1
|
Kanuri B, Maremanda KP, Chattopadhyay D, Essop MF, Lee MKS, Murphy AJ, Nagareddy PR. Redefining Macrophage Heterogeneity in Atherosclerosis: A Focus on Possible Therapeutic Implications. Compr Physiol 2025; 15:e70008. [PMID: 40108774 DOI: 10.1002/cph4.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/20/2025] [Accepted: 03/08/2025] [Indexed: 03/22/2025]
Abstract
Atherosclerosis is a lipid disorder where modified lipids (especially oxidized LDL) induce macrophage foam cell formation in the aorta. Its pathogenesis involves a continuum of persistent inflammation accompanied by dysregulated anti-inflammatory responses. Changes in the immune cell status due to differences in the lesional microenvironment are crucial in terms of plaque development, its progression, and plaque rupture. Ly6Chi monocytes generated through both medullary and extramedullary cascades act as one of the major sources of plaque macrophages and thereby foam cells. Both monocytes and monocyte-derived macrophages also participate in pathological events in atherosclerosis-associated multiple organ systems through inter-organ communications. For years, macrophage phenotypes M1 and M2 have been shown to perpetuate inflammatory and resolution responses; nevertheless, such a dualistic classification is too simplistic and contains severe drawbacks. As the lesion microenvironment is enriched with multiple mediators that possess the ability to activate macrophages to diverse phenotypes, it is obvious that such cells should demonstrate substantial heterogeneity. Considerable research in this regard has indicated the presence of additional macrophage phenotypes that are exclusive to atherosclerotic plaques, namely Mox, M4, Mhem, and M(Hb) type. Furthermore, although the concept of macrophage clusters has come to the fore in recent years with the evolution of high-dimensional techniques, classifications based on such 'OMICS' approaches require extensive functional validation as well as metabolic phenotyping. Bearing this in mind, the current review provides an overview of the status of different macrophage populations and their role during atherosclerosis and also outlines possible therapeutic implications.
Collapse
Affiliation(s)
- Babunageswararao Kanuri
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| | - Krishna P Maremanda
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| | - Dipanjan Chattopadhyay
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| | - M Faadiel Essop
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Man Kit Sam Lee
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Andrew J Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Prabhakara R Nagareddy
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| |
Collapse
|
2
|
van Tuijl J, van Heck JI, Bahrar H, Broeders W, Wijma J, ten Have YM, Giera M, Zweers-van Essen H, Rodwell L, Joosten LA, Netea MG, Afman LA, Bekkering S, Riksen NP. Single high-fat challenge and trained innate immunity: A randomized controlled cross-over trial. iScience 2024; 27:111103. [PMID: 39493874 PMCID: PMC11530819 DOI: 10.1016/j.isci.2024.111103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/11/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024] Open
Abstract
Brief exposure of monocytes to atherogenic molecules, such as oxidized lipoproteins, triggers a persistent pro-inflammatory phenotype, named trained immunity. In mice, transient high-fat diet leads to trained immunity, which aggravates atherogenesis. We hypothesized that a single high-fat challenge in humans induces trained immunity. In a randomized controlled cross-over study, 14 healthy individuals received a high-fat or reference shake, and blood was drawn before and after 1, 2, 4, 6, 24, and 72 h. Incubation of donor monocytes with the post-high-fat-shake serum induced trained immunity, regulated via Toll-like receptor 4. This was not mediated via triglyceride-rich lipoproteins, C12, 14, and 16, or metabolic endotoxemia. In vivo, however, the high-fat challenge did not affect monocyte phenotype and function. We conclude that a high-fat challenge leads to alterations in the serum composition that have the potential to induce trained immunity in vitro. However, this does not translate into a (persistent) hyperinflammatory monocyte phenotype in vivo.
Collapse
Affiliation(s)
- Julia van Tuijl
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Julia I.P. van Heck
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Harsh Bahrar
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Wieteke Broeders
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Johan Wijma
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Yvonne M. ten Have
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Heidi Zweers-van Essen
- Department of Gastroenterology and Hepatology-Dietetics, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | - Laura Rodwell
- Section Biostatics, Department of Health Evidence, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | - Leo A.B. Joosten
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Lydia A. Afman
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University and Research, Wageningen 6700 HB, the Netherlands
| | - Siroon Bekkering
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC 3052, Australia
| | - Niels P. Riksen
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| |
Collapse
|
3
|
Qin X, Du J, He R, Li Y, Li H, Liang X. Potential mechanisms and therapeutic strategies for LPS-associated female fertility decline. J Assist Reprod Genet 2024; 41:2739-2758. [PMID: 39167249 PMCID: PMC11534943 DOI: 10.1007/s10815-024-03226-2] [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: 05/08/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Abstract
As a major component of the outer membrane of Gram-negative bacteria, lipopolysaccharide (LPS) can be recognized by toll-like receptors (TLRs) and induce inflammation through MyD88 or the TIR domain-containing adapter-inducing interferon-β (TRIF) pathway. Previous studies have found that LPS-associated inflammatory/immune challenges were associated with ovarian dysfunction and reduced female fertility. However, the etiology and pathogenesis of female fertility decline associated with LPS are currently complex and multifaceted. In this review, PubMed was used to search for references on LPS and fertility decline so as to elucidate the potential mechanisms of LPS-associated female fertility decline and summarize therapeutic strategies that may improve LPS-associated fertility decline.
Collapse
Affiliation(s)
- Xue Qin
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Junhong Du
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Ruifen He
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yaxi Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Hongli Li
- Department of Obstetrics and Gynecology, Key Laboratory for Gynecologic Oncology Gansu Province, The First Hospital of Lanzhou University, No.1, Donggangxi Rd, Chengguan District, Lanzhou, 730000, China
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, Key Laboratory for Gynecologic Oncology Gansu Province, The First Hospital of Lanzhou University, No.1, Donggangxi Rd, Chengguan District, Lanzhou, 730000, China.
| |
Collapse
|
4
|
Lombardo GE, Navarra M, Cremonini E. A flavonoid-rich extract of bergamot juice improves high-fat diet-induced intestinal permeability and associated hepatic damage in mice. Food Funct 2024; 15:9941-9953. [PMID: 39263833 DOI: 10.1039/d4fo02538e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Consumption of high-fat diets (HFDs) is a contributing factor to obesity, insulin resistance and non-alcoholic fatty liver disease (NAFLD). Several studies suggested the protective role of bioactives present in Citrus fruits against the above mentioned chronic metabolic conditions. In this study, we evaluated if a flavonoid-rich extract of Citrus bergamia (bergamot) juice (BJe) could inhibit HFD-induced intestinal permeability and endotoxemia and, through this mechanism, mitigate the associated hepatic damage in C57BL/6J mice. After 12 weeks of the treatment, HFD consumption caused high body weight (BW) gain, hyperinsulinemia, hyperglycemia, and dyslipidemia, which were mitigated by BJe (50 mg per kg BW) supplementation. Furthermore, supplementation with BJe prevented HFD-induced liver alterations, including increased plasma alanine aminotransferase (ALT) activity, increased hepatic lipid deposition, high NAS, and fibrosis. Mice fed a HFD for 12 weeks showed (i) a decrease in small intestine tight junction protein levels (ZO-1, occludin, and claudin-1), (ii) increased intestinal permeability, and (iii) endotoxemia. All these adverse events were mitigated by BJe supplementation. Linking the capacity of BJe to prevent HFD-associated endotoxemia, supplementation with this extract decreased the HFD-induced overexpression of hepatic TLR-4, downstream signaling pathways (MyD88, NF-κB and MAPK), and the associated inflammation, evidenced by increased MCP-1, TNF-α, IL-6, iNOS, and F4/80 levels. Overall, we suggest that BJe could mitigate the harmful consequences of western style diet consumption on liver physiology by protecting the gastrointestinal tract from permeabilization and associated metabolic endotoxemia.
Collapse
Affiliation(s)
- Giovanni E Lombardo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy.
- Prof. Antonio Imbesi Foundation, Messina, Italy
- Department of Medicine and Surgery, "Kore" University of Enna, Enna, Italy
- Department of Nutrition, University of California, Davis, USA.
| | - Michele Navarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy.
| | | |
Collapse
|
5
|
Beckers KF, Flanagan JP, Sones JL. Microbiome and pregnancy: focus on microbial dysbiosis coupled with maternal obesity. Int J Obes (Lond) 2024; 48:439-448. [PMID: 38145995 PMCID: PMC10978494 DOI: 10.1038/s41366-023-01438-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/22/2023] [Accepted: 12/01/2023] [Indexed: 12/27/2023]
Abstract
Obesity is becoming a worldwide pandemic with over one billion people affected. Of women in the United States, who are of childbearing age, two-thirds of them are considered overweight/obese. Offspring of women with obesity have a greater likelihood of developing cardiometabolic disease later in life, therefore making obesity a transgenerational issue. Emerging topics such as maternal microbial dysbiosis with altered levels of bacterial phyla and maternal obesity programming offspring cardiometabolic disease are a novel area of research discussed in this review. In the authors' opinion, beneficial therapeutics will be developed from knowledge of bacterial-host interactions at the most specific level possible. Although there is an abundance of obesity-related microbiome research, it is not concise, readily available, nor easy to interpret at this time. This review details the current knowledge regarding the relationship between obesity and the gut microbiome, with an emphasis on maternal obesity.
Collapse
Affiliation(s)
- Kalie F Beckers
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Juliet P Flanagan
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Jenny L Sones
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA.
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA.
- Clinical Sciences, Colorado State University College of Veterinary Medicine and Biomedical Sciences, Fort Collins, CO, USA.
| |
Collapse
|
6
|
Parvaneh RR, Vajdi M, Shiraz AN, Khani M, Farshbaf SE, Farhangi MA. Prognostic value of circulating macrophage inhibitory cytokine 1-growth differentiation factor 15 (MIC-1/GDF15) in obesity: Relation with vascular endothelial growth factor (VEGF) and markers of oxidative stress. Nutr Health 2023; 29:707-713. [PMID: 35549472 DOI: 10.1177/02601060221099716] [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] [Indexed: 11/16/2022]
Abstract
Background: Macrophage inhibitory cytokine 1-Growth differentiation Factor 15 (MIC-1/GDF15) and vascular endothelial growth factor (VEGF) are novel regulators of obesity and energy homeostasis and food intake. Aims: In the current cross-sectional study, we aimed to evaluate MIC-1 and VEGF concentrations and their association with serum lipids and biomarkers of oxidative stress in obese individuals. Methods: Fifty six obese subjects, aged between 20-50 years old, participated in the current study. Anthropometric and nutritional parameters were assessed and serum and blood concentrations of MIC-1/GDF15, VEGF, markers of oxidative stress and serum lipids were evaluated. Results: Serum VEGF was strongly associated with serum lipids and MIC-1/GDF15 concentrations while serum MIC-1/GDF15 was associated with total cholesterol (TC) and VEGF levels. In multivariate regression analysis, serum VEGF, appetite and GPX were potent determinants of MIC-1/GDF15 concentrations while VEGF was only associated with serum MIC-1/GDF15. Conclusion: The findings of the current study demonstrated the association between MIC-1/GDF15 and VEGF. Moreover, a positive association between these cytokines and serum lipids, was also observed. The results suggest that MIC-1/GDF15 and VEGF might be considered as prognostic markers in obesity-related metabolic disorders. Although further mechanistic studies are needed to better clarify the underlying mechanism.
Collapse
Affiliation(s)
- Roghayeh Rahbar Parvaneh
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahdi Vajdi
- Department of Community Nutrition, Faculty of Nutrition, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ata Nikfam Shiraz
- School of Nursing and Midwifery, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Khani
- Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Ebadpour Farshbaf
- Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
7
|
Omran F, Murphy AM, Younis AZ, Kyrou I, Vrbikova J, Hainer V, Sramkova P, Fried M, Ball G, Tripathi G, Kumar S, McTernan PG, Christian M. The impact of metabolic endotoxaemia on the browning process in human adipocytes. BMC Med 2023; 21:154. [PMID: 37076885 PMCID: PMC10116789 DOI: 10.1186/s12916-023-02857-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 04/03/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND Dysfunctional adipose tissue (AT) is known to contribute to the pathophysiology of metabolic disease, including type 2 diabetes mellitus (T2DM). This dysfunction may occur, in part, as a consequence of gut-derived endotoxaemia inducing changes in adipocyte mitochondrial function and reducing the proportion of BRITE (brown-in-white) adipocytes. Therefore, the present study investigated whether endotoxin (lipopolysaccharide; LPS) directly contributes to impaired human adipocyte mitochondrial function and browning in human adipocytes, and the relevant impact of obesity status pre and post bariatric surgery. METHODS Human differentiated abdominal subcutaneous (AbdSc) adipocytes from participants with obesity and normal-weight participants were treated with endotoxin to assess in vitro changes in mitochondrial function and BRITE phenotype. Ex vivo human AbdSc AT from different groups of participants (normal-weight, obesity, pre- and 6 months post-bariatric surgery) were assessed for similar analyses including circulating endotoxin levels. RESULTS Ex vivo AT analysis (lean & obese, weight loss post-bariatric surgery) identified that systemic endotoxin negatively correlated with BAT gene expression (p < 0.05). In vitro endotoxin treatment of AbdSc adipocytes (lean & obese) reduced mitochondrial dynamics (74.6% reduction; p < 0.0001), biogenesis (81.2% reduction; p < 0.0001) and the BRITE phenotype (93.8% reduction; p < 0.0001). Lean AbdSc adipocytes were more responsive to adrenergic signalling than obese AbdSc adipocytes; although endotoxin mitigated this response (92.6% reduction; p < 0.0001). CONCLUSIONS Taken together, these data suggest that systemic gut-derived endotoxaemia contributes to both individual adipocyte dysfunction and reduced browning capacity of the adipocyte cell population, exacerbating metabolic consequences. As bariatric surgery reduces endotoxin levels and is associated with improving adipocyte functionality, this may provide further evidence regarding the metabolic benefits of such surgical interventions.
Collapse
Affiliation(s)
- Farah Omran
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV2 2DX, UK
| | - Alice M Murphy
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Awais Z Younis
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, Coventry, CV1 5FB, UK
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
- Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | | | | | | | | | - Graham Ball
- Medical Technology Research Centre, Anglia Ruskin University, Cambridge, UK
| | - Gyanendra Tripathi
- Human Sciences Research Centre, College of Life and Natural Sciences, University of Derby, Derby, DE22 1GB, UK
| | - Sudhesh Kumar
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Philip G McTernan
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK.
| | - Mark Christian
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK.
| |
Collapse
|
8
|
Zhang L, Zhang T, Sun J, Huang Y, Liu T, Ye Z, Hu J, Zhang G, Chen H, Ye Z, He Y, Qin J. Calorie restriction ameliorates hyperglycemia, modulates the disordered gut microbiota, and mitigates metabolic endotoxemia and inflammation in type 2 diabetic rats. J Endocrinol Invest 2023; 46:699-711. [PMID: 36219316 DOI: 10.1007/s40618-022-01914-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/29/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE The effects of calorie restriction (CR) on gut microbiota and the mechanism of CR ameliorating hyperglycemia in streptozotocin (STZ)-induced T2DM model rats were explored. METHODS High-fat diet and STZ injection were applied to induce T2DM model rats. Rats were divided into the following three groups: the control-diet ad libitum group, the T2DM model group fed with ad libitum diet, and the T2DM group fed with 30% restriction diet. 16S rRNA sequencing was used to determine the bacterial communities. Lipopolysaccharide (LPS)-binding protein (LBP), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) were measured. RESULTS Glucose tolerance and insulin sensitivity were improved by CR, as well as the levels of fasting and random plasma glucose. Besides, CR not only modulated the overall structure of gut microbiota but also had selective enrichment in anti-inflammatory bacteria such as Lachnospiraceae_NK4A136_group, Ruminococcaceae_9, Allobaculum, Alistipes, and Oscillibacter, and decreased pro-inflammatory pathogenic bacteria such as Bacteroides, Lachnoclostridium, and Bifidobacterium. Tax4Fun indicated that CR could regulate related functional pathways such as lipopolysaccharide biosynthesis, and the plasma levels of LBP, IL-6, and TNF-α were markedly reduced by CR, suggesting the mechanism of CR ameliorating hyperglycemia may associate with the modulation of disordered gut microbiota and the reduction of metabolic endotoxemia and inflammation. CONCLUSION CR could ameliorate hyperglycemia, the mechanism of which may associate with the alteration of the overall structure of gut microbiota, restoration of disordered microbiota function, and the downregulation of metabolic endotoxemia and inflammation in diabetic rats.
Collapse
Affiliation(s)
- L Zhang
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - T Zhang
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - J Sun
- Peking University Shenzhen Hospital, Shenzhen, 518035, China
| | - Y Huang
- The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - T Liu
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Z Ye
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - J Hu
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - G Zhang
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - H Chen
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Z Ye
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Y He
- School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - J Qin
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
9
|
Su S, Zhen M, Zhou C, Cao X, Sun Z, Xu Y, Li L, Jia W, Wu Z, Wang C. Efficiently Inhibiting Systemic Inflammatory Cascades by Fullerenes for Retarding HFD-Fueled Atherosclerosis. Adv Healthc Mater 2023; 12:e2202161. [PMID: 36623263 DOI: 10.1002/adhm.202202161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/31/2022] [Indexed: 01/11/2023]
Abstract
Atherosclerosis accounts for major mortality of cardiac-cerebral vascular diseases worldwide. Pathologically, persistent inflammation dominates the progression of atherosclerosis, which can be accelerated by a high-fat diet (HFD), possibly through triggering local intestinal oxidative stress and ensuing gut barrier dysfunction. Current pharmacotherapy has been disappointing, ascribed to limited therapeutic efficacy and undesirable side effects. Hence it is compelling to explore novel efficient anti-atherosclerotic drugs with minimal toxicity. Herein, two fullerene-based therapies with exceptional antioxidant capacity, in the form of water-soluble injectable fullerene nanoparticles (IFNPs) and oral fullerene tablets (OFTs), are demonstrated to retard HFD-fueled atherosclerosis in ApoE-/- mice with favorable biosafety. Especially, OFTs afford robust anti-atherosclerotic therapeutic even against advanced plaques, besides stabilizing plaques with less lipid deposition and improved collagen expression. Specifically, it is identified that OFTs can ameliorate HFD-induced dysregulated intestinal redox homeostasis and restore gut barrier integrity, thereby restraining the translocation of luminal lipopolysaccharide (LPS) into the bloodstream. Furthermore, significantly reduced circulating LPS after OFTs treatment contributes to down-regulated LPS/TLR4/NF-κB signaling in aortic focal, which further mitigates local inflammation and disease development. Overall, this study confirms the universal anti-atherosclerotic effect of fullerenes and provides a novel therapeutic mechanism via modulating intestinal barrier to attenuate atherosclerosis.
Collapse
Affiliation(s)
- Sheng'e Su
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- Department of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mingming Zhen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- Department of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chen Zhou
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- Department of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinran Cao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- Department of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zihao Sun
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- Department of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuan Xu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- Department of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lei Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- Department of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wang Jia
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- Department of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhanfeng Wu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- Department of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chunru Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- Department of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
10
|
Oteiza PI, Cremonini E, Fraga CG. Anthocyanin actions at the gastrointestinal tract: Relevance to their health benefits. Mol Aspects Med 2023; 89:101156. [PMID: 36379746 DOI: 10.1016/j.mam.2022.101156] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/15/2022]
Abstract
Anthocyanins (AC) are flavonoids abundant in the human diet, which consumption has been associated to several health benefits, including the mitigation of cardiovascular disease, type 2 diabetes, non-alcoholic fatty liver disease, and neurological disorders. It is widely recognized that the gastrointestinal (GI) tract is not only central for food digestion but actively participates in the regulation of whole body physiology. Given that AC, and their metabolites reach high concentrations in the intestinal lumen after food consumption, their biological actions at the GI tract can in part explain their proposed local and systemic health benefits. In terms of mechanisms of action, AC have been found to: i) inhibit GI luminal enzymes that participate in the absorption of lipids and carbohydrates; ii) preserve intestinal barrier integrity and prevent endotoxemia, inflammation and oxidative stress; iii) sustain goblet cell number, immunological functions, and mucus production; iv) promote a healthy microbiota; v) be metabolized by the microbiota to AC metabolites which will be absorbed and have systemic effects; and vi) modulate the metabolism of GI-generated hormones. This review will summarize and discuss the latest information on AC actions at the GI tract and their relationship to overall health benefits.
Collapse
Affiliation(s)
- Patricia I Oteiza
- Department of Nutrition, University of California, Davis, USA; Department of Environmental Toxicology, University of California, Davis, USA.
| | - Eleonora Cremonini
- Department of Nutrition, University of California, Davis, USA; Department of Environmental Toxicology, University of California, Davis, USA
| | - Cesar G Fraga
- Department of Nutrition, University of California, Davis, USA; Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Bioquímica y Medicina Molecular (IBIMOL), UBA-CONICET, Buenos Aires, Argentina
| |
Collapse
|
11
|
Tonelli A, Lumngwena EN, Ntusi NAB. The oral microbiome in the pathophysiology of cardiovascular disease. Nat Rev Cardiol 2023; 20:386-403. [PMID: 36624275 DOI: 10.1038/s41569-022-00825-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/01/2022] [Indexed: 01/11/2023]
Abstract
Despite advances in our understanding of the pathophysiology of many cardiovascular diseases (CVDs) and expansion of available therapies, the global burden of CVD-associated morbidity and mortality remains unacceptably high. Important gaps remain in our understanding of the mechanisms of CVD and determinants of disease progression. In the past decade, much research has been conducted on the human microbiome and its potential role in modulating CVD. With the advent of high-throughput technologies and multiomics analyses, the complex and dynamic relationship between the microbiota, their 'theatre of activity' and the host is gradually being elucidated. The relationship between the gut microbiome and CVD is well established. Much less is known about the role of disruption (dysbiosis) of the oral microbiome; however, interest in the field is growing, as is the body of literature from basic science and animal and human investigations. In this Review, we examine the link between the oral microbiome and CVD, specifically coronary artery disease, stroke, peripheral artery disease, heart failure, infective endocarditis and rheumatic heart disease. We discuss the various mechanisms by which oral dysbiosis contributes to CVD pathogenesis and potential strategies for prevention and treatment.
Collapse
Affiliation(s)
- Andrea Tonelli
- Division of Cardiology, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa.,Cardiovascular Research Unit, Christiaan Barnard Division of Cardiothoracic Surgery, Department of Surgery, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa.,Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Extramural Research Unit on the Intersection of Noncommunicable Diseases and Infectious Disease, South African Medical Research Council, Cape Town, South Africa
| | - Evelyn N Lumngwena
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa.,Centre for the Study of Emerging and Re-emerging Infections, Institute for Medical Research and Medicinal Plant Studies, Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
| | - Ntobeko A B Ntusi
- Division of Cardiology, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa. .,Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa. .,Extramural Research Unit on the Intersection of Noncommunicable Diseases and Infectious Disease, South African Medical Research Council, Cape Town, South Africa. .,Cape Universities Body Imaging Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa. .,Wellcome Centre for Infectious Disease Research, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
12
|
Potruch A, Schwartz A, Ilan Y. The role of bacterial translocation in sepsis: a new target for therapy. Therap Adv Gastroenterol 2022; 15:17562848221094214. [PMID: 35574428 PMCID: PMC9092582 DOI: 10.1177/17562848221094214] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/28/2022] [Indexed: 02/04/2023] Open
Abstract
Sepsis is a leading cause of death in critically ill patients, primarily due to multiple organ failures. It is associated with a systemic inflammatory response that plays a role in the pathogenesis of the disease. Intestinal barrier dysfunction and bacterial translocation (BT) play pivotal roles in the pathogenesis of sepsis and associated organ failure. In this review, we describe recent advances in understanding the mechanisms by which the gut microbiome and BT contribute to the pathogenesis of sepsis. We also discuss several potential treatment modalities that target the microbiome as therapeutic tools for patients with sepsis.
Collapse
|
13
|
Roy P, Orecchioni M, Ley K. How the immune system shapes atherosclerosis: roles of innate and adaptive immunity. Nat Rev Immunol 2022; 22:251-265. [PMID: 34389841 PMCID: PMC10111155 DOI: 10.1038/s41577-021-00584-1] [Citation(s) in RCA: 254] [Impact Index Per Article: 84.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is the root cause of many cardiovascular diseases. Extensive research in preclinical models and emerging evidence in humans have established the crucial roles of the innate and adaptive immune systems in driving atherosclerosis-associated chronic inflammation in arterial blood vessels. New techniques have highlighted the enormous heterogeneity of leukocyte subsets in the arterial wall that have pro-inflammatory or regulatory roles in atherogenesis. Understanding the homing and activation pathways of these immune cells, their disease-associated dynamics and their regulation by microbial and metabolic factors will be crucial for the development of clinical interventions for atherosclerosis, including potentially vaccination-based therapeutic strategies. Here, we review key molecular mechanisms of immune cell activation implicated in modulating atherogenesis and provide an update on the contributions of innate and adaptive immune cell subsets in atherosclerosis.
Collapse
Affiliation(s)
- Payel Roy
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Marco Orecchioni
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
14
|
Keeter WC, Ma S, Stahr N, Moriarty AK, Galkina EV. Atherosclerosis and multi-organ-associated pathologies. Semin Immunopathol 2022; 44:363-374. [PMID: 35238952 PMCID: PMC9069968 DOI: 10.1007/s00281-022-00914-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/13/2022] [Indexed: 12/31/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the vascular system that is characterized by the deposition of modified lipoproteins, accumulation of immune cells, and formation of fibrous tissue within the vessel wall. The disease occurs in vessels throughout the body and affects the functions of almost all organs including the lymphoid system, bone marrow, heart, brain, pancreas, adipose tissue, liver, kidneys, and gastrointestinal tract. Atherosclerosis and associated factors influence these tissues via the modulation of local vascular functions, induction of cholesterol-associated pathologies, and regulation of local immune responses. In this review, we discuss how atherosclerosis interferers with functions of different organs via several common pathways and how the disturbance of immunity in atherosclerosis can result in disease-provoking dysfunctions in multiple tissues. Our growing appreciation of the implication of atherosclerosis and associated microenvironmental conditions in the multi-organ pathology promises to influence our understanding of CVD-associated disease pathologies and to provide new therapeutic opportunities.
Collapse
Affiliation(s)
- W Coles Keeter
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Shelby Ma
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Natalie Stahr
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Alina K Moriarty
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Elena V Galkina
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA.
| |
Collapse
|
15
|
The role of SCFAs to reduce endotoxin and asprosin induced inflammation in human lung epithelial and adipocyte cells. Proc Nutr Soc 2022. [DOI: 10.1017/s0029665122001458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
16
|
Lad N, Murphy A, Parenti C, Nelson C, Williams N, Sharpe G, McTernan P. Asthma and obesity: endotoxin another insult to add to injury? Clin Sci (Lond) 2021; 135:2729-2748. [PMID: 34918742 PMCID: PMC8689194 DOI: 10.1042/cs20210790] [Citation(s) in RCA: 9] [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: 08/05/2021] [Revised: 11/29/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022]
Abstract
Low-grade inflammation is often an underlying cause of several chronic diseases such as asthma, obesity, cardiovascular disease, and type 2 diabetes mellitus (T2DM). Defining the mediators of such chronic low-grade inflammation often appears dependent on which disease is being investigated. However, downstream systemic inflammatory cytokine responses in these diseases often overlap, noting there is no doubt more than one factor at play to heighten the inflammatory response. Furthermore, it is increasingly believed that diet and an altered gut microbiota may play an important role in the pathology of such diverse diseases. More specifically, the inflammatory mediator endotoxin, which is a complex lipopolysaccharide (LPS) derived from the outer membrane cell wall of Gram-negative bacteria and is abundant within the gut microbiota, and may play a direct role alongside inhaled allergens in eliciting an inflammatory response in asthma. Endotoxin has immunogenic effects and is sufficiently microscopic to traverse the gut mucosa and enter the systemic circulation to act as a mediator of chronic low-grade inflammation in disease. Whilst the role of endotoxin has been considered in conditions of obesity, cardiovascular disease and T2DM, endotoxin as an inflammatory trigger in asthma is less well understood. This review has sought to examine the current evidence for the role of endotoxin in asthma, and whether the gut microbiota could be a dietary target to improve disease management. This may expand our understanding of endotoxin as a mediator of further low-grade inflammatory diseases, and how endotoxin may represent yet another insult to add to injury.
Collapse
Affiliation(s)
- Nikita Lad
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Alice M. Murphy
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Cristina Parenti
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Carl P. Nelson
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Neil C. Williams
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Graham R. Sharpe
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Philip G. McTernan
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| |
Collapse
|
17
|
Cabral F, Al-Rahem M, Skaggs J, Thomas TA, Kumar N, Wu Q, Fadda P, Yu L, Robinson JM, Kim J, Pandey E, Sun X, Jarjour WN, Rajaram MV, Harris EN, Ganesan LP. Stabilin receptors clear LPS and control systemic inflammation. iScience 2021; 24:103337. [PMID: 34816100 PMCID: PMC8591421 DOI: 10.1016/j.isci.2021.103337] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/17/2021] [Accepted: 10/20/2021] [Indexed: 01/17/2023] Open
Abstract
Lipopolysaccharides (LPSs) cause lethal endotoxemia if not rapidly cleared from blood circulation. Liver sinusoidal endothelial cells (LSEC) systemically clear LPS by unknown mechanisms. We discovered that LPS clearance through LSEC involves endocytosis and lysosomal inactivation via Stabilin-1 and 2 (Stab1 and Stab2) but does not involve TLR4. Cytokine production was inversely related to clearance/endocytosis of LPS by LSEC. When exposed to LPS, Stabilin double knockout mice (Stab DK) and Stab1 KO, but not Stab2 KO, showed significantly enhanced systemic inflammatory cytokine production and early death compared with WT mice. Stab1 KO is not significantly different from Stab DK in circulatory LPS clearance, LPS uptake and endocytosis by LSEC, and cytokine production. These data indicate that (1) Stab1 receptor primarily facilitates the proactive clearance of LPS and limits TLR4-mediated inflammation and (2) TLR4 and Stab1 are functionally opposing LPS receptors. These findings suggest that endotoxemia can be controlled by optimizing LPS clearance by Stab1.
Collapse
Affiliation(s)
- Fatima Cabral
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, USA
| | - Mustafa Al-Rahem
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - John Skaggs
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Thushara A. Thomas
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Naresh Kumar
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Qian Wu
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Paolo Fadda
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Lianbo Yu
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - John M. Robinson
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Jonghan Kim
- Department of Biomedical & Nutritional Sciences, University of Massachusetts Lowell, MA 01854, USA
| | - Ekta Pandey
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, USA
| | - Xinghui Sun
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, USA
| | - Wael N. Jarjour
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Murugesan V.S. Rajaram
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Edward N. Harris
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, USA
| | - Latha P. Ganesan
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
18
|
Ashok T, Puttam H, Tarnate VCA, Jhaveri S, Avanthika C, Trejo Treviño AG, Sl S, Ahmed NT. Role of Vitamin B12 and Folate in Metabolic Syndrome. Cureus 2021; 13:e18521. [PMID: 34754676 PMCID: PMC8569690 DOI: 10.7759/cureus.18521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 12/11/2022] Open
Abstract
Metabolic syndrome (MS) is a collection of pathological metabolic conditions that includes insulin resistance, central or abdominal obesity, dyslipidemia, and hypertension. It affects large populations worldwide, and its prevalence is rising exponentially. There is no specific mechanism that leads to the development of MS. Proposed hypotheses range from visceral adiposity being a key factor to an increase in very-low-density lipoprotein and fatty acid synthesis as the primary cause of MS. Numerous pharmaceutical therapies are widely available in the market for the treatment of the individual components of MS. The relationship between MS and vitamin B complex supplementation, specifically folic acid and vitamin B12, has been a subject of investigation worldwide, with several trials reporting a positive impact with vitamin supplementation on MS. In this study, an all-language literature search was conducted on Medline, Cochrane, Embase, and Google Scholar till September 2021. The following search strings and Medical Subject Headings (MeSH) terms were used: “Vitamin B12,” “Folate,” “Metabolic Syndrome,” and “Insulin Resistance.” We explored the literature on MS for its epidemiology, pathophysiology, newer treatment options, with a special focus on the effectiveness of supplementation with vitamins B9 and B12. According to the literature, vitamin B12 and folate supplementation, along with a host of novel therapies, has a considerable positive impact on MS. These findings must be kept in mind while designing newer treatment protocols in the future.
Collapse
Affiliation(s)
- Tejaswini Ashok
- Internal Medicine, Jagadguru Sri Shivarathreeshwara Medical College, Mysore, IND
| | - Harivarsha Puttam
- Internal Medicine, Employees' State Insurance Corporation Medical College and Hospital, Hyderabad, IND
| | | | - Sharan Jhaveri
- Internal Medicine, Smt. Nathiba Hargovandas Lakhmichand Municipal Medical College, Ahmedabad, IND
| | - Chaithanya Avanthika
- Medicine and Surgery, Karnataka Institute of Medical Sciences, Hubli, IND.,Pediatrics, Karnataka Institute of Medical Sciences, Hubli, IND
| | | | - Sandeep Sl
- Internal Medicine, SRM Medical College Hospital & Research Centre, Kattankulathur, IND
| | - Nazia T Ahmed
- Medicine, Shahabuddin Medical College and Hospital, Dhaka, BGD
| |
Collapse
|
19
|
Mehmood K, Moin A, Hussain T, Rizvi SMD, Gowda DV, Shakil S, Kamal MA. Can manipulation of gut microbiota really be transformed into an intervention strategy for cardiovascular disease management? Folia Microbiol (Praha) 2021; 66:897-916. [PMID: 34699042 DOI: 10.1007/s12223-021-00926-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 10/03/2021] [Indexed: 02/08/2023]
Abstract
Recent advancement in manipulation techniques of gut microbiota either ex vivo or in situ has broadened its plausible applicability for treating various diseases including cardiovascular disease. Several reports suggested that altering gut microbiota composition is an effective way to deal with issues associated with managing cardiovascular diseases. However, actual translation of gut microbiota manipulation-based techniques into cardiovascular-therapeutic approach is still questionable. This review summarized the evidence on challenges, opportunities, recent development, and future prospects of gut microbiota manipulation for targeting cardiovascular diseases. Initially, issues associated with current cardiovascular diseases treatment strategy, association of gut microbiota with cardiovascular disease, and its influence on cardiovascular drugs were discussed, followed by applicability of gut microbiota manipulation as a cardiovascular disease intervention strategy along with its challenges and future prospects. Despite the fact that the gut microbiota is rugged, interventions like probiotics, prebiotics, synbiotics, fecal microbiota transplantation, fecal virome transplantation, antibiotics, diet changes, and exercises could manipulate it. Advanced techniques like administration of engineered bacteriophages and bacteria could also be employed. Intensive exploration revealed that if sufficiently controlled approach and proper monitoring were applied, gut microbiota could provide a compelling answer for cardiovascular therapy.
Collapse
Affiliation(s)
- Khalid Mehmood
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail, KSA, Saudi Arabia.,Department of Pharmacy, Abbottabad University of Science and Technology, Havelian, Pakistan
| | - Afrasim Moin
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail, KSA, Saudi Arabia
| | - Talib Hussain
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail, KSA, Saudi Arabia
| | - Syed Mohd Danish Rizvi
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail, KSA, Saudi Arabia.
| | - D V Gowda
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, India
| | - Shazi Shakil
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - M A Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Enzymoics 7 Peterlee Place, NSW, 2770, Hebersham, Australia.,Novel Global Community, Educational Foundation, Hebersham, Australia
| |
Collapse
|
20
|
Kao TW, Huang CC. Recent Progress in Metabolic Syndrome Research and Therapeutics. Int J Mol Sci 2021; 22:6862. [PMID: 34202257 PMCID: PMC8269131 DOI: 10.3390/ijms22136862] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
Metabolic syndrome (MetS) is a well-defined yet difficult-to-manage disease entity. Both the precipitous rise in its incidence due to contemporary lifestyles and the growing heterogeneity among affected populations present unprecedented challenges. Moreover, the predisposed risk for developing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in populations with MetS, and the viral impacts on host metabolic parameters, underscores the need to investigate this mechanism thoroughly. Recent investigations of metabolomics and proteomics have revealed not only differentially expressed substances in MetS, but also the consequences of diet consumption and physical activity on energy metabolism. These variations in metabolites, as well as protein products, also influence a wide spectrum of host characteristics, from cellular behavior to phenotype. Research on the dysregulation of gut microbiota and the resultant inflammatory status has also contributed to our understanding of the underlying pathogenic mechanisms. As for state-of-the-art therapies, advancing depictions of the bio-molecular landscape of MetS have emerged and now play a key role in individualized precision medicine. Fecal microbiota transplantation, aiming to restore the host's homeostasis, and targeting of the bile acid signaling pathway are two approaches to combatting MetS. Comprehensive molecular inquiries about MetS by omics measures are mandatory to facilitate the development of novel therapeutic modalities.
Collapse
Affiliation(s)
- Ting-Wei Kao
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan;
| | - Chin-Chou Huang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| |
Collapse
|
21
|
Rapa SF, Di Paola R, Cordaro M, Siracusa R, D’Amico R, Fusco R, Autore G, Cuzzocrea S, Stuppner H, Marzocco S. Plumericin Protects against Experimental Inflammatory Bowel Disease by Restoring Intestinal Barrier Function and Reducing Apoptosis. Biomedicines 2021; 9:biomedicines9010067. [PMID: 33445622 PMCID: PMC7826791 DOI: 10.3390/biomedicines9010067] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 02/07/2023] Open
Abstract
Intestinal epithelial barrier impairment plays a key pathogenic role in inflammatory bowel diseases (IBDs). In particular, together with oxidative stress, intestinal epithelial barrier alteration is considered as upstream event in ulcerative colitis (UC). In order to identify new products of natural origin with a potential activity for UC treatment, this study evaluated the effects of plumericin, a spirolactone iridoid, present as one of the main bioactive components in the bark of Himatanthus sucuuba (Woodson). Plumericin was evaluated for its ability to improve barrier function and to reduce apoptotic parameters during inflammation, both in intestinal epithelial cells (IEC-6), and in an animal experimental model of 2, 4, 6-dinitrobenzene sulfonic acid (DNBS)-induced colitis. Our results indicated that plumericin increased the expression of adhesion molecules, enhanced IEC-6 cells actin cytoskeleton rearrangement, and promoted their motility. Moreover, plumericin reduced apoptotic parameters in IEC-6. These results were confirmed in vivo. Plumericin reduced the activity of myeloperoxidase, inhibited the expression of ICAM-1, P-selectin, and the formation of PAR, and reduced apoptosis parameters in mice colitis induced by DNBS. These results support a pharmacological potential of plumericin in the treatment of UC, due to its ability to improve the structural integrity of the intestinal epithelium and its barrier function.
Collapse
Affiliation(s)
- Shara Francesca Rapa
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (S.F.R.); (G.A.)
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy; (R.D.P.); (R.S.); (R.D.); (R.F.); (S.C.)
| | - Marika Cordaro
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy; (R.D.P.); (R.S.); (R.D.); (R.F.); (S.C.)
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy; (R.D.P.); (R.S.); (R.D.); (R.F.); (S.C.)
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy; (R.D.P.); (R.S.); (R.D.); (R.F.); (S.C.)
| | - Giuseppina Autore
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (S.F.R.); (G.A.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy; (R.D.P.); (R.S.); (R.D.); (R.F.); (S.C.)
| | - Hermann Stuppner
- Institute of Pharmacy/Pharmacognosy, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria;
| | - Stefania Marzocco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (S.F.R.); (G.A.)
- Correspondence: ; Tel.: +89-969159
| |
Collapse
|
22
|
Koszewicz M, Jaroch J, Brzecka A, Ejma M, Budrewicz S, Mikhaleva LM, Muresanu C, Schield P, Somasundaram SG, Kirkland CE, Avila-Rodriguez M, Aliev G. Dysbiosis is one of the risk factor for stroke and cognitive impairment and potential target for treatment. Pharmacol Res 2020; 164:105277. [PMID: 33166735 DOI: 10.1016/j.phrs.2020.105277] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
More than 50 million people have various forms of cognitive impairment basically caused by neurodegenerative diseases, such as Alzheimer's, Parkinson's, and cerebrovascular diseases as well as stroke. Often these conditions coexist and exacerbate one another. The damaged area in post-stroke dementia may lead to neurodegenerative lesions. Gut microbiome functions like an endocrine organ by generating bioactive metabolites that can directly or indirectly impact human physiology. An alteration in the composition and function of intestinal flora, i.e. gut dysbiosis, is implicated in neurodegenerative and cerebrovascular diseases. Additionally, gut dysbiosis may accelerate the progression of cognitive impairment. Dysbiosis may result from obesity; metabolic disorders, cardiovascular disease, and sleep disorders, Lack of physical activity is associated with dysbiosis as well. These may coexist in various patterns in older people, enhancing the risk, incidence, and progression of cerebrovascular lesions, neurodegenerative disorders, and cognitive impairment, creating a vicious circle. Recently, it has been reported that several metabolites produced by gut microbiota (e.g., trimethylamine/trimethylamine N-oxide, short-chain fatty acids, secondary bile acids) may be linked to neurodegenerative and cerebrovascular diseases. New treatment modalities, including prebiotic and probiotics, may normalize the gut microbiota composition, change the brain-gut barrier, and decrease the risk of the pathology development. Fecal microbiota transplantation, sometimes in combination with other methods, is used for remodeling and replenishing the symbiotic gut microbiome. This promising field of research is associated with basic findings of bidirectional communication between body organs and gut microbiota that creates new possibilities of pharmacological treatments of many clinical conditions. The authors present the role of gut microbiota in physiology, and the novel therapeutic targets in modulation of intestinal microbiota Personalized therapies based on their personal genome make up could offer benefits by modulating microbiota cross-talk with brain and cardiovascular system. A healthy lifestyle, including pre and probiotic nutrition is generally recommended. Prevention may also be enhanced by correcting gut dysbiosis resulting a reduced risk of post-stroke cognitive impairment including dementia.
Collapse
Affiliation(s)
- Magdalena Koszewicz
- Department of Neurology, Wroclaw Medical University, 50-556 Wrocław, Borowska 213, Poland
| | - Joanna Jaroch
- Faculty of Health Sciences, Wroclaw Medical University, 51-618 Wrocław, Bartla 5, Poland; Department of Cardiology, Lower Silesian Specialist Hospital, Fieldorfa 2, 54-049 Wroclaw, Poland
| | - Anna Brzecka
- Department of Pulmonology and Lung Oncology, Wroclaw Medical University, 53-439, Wroclaw, Grabiszynska 105, Poland
| | - Maria Ejma
- Department of Neurology, Wroclaw Medical University, 50-556 Wrocław, Borowska 213, Poland
| | - Slawomir Budrewicz
- Department of Neurology, Wroclaw Medical University, 50-556 Wrocław, Borowska 213, Poland
| | - Liudmila M Mikhaleva
- Federal State Budgetary Institution «Research Institute of Human Morphology», 3, Tsyurupy Str., Moscow, 117418, Russian Federation
| | - Cristian Muresanu
- Research Center for Applied Biotechnology in Diagnosis and Molecular Therapies, Str. Trifoiului nr. 12 G, 400478, Cluj-Napoca, Romania
| | - Pamela Schield
- School of Education & Athletics, Salem University, Salem, WV 26426, United States
| | | | - Cecil E Kirkland
- Department of Biological Sciences, Salem University, Salem, WV, USA
| | - Marco Avila-Rodriguez
- Health Sciences Faculty, Clinic Sciences Department, University of Tolima, 730006 Ibague, Colombia
| | - Gjumrakch Aliev
- Federal State Budgetary Institution «Research Institute of Human Morphology», 3, Tsyurupy Str., Moscow, 117418, Russian Federation; I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow, 119991, Russia; Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, 142432, Russia; GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX, 78229, USA.
| |
Collapse
|
23
|
Meleshko TV. EDIBLE FRUITS EXTRACTS AFFECT INTESTINAL MICROBIOTA ISOLATED FROM PATIENTS WITH NONCOMMUNICABLE DISEASES ASSOCIATED WITH CHRONIC INFLAMMATION. BIOTECHNOLOGIA ACTA 2020. [DOI: 10.15407/biotech13.05.087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The aim of our study was to investigate the gut microbiota in patients with noncommunicable diseases associated with chronic inflammation, namely obesity, type 2 diabetes, atherosclerosis, and cardiovascular disease as well as to find out potential ability of edible plants’ fruits extracts to inhibit the growth of selected conditionally pathogenic microorganisms. Limited clinical trial was performed and gut microbiota analysis was done using routine methods and by qPCR. The antibacterial properties of edible plants’ fruits in relation to the selected potentially pathogenic microorganisms were studied. The composition of the intestinal microbiota of obese patients was characterized by an increase in the number of Enterococcus spp. and Lactobacillus spp. along with a decrease in the amount of Escherichia coli. Decreases in E. coli and lactobacilli were observed in patients with type 2 diabetes. In atherosclerosis, an increase in streptococci, enterococci, and enterobacteria was observed, whereas in patients with cardiovascular disease there was an additional increase in staphylococci and candida along with a decrease in E. coli. Decreases in Bifidobacterium spp., Bacteroides spp., Roseburia intestinalis and Akkermansia muciniphila were observed in patients of all groups. The growth of Klebsiella spp. was inhibited by red currant (Ribes rubrum) and plum (Prunus domestica) extracts; Enterobacter spp. – cherry (Prunus avium) extract; Proteus spp. – extracts of blueberry (Vaccinium myrtillus) and dogwood (Cornus mas); Staphylococcus spp. – the extracts of black currant (Ribes nigrum), cherry (Prunus avium), plum (Prunus domestica), jostaberry (Ribes nigrum × Ribes divaricatum × Ribes uva-crispa), cherry plum (Prunus cerasifera) and dogwood (Cornus mas) The obtained data can be used for early diagnosis of noncommunicable diseases and for their prevention with the help of personalized nutrition.
Collapse
|
24
|
Kashtanova DA, Tkacheva ON. The phenomenon of intestinal permeability and its association with cardiovascular disease. Current status. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2020. [DOI: 10.15829/1728-8800-2020-2474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Changes in the intestinal permeability in various pathologies are widely discussed in the scientific community. There is still no consensus on whether high intestinal permeability can lead to chronic noncommunicable diseases, but there is much evidence that increased permeability can aggravate some of them. The article discusses a modern vision of the intestinal permeability including its potential contribution to the development of cardiovascular pathologies, which are the number one mortality cause both in Russia and around the world.
Collapse
Affiliation(s)
- D. A. Kashtanova
- Russian Clinical and Research Center of Gerontology, Pirogov Russian National Research Medical University
| | - O. N. Tkacheva
- Russian Clinical and Research Center of Gerontology, Pirogov Russian National Research Medical University
| |
Collapse
|
25
|
Pearce K, Estanislao D, Fareed S, Tremellen K. Metabolic Endotoxemia, Feeding Studies and the Use of the Limulus Amebocyte (LAL) Assay; Is It Fit for Purpose? Diagnostics (Basel) 2020; 10:diagnostics10060428. [PMID: 32599766 PMCID: PMC7345849 DOI: 10.3390/diagnostics10060428] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022] Open
Abstract
The Limulus amebocyte assay (LAL) is increasingly used to quantify metabolic endotoxemia (ME), particularly in feeding studies. However, the assay was not intended to assess plasma at levels typically seen in ME. We aimed to optimize and validate the LAL assay under a range of pre-treatment conditions against the well-established lipopolysaccharide binding protein assay (LBP). Fifteen healthy overweight and obese males aged 28.8 ± 9.1years provided plasma. The LAL assay employed a range of pre-treatments; 70 °C for 15 and 30 min and 80 °C for 15 and 30 min, ultrasonication (70 °C for 10 min and then 40 °C for 10 min), and dilution (1:50, 1:75, 1:100, and 1:200 parts) or diluted using 0.5% pyrosperse. Seventeen different plasma pre-treatment methods employed prior to the use of the LAL analytical technique failed to show any relationships with either LBP, or body mass index (BMI; obesity), the biological trigger for ME (p > 0.05 for all). As expected, BMI positively correlated with LBP (r = 0.523, p = 0.052. No relationships were observed between LAL with any of the sample pre-treatments and LBP or BMI. In its current form, the LAL assay is unsuitable for detecting levels of endotoxin typically seen in ME.
Collapse
Affiliation(s)
- Karma Pearce
- Division of Health Sciences, School of Pharmacy and Medical Sciences & Alliance for Research in Exercise Nutrition and Activity (ARENA), University of South Australia, Adelaide SA 5001, Australia; (D.E.); (S.F.)
- Correspondence: ; Tel.: +61-8-830-21133 or +61-8-8302-2389
| | - Dianne Estanislao
- Division of Health Sciences, School of Pharmacy and Medical Sciences & Alliance for Research in Exercise Nutrition and Activity (ARENA), University of South Australia, Adelaide SA 5001, Australia; (D.E.); (S.F.)
| | - Sinan Fareed
- Division of Health Sciences, School of Pharmacy and Medical Sciences & Alliance for Research in Exercise Nutrition and Activity (ARENA), University of South Australia, Adelaide SA 5001, Australia; (D.E.); (S.F.)
| | - Kelton Tremellen
- Department of Obstetrics Gynaecology and Reproductive Medicine, Flinders University, Bedford Park SA 5042, Australia;
- Repromed IVF Adelaide, 180 Fullarton Road, Dulwich SA 5065, Australia
| |
Collapse
|
26
|
Nallathambi R, Poulev A, Zuk JB, Raskin I. Proanthocyanidin-Rich Grape Seed Extract Reduces Inflammation and Oxidative Stress and Restores Tight Junction Barrier Function in Caco-2 Colon Cells. Nutrients 2020; 12:nu12061623. [PMID: 32492806 PMCID: PMC7352846 DOI: 10.3390/nu12061623] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 02/07/2023] Open
Abstract
Grape polyphenols have previously been shown to improve gut health and attenuate the symptoms of metabolic syndrome; however, the mechanism of these beneficial effects is still debated. In this study, we investigated the protective effect of proanthocyanidin-rich grape seed extract (GSE) on bacterial lipopolysaccharide (LPS)-induced oxidative stress, inflammation, and barrier integrity of human Caco-2 colon cells. GSE significantly reduced the LPS-induced intracellular reactive oxygen species (ROS) production and mitochondrial superoxide production, and upregulated the expression of antioxidant enzyme genes. GSE also restored the LPS-damaged mitochondrial function by increasing mitochondrial membrane potential. In addition, GSE increased the expression of tight junction proteins in the LPS-treated Caco-2 cells, increased the expression of anti-inflammatory cytokines, and decreased pro-inflammatory cytokine gene expression. Our findings suggest that GSE exerts its beneficial effects on metabolic syndrome by scavenging intestinal ROS, thus reducing oxidative stress, increasing epithelial barrier integrity, and decreasing intestinal inflammation.
Collapse
|
27
|
Iglesias DE, Cremonini E, Fraga CG, Oteiza PI. Ellagic acid protects Caco-2 cell monolayers against inflammation-induced permeabilization. Free Radic Biol Med 2020; 152:776-786. [PMID: 31981623 DOI: 10.1016/j.freeradbiomed.2020.01.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/16/2020] [Accepted: 01/18/2020] [Indexed: 02/07/2023]
Abstract
Chronic intestinal inflammation involves a cycle of oxidative stress, activation of redox sensitive transcription factors, and barrier permeabilization. The latter can lead to systemic inflammation and its associated co-morbidities. Diet can play a major role in the modulation of intestinal inflammation. Among plant bioactives, ellagic acid (EA) was reported to inhibit inflammatory bowel disease in animal models. This work investigated the mechanisms by which EA inhibits tumor necrosis factor alpha (TNFα)-induced inflammation, oxidative stress, and loss of barrier integrity. Caco-2 cells differentiated into an intestinal epithelial cell monolayer were incubated with TNFα (10 ng/ml), in the presence of different EA concentrations. TNFα triggered interleukin (IL) 6 and 8 release into the medium, which was inhibited by EA in a dose-dependent manner (IC50 = 17.3 μM for IL-6). TNFα also led to: i) increased ICAM-1 and NLRP3 expression; ii) loss of epithelial barrier function; iii) increased oxidant production from NOX and mitochondrial origin; iv) NF-κB and ERK1/2 activation; and v) increased MLCK gene expression and MLC phosphorylation. EA (10-40 μM) inhibited all these adverse effects of TNFα. EA mainly acted through NF-κB and ERK1/2 inhibition, breaking the cycle of inflammation, oxidative stress, redox-sensitive pathway (e.g. NF-κB, ERK1/2) activation and intestinal permeabilization. This suggests that consumption of EA, via foods or supplements, may afford a strategy to mitigate intestinal inflammation and its associated co-morbidities.
Collapse
Affiliation(s)
- Dario E Iglesias
- Departments of Nutrition and Environmental Toxicology, University of California, Davis, CA, USA; Physical Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina; IBIMOL, University of Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Eleonora Cremonini
- Departments of Nutrition and Environmental Toxicology, University of California, Davis, CA, USA
| | - Cesar G Fraga
- Physical Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina; IBIMOL, University of Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Patricia I Oteiza
- Departments of Nutrition and Environmental Toxicology, University of California, Davis, CA, USA.
| |
Collapse
|
28
|
Reis DJ, Ilardi SS, Namekata MS, Wing EK, Fowler CH. The depressogenic potential of added dietary sugars. Med Hypotheses 2020; 134:109421. [DOI: 10.1016/j.mehy.2019.109421] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/29/2019] [Accepted: 10/09/2019] [Indexed: 12/12/2022]
|
29
|
Komazin G, Maybin M, Woodard RW, Scior T, Schwudke D, Schombel U, Gisch N, Mamat U, Meredith TC. Substrate structure-activity relationship reveals a limited lipopolysaccharide chemotype range for intestinal alkaline phosphatase. J Biol Chem 2019; 294:19405-19423. [PMID: 31704704 DOI: 10.1074/jbc.ra119.010836] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/31/2019] [Indexed: 12/21/2022] Open
Abstract
Lipopolysaccharide (LPS) from the Gram-negative bacterial outer membrane potently activates the human innate immune system. LPS is recognized by the Toll-like receptor 4/myeloid differentiation factor-2 (TLR4/MD2) complex, leading to the release of pro-inflammatory cytokines. Alkaline phosphatase (AP) is currently being investigated as an anti-inflammatory agent for detoxifying LPS through dephosphorylating lipid A, thus providing a potential treatment for managing both acute (sepsis) and chronic (metabolic endotoxemia) pathologies wherein aberrant TLR4/MD2 activation has been implicated. Endogenous LPS preparations are chemically heterogeneous, and little is known regarding the LPS chemotype substrate range of AP. Here, we investigated the activity of AP on a panel of structurally defined LPS chemotypes isolated from Escherichia coli and demonstrate that calf intestinal AP (cIAP) has only minimal activity against unmodified enteric LPS chemotypes. Pi was only released from a subset of LPS chemotypes harboring spontaneously labile phosphoethanolamine (PEtN) modifications connected through phosphoanhydride bonds. We demonstrate that the spontaneously hydrolyzed O-phosphorylethanolamine is the actual substrate for AP. We found that the 1- and 4'-lipid A phosphate groups critical in TLR4/MD2 signaling become susceptible to hydrolysis only after de-O-acylation of ester linked primary acyl chains on lipid A. Furthermore, PEtN modifications on lipid A specifically enhanced hTLR4 agonist activity of underacylated LPS preparations. Computational binding models are proposed to explain the limitation of AP substrate specificity imposed by the acylation state of lipid A, and the mechanism of PEtN in enhancing hTLR4/MD2 signaling.
Collapse
Affiliation(s)
- Gloria Komazin
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Michael Maybin
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Ronald W Woodard
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Thomas Scior
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72000, Mexico
| | - Dominik Schwudke
- Bioanalytical Chemistry, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Ursula Schombel
- Bioanalytical Chemistry, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Nicolas Gisch
- Bioanalytical Chemistry, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Uwe Mamat
- Cellular Microbiology, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Timothy C Meredith
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802
| |
Collapse
|
30
|
Fang TJ, Guo JT, Lin MK, Lee MS, Chen YL, Lin WH. Protective effects of Lactobacillus plantarum against chronic alcohol-induced liver injury in the murine model. Appl Microbiol Biotechnol 2019; 103:8597-8608. [PMID: 31515596 DOI: 10.1007/s00253-019-10122-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/26/2019] [Accepted: 09/04/2019] [Indexed: 02/07/2023]
Abstract
Long-term alcohol consumption causes liver injuries such as alcoholic hepatitis, fatty liver, and endotoxemia. Some probiotics were demonstrated to exert beneficial effects in the gastrointestinal tract. The present study was aimed to evaluate the protective effects of Lactobacillus plantarum CMU995 against alcohol-induced liver injury. The mice were orally administered L. plantarum CMU995 for 1 week, followed by the administration of alcohol and different tested substances daily for 6 weeks. The liver injury was examined by measuring the levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), malondialdehyde (MDA), anti-oxidative enzyme, endotoxin, inflammatory cytokines, and lipid accumulation in the liver or serum among different groups. L. plantarum CMU995 exhibited beneficial effects on alcohol-induced liver injury via reduction in the serum concentration of AST, ALT, cholesterol, triglycerides, endotoxin, TNF-α, IL-1β, and oxidative stress. Furthermore, we also found that the levels of glutathione (GSH), superoxide dismutase (SOD), and intestinal tight junction protein zonula occludens-1 (ZO-1) were considerably higher in L. plantarum CMU995-fed groups when compared with placebo group. Meanwhile, the protective effects were demonstrated biological gradients as controversial dose-dependent. We speculate that L. plantarum CMU995 inhibited the migration of alcohol-derived endotoxin into the blood and liver, thereby improving the intestinal barrier. The present evidence may provide a novel microbiota-based strategy to prevent the alcohol-induced liver injury.
Collapse
Affiliation(s)
- Tony J Fang
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Jiun-Ting Guo
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan, Republic of China.,Department of Pharmacy, China Medical University, Taichung, Taiwan, Republic of China
| | - Ming-Kuem Lin
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Pharmacy, China Medical University, Taichung, Taiwan, Republic of China
| | - Meng-Shiou Lee
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Pharmacy, China Medical University, Taichung, Taiwan, Republic of China
| | - Yen-Lien Chen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Wen-Hsin Lin
- Department of Pharmacy, China Medical University, Taichung, Taiwan, Republic of China. .,College of Pharmacy, China Medical University, No. 91, Hsueh Shih Road, Taichung, 404, Taiwan, Republic of China.
| |
Collapse
|
31
|
Shariat A, Abbasalizad Farhangi M, Zeinalian R. Spirulina platensis supplementation, macrophage inhibitory cytokine-1 (MIC-1), oxidative stress markers and anthropometric features in obese individuals: A randomized controlled trial. J Herb Med 2019. [DOI: 10.1016/j.hermed.2019.100264] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
32
|
Connection between gut microbiome and the development of obesity. Eur J Clin Microbiol Infect Dis 2019; 38:1987-1998. [PMID: 31367997 DOI: 10.1007/s10096-019-03623-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 06/26/2019] [Indexed: 02/08/2023]
Abstract
The potential role of the gut microbiota in various human diseases has attracted considerable attention worldwide. Here, we discuss the vital role of the intestinal microbiota in the development of obesity. First, we describe how the gut microbiota promotes fat accumulation. Additionally, a high-fat diet leads to structural instability among in the gut microbiota, further leading to an increase in endotoxins, which aggravates obesity. We then discuss how gut microbiota metabolites, including short-chain fatty acids and lipopolysaccharides, affect the host. Finally, we review several strategies for regulating the intestinal flora.
Collapse
|
33
|
Cremonini E, Daveri E, Mastaloudis A, Adamo AM, Mills D, Kalanetra K, Hester SN, Wood SM, Fraga CG, Oteiza PI. Anthocyanins protect the gastrointestinal tract from high fat diet-induced alterations in redox signaling, barrier integrity and dysbiosis. Redox Biol 2019; 26:101269. [PMID: 31330482 PMCID: PMC6646927 DOI: 10.1016/j.redox.2019.101269] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 07/01/2019] [Accepted: 07/03/2019] [Indexed: 02/07/2023] Open
Abstract
The gastrointestinal (GI) tract can play a critical role in the development of pathologies associated with overeating, overweight and obesity. We previously observed that supplementation with anthocyanins (AC) (particularly glycosides of cyanidin and delphinidin) mitigated high fat diet (HFD)-induced development of obesity, dyslipidemia, insulin resistance and steatosis in C57BL/6J mice. This paper investigated whether these beneficial effects could be related to AC capacity to sustain intestinal monolayer integrity, prevent endotoxemia, and HFD-associated dysbiosis. The involvement of redox-related mechanisms were further investigated in Caco-2 cell monolayers. Consumption of a HFD for 14 weeks caused intestinal permeabilization and endotoxemia, which were associated with a decreased ileum expression of tight junction (TJ) proteins (occludin, ZO-1 and claudin-1), increased expression of NADPH oxidase (NOX1 and NOX4) and NOS2 and oxidative stress, and activation of redox sensitive signals (NF-κB and ERK1/2) that regulate TJ dynamics. AC supplementation mitigated all these events and increased GLP-2 levels, the intestinal hormone that upregulates TJ protein expression. AC also prevented, in vitro, tumor necrosis factor alpha-induced Caco-2 monolayer permeabilization, NOX1/4 upregulation, oxidative stress, and NF-κB and ERK activation. HFD-induced obesity in mice caused dysbiosis and affected the levels and secretion of MUC2, a mucin that participates in intestinal cell barrier protection and immune response. AC supplementation restored microbiota composition and MUC2 levels and distribution in HFD-fed mice. Thus, AC, particularly delphinidin and cyanidin, can preserve GI physiology in HFD-induced obesity in part through redox-regulated mechanisms. This can in part explain AC capacity to mitigate pathologies, i.e. insulin resistance and steatosis, associated with HFD-associated obesity. Anthocyanidins (AC) mitigate high fat diet (HFD)-induced intestinal permeabilization and endotoxemia. AC inhibit HFD-induced ileum NADPH oxidase upregulation and oxidative stress. AC inhibit the activation of redox sensitive signals that cause intestinal permeabilization. AC mitigate HFD-induced dysbiosis and improve ileum endocrine/immunological responses. AC's beneficial systemic effects in HFD-mice could begin at the GI tract.
Collapse
Affiliation(s)
- Eleonora Cremonini
- Departments of Nutrition, University of California, Davis, CA, USA; Environmental Toxicology, University of California, Davis, CA, USA
| | - Elena Daveri
- Departments of Nutrition, University of California, Davis, CA, USA; Environmental Toxicology, University of California, Davis, CA, USA
| | | | - Ana M Adamo
- Quimica Biológica Patológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química y Fisicoquímica Biológicas (IQUIFYB), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - David Mills
- Food Science and Technology, University of California, Davis, CA, USA; Viticulture and Enology, University of California, Davis, CA, USA
| | - Karen Kalanetra
- Food Science and Technology, University of California, Davis, CA, USA; Viticulture and Enology, University of California, Davis, CA, USA
| | | | - Steve M Wood
- Pharmanex Research, NSE Products, Inc., Provo, UT, USA
| | - Cesar G Fraga
- Departments of Nutrition, University of California, Davis, CA, USA; Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Bioquímica y Medicina Molecular (IBIMOL), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Patricia I Oteiza
- Departments of Nutrition, University of California, Davis, CA, USA; Environmental Toxicology, University of California, Davis, CA, USA.
| |
Collapse
|
34
|
Wensveen FM, Šestan M, Turk Wensveen T, Polić B. 'Beauty and the beast' in infection: How immune-endocrine interactions regulate systemic metabolism in the context of infection. Eur J Immunol 2019; 49:982-995. [PMID: 31106860 DOI: 10.1002/eji.201847895] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/28/2019] [Accepted: 05/17/2019] [Indexed: 02/07/2023]
Abstract
The immune and endocrine systems ensure two vital functions in the body. The immune system protects us from lethal pathogens, whereas the endocrine system ensures proper metabolic function of peripheral organs by regulating systemic homeostasis. These two systems were long thought to operate independently. The immune system uses cytokines and immune receptors, whereas the endocrine system uses hormones to regulate metabolism. However, recent findings show that the immune and endocrine systems closely interact, especially regarding regulation of glucose metabolism. In response to pathogen encounter, cytokines modify responsiveness of peripheral organs to endocrine signals, resulting in altered levels of blood hormones such as insulin, which promotes the ability of the body to fight infection. Here we provide an overview of recent literature describing various mechanisms, which the immune system utilizes to modify endocrine regulation of systemic metabolism. Moreover, we will describe how these immune-endocrine interactions derail in the context of obesity. From a clinical perspective we will elaborate how infection and obesity aggravate the development of metabolic diseases such as diabetes mellitus type 2 in humans. In summary, this review provides a comprehensive overview of immune-induced changes in systemic metabolism following infection, with a focus on regulation of glucose metabolism.
Collapse
Affiliation(s)
- Felix M Wensveen
- Department of Histology and Embryology, University of Rijeka School of Medicine, Rijeka, Croatia
| | - Marko Šestan
- Department of Histology and Embryology, University of Rijeka School of Medicine, Rijeka, Croatia
| | - Tamara Turk Wensveen
- Department of Endocrinology, Diabetes and Metabolic Diseases, Clinical hospital center Rijeka, Rijeka, Croatia
| | - Bojan Polić
- Department of Histology and Embryology, University of Rijeka School of Medicine, Rijeka, Croatia
| |
Collapse
|
35
|
Lew KN, Starkweather A, Cong X, Judge M. A Mechanistic Model of Gut-Brain Axis Perturbation and High-Fat Diet Pathways to Gut Microbiome Homeostatic Disruption, Systemic Inflammation, and Type 2 Diabetes. Biol Res Nurs 2019; 21:384-399. [PMID: 31113222 DOI: 10.1177/1099800419849109] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Type 2 diabetes (T2D) is a highly prevalent metabolic disease, affecting nearly 10% of the American population. Although the etiopathogenesis of T2D remains poorly understood, advances in DNA sequencing technologies have allowed for sophisticated interrogation of the human microbiome, providing insight into the role of the gut microbiome in the development and progression of T2D. An emerging body of research reveals that gut-brain axis (GBA) perturbations and a high-fat diet (HFD), along with other modifiable and nonmodifiable risk factors, contribute to gut microbiome homeostatic imbalance. Homeostatic imbalance or disruption increases gut wall permeability and facilitates translocation of endotoxins (lipopolysaccharides) into the circulation with resultant systemic inflammation. Chronic, low-grade systemic inflammation ensues with pro-inflammatory pathways activated, contributing to obesity, insulin resistance (IR), pancreatic β-cell decline, and, thereby, T2D. While GBA perturbations and HFD are implicated in provoking these conditions, prior mechanistic models have tended to examine HFD and GBA pathways exclusively without considering their shared pathways to T2D. Addressing this gap, this article proposes a mechanistic model informed by animal and human studies to advance scientific understanding of (1) modifiable and nonmodifiable risk factors for gut microbiome homeostatic disruption, (2) HFD and GBA pathways contributing to homeostatic disruption, and (3) shared GBA and HFD pro-inflammatory pathways to obesity, IR, β-cell decline, and T2D. The proposed mechanistic model, based on the extant literature, proposes a framework for studying the complex relationships of the gut microbiome to T2D to advance study in this promising area of research.
Collapse
Affiliation(s)
| | | | - Xiaomei Cong
- 1 School of Nursing, University of Connecticut, Storrs, CT, USA
| | - Michelle Judge
- 1 School of Nursing, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
36
|
Tang ZZ, Chen G, Hong Q, Huang S, Smith HM, Shah RD, Scholz M, Ferguson JF. Multi-Omic Analysis of the Microbiome and Metabolome in Healthy Subjects Reveals Microbiome-Dependent Relationships Between Diet and Metabolites. Front Genet 2019; 10:454. [PMID: 31164901 PMCID: PMC6534069 DOI: 10.3389/fgene.2019.00454] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/30/2019] [Indexed: 12/22/2022] Open
Abstract
The human microbiome has been associated with health status, and risk of disease development. While the etiology of microbiome-mediated disease remains to be fully elucidated, one mechanism may be through microbial metabolism. Metabolites produced by commensal organisms, including in response to host diet, may affect host metabolic processes, with potentially protective or pathogenic consequences. We conducted multi-omic phenotyping of healthy subjects (N = 136), in order to investigate the interaction between diet, the microbiome, and the metabolome in a cross-sectional sample. We analyzed the nutrient composition of self-reported diet (3-day food records and food frequency questionnaires). We profiled the gut and oral microbiome (16S rRNA) from stool and saliva, and applied metabolomic profiling to plasma and stool samples in a subset of individuals (N = 75). We analyzed these multi-omic data to investigate the relationship between diet, the microbiome, and the gut and circulating metabolome. On a global level, we observed significant relationships, particularly between long-term diet, the gut microbiome and the metabolome. Intake of plant-derived nutrients as well as consumption of artificial sweeteners were associated with significant differences in circulating metabolites, particularly bile acids, which were dependent on gut enterotype, indicating that microbiome composition mediates the effect of diet on host physiology. Our analysis identifies dietary compounds and phytochemicals that may modulate bacterial abundance within the gut and interact with microbiome composition to alter host metabolism.
Collapse
Affiliation(s)
- Zheng-Zheng Tang
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, WI, United States
- Wisconsin Institute for Discovery, Madison, WI, United States
| | - Guanhua Chen
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, WI, United States
| | - Qilin Hong
- Department of Statistics, University of Wisconsin–Madison, Madison, WI, United States
| | - Shi Huang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Holly M. Smith
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Rachana D. Shah
- Division of Pediatric Endocrinology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Matthew Scholz
- Vanderbilt Technologies for Advanced Genomics (VANTAGE), Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jane F. Ferguson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Translational and Clinical Cardiovascular Research Center (VTRACC), Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
37
|
Forkosh E, Ilan Y. The heart-gut axis: new target for atherosclerosis and congestive heart failure therapy. Open Heart 2019; 6:e000993. [PMID: 31168383 PMCID: PMC6519415 DOI: 10.1136/openhrt-2018-000993] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/22/2019] [Accepted: 03/29/2019] [Indexed: 02/06/2023] Open
Abstract
The human gut microbiota has been identified as a possible novel risk factor for cardiovascular disease. The intestinal microbiome plays a role in the pathogenesis of atherosclerosis and heart failure. Even though studies in rodents suggested that gut microbes may affect the risk of heart disease, this link has not been shown in humans. In the present study, we review several potential mechanisms by which the gut microbiome and bacterial translocation are associated with the development of cardiac disorders making them potential targets for novel therapeutic measures for these conditions. Modulation of the gut microbiota as a mechanism for altering the pathogenesis of disorders is an area of growing interest. Alteration in the gut microbiota is being explored as a method of reducing risk factors associated with cardiac diseases.
Collapse
Affiliation(s)
- Esther Forkosh
- Medicine, Hebrew University Hadassah Medical Center, Jerusalem, Israel
| | - Yaron Ilan
- Medicine, Hebrew University Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
38
|
Shi J, Zhao XH. Influence of the Maillard-type caseinate glycation with lactose on the intestinal barrier activity of the caseinate digest in IEC-6 cells. Food Funct 2019; 10:2010-2021. [DOI: 10.1039/c8fo02607f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The glycated caseinate digest of the Maillard-type shows lower capability than the caseinate digest to enhance the intestinal barrier function of IEC-6 cells.
Collapse
Affiliation(s)
- Jia Shi
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- PR China
| | - Xin-Huai Zhao
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- PR China
| |
Collapse
|
39
|
Mohammadkhah AI, Simpson EB, Patterson SG, Ferguson JF. Development of the Gut Microbiome in Children, and Lifetime Implications for Obesity and Cardiometabolic Disease. CHILDREN (BASEL, SWITZERLAND) 2018; 5:E160. [PMID: 30486462 PMCID: PMC6306821 DOI: 10.3390/children5120160] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/19/2018] [Accepted: 11/22/2018] [Indexed: 02/07/2023]
Abstract
Emerging evidence suggests that microbiome composition and function is associated with development of obesity and metabolic disease. Microbial colonization expands rapidly following birth, and microbiome composition is particularly variable during infancy. Factors that influence the formation of the gut microbiome during infancy and childhood may have a significant impact on development of obesity and metabolic dysfunction, with life-long consequences. In this review, we examine the determinants of gut microbiome composition during infancy and childhood, and evaluate the potential impact on obesity and cardiometabolic risk.
Collapse
Affiliation(s)
- Anica I Mohammadkhah
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Eoin B Simpson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Stephanie G Patterson
- Division of Critical Care Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Jane F Ferguson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
40
|
Rajendrakumar SK, Revuri V, Samidurai M, Mohapatra A, Lee JH, Ganesan P, Jo J, Lee YK, Park IK. Peroxidase-Mimicking Nanoassembly Mitigates Lipopolysaccharide-Induced Endotoxemia and Cognitive Damage in the Brain by Impeding Inflammatory Signaling in Macrophages. NANO LETTERS 2018; 18:6417-6426. [PMID: 30247915 DOI: 10.1021/acs.nanolett.8b02785] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Oxidative stress during sepsis pathogenesis remains the most-important factor creating imbalance and dysregulation in immune-cell function, usually observed following initial infection. Hydrogen peroxide (H2O2), a potentially toxic reactive oxygen species (ROS), is excessively produced by pro-inflammatory immune cells during the initial phases of sepsis and plays a dominant role in regulating the pathways associated with systemic inflammatory immune activation. In the present study, we constructed a peroxide scavenger mannosylated polymeric albumin manganese dioxide (mSPAM) nanoassembly to catalyze the decomposition of H2O2 responsible for the hyper-activation of pro-inflammatory immune cells. In a detailed manner, we investigated the role of mSPAM nanoassembly in modulating the expression and secretion of pro-inflammatory markers elevated in bacterial lipopolysaccharide (LPS)-mediated endotoxemia during sepsis. Through a facile one-step solution-phase approach, hydrophilic bovine serum albumin reduced manganese dioxide (BM) nanoparticles were synthesized and subsequently self-assembled with cationic mannosylated disulfide cross-linked polyethylenimine (mSP) to formulate mSPAM nanoassembly. In particular, we observed that the highly stable mSPAM nanoassembly suppressed HIF1α expression by scavenging H2O2 in LPS-induced macrophage cells. Initial investigation revealed that a significant reduction of free radicals by the treatment of mSPAM nanoassembly has reduced the infiltration of neutrophils and other leukocytes in a local endotoxemia animal model. Furthermore, therapeutic studies in a systemic endotoxemia model demonstrated that mSPAM treatment reduced TNF-α and IL-6 inflammatory cytokines in serum, in turn circumventing organ damage done by the inflammatory macrophages. Interestingly, we also observed that the reduction of these inflammatory cytokines by mSPAM nanoassembly further prevented IBA-1 immuno-positive microglial cell activation in the brain and consequently improved the cognitive function of the animals. Altogether, the administration of mSPAM nanoassembly scavenged H2O2 and suppressed HIF1α expression in LPS-stimulated macrophages and thereby inhibited the progression of local and systemic inflammation as well as neuroinflammation in an LPS-induced endotoxemia model. This mSPAM nanoassembly system could serve as a potent anti-inflammatory agent, and we further anticipate its successful application in treating various inflammation-related diseases.
Collapse
Affiliation(s)
- Santhosh Kalash Rajendrakumar
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University , Chonnam National University Medical School , Gwangju 61469 , Republic of Korea
| | - Vishnu Revuri
- Department of Green Bioengineering , Korea National University of Transportation , Chungju 27469 , Republic of Korea
| | - Manikandan Samidurai
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University , Chonnam National University Medical School , Gwangju 61469 , Republic of Korea
- NeuroMedical Convergence Lab, Biomedical Research Institute , Chonnam National University Hospital , Jebong-ro, Gwangju 501-757 , Republic of Korea
| | - Adityanarayan Mohapatra
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University , Chonnam National University Medical School , Gwangju 61469 , Republic of Korea
| | - Jae Hyuk Lee
- Department of Pathology , Chonnam National University Hwasun Hospital, Chonnam National University Medical School , Hwasun 58128 , Republic of Korea
| | - Palanivel Ganesan
- Nanotechnology Research Center, Department of Biotechnology and Applied Life Science, College of Biomedical and Health Science , Konkuk University GLOCAL Campus , Chungju 380-701 , Republic of Korea
| | - Jihoon Jo
- NeuroMedical Convergence Lab, Biomedical Research Institute , Chonnam National University Hospital , Jebong-ro, Gwangju 501-757 , Republic of Korea
| | - Yong-Kyu Lee
- Department of Green Bioengineering , Korea National University of Transportation , Chungju 27469 , Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University , Chonnam National University Medical School , Gwangju 61469 , Republic of Korea
| |
Collapse
|
41
|
Stephens RW, Arhire L, Covasa M. Gut Microbiota: From Microorganisms to Metabolic Organ Influencing Obesity. Obesity (Silver Spring) 2018; 26:801-809. [PMID: 29687647 DOI: 10.1002/oby.22179] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/27/2017] [Accepted: 01/03/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This review summarizes the current understanding of the relationship between gut microbiota and the host as it pertains to the regulation of energy balance and obesity. METHODS The paper begins with a brief description of the gut microbiota environment, distribution, and its unique symbiotic relationship with the host. The way that enviromental factors influence microbiota composition and subsequent impact on the host are then described. Next, the mechanisms linking gut dysbiosis with obesity are discussed, and finally current challenges and limitations in understanding the role of gut microbiota in control of obesity are presented. RESULTS Gut microbiota has been implicated in regulation of fat storage, as well as gut dysbiosis, thus contributing to the development of obesity, insulin resistance, hyperglycemia and hyperlipidemia. However, the underlying mechanisms of these processes are far from being clear and will require complex preclinical and clinical interdisciplinary studies of bacteria and host cell-to-cell interactions. CONCLUSIONS There is a need for a better understanding of how changes in gut microbiota composition can impact energy balance and thus control weight gain. This may represent a promising avenue in the race to develop nonsurgical treatments for obesity.
Collapse
Affiliation(s)
- Richard W Stephens
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, California, USA
| | - Lidia Arhire
- Department of Internal Medicine, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- Clinical Hospital Sf. Spiridon, Iasi, Romania
| | - Mihai Covasa
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, California, USA
- Department of Health and Human Development, University of Suceava, Romania
| |
Collapse
|
42
|
Abstract
Sepsis is a life-threatening organ dysfunction caused by a deregulated host response to infection. This inappropriate response to micro-organism invasion is characterized by an overwhelmed systemic inflammatory response and cardiovascular collapse that culminate in high mortality and morbidity in critical care units. The occurrence of sepsis in diabetes mellitus (DM) patients has become more frequent, as the prevalence of DM has increased dramatically worldwide. These two important diseases represent a global public health concern and highlight the importance of increasing our knowledge of the key elements of the immune response related to both conditions. In this context, it is well established that the cells taking part in the innate and adaptive immune responses in diabetic patients have compromised function. These altered responses favor micro-organism growth, a process that contributes to sepsis progression. The present review provides an update on the characteristics of the immune system in diabetic and septic subjects. We also explore the beneficial effects of insulin on the immune response in a glycemic control-dependent and independent manner.
Collapse
|
43
|
Bruce-Keller AJ, Salbaum JM, Berthoud HR. Harnessing Gut Microbes for Mental Health: Getting From Here to There. Biol Psychiatry 2018; 83:214-223. [PMID: 29031410 PMCID: PMC5859957 DOI: 10.1016/j.biopsych.2017.08.014] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 07/26/2017] [Accepted: 08/20/2017] [Indexed: 12/15/2022]
Abstract
There has been an explosion of interest in the study of microorganisms inhabiting the gastrointestinal tract (gut microbiota) and their impact on host health and physiology. Accumulating data suggest that altered communication between gut microbiota and host systems could participate in disorders such as obesity, diabetes mellitus, and autoimmune disorders as well as neuropsychiatric disorders, including autism, anxiety, and major depressive disorders. The conceptual development of the microbiome-gut-brain axis has facilitated understanding of the complex and bidirectional networks between gastrointestinal microbiota and their host, highlighting potential mechanisms through which this environment influences central nervous system physiology. Communication pathways between gut microbiota and the central nervous system could include autonomic, neuroendocrine, enteric, and immune systems, with pathology resulting in disruption to neurotransmitter balance, increases in chronic inflammation, or exacerbated hypothalamic-pituitary-adrenal axis activity. However, uncertainty remains regarding the generalizability of controlled animal studies to the more multifaceted pattern of human pathophysiology, especially with regard to the therapeutic potential for neuropsychiatric health. This narrative review summarizes current understanding of gut microbial influence over physiological function, with an emphasis on neurobehavioral and neurological impairment based on growing understanding of the gut-brain axis. Experimental and clinical data regarding means of therapeutic manipulation of gut microbiota as a novel treatment option for mental health are described, and important knowledge gaps are identified and discussed.
Collapse
Affiliation(s)
- Annadora J Bruce-Keller
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana.
| | - J Michael Salbaum
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Hans-Rudolf Berthoud
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| |
Collapse
|
44
|
Wang Z, Ka SO, Han YT, Bae EJ. Dihydropyranoaurone compound damaurone D inhibits LPS-induced inflammation and liver injury by inhibiting NF-κB and MAPK signaling independent of AMPK. Arch Pharm Res 2017; 41:314-323. [DOI: 10.1007/s12272-017-1001-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 12/14/2017] [Indexed: 12/25/2022]
|
45
|
Cremonini E, Wang Z, Bettaieb A, Adamo AM, Daveri E, Mills DA, Kalanetra KM, Haj FG, Karakas S, Oteiza PI. (-)-Epicatechin protects the intestinal barrier from high fat diet-induced permeabilization: Implications for steatosis and insulin resistance. Redox Biol 2017; 14:588-599. [PMID: 29154190 PMCID: PMC5691220 DOI: 10.1016/j.redox.2017.11.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 10/31/2017] [Accepted: 11/03/2017] [Indexed: 02/09/2023] Open
Abstract
Increased permeability of the intestinal barrier is proposed as an underlying factor for obesity-associated pathologies. Consumption of high fat diets (HFD) is associated with increased intestinal permeabilization and increased paracellular transport of endotoxins which can promote steatosis and insulin resistance. This study investigated whether dietary (-)-epicatechin (EC) supplementation can protect the intestinal barrier against HFD-induced permeabilization and endotoxemia, and mitigate liver damage and insulin resistance. Mechanisms leading to loss of integrity and function of the tight junction (TJ) were characterized. Consumption of a HFD for 15 weeks caused obesity, steatosis, and insulin resistance in male C57BL/6J mice. This was associated with increased intestinal permeability, decreased expression of ileal TJ proteins, and endotoxemia. Supplementation with EC (2-20mg/kg body weight) mitigated all these adverse effects. EC acted modulating cell signals and the gut hormone GLP-2, which are central to the regulation of intestinal permeability. Thus, EC prevented HFD-induced ileum NOX1/NOX4 upregulation, protein oxidation, and the activation of the redox-sensitive NF-κB and ERK1/2 pathways. Supporting NADPH oxidase as a target of EC actions, in Caco-2 cells EC and apocynin inhibited tumor necrosis alpha (TNFα)-induced NOX1/NOX4 overexpression, protein oxidation and monolayer permeabilization. Together, our findings demonstrate protective effects of EC against HFD-induced increased intestinal permeability and endotoxemia. This can in part underlie EC capacity to prevent steatosis and insulin resistance occurring as a consequence of HFD consumption.
Collapse
Affiliation(s)
- Eleonora Cremonini
- Department of Nutrition, University of California, Davis, USA; Department of Environmental Toxicology, University of California, Davis, USA
| | - Ziwei Wang
- Department of Nutrition, University of California, Davis, USA; Department of Environmental Toxicology, University of California, Davis, USA
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee-Knoxville, Knoxville, TN, USA
| | - Ana M Adamo
- Department of Biological Chemistry and IQUIFIB (UBA-CONICET), School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - Elena Daveri
- Department of Nutrition, University of California, Davis, USA; Department of Environmental Toxicology, University of California, Davis, USA
| | - David A Mills
- Department of Food Science and Technology, University of California, Davis, USA; Department of Viticulture and Enology, University of California, Davis, USA
| | - Karen M Kalanetra
- Department of Food Science and Technology, University of California, Davis, USA; Department of Viticulture and Enology, University of California, Davis, USA
| | - Fawaz G Haj
- Department of Nutrition, University of California, Davis, USA; Department of Environmental Toxicology, University of California, Davis, USA
| | - Sidika Karakas
- Department of Internal Medicine, University of California, Davis, USA
| | - Patricia I Oteiza
- Department of Nutrition, University of California, Davis, USA; Department of Environmental Toxicology, University of California, Davis, USA.
| |
Collapse
|
46
|
Deehan EC, Duar RM, Armet AM, Perez-Muñoz ME, Jin M, Walter J. Modulation of the Gastrointestinal Microbiome with Nondigestible Fermentable Carbohydrates To Improve Human Health. Microbiol Spectr 2017; 5:10.1128/microbiolspec.bad-0019-2017. [PMID: 28936943 PMCID: PMC11687544 DOI: 10.1128/microbiolspec.bad-0019-2017] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Indexed: 02/07/2023] Open
Abstract
There is a clear association between the gastrointestinal (GI) microbiome and the development of chronic noncommunicable diseases, providing a rationale for the development of strategies that target the GI microbiota to improve human health. In this article, we discuss the potential of supplementing the human diet with nondigestible fermentable carbohydrates (NDFCs) to modulate the composition, structure, diversity, and metabolic potential of the GI microbiome in an attempt to prevent or treat human disease. The current concepts by which NDFCs can be administered to humans, including prebiotics, fermentable dietary fibers, and microbiota-accessible carbohydrates, as well as the mechanisms by which these carbohydrates exert their health benefits, are discussed. Epidemiological research presents compelling evidence for the health effects of NDFCs, with clinical studies providing further support for some of these benefits. However, rigorously designed human intervention studies with well-established clinical markers and microbial endpoints are still essential to establish (i) the clinical efficiency of specific NDFCs, (ii) the causal role of the GI microbiota in these effects, (iii) the underlying mechanisms involved, and (iv) the degree by which inter-individual differences between GI microbiomes influence these effects. Such studies would provide the mechanistic understanding needed for a systematic application of NDFCs to improve human health via GI microbiota modulation while also allowing the personalization of these dietary strategies.
Collapse
Affiliation(s)
- Edward C Deehan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
| | - Rebbeca M Duar
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
| | - Anissa M Armet
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
| | - Maria Elisa Perez-Muñoz
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
| | - Mingliang Jin
- Department of Microbiology and Immunology, Northwestern Polytechnical University, Xi'an, Shaanxi, China 710065
| | - Jens Walter
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
| |
Collapse
|
47
|
Liljestrand JM, Paju S, Buhlin K, Persson GR, Sarna S, Nieminen MS, Sinisalo J, Mäntylä P, Pussinen PJ. Lipopolysaccharide, a possible molecular mediator between periodontitis and coronary artery disease. J Clin Periodontol 2017; 44:784-792. [PMID: 28556187 DOI: 10.1111/jcpe.12751] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2017] [Indexed: 12/11/2022]
Abstract
AIM We aimed to study how lipopolysaccharide (LPS) in saliva and serum associates with each other, periodontal microbial burden, periodontitis and coronary artery disease (CAD). MATERIALS AND METHODS The used Parogene cohort comprised N = 505 Finnish adults. Coronary diagnosis was acquired by coronary angiography, and the main outcomes were as follows: no significant CAD (n = 123), stable CAD (n = 184) and acute coronary syndrome (n = 169). Periodontitis was defined according to clinical and radiographic examinations. Levels for 75 strains of subgingival bacteria were determined by checkerboard DNA-DNA hybridization. Saliva and serum LPS activity was analysed by Limulus amebocyte lysate assay. RESULTS The level of 11 bacterial strains, which were mainly oral and respiratory Gram-negative species, associated with salivary LPS levels in an age- and gender-adjusted linear regression. A total of 4.9% of the serum LPS, that is endotoxemia, variation was explainable by saliva LPS among patients with periodontitis (n = 247, R2 = .049, Pearson's r = .222, p < .001). Endotoxemia associated with stable CAD in a confounder adjusted multinomial logistic regression model (OR 1.99, 95% CI 1.04-3.81, p = .039, 3rd tertile). CONCLUSIONS In particular in periodontitis patients, subgingival microbial burden contributes to endotoxemia. LPS is a possible molecular mediator between periodontitis and CAD.
Collapse
Affiliation(s)
- John M Liljestrand
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Susanna Paju
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kåre Buhlin
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - G Rutger Persson
- Departments of Oral Medicine and Periodontics, University of Washington, Seattle, WA, USA
| | - Seppo Sarna
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Markku S Nieminen
- Department of Cardiology, Heart and Lung Center, Department of Medicine, Helsinki University Hospital, Helsinki, Finland
| | - Juha Sinisalo
- Department of Cardiology, Heart and Lung Center, Department of Medicine, Helsinki University Hospital, Helsinki, Finland
| | - Päivi Mäntylä
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pirkko J Pussinen
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
48
|
Zeinalian R, Farhangi MA, Shariat A, Saghafi-Asl M. The effects of Spirulina Platensis on anthropometric indices, appetite, lipid profile and serum vascular endothelial growth factor (VEGF) in obese individuals: a randomized double blinded placebo controlled trial. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:225. [PMID: 28431534 PMCID: PMC5399840 DOI: 10.1186/s12906-017-1670-y] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 03/08/2017] [Indexed: 12/27/2022]
Abstract
Background In recent years, a great attention has been focused on Spirulina platensis as a source of potential valuable nutrients for prevention and treatment of chronic diseases. The objectives of the current study were to determine the effects of Spirulina platensis on anthropometric parameters, serum lipids, appetite and serum Vascular Endothelial Growth Factor (VEGF) in obese individuals. Methods In the current study sixty four obese individuals aged 20–50 years were enrolled and randomly allocated into two groups of intervention and placebo. Intervention group (n = 29) received each 500 mg of the Spirulina platensis a twice-daily dosage while the control group (n = 27) received two pills daily starch for 12 weeks. Anthropometric parameters and serum VEGF and lipid profile were measured in fasting blood samples at the beginning and end of the study period. Dietary intakes were assessed by a 24-h recall method and appetite was measured using standard visual analogue scale (VAS). Results Body weight and body mass index (BMI) were decreased in intervention and placebo treated groups although the mean reduction in Spirulina platensis-treated group was significantly higher (P < 0.05). Serum total cholesterol (TC) significantly reduced in intervention group (P < 0.05). Also, treatment with Spirulina platensis significantly reduced appetite (P = 0.008). Mean serum VEGF, low density lipoprotein-cholesterol, and triglycerides did not change significantly after intervention. Serum high density lipoprotein-cholesterol concentrations (HDL-c) significantly increased in both groups while no difference in mean difference of this change has been observed. Conclusion Spirulina supplementation at a dose of 1 g/d for 12 weeks is effective in modulating body weight and appetite and partly modifies serum lipids. This can further confirm the efficacy of this herbal supplement in control and prevention of obesity and obesity- related disorders. Trial registration Iranian registry of clinical trials (IRCT registration number: IRCT2015071219082N7; Date registered: September 12, 2015).
Collapse
|
49
|
A Fermented Whole Grain Prevents Lipopolysaccharides-Induced Dysfunction in Human Endothelial Progenitor Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1026268. [PMID: 28386305 PMCID: PMC5366772 DOI: 10.1155/2017/1026268] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/08/2017] [Accepted: 02/20/2017] [Indexed: 02/06/2023]
Abstract
Endogenous and exogenous signals derived by the gut microbiota such as lipopolysaccharides (LPS) orchestrate inflammatory responses contributing to development of the endothelial dysfunction associated with atherosclerosis in obesity, metabolic syndrome, and diabetes. Endothelial progenitor cells (EPCs), bone marrow derived stem cells, promote recovery of damaged endothelium playing a pivotal role in cardiovascular repair. Since healthy nutrition improves EPCs functions, we evaluated the effect of a fermented grain, Lisosan G (LG), on early EPCs exposed to LPS. The potential protective effect of LG against LPS-induced alterations was evaluated as cell viability, adhesiveness, ROS production, gene expression, and NF-kB signaling pathway activation. Our results showed that LPS treatment did not affect EPCs viability and adhesiveness but induced endothelial alterations via activation of NF-kB signaling. LG protects EPCs from inflammation as well as from LPS-induced oxidative and endoplasmic reticulum (ER) stress reducing ROS levels, downregulating proinflammatory and proapoptotic factors, and strengthening antioxidant defense. Moreover, LG pretreatment prevented NF-kB translocation from the cytoplasm into the nucleus caused by LPS exposure. In human EPCs, LPS increases ROS and upregulates proinflammatory tone, proapoptotic factors, and antioxidants. LG protects EPCs exposed to LPS reducing ROS, downregulating proinflammatory and proapoptotic factors, and strengthening antioxidant defenses possibly by inhibiting NF-κB nuclear translocation.
Collapse
|
50
|
de Groot PF, Frissen MN, de Clercq NC, Nieuwdorp M. Fecal microbiota transplantation in metabolic syndrome: History, present and future. Gut Microbes 2017; 8:253-267. [PMID: 28609252 PMCID: PMC5479392 DOI: 10.1080/19490976.2017.1293224] [Citation(s) in RCA: 201] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The history of fecal microbiota transplantation (FMT) dates back even to ancient China. Recently, scientific studies have been looking into FMT as a promising treatment of various diseases, while in the process teaching us about the interaction between the human host and its resident microbial communities. Current research focuses mainly on Clostridium difficile infections, however interest is rising in other areas such as inflammatory bowel disease (IBD) and the metabolic syndrome. With regard to the latter, the intestinal microbiota might be causally related to the progression of insulin resistance and diabetes. FMT in metabolic syndrome has proven to be an intriguing method to study the role of the gut microbiota and open the way to new therapies by dissecting in whom insulin resistance is driven by microbiota. In this article we review the history of FMT, the present evidence on its role in the pathophysiology of metabolic syndrome and its efficacy, limitations and future prospects.
Collapse
Affiliation(s)
- P. F. de Groot
- Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - M. N. Frissen
- Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - N. C. de Clercq
- Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - M. Nieuwdorp
- Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands,Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Department of Internal medicine, VU University Medical Center, Amsterdam, The Netherlands,ICAR, VU University Medical Center, Amsterdam, The Netherlands,CONTACT M. Nieuwdorp , Department of Internal and Vascular Medicine, Academic Medical Center Meibergdreef 9, room F4–159.2, 1105 AZ Amsterdam, the Netherlands
| |
Collapse
|