, The Ohio State University, 1680 Madison Ave, Wooster, OH, 44691, USA
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Patricia A Boley
- Center for Food Animal Health (CFAH), Department of Animal Sciences, The Ohio State University, 1680 Madison Ave, Wooster, OH, 44691, USA
| | - Saroj Khatiwada
- Center for Food Animal Health (CFAH), Department of Animal Sciences, The Ohio State University, 1680 Madison Ave, Wooster, OH, 44691, USA
| | - Carolyn M Lee
- Center for Food Animal Health (CFAH), Department of Animal Sciences, The Ohio State University, 1680 Madison Ave, Wooster, OH, 44691, USA
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Menuka Bhandari
- Center for Food Animal Health (CFAH), Department of Animal Sciences, The Ohio State University, 1680 Madison Ave, Wooster, OH, 44691, USA
| | - Scott P Kenney
- Center for Food Animal Health (CFAH), Department of Animal Sciences, The Ohio State University, 1680 Madison Ave, Wooster, OH, 44691, USA.
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
8
|
Burmeister Y, Weyer K, Dörre A, Seilheimer B. The Multicomponent Medicinal Product Hepar Compositum Reduces Hepatic Inflammation and Fibrosis in a Streptozotocin- and High-Fat Diet-Induced Model of Metabolic Dysfunction-Associated Steatotic Liver Disease/Metabolic Dysfunction-Associated Steatohepatitis. Biomedicines 2023; 11:3216. [PMID: 38137437 PMCID: PMC10740479 DOI: 10.3390/biomedicines11123216] [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: 09/18/2023] [Revised: 11/22/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD)-formerly known as non-alcoholic fatty liver disease (NAFLD)-is the most common chronic liver disease worldwide. Since there is currently no approved pharmacotherapy for MASLD, there is an urgent unmet need for efficacious therapeutics for this disease. Hepar compositum (HC-24) is a multicomponent medicinal product that consists of 24 natural ingredients. It has been shown to have anti-inflammatory properties in an obesity-associated MASLD mouse model, but its potential to reduce MASLD-associated fibrosis had not been explored before this study. Here, we investigated the hepatic anti-inflammatory and anti-fibrotic potential of HC-24 in a streptozotocin (STZ)- and high-fat diet (HFD)-induced model of MASLD. Mice received a single injection of low-dose STZ at 2 days of age, followed by HFD feeding from 4 to 9 weeks of age. Mice were treated every second day with HC-24 or daily with the positive control telmisartan from 6 to 9 weeks of age. A non-diseased control group was included as a healthy reference. An explorative small-scale pilot study demonstrated that HC-24 improved liver histology, resulting in a lower NAFLD activity score and reduced liver fibrosis. A subsequent full study confirmed these effects and showed that HC-24 reduced hepatic inflammation, specifically reducing T helper cell and neutrophil influx, and decreased hepatic fibrosis (with qualitatively reduced collagen type I and type III immunopositivity) in the absence of an effect on body and liver weight, blood glucose or liver steatosis. These results show that HC-24 has hepatoprotective, anti-inflammatory, and anti-fibrotic properties in an STZ- and HFD-induced model of MASLD/MASH, suggesting that this multicomponent medicine has therapeutic potential for MASLD patients.
Collapse
Affiliation(s)
| | - Kathrin Weyer
- Heel GmbH, 76532 Baden-Baden, Germany; (Y.B.); (B.S.)
| | - Achim Dörre
- Independent Researcher, 14641 Nauen, Germany;
| | | |
Collapse
|
9
|
Spoelder M, Bright Y, Morrison MC, van Kempen V, de Groodt L, Begalli M, Schuijt N, Kruiger E, Bulthuis R, Gross G, Kleemann R, van Diepen JA, Homberg JR. Cognitive Performance during the Development of Diabetes in the Zucker Diabetic Fatty Rat. Cells 2023; 12:2463. [PMID: 37887307 PMCID: PMC10605915 DOI: 10.3390/cells12202463] [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: 08/25/2023] [Revised: 09/30/2023] [Accepted: 10/05/2023] [Indexed: 10/28/2023] Open
Abstract
Increased insulin levels may support the development of neural circuits involved in cognition, while chronic mild inflammation may also result in cognitive impairment. This study aimed to gain more insight into whether cognition is already impacted during adolescence in a genetic rat model for obesity and type 2 diabetes. Visual discrimination learning throughout adolescence and the level of motivation during early adulthood were investigated in Zucker Diabetic Fatty (ZDF) obese and ZDF lean rats using operant touchscreens. Blood glucose, insulin, and lipids were longitudinally analyzed. Histological analyses were performed in the liver, white adipose tissues, and the prefrontal cortex. Prior to the experiments with the genetic ZDF research model, all experimental assays were performed in two groups of outbred Long Evans rats to investigate the effect of different feeding circumstances. Adolescent ZDF obese rats outperformed ZDF lean rats on visual discrimination performance. During the longitudinal cognitive testing period, insulin levels sharply increased over weeks in ZDF obese rats and were significantly enhanced from 6 weeks of age onwards. Early signs of liver steatosis and enlarged adipocytes in white adipose tissue were observed in early adult ZDF obese rats. Histological analyses in early adulthood showed no group differences in the number of prefrontal cortex neurons and microglia, nor PSD95 and SIRT1 mRNA expression levels. Together, our data show that adolescent ZDF obese rats even display enhanced cognition despite their early diabetic profile.
Collapse
Affiliation(s)
- Marcia Spoelder
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| | - Yami Bright
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| | - Martine C. Morrison
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 CE Leiden, The Netherlands
| | - Veerle van Kempen
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| | - Lilian de Groodt
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| | - Malvina Begalli
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| | - Nikita Schuijt
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| | - Eva Kruiger
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| | - Ronald Bulthuis
- Metris B.V., Kruisweg 829c, 2132 NG Hoofddorp, The Netherlands
| | - Gabriele Gross
- Medical and Scientific Affairs, Reckitt|Mead Johnson Nutrition Institute, Middenkampweg 2, 6545 CJ Nijmegen, The Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 CE Leiden, The Netherlands
| | - Janna A. van Diepen
- Medical and Scientific Affairs, Reckitt|Mead Johnson Nutrition Institute, Middenkampweg 2, 6545 CJ Nijmegen, The Netherlands
| | - Judith R. Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| |
Collapse
|
10
|
Ullah A, Ud Din A, Ding W, Shi Z, Pervaz S, Shen B. A narrative review: CXC chemokines influence immune surveillance in obesity and obesity-related diseases: Type 2 diabetes and nonalcoholic fatty liver disease. Rev Endocr Metab Disord 2023; 24:611-631. [PMID: 37000372 PMCID: PMC10063956 DOI: 10.1007/s11154-023-09800-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2023] [Indexed: 04/01/2023]
Abstract
Adipose tissue develops lipids, aberrant adipokines, chemokines, and pro-inflammatory cytokines as a consequence of the low-grade systemic inflammation that characterizes obesity. This low-grade systemic inflammation can lead to insulin resistance (IR) and metabolic complications, such as type 2 diabetes (T2D) and nonalcoholic fatty liver disease (NAFLD). Although the CXC chemokines consists of numerous regulators of inflammation, cellular function, and cellular migration, it is still unknown that how CXC chemokines and chemokine receptors contribute to the development of metabolic diseases (such as T2D and NAFLD) during obesity. In light of recent research, the objective of this review is to provide an update on the linkage between the CXC chemokine, obesity, and obesity-related metabolic diseases (T2D and NAFLD). We explore the differential migratory and immunomodulatory potential of CXC chemokines and their mechanisms of action to better understand their role in clinical and laboratory contexts. Besides that, because CXC chemokine profiling is strongly linked to leukocyte recruitment, macrophage recruitment, and immunomodulatory potential, we hypothesize that it could be used to predict the therapeutic potential for obesity and obesity-related diseases (T2D and NAFLD).
Collapse
Affiliation(s)
- Amin Ullah
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Xinchuan Road 2222, Chengdu, Sichuan, China.
| | - Ahmad Ud Din
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Xinchuan Road 2222, Chengdu, Sichuan, China
| | - Wen Ding
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Xinchuan Road 2222, Chengdu, Sichuan, China
| | - Zheng Shi
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated hospital, Chengdu University, 610106, Chengdu, China
| | - Sadaf Pervaz
- Joint International Research Laboratory of Reproduction and Development, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Xinchuan Road 2222, Chengdu, Sichuan, China.
| |
Collapse
|
11
|
Gart E, van Duyvenvoorde W, Snabel JM, de Ruiter C, Attema J, Caspers MPM, Lek S, van Heuven BJ, Speksnijder AGCL, Giera M, Menke A, Salic K, Bence KK, Tesz GJ, Keijer J, Kleemann R, Morrison MC. Translational characterization of the temporal dynamics of metabolic dysfunctions in liver, adipose tissue and the gut during diet-induced NASH development in Ldlr-/-.Leiden mice. Heliyon 2023; 9:e13985. [PMID: 36915476 PMCID: PMC10006542 DOI: 10.1016/j.heliyon.2023.e13985] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/17/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Background NAFLD progression, from steatosis to inflammation and fibrosis, results from an interplay of intra- and extrahepatic mechanisms. Disease drivers likely include signals from white adipose tissue (WAT) and gut. However, the temporal dynamics of disease development remain poorly understood. Methods High-fat-diet (HFD)-fed Ldlr-/-.Leiden mice were compared to chow-fed controls. At t = 0, 8, 16, 28 and 38w mice were euthanized, and liver, WAT depots and gut were analyzed biochemically, histologically and by lipidomics and transcriptomics together with circulating factors to investigate the sequence of pathogenic events and organ cross-talk during NAFLD development. Results HFD-induced obesity was associated with an increase in visceral fat, plasma lipids and hyperinsulinemia at t = 8w, along with increased liver steatosis and circulating liver damage biomarkers. In parallel, upstream regulator analysis predicted that lipid catabolism regulators were deactivated and lipid synthesis regulators were activated. Subsequently, hepatocyte hypertrophy, oxidative stress and hepatic inflammation developed. Hepatic collagen accumulated from t = 16 w and became pronounced at t = 28-38 w. Epididymal WAT was maximally hypertrophic from t = 8 w, which coincided with inflammation development. Mesenteric and subcutaneous WAT hypertrophy developed slower and did not appear to reach a maximum, with minimal inflammation. In gut, HFD significantly increased permeability, induced a shift in microbiota composition from t = 8 w and changed circulating gut-derived metabolites. Conclusion HFD-fed Ldlr-/-.Leiden mice develop obesity, dyslipidemia and insulin resistance, essentially as observed in obese NAFLD patients, underlining their translational value. We demonstrate that marked epididymal-WAT inflammation, and gut permeability and dysbiosis precede the development of NAFLD stressing the importance of a multiple-organ approach in the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Eveline Gart
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands.,Human and Animal Physiology, Wageningen University, 6708 WD Wageningen, the Netherlands
| | - Wim van Duyvenvoorde
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| | - Jessica M Snabel
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| | - Christa de Ruiter
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| | - Joline Attema
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| | - Martien P M Caspers
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, the Netherlands
| | - Serene Lek
- Clinnovate Health UK Ltd, Glasgow, United Kingdom
| | | | | | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Aswin Menke
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| | - Kanita Salic
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| | - Kendra K Bence
- Pfizer Worldwide Research, Development & Medical, Internal Medicine Research Unit, Cambridge, MA, USA
| | - Gregory J Tesz
- Pfizer Worldwide Research, Development & Medical, Internal Medicine Research Unit, Cambridge, MA, USA
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, 6708 WD Wageningen, the Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| | - Martine C Morrison
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| |
Collapse
|
12
|
Seidel F, Kleemann R, van Duyvenvoorde W, van Trigt N, Keijzer N, van der Kooij S, van Kooten C, Verschuren L, Menke A, Kiliaan AJ, Winter J, Hughes TR, Morgan BP, Baas F, Fluiter K, Morrison MC. Therapeutic Intervention with Anti-Complement Component 5 Antibody Does Not Reduce NASH but Does Attenuate Atherosclerosis and MIF Concentrations in Ldlr-/-.Leiden Mice. Int J Mol Sci 2022; 23:ijms231810736. [PMID: 36142647 PMCID: PMC9506266 DOI: 10.3390/ijms231810736] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/07/2022] [Accepted: 09/10/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Chronic inflammation is an important driver in the progression of non-alcoholic steatohepatitis (NASH) and atherosclerosis. The complement system, one of the first lines of defense in innate immunity, has been implicated in both diseases. However, the potential therapeutic value of complement inhibition in the ongoing disease remains unclear. Methods: After 20 weeks of high-fat diet (HFD) feeding, obese Ldlr-/-.Leiden mice were treated twice a week with an established anti-C5 antibody (BB5.1) or vehicle control. A separate group of mice was kept on a chow diet as a healthy reference. After 12 weeks of treatment, NASH was analyzed histopathologically, and genome-wide hepatic gene expression was analyzed by next-generation sequencing and pathway analysis. Atherosclerotic lesion area and severity were quantified histopathologically in the aortic roots. Results: Anti-C5 treatment considerably reduced complement system activity in plasma and MAC deposition in the liver but did not affect NASH. Anti-C5 did, however, reduce the development of atherosclerosis, limiting the total lesion size and severity independently of an effect on plasma cholesterol but with reductions in oxidized LDL (oxLDL) and macrophage migration inhibitory factor (MIF). Conclusion: We show, for the first time, that treatment with an anti-C5 antibody in advanced stages of NASH is not sufficient to reduce the disease, while therapeutic intervention against established atherosclerosis is beneficial to limit further progression.
Collapse
Affiliation(s)
- Florine Seidel
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands
- Department Medical Imaging, Anatomy, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, 6525 EZ Nijmegen, The Netherlands
- Correspondence:
| | - Robert Kleemann
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands
| | - Wim van Duyvenvoorde
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands
| | - Nikki van Trigt
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands
| | - Nanda Keijzer
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands
| | - Sandra van der Kooij
- Department of Internal Medicine (Nephrology) and Transplant Center, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Cees van Kooten
- Department of Internal Medicine (Nephrology) and Transplant Center, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), 3704 HE Zeist, The Netherlands
| | - Aswin Menke
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands
| | - Amanda J. Kiliaan
- Department Medical Imaging, Anatomy, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, 6525 EZ Nijmegen, The Netherlands
| | - Johnathan Winter
- Complement Biology Group, Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
- UK Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Timothy R. Hughes
- Complement Biology Group, Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
- UK Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - B. Paul Morgan
- Complement Biology Group, Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
- UK Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Frank Baas
- Department of Clinical Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Kees Fluiter
- Department of Clinical Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Martine C. Morrison
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands
| |
Collapse
|
13
|
Yang M, Liu S, Zhang C. The Related Metabolic Diseases and Treatments of Obesity. Healthcare (Basel) 2022; 10:1616. [PMID: 36141228 PMCID: PMC9498506 DOI: 10.3390/healthcare10091616] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Obesity is a chronic disease characterized by the abnormal or excessive accumulation of body fat, affecting more than 1 billion people worldwide. Obesity is commonly associated with other metabolic disorders, such as type 2 diabetes, non-alcoholic fatty liver disease, cardiovascular diseases, chronic kidney disease, and cancers. Factors such as a sedentary lifestyle, overnutrition, socioeconomic status, and other environmental and genetic conditions can cause obesity. Many molecules and signaling pathways are involved in the pathogenesis of obesity, such as nuclear factor (NF)-κB, Toll-like receptors (TLRs), adhesion molecules, G protein-coupled receptors (GPCRs), programmed cell death 1 (PD-1)/programmed death-ligand 1 (PD-L1), and sirtuin 1 (SIRT1). Commonly used strategies of obesity management and treatment include exercise and dietary change or restriction for the early stage of obesity, bariatric surgery for server obesity, and Food and Drug Administration (FDA)-approved medicines such as semaglutide and liraglutide that can be used as monotherapy or as a synergistic treatment. In addition, psychological management, especially for patients with obesity and distress, is a good option. Gut microbiota plays an important role in obesity and its comorbidities, and gut microbial reprogramming by fecal microbiota transplantation (FMT), probiotics, prebiotics, or synbiotics shows promising potential in obesity and metabolic syndrome. Many clinical trials are ongoing to evaluate the therapeutic effects of different treatments. Currently, prevention and early treatment of obesity are the best options to prevent its progression to many comorbidities.
Collapse
Affiliation(s)
- Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
| | - Shuai Liu
- The First Affiliated Hospital, Zhejiang University, Hangzhou 310006, China
| | - Chunye Zhang
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
14
|
Gart E, van Duyvenvoorde W, Caspers MPM, van Trigt N, Snabel J, Menke A, Keijer J, Salic K, Morrison MC, Kleemann R. Intervention with isoleucine or valine corrects hyperinsulinemia and reduces intrahepatic diacylglycerols, liver steatosis, and inflammation in Ldlr-/-.Leiden mice with manifest obesity-associated NASH. FASEB J 2022; 36:e22435. [PMID: 35830259 DOI: 10.1096/fj.202200111r] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 11/11/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) is associated with a disturbed metabolism in liver, insulin resistance, and excessive accumulation of ectopic fat. Branched-chain amino acids (BCAAs) may beneficially modulate hepatic lipids, however, it remains unclear whether individual BCAAs can attenuate already established NASH and associated oxidative-inflammatory stress. After a 26 weeks run-in on fast food diet (FFD), obese Ldlr-/-.Leiden mice were treated for another 12 weeks with either valine or isoleucine (3% of FFD) and then compared to FFD controls. Valine and isoleucine did not affect obesity, dyslipidemia, gut permeability, or fecal fatty acid excretion, but significantly reduced hyperinsulinemia. Valine and isoleucine reduced ALT, CK18-M30, and liver steatosis with a particularly pronounced suppression of the microvesicular component (-61% by valine and -71% by isoleucine). Both BCAAs decreased intrahepatic diacylglycerols and 4-hydroxynonenal immunoreactivity, a marker for oxidative stress-induced lipid peroxidation. Functional genomics analysis demonstrated that valine and isoleucine affected BCAA metabolism genes, deactivated master regulators of anabolic pathways related to steatosis (e.g., SREBPF1), and activated master regulators of mitochondrial biogenesis (e.g., PPARGC1A) and lipid catabolism (e.g., ACOX1, AMPK). This correction of critical metabolic pathways on gene expression level was accompanied by a significant decrease in histological liver inflammation, and suppression of FFD-stimulated cytokine and chemokine proteins KC/CXCL1, MCP-1/CCL2, and MIP-2/CXCL2 and their pathways. In conclusion, dietary intervention with either valine or isoleucine corrected liver diacylglycerols, gene expression of multiple metabolic processes, and reduced NASH histology with profound hepatoprotective effects on oxidative stress and inflammatory proteins.
Collapse
Affiliation(s)
- Eveline Gart
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands.,Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Wim van Duyvenvoorde
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Martien P M Caspers
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, the Netherlands
| | - Nikki van Trigt
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Jessica Snabel
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Aswin Menke
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Kanita Salic
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Martine C Morrison
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands.,Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands.,Department of Vascular Surgery, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
15
|
Gart E, Salic K, Morrison MC, Giera M, Attema J, de Ruiter C, Caspers M, Schuren F, Bobeldijk-Pastorova I, Heer M, Qin Y, Kleemann R. The Human Milk Oligosaccharide 2′-Fucosyllactose Alleviates Liver Steatosis, ER Stress and Insulin Resistance by Reducing Hepatic Diacylglycerols and Improved Gut Permeability in Obese Ldlr-/-.Leiden Mice. Front Nutr 2022; 9:904740. [PMID: 35782914 PMCID: PMC9248376 DOI: 10.3389/fnut.2022.904740] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/17/2022] [Indexed: 12/19/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a complex multifactorial disorder that is associated with gut dysbiosis, enhanced gut permeability, adiposity and insulin resistance. Prebiotics such as human milk oligosaccharide 2′-fucosyllactose are thought to primarily improve gut health and it is uncertain whether they would affect more distant organs. This study investigates whether 2′-fucosyllactose can alleviate NAFLD development in manifest obesity. Obese hyperinsulinemic Ldlr-/-.Leiden mice, after an 8 week run-in on a high-fat diet (HFD), were treated with 2′-fucosyllactose by oral gavage until week 28 and compared to HFD-vehicle controls. 2′-fucosyllactose did not affect food intake, body weight, total fat mass or plasma lipids. 2′-fucosyllactose altered the fecal microbiota composition which was paralleled by a suppression of HFD-induced gut permeability at t = 12 weeks. 2′-fucosyllactose significantly attenuated the development of NAFLD by reducing microvesicular steatosis. These hepatoprotective effects were supported by upstream regulator analyses showing that 2′-fucosyllactose activated ACOX1 (involved in lipid catabolism), while deactivating SREBF1 (involved in lipogenesis). Furthermore, 2′-fucosyllactose suppressed ATF4, ATF6, ERN1, and NUPR1 all of which participate in endoplasmic reticulum stress. 2′-fucosyllactose reduced fasting insulin concentrations and HOMA-IR, which was corroborated by decreased intrahepatic diacylglycerols. In conclusion, long-term supplementation with 2′-fucosyllactose can counteract the detrimental effects of HFD on gut dysbiosis and gut permeability and attenuates the development of liver steatosis. The observed reduction in intrahepatic diacylglycerols provides a mechanistic rationale for the improvement of hyperinsulinemia and supports the use of 2′-fucosyllactose to correct dysmetabolism and insulin resistance.
Collapse
Affiliation(s)
- Eveline Gart
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, Netherlands
- *Correspondence: Eveline Gart,
| | - Kanita Salic
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Martine C. Morrison
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Joline Attema
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Christa de Ruiter
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Martien Caspers
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Zeist, Netherlands
| | - Frank Schuren
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Zeist, Netherlands
| | - Ivana Bobeldijk-Pastorova
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, Netherlands
| | | | - Yan Qin
- Human Nutrition, BASF Pte Ltd., Singapore, Singapore
| | - Robert Kleemann
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, Netherlands
| |
Collapse
|
16
|
Morrison MC, Gart E, van Duyvenvoorde W, Snabel J, Nielsen MJ, Leeming DJ, Menke A, Kleemann R. Heat-Inactivated Akkermansia muciniphila Improves Gut Permeability but Does Not Prevent Development of Non-Alcoholic Steatohepatitis in Diet-Induced Obese Ldlr-/-.Leiden Mice. Int J Mol Sci 2022; 23:ijms23042325. [PMID: 35216439 PMCID: PMC8878538 DOI: 10.3390/ijms23042325] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/02/2022] [Accepted: 02/13/2022] [Indexed: 02/07/2023] Open
Abstract
The development of non-alcoholic steatohepatitis (NASH) has been associated with alterations in gut microbiota composition and reduced gut barrier function. Akkermansia muciniphila is a gut microbe that is thought to have health-promoting properties, including the ability to improve gut barrier function and host metabolism, both when administered live and after heat-inactivation. We questioned whether heat-inactivated A. muciniphila may reduce NASH development. Ldlr−/−.Leiden mice, a translational, diet-induced model for NASH, were fed a NASH-inducing high-fat diet (HFD) supplemented with heat-inactivated A. muciniphila. After 28 weeks, effects of the treatment on obesity and associated metabolic dysfunction in the gut (microbiota composition and permeability), adipose tissue, and liver were studied relative to an untreated HFD control. Treatment with heat-inactivated A. muciniphila did not affect body weight or adiposity and had no effect on plasma lipids, blood glucose, or plasma insulin. Heat-inactivated A. muciniphila had some minor effects on mucosal microbiota composition in ileum and colon and improved gut barrier function, as assessed by an in vivo functional gut permeability test. Epidydimal white adipose tissue (WAT) hypertrophy and inflammation were not affected, but heat-inactivated A. muciniphila did reduce hypertrophy in the mesenteric WAT which is in close proximity to the intestine. Heat-inactivated A. muciniphila did not affect the development of NASH or associated fibrosis in the liver and did not affect circulating bile acids or markers of liver fibrosis, but did reduce PRO-C4, a type IV collagen synthesis marker, which may be associated with gut integrity. In conclusion, despite beneficial effects in the gut and mesenteric adipose tissue, heat-inactivated A. muciniphila did not affect the development of NASH and fibrosis in a chronic disease setting that mimics clinically relevant disease stages.
Collapse
Affiliation(s)
- Martine C. Morrison
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (E.G.); (W.v.D.); (J.S.); (A.M.); (R.K.)
- Human and Animal Physiology, Wageningen University, 6708 WD Wageningen, The Netherlands
- Correspondence:
| | - Eveline Gart
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (E.G.); (W.v.D.); (J.S.); (A.M.); (R.K.)
- Human and Animal Physiology, Wageningen University, 6708 WD Wageningen, The Netherlands
| | - Wim van Duyvenvoorde
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (E.G.); (W.v.D.); (J.S.); (A.M.); (R.K.)
| | - Jessica Snabel
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (E.G.); (W.v.D.); (J.S.); (A.M.); (R.K.)
| | - Mette Juul Nielsen
- Nordic Bioscience, Biomarkers and Research, 2730 Herlev, Denmark; (M.J.N.); (D.J.L.)
| | - Diana Julie Leeming
- Nordic Bioscience, Biomarkers and Research, 2730 Herlev, Denmark; (M.J.N.); (D.J.L.)
| | - Aswin Menke
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (E.G.); (W.v.D.); (J.S.); (A.M.); (R.K.)
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (E.G.); (W.v.D.); (J.S.); (A.M.); (R.K.)
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
17
|
Arnoldussen IAC, Morrison MC, Wiesmann M, van Diepen JA, Worms N, Voskuilen M, Verweij V, Geenen B, Gualdo NP, van der Logt L, Gross G, Kleemann R, Kiliaan AJ. Milk fat globule membrane attenuates high fat diet-induced neuropathological changes in obese Ldlr-/-.Leiden mice. Int J Obes (Lond) 2022; 46:342-349. [PMID: 34716425 DOI: 10.1038/s41366-021-00998-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/02/2021] [Accepted: 10/12/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Milk-fat globule membrane (MFGM) is a complex structure secreted by the mammary gland and present in mammalian milk. MFGM contains lipids and glycoproteins as well as gangliosides, which may be involved in myelination processes. Notably, myelination and thereby white matter integrity are often altered in obesity. Furthermore, MFGM interventions showed beneficial effects in obesity by affecting inflammatory processes and the microbiome. In this study, we investigated the impact of a dietary MFGM intervention on fat storage, neuroinflammatory processes and myelination in a rodent model of high fat diet (HFD)-induced obesity. METHODS 12-week-old male low density lipoprotein receptor-deficient Leiden mice were exposed to a HFD, a HFD enriched with 3% whey protein lipid concentrate (WPC) high in MFGM components, or a low fat diet. The impact of MFGM supplementation during 24-weeks of HFD-feeding was examined over time by analyzing body weight and fat storage, assessing cognitive tasks and MRI scanning, analyzing myelinization with polarized light imaging and examining neuroinflammation using immunohistochemistry. RESULTS We found in this study that 24 weeks of HFD-feeding induced excessive fat storage, increased systolic blood pressure, altered white matter integrity, decreased functional connectivity, induced neuroinflammation and impaired spatial memory. Notably, supplementation with 3% WPC high in MFGM components restored HFD-induced neuroinflammation and attenuated the reduction in hippocampal-dependent spatial memory and hippocampal functional connectivity. CONCLUSIONS We showed that supplementation with WPC high in MFGM components beneficially contributed to hippocampal-dependent spatial memory, functional connectivity in the hippocampus and anti-inflammatory processes in HFD-induced obesity in rodents. Current knowledge regarding exact biological mechanisms underlying these effects should be addressed in future studies.
Collapse
Affiliation(s)
- Ilse A C Arnoldussen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Preclinical Imaging Center PRIME, Nijmegen, the Netherlands
| | - Martine C Morrison
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands.,Human and Animal Physiology, Wageningen University, Wageningen, the Netherlands
| | - Maximilian Wiesmann
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Preclinical Imaging Center PRIME, Nijmegen, the Netherlands
| | - Janna A van Diepen
- Medical and Scientific Affairs, Reckitt Mead Johnson Nutrition Institute, Nijmegen, the Netherlands
| | - Nicole Worms
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Marijke Voskuilen
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Vivienne Verweij
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Preclinical Imaging Center PRIME, Nijmegen, the Netherlands
| | - Bram Geenen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Preclinical Imaging Center PRIME, Nijmegen, the Netherlands
| | - Natàlia Pujol Gualdo
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Preclinical Imaging Center PRIME, Nijmegen, the Netherlands
| | - Lonneke van der Logt
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Preclinical Imaging Center PRIME, Nijmegen, the Netherlands
| | - Gabriele Gross
- Medical and Scientific Affairs, Reckitt Mead Johnson Nutrition Institute, Nijmegen, the Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands.,Department of Vascular Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Amanda J Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Preclinical Imaging Center PRIME, Nijmegen, the Netherlands.
| |
Collapse
|
18
|
Gart E, van Duyvenvoorde W, Toet K, Caspers MPM, Verschuren L, Nielsen MJ, Leeming DJ, Souto Lima E, Menke A, Hanemaaijer R, Keijer J, Salic K, Kleemann R, Morrison MC. Butyrate Protects against Diet-Induced NASH and Liver Fibrosis and Suppresses Specific Non-Canonical TGF-β Signaling Pathways in Human Hepatic Stellate Cells. Biomedicines 2021; 9:biomedicines9121954. [PMID: 34944770 PMCID: PMC8698820 DOI: 10.3390/biomedicines9121954] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/18/2022] Open
Abstract
In obesity-associated non-alcoholic steatohepatitis (NASH), persistent hepatocellular damage and inflammation are key drivers of fibrosis, which is the main determinant of NASH-associated mortality. The short-chain fatty acid butyrate can exert metabolic improvements and anti-inflammatory activities in NASH. However, its effects on NASH-associated liver fibrosis remain unclear. Putative antifibrotic effects of butyrate were studied in Ldlr-/-.Leiden mice fed an obesogenic diet (HFD) containing 2.5% (w/w) butyrate for 38 weeks and compared with a HFD-control group. Antifibrotic mechanisms of butyrate were further investigated in TGF-β-stimulated primary human hepatic stellate cells (HSC). HFD-fed mice developed obesity, insulin resistance, increased plasma leptin levels, adipose tissue inflammation, gut permeability, dysbiosis, and NASH-associated fibrosis. Butyrate corrected hyperinsulinemia, lowered plasma leptin levels, and attenuated adipose tissue inflammation, without affecting gut permeability or microbiota composition. Butyrate lowered plasma ALT and CK-18M30 levels and attenuated hepatic steatosis and inflammation. Butyrate inhibited fibrosis development as demonstrated by decreased hepatic collagen content and Sirius-red-positive area. In TGF-β-stimulated HSC, butyrate dose-dependently reduced collagen deposition and decreased procollagen1α1 and PAI1 protein expression. Transcriptomic analysis and subsequent pathway and upstream regulator analysis revealed deactivation of specific non-canonical TGF-β signaling pathways Rho-like GTPases and PI3K/AKT and other important pro-fibrotic regulators (e.g., YAP/TAZ, MYC) by butyrate, providing a potential rationale for its antifibrotic effects. In conclusion, butyrate protects against obesity development, insulin resistance-associated NASH, and liver fibrosis. These antifibrotic effects are at least partly attributable to a direct effect of butyrate on collagen production in hepatic stellate cells, involving inhibition of non-canonical TGF-β signaling pathways.
Collapse
Affiliation(s)
- Eveline Gart
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (W.v.D.); (K.T.); (A.M.); (R.H.); (K.S.); (R.K.); (M.C.M.)
- Human and Animal Physiology, Wageningen University, 6708 WD Wageningen, The Netherlands;
- Correspondence:
| | - Wim van Duyvenvoorde
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (W.v.D.); (K.T.); (A.M.); (R.H.); (K.S.); (R.K.); (M.C.M.)
| | - Karin Toet
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (W.v.D.); (K.T.); (A.M.); (R.H.); (K.S.); (R.K.); (M.C.M.)
| | - Martien P. M. Caspers
- Department of Microbiology and Systems Biology, TNO, 3704 HE Zeist, The Netherlands; (M.P.M.C.); (L.V.); (E.S.L.)
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, TNO, 3704 HE Zeist, The Netherlands; (M.P.M.C.); (L.V.); (E.S.L.)
| | - Mette Juul Nielsen
- Nordic Bioscience, Biomarkers and Research, 2730 Herlev, Denmark; (M.J.N.); (D.J.L.)
| | - Diana Julie Leeming
- Nordic Bioscience, Biomarkers and Research, 2730 Herlev, Denmark; (M.J.N.); (D.J.L.)
| | - Everton Souto Lima
- Department of Microbiology and Systems Biology, TNO, 3704 HE Zeist, The Netherlands; (M.P.M.C.); (L.V.); (E.S.L.)
| | - Aswin Menke
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (W.v.D.); (K.T.); (A.M.); (R.H.); (K.S.); (R.K.); (M.C.M.)
| | - Roeland Hanemaaijer
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (W.v.D.); (K.T.); (A.M.); (R.H.); (K.S.); (R.K.); (M.C.M.)
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, 6708 WD Wageningen, The Netherlands;
| | - Kanita Salic
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (W.v.D.); (K.T.); (A.M.); (R.H.); (K.S.); (R.K.); (M.C.M.)
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (W.v.D.); (K.T.); (A.M.); (R.H.); (K.S.); (R.K.); (M.C.M.)
| | - Martine C. Morrison
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (W.v.D.); (K.T.); (A.M.); (R.H.); (K.S.); (R.K.); (M.C.M.)
| |
Collapse
|
19
|
Pieterman EJ, Princen HMG, Jarke A, Nilsson R, Cavallin A, Bergenholm L, Henricsson M, Gopaul VS, Agrawal R, Nissen SE, Hurt-Camejo E. Chronic Oral Administration of Mineral Oil Compared With Corn Oil: Effects on Gut Permeability and Plasma Inflammatory and Lipid Biomarkers. Front Pharmacol 2021; 12:681455. [PMID: 34483899 PMCID: PMC8415260 DOI: 10.3389/fphar.2021.681455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/02/2021] [Indexed: 12/02/2022] Open
Abstract
We investigated the effects of chronic oral administration of mineral oil, versus corn oil as control, on intestinal permeability, inflammatory markers, and plasma lipids in APOE*3-Leiden.CETP mice. Mice received mineral oil or corn oil 15 or 30 μL/mouse/day for 16 weeks (15 mice/group). Intestinal permeability was increased with mineral versus corn oil 30 µL/day, shown by increased mean plasma FITC-dextran concentrations 2 h post-administration (11 weeks: 1.5 versus 1.1 μg/ml, p = 0.02; 15 weeks: 1.7 versus 1.3 μg/ml, p = 0.08). Mean plasma lipopolysaccharide-binding protein levels were raised with mineral versus corn oil 30 µL/day (12 weeks: 5.8 versus 4.4 μg/ml, p = 0.03; 16 weeks: 5.8 versus 4.5 μg/ml, p = 0.09), indicating increased intestinal bacterial endotoxin absorption and potential pro-inflammatory effects. Plasma cholesterol and triglyceride concentrations were decreased with mineral oil, without affecting liver lipids among treated groups. Fecal neutral sterol measurements indicated increased fecal cholesterol excretion with mineral oil 30 µL/day (+16%; p = 0.04). Chronic oral administration of mineral oil in APOE*3-Leiden.CETP mice increased intestinal permeability, with potential pro-inflammatory effects, and decreased plasma cholesterol and triglyceride levels. Our findings may raise concerns about the use of mineral oil as a placebo in clinical studies.
Collapse
Affiliation(s)
- Elsbet J Pieterman
- The Netherlands Organisation for Applied Scientific Research (TNO), Metabolic Health Research, Leiden, Netherlands
| | - Hans M G Princen
- The Netherlands Organisation for Applied Scientific Research (TNO), Metabolic Health Research, Leiden, Netherlands
| | - Annica Jarke
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ralf Nilsson
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anders Cavallin
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Linnéa Bergenholm
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Marcus Henricsson
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - V Sashi Gopaul
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rahul Agrawal
- Global Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Steven E Nissen
- Department of Cardiovascular Medicine and Cleveland Clinic Coordinating Center for Clinical Research, Cleveland Clinic, Cleveland, OH, United States
| | - Eva Hurt-Camejo
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
20
|
Krill Oil Treatment Increases Distinct PUFAs and Oxylipins in Adipose Tissue and Liver and Attenuates Obesity-Associated Inflammation via Direct and Indirect Mechanisms. Nutrients 2021; 13:nu13082836. [PMID: 34444996 PMCID: PMC8401900 DOI: 10.3390/nu13082836] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/31/2022] Open
Abstract
The development of obesity is characterized by the metabolic overload of tissues and subsequent organ inflammation. The health effects of krill oil (KrO) on obesity-associated inflammation remain largely elusive, because long-term treatments with KrO have not been performed to date. Therefore, we examined the putative health effects of 28 weeks of 3% (w/w) KrO supplementation to an obesogenic diet (HFD) with fat derived mostly from lard. The HFD with KrO was compared to an HFD control group to evaluate the effects on fatty acid composition and associated inflammation in epididymal white adipose tissue (eWAT) and the liver during obesity development. KrO treatment increased the concentrations of EPA and DHA and associated oxylipins, including 18-HEPE, RvE2 and 14-HDHA in eWAT and the liver. Simultaneously, KrO decreased arachidonic acid concentrations and arachidonic-acid-derived oxylipins (e.g., HETEs, PGD2, PGE2, PGF2α, TXB2). In eWAT, KrO activated regulators of adipogenesis (e.g., PPARγ, CEBPα, KLF15, STAT5A), induced a shift towards smaller adipocytes and increased the total adipocyte numbers indicative for hyperplasia. KrO reduced crown-like structures in eWAT, and suppressed HFD-stimulated inflammatory pathways including TNFα and CCL2/MCP-1 signaling. The observed eWAT changes were accompanied by reduced plasma leptin and increased plasma adiponectin levels over time, and improved insulin resistance (HOMA-IR). In the liver, KrO suppressed inflammatory signaling pathways, including those controlled by IL-1β and M-CSF, without affecting liver histology. Furthermore, KrO deactivated hepatic REL-A/p65-NF-κB signaling, consistent with increased PPARα protein expression and a trend towards an increase in IkBα. In conclusion, long-term KrO treatment increased several anti-inflammatory PUFAs and oxylipins in WAT and the liver. These changes were accompanied by beneficial effects on general metabolism and inflammatory tone at the tissue level. The stimulation of adipogenesis by KrO allows for safe fat storage and may, together with more direct PPAR-mediated anti-inflammatory mechanisms, attenuate inflammation.
Collapse
|