1
|
Mamun MAA, Rakib A, Mandal M, Singh UP. Impact of a High-Fat Diet on the Gut Microbiome: A Comprehensive Study of Microbial and Metabolite Shifts During Obesity. Cells 2025; 14:463. [PMID: 40136712 PMCID: PMC11940932 DOI: 10.3390/cells14060463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Over the last few decades, the prevalence of metabolic diseases such as obesity, diabetes, non-alcoholic fatty liver disease, hypertension, and hyperuricemia has surged, primarily due to high-fat diet (HFD). The pathologies of these metabolic diseases show disease-specific alterations in the composition and function of their gut microbiome. How HFD alters the microbiome and its metabolite to mediate adipose tissue (AT) inflammation and obesity is not well known. Thus, this study aimed to identify the changes in the gut microbiome and metabolomic signatures induced by an HFD to alter obesity. To explore the changes in the gut microbiota and metabolites, 16S rRNA gene amplicon sequencing and metabolomic analyses were performed after HFD and normal diet (ND) feeding. We noticed that, at taxonomic levels, the number of operational taxonomic units (OTUs), along with the Chao and Shannon indexes, significantly shifted in HFD-fed mice compared to those fed a ND. Similarly, at the phylum level, an increase in Firmicutes and a decrease in Bacteroidetes were noticed in HFD-fed mice. At the genus level, an increase in Lactobacillus and Ruminococcus was observed, while Allobaculum, Clostridium, and Akkermansia were markedly reduced in the HFD group. Many bacteria from the Ruminococcus genus impair bile acid metabolism and restrict weight loss. Firmicutes are efficient in breaking down complex carbohydrates into short-chain fatty acids (SCFAs) and other metabolites, whereas Bacteroidetes are involved in a more balanced or efficient energy extraction. Thus, an increase in Firmicutes over Bacteroidetes enhances the absorption of more calories from food, which may contribute to obesity. Taken together, the altered gut microbiota and metabolites trigger AT inflammation, which contributes to metabolic dysregulation and disease progression. Thus, this study highlights the potential of the gut microbiome in the development of therapeutic strategies for obesity and related metabolic disorders.
Collapse
Affiliation(s)
| | | | | | - Udai P. Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163, USA; (M.A.A.M.); (A.R.); (M.M.)
| |
Collapse
|
2
|
Han EJ, Ahn JS, Choi YJ, Kim DH, Chung HJ. Changes in Gut Microbiota According to Disease Severity in a Lupus Mouse Model. Int J Mol Sci 2025; 26:1006. [PMID: 39940777 PMCID: PMC11817498 DOI: 10.3390/ijms26031006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/15/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifaceted autoimmune disease driven by immune dysregulation. This study investigated the relationship between gut microbiota and lupus severity using the MRL/lpr lupus mouse model. Mice were grouped based on total immunoglobulin (Ig)G, IgG2a levels, and urine albumin-to-creatinine ratio (ACR), allowing for the comparison of gut microbiota profiles across different disease severities. Interestingly, severe lupus mice exhibited significant reductions in Ruminiclostridium cellulolyticum, Lactobacillus johnsonii, and Kineothrix alysoides, while Clostridium saudiense, Pseudoflavonifractor phocaeensis, and Intestinimonas butyriciproducens were enriched. These microbial shifts correlated with elevated IgG, IgG2a, and ACR levels, indicating that changes in the gut microbiome may directly influence key immunological markers associated with lupus severity. The depletion of beneficial species and the enrichment of potentially pathogenic bacteria appear to contribute to immune activation and disease progression. This study suggests that gut microbiota dysbiosis plays a critical role in exacerbating lupus by modulating immune responses, reinforcing the link between microbial composition and lupus pathogenesis. Our findings provide the first evidence identifying these distinct gut microbial species as potential contributors to lupus severity, highlighting their role as key factors in disease progression.
Collapse
Affiliation(s)
| | | | | | | | - Hea-Jong Chung
- Gwangju Center, Korea Basic Science Institute, Gwangju 61751, Republic of Korea; (E.-J.H.); (J.-S.A.); (Y.-J.C.); (D.-H.K.)
| |
Collapse
|
3
|
Guo L, An T. Investigating the correlation between gut microbiota and prostate cancer through a two-sample Mendelian randomization analysis. Medicine (Baltimore) 2025; 104:e41141. [PMID: 40184142 PMCID: PMC11709207 DOI: 10.1097/md.0000000000041141] [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: 10/02/2024] [Revised: 11/25/2024] [Accepted: 12/12/2024] [Indexed: 04/05/2025] Open
Abstract
Previous studies in observational epidemiology have suggested a potential correlation between the gastrointestinal tract microbiota and prostate cancer. However, the causal relationship between the 2 remains uncertain, our objective was to thoroughly examine the influence of the gut microbiome on the progression of prostate cancer. In this study, we focused on investigating the gut microbiome as an exposure factor, specifically analyzing data from the MiBioGen consortium, which had a substantial sample size of 18,340 participants. As our disease outcome, we utilized prostate cancer data from the FinnGen genome-wide association study, which involved 13,216 participants. To establish causal relationships, we conducted a comprehensive Mendelian randomization analysis employing multiple methods, including inverse variance-weighted, Mendelian randomization-Egger, maximum likelihood, and weighted median approaches. Additionally, we performed sensitivity analysis to address issues such as heterogeneity and horizontal pleiotropy, ensuring the robustness of our findings. The results obtained through inverse variance-weighted analysis revealed that certain microbial groups exhibited a protective effect on prostate cancer. Specifically, the phylum Verrucomicrobia, particularly the family Rikenellaceae, and the genera Anaerotruncus, Eisenbergiella, Olsenella, and Parabacteroides were found to have a beneficial impact. Conversely, the class Bacilli, class Erysipelotrichia, order Erysipelotrichales, order Lactobacillales, family Erysipelotrichaceae, and the genera Marvinbryantia, Romboutsia, Ruminococcaceae UCG002, and Sutterella had an adverse influence on prostate cancer. The sensitivity analysis did not reveal any such outliers, further strengthening the validity of our results. To summarize, a cause-and-effect connection was discovered between various types and prostate cancer. Nevertheless, additional randomized controlled experiments are required for validation.
Collapse
Affiliation(s)
- Lingyu Guo
- Department of Urology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Tian An
- Department of Dermatology and Plastic Surgery, The Second Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xianyang, China
| |
Collapse
|
4
|
Wang YT, Moura AK, Zuo R, Zhou W, Wang Z, Roudbari K, Hu JZ, Li PL, Zhang Y, Li X. Coronary Microvascular Dysfunction Is Associated With Augmented Lysosomal Signaling in Hypercholesterolemic Mice. J Am Heart Assoc 2024; 13:e037460. [PMID: 39604023 DOI: 10.1161/jaha.124.037460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/08/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Accumulating evidence indicates that coronary microvascular dysfunction (CMD) caused by hypercholesterolemia can lead to myocardial ischemia, with or without obstructive atherosclerotic coronary artery disease. However, the molecular pathways associated with compromised coronary microvascular function before the development of myocardial ischemic injury remain poorly defined. In this study, we investigated the effects of hypercholesterolemia on the function and integrity of the coronary microcirculation in mice and the underlying mechanisms. METHODS AND RESULTS Mice were fed a hypercholesterolemic Paigen's diet for 8 weeks. Echocardiography data showed that Paigen's diet caused CMD, characterized by significant reductions in coronary blood flow and coronary flow reserve, but did not affect cardiac remodeling or dysfunction. Immunofluorescence studies revealed that Paigen's diet-induced CMD was associated with activation of coronary arterioles inflammation and increased myocardial inflammatory cell infiltration. These pathological changes occurred in parallel with the upregulation of lysosomal signaling pathways in endothelial cells (ECs). Treating hypercholesterolemic mice with the cholesterol-lowering drug ezetimibe significantly ameliorated Paigen's diet-induced adverse effects, including hypercholesterolemia, steatohepatitis, reduced coronary flow reserve, coronary endothelial cell inflammation, and myocardial inflammatory cell infiltration. In cultured mouse cardiac ECs, 7-ketocholesterol increased mitochondrial reactive oxygen species and inflammatory responses. Meanwhile, 7-ketocholesterol induced the activation of transcriptional factor EB and lysosomal signaling in mouse cardiac ECs, whereas the lysosome inhibitor bafilomycin A1 blocked 7-ketocholesterol-induced transcriptional factor EB activation and exacerbated 7-ketocholesterol-induced inflammation and cell death. Interestingly, ezetimibe synergistically enhanced 7-ketocholesterol-induced transcriptional factor EB activation and attenuated 7-ketocholesterol-induced mitochondrial reactive oxygen species and inflammatory responses in mouse cardiac ECs. CONCLUSIONS These results suggest that CMD can develop and precede detectable cardiac functional or structural changes in the setting of hypercholesterolemia and that upregulation of transcriptional factor EB-mediated lysosomal signaling in endothelial cells plays a protective role against CMD.
Collapse
Affiliation(s)
- Yun-Ting Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy University of Houston TX
| | - Alexandra K Moura
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy University of Houston TX
| | - Rui Zuo
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy University of Houston TX
| | - Wei Zhou
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy University of Houston TX
- Department of Medical Ultrasound Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Zhengchao Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy University of Houston TX
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences Fujian Normal University Fuzhou China
| | - Kiana Roudbari
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy University of Houston TX
| | - Jenny Z Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy University of Houston TX
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology Virginia Commonwealth University, School of Medicine Richmond VA
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy University of Houston TX
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy University of Houston TX
| |
Collapse
|
5
|
Khalil M, Di Ciaula A, Mahdi L, Jaber N, Di Palo DM, Graziani A, Baffy G, Portincasa P. Unraveling the Role of the Human Gut Microbiome in Health and Diseases. Microorganisms 2024; 12:2333. [PMID: 39597722 PMCID: PMC11596745 DOI: 10.3390/microorganisms12112333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
The human gut is a complex ecosystem that supports billions of living species, including bacteria, viruses, archaea, phages, fungi, and unicellular eukaryotes. Bacteria give genes and enzymes for microbial and host-produced compounds, establishing a symbiotic link between the external environment and the host at both the gut and systemic levels. The gut microbiome, which is primarily made up of commensal bacteria, is critical for maintaining the healthy host's immune system, aiding digestion, synthesizing essential nutrients, and protecting against pathogenic bacteria, as well as influencing endocrine, neural, humoral, and immunological functions and metabolic pathways. Qualitative, quantitative, and/or topographic shifts can alter the gut microbiome, resulting in dysbiosis and microbial dysfunction, which can contribute to a variety of noncommunicable illnesses, including hypertension, cardiovascular disease, obesity, diabetes, inflammatory bowel disease, cancer, and irritable bowel syndrome. While most evidence to date is observational and does not establish direct causation, ongoing clinical trials and advanced genomic techniques are steadily enhancing our understanding of these intricate interactions. This review will explore key aspects of the relationship between gut microbiota, eubiosis, and dysbiosis in human health and disease, highlighting emerging strategies for microbiome engineering as potential therapeutic approaches for various conditions.
Collapse
Affiliation(s)
- Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Laura Mahdi
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Nour Jaber
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Domenica Maria Di Palo
- Division of Hygiene, Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Annarita Graziani
- Institut AllergoSan Pharmazeutische Produkte Forschungs- und Vertriebs GmbH, 8055 Graz, Austria;
| | - Gyorgy Baffy
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02130, USA;
- Section of Gastroenterology, Department of Medicine, VA Boston Healthcare System, Boston, MA 02130, USA
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| |
Collapse
|
6
|
Liu T, Lei C, Huang Q, Song W, Li C, Sun N, Liu Z. Hesperidin and Fecal Microbiota Transplantation Modulate the Composition of the Gut Microbiota and Reduce Obesity in High Fat Diet Mice. Diabetes Metab Syndr Obes 2024; 17:3643-3656. [PMID: 39398388 PMCID: PMC11468570 DOI: 10.2147/dmso.s474034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024] Open
Abstract
Introduction Obesity, which is associated with gut microbiota dysbiosis, low-grade chronic inflammation and intestinal barrier dysfunction, can cause a variety of chronic metabolic diseases. Phytochemical flavonoids have a variety of biological activities, among which there may be safe and effective anti-obesity solutions. Methods We tested a plant-derived flavonoid hesperidin and fecal microbiota transplantation (FMT) to alleviate diet-induced obesity. High-fat diet (HFD)-fed mice were treated with hesperidin (100 and 200 mg/kg BW) and FMT. Results Results indicated that hesperidin had the effects of reducing obesity as indicated by reduction of body weight, fat accumulation and blood lipids, reducing inflammation as indicated by reduction of pro-inflammation factors including TNFα, IL-6, IL-1βand iNOS, and improving gut integrity as indicated by increasing colon length, reducing plasma gut permeability indicators iFABP and LBP, increased mRNA expression of mucus protein Muc2, tight junction p Claudin 2, Occludin and ZO-1 in the HFD-fed mice. The anti-obesity effects of hesperidin treatment have a dose-dependent manner. In addition, 16S rRNA-based gut microbiota analysis revealed that hesperidin selectively promoted the growth of Lactobacillus salivarius, Staphylococcus sciuri and Desulfovibrio C21_c20 while inhibiting Bifidobacterium pseudolongum, Mucispirillum schaedleri, Helicobacter ganmani and Helicobacter hepaticus in the HFD-fed mice. Horizontal feces transfer from the normal diet (ND)-fed mice to the HFD-fed mice conferred anti-obesity effects and transmitted some of the HFD-modulated microbes. Conclusion We concluded that hesperidin and FMT both affect the reduction of body weight and improve HFD-related disorders in the HFD-fed mice possibly through modulating the composition of the gut microbiota.
Collapse
Affiliation(s)
- Ting Liu
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510799, People’s Republic of China
| | - Chao Lei
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510799, People’s Republic of China
| | - Qinhong Huang
- The First Clinical College, Guangzhou Medical University, Guangzhou, 511400, People’s Republic of China
| | - Weiqi Song
- Department of Public Health, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Chen Li
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510799, People’s Republic of China
| | - Ning Sun
- Guangzhou 11th People’s Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, 510530, People’s Republic of China
- The State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, the Hong Kong Polytechnic University, 999077, Hong Kong, China
| | - Zhihua Liu
- Department of Anorectal Surgery, the Tenth Affiliated Hospital of Southern Medical University (Dongguan People’s Hospital), Dongguan, People’s Republic of China
| |
Collapse
|
7
|
Lu M, Feng R, Li M, Liu L, Xiao Y, Liu Y, Yin C. Causal relationship between gut microbiota and childhood obesity: A Mendelian randomization study and case-control study. Clin Nutr ESPEN 2024; 63:197-206. [PMID: 38963766 DOI: 10.1016/j.clnesp.2024.05.012] [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: 12/03/2023] [Revised: 03/28/2024] [Accepted: 05/17/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Gut microbiota and obesity are deeply interconnected. However, the causality in the relationship between these factors remains unclear. Therefore, this study aimed to elucidate the genetic relationship between gut microbiota and childhood obesity. METHODS Genetic summary statistics for the gut microbiota were obtained from the MiBioGen consortium. Genome-wide association studies (GWAS) summary data for childhood obesity were obtained from North American, Australian, and European collaborative genome-wide meta-analyses. Mendelian randomization (MR) analyses were performed using the inverse variance weighting method. 16 children with obesity and 16 without obesity were included for clinical observation, and their weight, body mass index, blood lipid levels, and gut microbiology were assessed. Paired t-test was the primary method of data analysis, and statistical significance was set at P < 0.05. RESULTS MR identified 16 causal relationships between the gut microbiome and childhood obesity. In the case-control study, we found that five gut microorganisms differed between children with and without obesity, whereas three gut microorganisms changed after weight loss in children with obesity. CONCLUSION Our study provides new insights into the genetic mechanisms underlying gut microbiota and childhood obesity. TRIAL REGISTRATION NUMBER ChiCTR2300072179. NAME OF REGISTRY Change of intestinal flora and plasma metabolome in obese children and their weight loss intervention: a randomized controlled tria URL OF REGISTRY: https://www.chictr.org.cn/showproj.html. DATE OF REGISTRATION 2023-06-06. DATE OF ENROLMENT OF THE FIRST PARTICIPANT TO THE TRIAL 2023-06-07.
Collapse
Affiliation(s)
- Mengnan Lu
- Department of Pediatrics, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an, Shanxi, 710054, China
| | - Ruoyang Feng
- Department of Joint Surgery, Xi'an Jiaotong University HongHui Hospital, Xi'an, Shanxi, 710054, China
| | - Meng Li
- Department of Pediatrics, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an, Shanxi, 710054, China
| | - Lujie Liu
- Department of Pediatrics, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an, Shanxi, 710054, China
| | - Yanfeng Xiao
- Department of Pediatrics, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an, Shanxi, 710054, China.
| | - Yuesheng Liu
- Department of Pediatrics, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an, Shanxi, 710054, China.
| | - Chunyan Yin
- Department of Pediatrics, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an, Shanxi, 710054, China.
| |
Collapse
|
8
|
Mahran A, Hosni AM, Farag NG, Elkhawaga AA, Mageed AAA. Role of Claudin- 3 as a biomarker of gut-skin axis integrity in patients with psoriasis. Arch Dermatol Res 2024; 316:476. [PMID: 39023797 DOI: 10.1007/s00403-024-03071-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 07/20/2024]
Abstract
Increased intestinal permeability and gut dysbiosis are important factors in the pathophysiology of psoriasis and its associated conditions. Claudin-3 is a protein that is found in tight junctions and may be used to assess the integrity of the gut barrier. The aim of this study was to investigate serum concentration of Claudin- 3 (CLDN3) in patients with psoriasis. Exploring its possible relations with patients' demographic, clinical and laboratory findings was another objective. Fifty psoriatic patients and thirty-five age- and sex-matched healthy volunteers served as the study's control group in this case-control, hospital-based research. The amount of serum CLDN3 was determined by means of an enzyme-linked immunosorbent test (ELISA). Concentration of serum CLDN3 was found to be significantly higher in patients with psoriasis. (p = 0.002). There was no statistically significant correlation between CLDN3 and patient's clinical & laboratory variables. We demonstrated that gut permeability is dysfunctional in patients with psoriasis as indicated by reduction of serum CLDN3. Further investigations are needed to determine whether modulation of gut barrier may represent a new therapeutic approach for psoriasis.
Collapse
Affiliation(s)
- Ayman Mahran
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Amal Mohammed Hosni
- Department of Clinical Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Nesma G Farag
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Amal A Elkhawaga
- Department of Medical Microbiology and immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed A Abdel Mageed
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
9
|
Wang YT, Moura AK, Zuo R, Zhou W, Wang Z, Roudbari K, Hu JZ, Li PL, Zhang Y, Li X. Coronary Microvascular Dysfunction is Associated with Augmented Lysosomal Signaling in Hypercholesterolemic Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.603000. [PMID: 39026774 PMCID: PMC11257577 DOI: 10.1101/2024.07.10.603000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Accumulating evidence indicates that coronary microvascular dysfunction (CMD) caused by hypercholesterolemia can lead to myocardial ischemia, with or without obstructive atherosclerotic coronary artery disease (CAD). However, the molecular pathways associated with compromised coronary microvascular function prior to the development of myocardial ischemic injury remain poorly defined. In this study, we investigated the effects of hypercholesterolemia on the function and integrity of the coronary microcirculation in mice and the underlying mechanisms. Mice were fed with a hypercholesterolemic Paigen's diet (PD) for 8 weeks. Echocardiography data showed that PD caused CMD, characterized by significant reductions in coronary blood flow and coronary flow reserve (CFR), but did not affect cardiac remodeling or dysfunction. Immunofluorescence studies revealed that PD-induced CMD was associated with activation of coronary arterioles inflammation and increased myocardial inflammatory cell infiltration. These pathological changes occurred in parallel with the upregulation of lysosomal signaling pathways in endothelial cells (ECs). Treating hypercholesterolemic mice with the cholesterol-lowering drug ezetimibe significantly ameliorated PD-induced adverse effects, including hypercholesterolemia, steatohepatitis, reduced CFR, coronary EC inflammation, and myocardial inflammatory cell infiltration. In cultured mouse cardiac endothelial cells (MCECs), 7-ketocholesterol (7K) increased mitochondrial reactive oxygen species (ROS) and inflammatory responses. Meanwhile, 7K induced the activation of TFEB and lysosomal signaling in MCECs, whereas the lysosome inhibitor bafilomycin A1 blocked 7K-induced TFEB activation and exacerbated 7K-induced inflammation and cell death. Interestingly, ezetimibe synergistically enhanced 7K-induced TFEB activation and attenuated 7K-induced mitochondrial ROS and inflammatory responses in MCECs. These results suggest that CMD can develop and precede detectable cardiac functional or structural changes in the setting of hypercholesterolemia, and that upregulation of TFEB-mediated lysosomal signaling in ECs plays a protective role against CMD.
Collapse
Affiliation(s)
- Yun-Ting Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
| | - Alexandra K. Moura
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
| | - Rui Zuo
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
| | - Wei Zhou
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengchao Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Kiana Roudbari
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
| | - Jenny Z. Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, USA
| |
Collapse
|
10
|
Montoya-Hernández D, Dufoo-Hurtado E, Cruz-Hernández A, Campos-Vega R. Spent coffee grounds and its antioxidant dietary fiber promote different colonic microbiome signatures: Benefits for subjects with chronodisruption. Microb Pathog 2023; 185:106431. [PMID: 37984489 DOI: 10.1016/j.micpath.2023.106431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/17/2023] [Accepted: 10/28/2023] [Indexed: 11/22/2023]
Abstract
Chronodisruption, commonly displayed by people living with obesity (PLO), is linked to colonic microbiota dysbiosis, and may increase the risk of many chronic non-communicable diseases, whereas dietary interventions-called chrononutrition may mitigate it. We evaluated the in vitro effects of spent coffee grounds (SCG), and their antioxidant dietary fiber (SCG-DF) on the colonic microbiota of an obese donor displaying dysbiosis and chronodisruption. Basal microbiota pattern was associated with an increased risk of non-communicable chronic diseases. Both samples decrease species richness and increase microbiota diversity (p < 0.05; Chao and Shannon index, respectively), positively enhancing Firmicutes/Bacteroidetes index (SCG, p < 0.04; SCG-DF, p < 0.02). SCG and SCG-DF modulated the microbiota, but SCG-DF induced greater changes, significantly increasing. p_Actonobacterias (SCG p < 0.04; SCG-DF, p < 0.02), and reducing g_Alistipes; s_putredinis, g_Prevotella;s_copri. The highest increase was displayed by p_Proteobacteria (f_Desulfovibrionaceae and f_Alcanigenaceae, p < 0.05), while g_Haemophilus; s_parainfluenzae decreased (p < 0.05). However, neither SCG nor SCG-DF modulated g_Alistipes (evening-type colonic microbial marker) beneficially. SCG and SCG-DF reduced (p < 0.05) g_Lachnospira, a microbial evening-type marker, among other microbial populations, of an obese donor displaying chronodisruption and dysbiosis. SCG and SCG-DF displayed a prebiotic effect with the potential to mitigate diseases linked to chronodisruption.
Collapse
Affiliation(s)
- Diego Montoya-Hernández
- Programa de Posgrado en Alimentos del Centro de la República (PROPAC), Research and Graduate Studies in Food Science, School of Chemistry, Universidad Autónoma de Querétaro, Santiago de Querétaro, 76010, Qro, Mexico.
| | - Elisa Dufoo-Hurtado
- Programa de Posgrado en Alimentos del Centro de la República (PROPAC), Research and Graduate Studies in Food Science, School of Chemistry, Universidad Autónoma de Querétaro, Santiago de Querétaro, 76010, Qro, Mexico.
| | - Andrés Cruz-Hernández
- Escuela de Agronomía, Universidad De La Salle Bajío Campus Campestre, Av. Universidad 602, Col. Lomas del Campestre, León, 37150, Mexico.
| | - Rocio Campos-Vega
- Programa de Posgrado en Alimentos del Centro de la República (PROPAC), Research and Graduate Studies in Food Science, School of Chemistry, Universidad Autónoma de Querétaro, Santiago de Querétaro, 76010, Qro, Mexico.
| |
Collapse
|
11
|
Qiao J, Xiao X, Wang K, Haubruge E, Dong J, Zhang H. Rapeseed bee pollen alleviates chronic non-bacterial prostatitis via regulating gut microbiota. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:7896-7904. [PMID: 37486857 DOI: 10.1002/jsfa.12878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/11/2023] [Accepted: 07/24/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND Rapeseed bee pollen has been recognized as a critical treatment for chronic non-bacterial prostatitis (CNP) and it also can modulate gut microbiota and improve gut health. This study aimed to explore the anti-prostatitis effects of rapeseed bee pollen with or without wall-disruption, and to investigate the connection between this treatment and gut microbiota. RESULTS The results reveal that rapeseed bee pollen can effectively alleviate chronic non-bacteria prostatitis by selectively regulating gut microbiota, with higher doses and wall-disrupted pollen showing greater efficacy. Treatment with a high dose of wall-disrupted rapeseed bee pollen (WDH, 1.26 g kg-1 body weight) reduced prostate wet weight and prostate index by approximately 32% and 36%, respectively, nearly the levels observed in the control group. Wall-disrupted rapeseed bee pollen treatment also reduced significantly (p < 0.05) the expression of proinflammatory cytokines (IL-6, IL-8, IL-1β, and TNF-α), as confirmed by immunofluorescence with laser scanning confocal microscope. Our results show that rapeseed bee pollen can inhibit pathogenic bacteria and enhance probiotics, particularly in the Firmicutes-to-Bacteroidetes (F/B) ratio and the abundance of Prevotella (genus). CONCLUSION This is the first study to investigate the alleviation of CNP with rapeseed bee pollen through gut microbiota. These results seem to provide better understanding for the development of rapeseed bee pollen as a complementary medicine. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jiangtao Qiao
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Terra Research Center, Gembloux Agro-Bio Tech, University of Liege, Gembloux, Belgium
| | - Xingying Xiao
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Kai Wang
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Terra Research Center, Gembloux Agro-Bio Tech, University of Liege, Gembloux, Belgium
| | - Eric Haubruge
- Terra Research Center, Gembloux Agro-Bio Tech, University of Liege, Gembloux, Belgium
| | - Jie Dong
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Bee Products for Quality and Safety Control, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Hongcheng Zhang
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Bee Products for Quality and Safety Control, Ministry of Agriculture and Rural Affairs, Beijing, China
| |
Collapse
|
12
|
Chiodi V, Rappa F, Lo Re O, Chaldakov GN, Lelouvier B, Micale V, Domenici MR, Vinciguerra M. Deficiency of histone variant macroH2A1.1 is associated with sexually dimorphic obesity in mice. Sci Rep 2023; 13:19123. [PMID: 37926763 PMCID: PMC10625986 DOI: 10.1038/s41598-023-46304-8] [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: 05/08/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023] Open
Abstract
Obesity has a major socio-economic health impact. There are profound sex differences in adipose tissue deposition and obesity-related conditions. The underlying mechanisms driving sexual dimorphism in obesity and its associated metabolic disorders remain unclear. Histone variant macroH2A1.1 is a candidate epigenetic mechanism linking environmental and dietary factors to obesity. Here, we used a mouse model genetically depleted of macroH2A1.1 to investigate its potential epigenetic role in sex dimorphic obesity, metabolic disturbances and gut dysbiosis. Whole body macroH2A1 knockout (KO) mice, generated with the Cre/loxP technology, and their control littermates were fed a high fat diet containing 60% of energy derived from fat. The diet was administered for three months starting from 10 to 12 weeks of age. We evaluated the progression in body weight, the food intake, and the tolerance to glucose by means of a glucose tolerance test. Gut microbiota composition, visceral adipose and liver tissue morphology were assessed. In addition, adipogenic gene expression patterns were evaluated in the visceral adipose tissue. Female KO mice for macroH2A1.1 had a more pronounced weight gain induced by high fat diet compared to their littermates, while the increase in body weight in male mice was similar in the two genotypes. Food intake was generally increased upon KO and decreased by high fat diet in both sexes, with the exception of KO females fed a high fat diet that displayed the same food intake of their littermates. In glucose tolerance tests, glucose levels were significantly elevated upon high fat diet in female KO compared to a standard diet, while this effect was absent in male KO. There were no differences in hepatic histology. Upon a high fat diet, in female adipocyte cross-sectional area was larger in KO compared to littermates: activation of proadipogenic genes (ACACB, AGT, ANGPT2, FASN, RETN, SLC2A4) and downregulation of antiadipogenic genes (AXIN1, E2F1, EGR2, JUN, SIRT1, SIRT2, UCP1, CCND1, CDKN1A, CDKN1B, EGR2) was detected. Gut microbiota profiling showed increase in Firmicutes and a decrease in Bacteroidetes in females, but not males, macroH2A1.1 KO mice. MacroH2A1.1 KO mice display sexual dimorphism in high fat diet-induced obesity and in gut dysbiosis, and may represent a useful model to investigate epigenetic and metabolic differences associated to the development of obesity-associated pathological conditions in males and females.
Collapse
Affiliation(s)
- Valentina Chiodi
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanita', Rome, Italy
| | - Francesca Rappa
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Oriana Lo Re
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, Varna, Bulgaria
- International Clinical Research Center (FNUSA-ICRC), St'Anne University Hospital, Brno, Czech Republic
| | - George N Chaldakov
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, Varna, Bulgaria
- Department of Anatomy and Cell Biology, Research Institute of the Medical University, Varna, Bulgaria
| | | | - Vincenzo Micale
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Maria Rosaria Domenici
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanita', Rome, Italy
| | - Manlio Vinciguerra
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, Varna, Bulgaria.
- International Clinical Research Center (FNUSA-ICRC), St'Anne University Hospital, Brno, Czech Republic.
- Liverpool Centre for Cardiovascular Science (LCCS), Liverpool John Moores University, Liverpool, UK.
| |
Collapse
|
13
|
Zhang M, Zhang M, Kou G, Li Y. The relationship between gut microbiota and inflammatory response, learning and memory in mice by sleep deprivation. Front Cell Infect Microbiol 2023; 13:1159771. [PMID: 37293204 PMCID: PMC10244646 DOI: 10.3389/fcimb.2023.1159771] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/27/2023] [Indexed: 06/10/2023] Open
Abstract
Objective Sleep deprivation has developed into a common phenomenon, which can lead to inflammatory responses and cognitive impairment, but the underlying mechanism is ambiguous. Emerging evidence shows that gut microbiota plays a crucial role in theoccurrence and development of inflammatory and psychiatric diseases, possibly through neuroinflammation and the brain-gut axis. The current study investigated the influence of sleep deprivation on gut microbiota composition, pro-inflammatory cytokines, learning and memory in mice. Further, it explored whether changes in gut microbiota increase pro-inflammatory cytokine and induce learning and memory impairment. Methods Healthy 8-week-old male C57BL/6J mice were randomly divided into the regular control group (RC), environmental control group (EC), and sleep deprivation group (SD). The sleep deprivation model was established by the Modified Multiple Platform Method. The experimental mice were subjected to sleep deprivation for 6h/d (8:00 am∼14:00 pm) in a sleep deprivation chamber, and the duration of sleep deprivation was 8 weeks. Morris water maze test to assess learning and memory in mice. Enzyme-Linked Immunosorbent Assay determined the concentrations of inflammatory cytokines. The changes in gut microbiota in mice were analyzed by 16S rRNA sequencing. Results We found that SD mice had elevated latency of exploration to reach the hidden platform (p>0.05) and significantly decreased traversing times, swimming distance, and swimming time in the target zone when the hidden platform was removed (p<0.05). Sleep deprivation caused dysregulated expression in serum IL-1β, IL-6, and TNF-α in mice, and the difference was significant (all p<0.001). Tannerellaceae, Rhodospirillales, Alistipes, and Parabacteroides were significantly increased in SD mice. Correlation analysis showed IL-1β was positively correlated with the abundance of Muribaculaceae (r=0.497, p<0.05) and negatively correlated with the abundance of Lachnospiraceae (r=-0.583, p<0.05). The TNF-α was positively correlated with the abundances of Erysipelotrichaceae, Burkholderiaceae, and Tannerellaceae (r=0.492, r=0.646, r=0.726, all p<0.05). Conclusion Sleep deprivation can increase pro-inflammatory cytokine responses and learning and memory impairment in mice and may be caused by the disorder of the microbiota. These findings of this study may open avenues for potential interventions that can relieve the detrimental consequences of sleep loss.
Collapse
Affiliation(s)
- Mengjie Zhang
- School of Physical Education and Sport Science, Fujian Normal University, Fuzhou, China
- Zhengzhou University, Zhengzhou, China
| | - Mengying Zhang
- Zhengzhou University, Zhengzhou, China
- Synergetic Innovation Center of Kinesis and Health, School of Physical Education (Main Campus), Zhengzhou University, Zhengzhou, China
| | - Guangning Kou
- Centre of Sport Nutrition and Health, School of Physical Education, Zhengzhou University, Zhengzhou, China
| | - Yan Li
- Zhengzhou University, Zhengzhou, China
- Synergetic Innovation Center of Kinesis and Health, School of Physical Education (Main Campus), Zhengzhou University, Zhengzhou, China
| |
Collapse
|
14
|
Hou JY, Xu H, Cao GZ, Tian LL, Wang LH, Zhu NQ, Zhang JJ, Yang HJ. Multi-omics reveals Dengzhan Shengmai formulation ameliorates cognitive impairments in D-galactose-induced aging mouse model by regulating CXCL12/CXCR4 and gut microbiota. Front Pharmacol 2023; 14:1175970. [PMID: 37101548 PMCID: PMC10123283 DOI: 10.3389/fphar.2023.1175970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/28/2023] [Indexed: 04/28/2023] Open
Abstract
Dengzhan Shengmai (DZSM), a traditional Chinese medicine formulation, has been administered extensively to elderly individuals with cognitive impairment (CI). However, the underlying mechanisms by which Dengzhan Shengmai improves cognitive impairment remains unknown. This study aimed to elucidate the underlying mechanism of the effect of Dengzhan Shengmai on aging-associated cognitive impairment via a comprehensive combination of transcriptomics and microbiota assessment. Dengzhan Shengmai was orally administered to a D-galactose-induced aging mouse model, and evaluation with an open field task (OFT), Morris water maze (MWM), and histopathological staining was performed. Transcriptomics and 16S rDNA sequencing were applied to elucidate the mechanism of Dengzhan Shengmai in alleviating cognitive deficits, and enzyme-linked immunosorbent assay (ELISA), quantitative real-time polymerase chain reaction (PCR), and immunofluorescence were employed to verify the results. The results first confirmed the therapeutic effects of Dengzhan Shengmai against cognitive defects; specifically, Dengzhan Shengmai improved learning and impairment, suppressed neuro loss, and increased Nissl body morphology repair. Comprehensive integrated transcriptomics and microbiota analysis indicated that chemokine CXC motif receptor 4 (CXCR4) and its ligand CXC chemokine ligand 12 (CXCL12) were targets for improving cognitive impairments with Dengzhan Shengmai and also indirectly suppressed the intestinal flora composition. Furthermore, in vivo results confirmed that Dengzhan Shengmai suppressed the expression of CXC motif receptor 4, CXC chemokine ligand 12, and inflammatory cytokines. This suggested that Dengzhan Shengmai inhibited CXC chemokine ligand 12/CXC motif receptor 4 expression and modulated intestinal microbiome composition by influencing inflammatory factors. Thus, Dengzhan Shengmai improves aging-related cognitive impairment effects via decreased CXC chemokine ligand 12/CXC motif receptor 4 and inflammatory factor modulation to improve gut microbiota composition.
Collapse
Affiliation(s)
- Jing-Yi Hou
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences and MEGAROBO, Beijing, China
- Postdoctoral Mobile Research Station of China Academy of Chinese Medicine Sciences, Beijing, China
| | - He Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guang-Zhao Cao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liang-Liang Tian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li-Han Wang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences and MEGAROBO, Beijing, China
- Postdoctoral Mobile Research Station of China Academy of Chinese Medicine Sciences, Beijing, China
| | - Nai-Qiang Zhu
- Postdoctoral Mobile Research Station of China Academy of Chinese Medicine Sciences, Beijing, China
| | - Jing-Jing Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Jing-Jing Zhang, ; Hong-Jun Yang,
| | - Hong-Jun Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences and MEGAROBO, Beijing, China
- *Correspondence: Jing-Jing Zhang, ; Hong-Jun Yang,
| |
Collapse
|
15
|
Zhang W, Zhang S, Zhao F, Du J, Wang Z. Causal relationship between gut microbes and cardiovascular protein expression. Front Cell Infect Microbiol 2022; 12:1048519. [PMID: 36544908 PMCID: PMC9760811 DOI: 10.3389/fcimb.2022.1048519] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/17/2022] [Indexed: 12/11/2022] Open
Abstract
Evidence supports associations between gut microbiota and cardiovascular protein levels in plasma. However, it is unclear whether these associations reflect a causal relationship. To reveal the causal relationship between gut microbiota and cardiovascular protein levels in plasma, we estimated their causal effects using two-sample Mendelian randomization (MR) analysis. Sensitivity analysis was also performed to assess the robustness of our results. Genome-wide association study (GWAS) of microbiomes in the MiBioGen study included 211 bacterial taxa (18,473 individuals), and GWAS of 90 cardiovascular proteins included 30,931 individuals. There were 196 bacterial taxa from five levels available for analysis. The following 14 causal relationships were identified: phylum Euryarchaeota and carbohydrate antigen 125 (β = 0.289), order Bacillales and CSF-1 (β = -0.211), genus Dorea and HSP-27 (β = 0.465), phylum Actinobacteria and IL-8 (β = 0.274), order Enterobacteriales and KIM-1 (β = -0.499), class Actinobacteria, genus Bifidobacterium, phylum Actinobacteria and LEP (β = -0.219, β = -0.201, and β = -0.221), genus Methanobrevibacter and NT-proBNP (β = 0.371), family Peptostreptococcaceae and SRC (β = 0.191), order Verrucomicrobiales, phylum Verrucomicrobia and TNF-R2 (β = 0.251 and β = 0.233), family Veillonellaceae and t-PA (β = 0.271), and class Erysipelotrichia and VEGF-D (β = 0.390). Sensitivity analysis showed no evidence of pleiotropy or heterogeneity. The results of the reverse MR analysis showed no reverse causality for any of the 13 gut microbes and 11 cardiovascular proteins. Mendelian randomization estimates provide strong evidence for a causal effect of gut microbiota-mediated alterations on cardiovascular protein expression.
Collapse
Affiliation(s)
- Wenchuan Zhang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shuwan Zhang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Feng Zhao
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, Shenyang, Liaoning, China
| | - Jinda Du
- Department of Gastroenterology, General Hospital of Northern Theatre Command, Shenyang, Liaoning, China
| | - Zhe Wang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China,*Correspondence: Zhe Wang,
| |
Collapse
|
16
|
Tsan L, Sun S, Hayes AMR, Bridi L, Chirala LS, Noble EE, Fodor AA, Kanoski SE. Early life Western diet-induced memory impairments and gut microbiome changes in female rats are long-lasting despite healthy dietary intervention. Nutr Neurosci 2022; 25:2490-2506. [PMID: 34565305 PMCID: PMC8957635 DOI: 10.1080/1028415x.2021.1980697] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Western diet consumption during adolescence results in hippocampus (HPC)-dependent memory impairments and gut microbiome dysbiosis. Whether these adverse outcomes persist in adulthood following healthy dietary intervention is unknown. Here we assessed the short- and long-term effects of adolescent consumption of a Western diet enriched with either sugar or both sugar and fat on metabolic outcomes, HPC function, and gut microbiota. METHODS Adolescent female rats (PN 26) were fed a standard chow diet (CHOW), chow with access to 11% sugar solution (SUG), or a junk food cafeteria-style diet (CAF) containing various foods high in fat and/or sugar. During adulthood (PN 65+), metabolic outcomes, HPC-dependent memory, and gut microbial populations were evaluated. In a subsequent experiment, these outcomes were evaluated following a 5-week dietary intervention where CAF and SUG groups were maintained on standard chow alone. RESULTS Both CAF and SUG groups demonstrated impaired HPC-dependent memory, increased adiposity, and altered gut microbial populations relative to the CHOW group. However, impaired peripheral glucose regulation was only observed in the SUG group. When examined following a healthy dietary intervention in a separate experiment, metabolic dysfunction was not observed in either the CAF or SUG group, whereas HPC-dependent memory impairments were observed in the CAF but not the SUG group. In both groups the composition of the gut microbiota remained distinct from CHOW rats after the dietary intervention. CONCLUSIONS While the metabolic impairments associated with adolescent junk food diet consumption are not present in adulthood following dietary intervention, the HPC-dependent memory impairments and the gut microbiome dysbiosis persist.
Collapse
Affiliation(s)
- Linda Tsan
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Shan Sun
- Department of Bioinformatics and Genomics at the University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Anna M. R. Hayes
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Lana Bridi
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Lekha S. Chirala
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Emily E. Noble
- Department of Foods and Nutrition, University of Georgia, Athens, GA, USA
| | - Anthony A. Fodor
- Department of Bioinformatics and Genomics at the University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Scott E. Kanoski
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
17
|
Márquez-Sánchez AC, Koltsova EK. Immune and inflammatory mechanisms of abdominal aortic aneurysm. Front Immunol 2022; 13:989933. [PMID: 36275758 PMCID: PMC9583679 DOI: 10.3389/fimmu.2022.989933] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening cardiovascular disease. Immune-mediated infiltration and a destruction of the aortic wall during AAA development plays significant role in the pathogenesis of this disease. While various immune cells had been found in AAA, the mechanisms of their activation and function are still far from being understood. A better understanding of mechanisms regulating the development of aberrant immune cell activation in AAA is essential for the development of novel preventive and therapeutic approaches. In this review we summarize current knowledge about the role of immune cells in AAA and discuss how pathogenic immune cell activation is regulated in this disease.
Collapse
|
18
|
Brettle H, Tran V, Drummond GR, Franks AE, Petrovski S, Vinh A, Jelinic M. Sex hormones, intestinal inflammation, and the gut microbiome: Major influencers of the sexual dimorphisms in obesity. Front Immunol 2022; 13:971048. [PMID: 36248832 PMCID: PMC9554749 DOI: 10.3389/fimmu.2022.971048] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity is defined as the excessive accumulation of body fat and is associated with an increased risk of developing major health problems such as cardiovascular disease, diabetes and stroke. There are clear sexual dimorphisms in the epidemiology, pathophysiology and sequelae of obesity and its accompanying metabolic disorders, with females often better protected compared to males. This protection has predominantly been attributed to the female sex hormone estrogen and differences in fat distribution. More recently, the sexual dimorphisms of obesity have also been attributed to the differences in the composition and function of the gut microbiota, and the intestinal immune system. This review will comprehensively summarize the pre-clinical and clinical evidence for these sexual dimorphisms and discuss the interplay between sex hormones, intestinal inflammation and the gut microbiome in obesity. Major gaps and limitations of this rapidly growing area of research will also be highlighted in this review.
Collapse
Affiliation(s)
- Holly Brettle
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Vivian Tran
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Grant R. Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Ashley E. Franks
- Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Steve Petrovski
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Maria Jelinic
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
- *Correspondence: Maria Jelinic,
| |
Collapse
|
19
|
Afzaal M, Saeed F, Shah YA, Hussain M, Rabail R, Socol CT, Hassoun A, Pateiro M, Lorenzo JM, Rusu AV, Aadil RM. Human gut microbiota in health and disease: Unveiling the relationship. Front Microbiol 2022; 13:999001. [PMID: 36225386 PMCID: PMC9549250 DOI: 10.3389/fmicb.2022.999001] [Citation(s) in RCA: 194] [Impact Index Per Article: 64.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/31/2022] [Indexed: 12/04/2022] Open
Abstract
The human gut possesses millions of microbes that define a complex microbial community. The gut microbiota has been characterized as a vital organ forming its multidirectional connecting axis with other organs. This gut microbiota axis is responsible for host-microbe interactions and works by communicating with the neural, endocrinal, humoral, immunological, and metabolic pathways. The human gut microorganisms (mostly non-pathogenic) have symbiotic host relationships and are usually associated with the host’s immunity to defend against pathogenic invasion. The dysbiosis of the gut microbiota is therefore linked to various human diseases, such as anxiety, depression, hypertension, cardiovascular diseases, obesity, diabetes, inflammatory bowel disease, and cancer. The mechanism leading to the disease development has a crucial correlation with gut microbiota, metabolic products, and host immune response in humans. The understanding of mechanisms over gut microbiota exerts its positive or harmful impacts remains largely undefined. However, many recent clinical studies conducted worldwide are demonstrating the relation of specific microbial species and eubiosis in health and disease. A comprehensive understanding of gut microbiota interactions, its role in health and disease, and recent updates on the subject are the striking topics of the current review. We have also addressed the daunting challenges that must be brought under control to maintain health and treat diseases.
Collapse
Affiliation(s)
- Muhammad Afzaal
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
- *Correspondence: Muhammad Afzaal,
| | - Farhan Saeed
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Yasir Abbas Shah
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muzzamal Hussain
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Roshina Rabail
- National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| | | | - Abdo Hassoun
- Sustainable AgriFoodtech Innovation & Research (SAFIR), Arras, France
- Syrian Academic Expertise (SAE), Gaziantep, Turkey
| | - Mirian Pateiro
- Centro Tecnológico de la Carne de Galicia, Ourense, Spain
| | - José M. Lorenzo
- Centro Tecnológico de la Carne de Galicia, Ourense, Spain
- Área de Tecnoloxía dos Alimentos, Faculdade de Ciências de Ourense, Universidade de Vigo, Ourense, Spain
| | - Alexandru Vasile Rusu
- Life Science Institute, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
- Faculty of Animal Science and Biotechnology, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
| | - Rana Muhammad Aadil
- National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
- Rana Muhammad Aadil,
| |
Collapse
|
20
|
Wan S, Sun N, Li H, Khan A, Zheng X, Sun Y, Fan R. Deoxynivalenol damages the intestinal barrier and biota of the broiler chickens. BMC Vet Res 2022; 18:311. [PMID: 35965338 PMCID: PMC9377127 DOI: 10.1186/s12917-022-03392-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
Background In the livestock feed industry, feed and feed raw materials are extremely susceptible to mycotoxin contamination. Deoxynivalenol (DON) is one of the main risk factors for mycotoxin contamination in broiler feed and feedstuff, however, there is still little knowledge about this. Hence, the purpose of this study was to explore the toxicity effect of DON on the intestinal barrier and the microecological balance of the biota in broiler chickens. Results In our present study, we compared the pathological scores of the small intestines of broilers on the 5th, 7th, and 10th day, and chose the 7th day to analyze the small intestine histomorphology, tight junctions, and cecal biota of the broilers. The results showed the damage to the small intestine worsened over time, the small intestinal villi of broilers were breakage, the tight junctions of the small intestine were destroyed, the cecal biota was unbalanced, and the growth performance of broilers was reduced on the 7th day. Conclusions DON could damage the functional and structural completeness of the intestinal tract, disorder the Intestinal biota, and finally lead to declined broiler performance. Our study provided a basis for the prevention and treatment of DON in broiler production. Supplementary Information The online version contains supplementary material available at 10.1186/s12917-022-03392-4.
Collapse
Affiliation(s)
- Shuangxiu Wan
- Shanxi Key Lab. for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China.,College of Pharmacy, Heze University, Heze, Shangdong, 274000, People's Republic of China
| | - Na Sun
- Shanxi Key Lab. for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Hongquan Li
- Shanxi Key Lab. for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Ajab Khan
- Shanxi Key Lab. for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Xiaozhong Zheng
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Yaogui Sun
- Shanxi Key Lab. for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Ruiwen Fan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, People's Republic of China.
| |
Collapse
|
21
|
Zubcevic J, Watkins J, Lin C, Bautista B, Hatch HM, Tevosian SG, Hayward LF. Nicotine Exposure during Rodent Pregnancy Alters the Composition of Maternal Gut Microbiota and Abundance of Maternal and Amniotic Short Chain Fatty Acids. Metabolites 2022; 12:metabo12080735. [PMID: 36005607 PMCID: PMC9414314 DOI: 10.3390/metabo12080735] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/28/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022] Open
Abstract
Tobacco smoking is the leading cause of preventable death. Numerous reports link smoking in pregnancy with serious adverse outcomes, such as miscarriage, stillbirth, prematurity, low birth weight, perinatal morbidity, and infant mortality. Corollaries of consuming nicotine in pregnancy, separate from smoking, are less explored, and the mechanisms of nicotine action on maternal–fetal communication are poorly understood. This study examined alterations in the maternal gut microbiome in response to nicotine exposure during pregnancy. We report that changes in the maternal gut microbiota milieu are an important intermediary that may mediate the prenatal nicotine exposure effects, affect gene expression, and alter fetal exposure to circulating short-chain fatty acids (SCFAs) and leptin during in utero development.
Collapse
Affiliation(s)
- Jasenka Zubcevic
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
- Correspondence: (J.Z.); (S.G.T.)
| | - Jacqueline Watkins
- Department of Physiological Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
| | - Cindy Lin
- Department of Physiological Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
| | - Byrell Bautista
- Department of Physiological Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
| | - Heather M. Hatch
- Department of Physiological Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
| | - Sergei G. Tevosian
- Department of Physiological Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
- Correspondence: (J.Z.); (S.G.T.)
| | - Linda F. Hayward
- Department of Physiological Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
| |
Collapse
|
22
|
Angelidi AM, Belanger MJ, Kokkinos A, Koliaki CC, Mantzoros CS. Novel Noninvasive Approaches to the Treatment of Obesity: From Pharmacotherapy to Gene Therapy. Endocr Rev 2022; 43:507-557. [PMID: 35552683 PMCID: PMC9113190 DOI: 10.1210/endrev/bnab034] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Indexed: 02/08/2023]
Abstract
Recent insights into the pathophysiologic underlying mechanisms of obesity have led to the discovery of several promising drug targets and novel therapeutic strategies to address the global obesity epidemic and its comorbidities. Current pharmacologic options for obesity management are largely limited in number and of modest efficacy/safety profile. Therefore, the need for safe and more efficacious new agents is urgent. Drugs that are currently under investigation modulate targets across a broad range of systems and tissues, including the central nervous system, gastrointestinal hormones, adipose tissue, kidney, liver, and skeletal muscle. Beyond pharmacotherapeutics, other potential antiobesity strategies are being explored, including novel drug delivery systems, vaccines, modulation of the gut microbiome, and gene therapy. The present review summarizes the pathophysiology of energy homeostasis and highlights pathways being explored in the effort to develop novel antiobesity medications and interventions but does not cover devices and bariatric methods. Emerging pharmacologic agents and alternative approaches targeting these pathways and relevant research in both animals and humans are presented in detail. Special emphasis is given to treatment options at the end of the development pipeline and closer to the clinic (ie, compounds that have a higher chance to be added to our therapeutic armamentarium in the near future). Ultimately, advancements in our understanding of the pathophysiology and interindividual variation of obesity may lead to multimodal and personalized approaches to obesity treatment that will result in safe, effective, and sustainable weight loss until the root causes of the problem are identified and addressed.
Collapse
Affiliation(s)
- Angeliki M Angelidi
- Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Matthew J Belanger
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alexander Kokkinos
- First Department of Propaedeutic Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Chrysi C Koliaki
- First Department of Propaedeutic Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Christos S Mantzoros
- Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Kennedy L, Francis H, Alpini G. Macrophage-Specific SCAP Promotes Liver and Adipose Tissue Damage in a Lean NAFLD Model: Lean, Mean, Proinflammatory Machine. Cell Mol Gastroenterol Hepatol 2022; 14:236-238. [PMID: 35500675 PMCID: PMC9254626 DOI: 10.1016/j.jcmgh.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 12/10/2022]
Affiliation(s)
| | | | - Gianfranco Alpini
- Correspondence Address correspondence to: Gianfranco Alpini, PhD, FAASLD, AGAF, Indiana Center for Liver Research, Indiana University School of Medicine, 702 Rotary Circle, Room 013C, Indianapolis, Indiana 46202.
| |
Collapse
|
24
|
Huang X, Yao Y, Hou X, Wei L, Rao Y, Su Y, Zheng G, Ruan XZ, Li D, Chen Y. Macrophage SCAP Contributes to Metaflammation and Lean NAFLD by Activating STING-NF-κB Signaling Pathway. Cell Mol Gastroenterol Hepatol 2022; 14:1-26. [PMID: 35367665 PMCID: PMC9117819 DOI: 10.1016/j.jcmgh.2022.03.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Sterol regulatory element binding protein cleavage-activating protein (SCAP) is a cholesterol sensor that confers a broad range of functional effects in metabolic diseases. Lean nonalcoholic fatty liver disease (NAFLD) is characterized by a decrease in subcutaneous fat and ectopic fat deposition in the liver. SCAP may mediate the development of lean NAFLD, but the mechanism of action remains unclear. METHODS C57BL/6J wild-type and macrophage SCAP-specific knockout mice (SCAPΔMϕ) were subjected to Paigen diet (PD) feeding induced lean NAFLD. Inflammation and lipid metabolism of adipose and liver were evaluated. The STING-NF-κB signaling pathway was examined in vivo and in vitro to explore the underlying mechanism of macrophage SCAP on metaflammation. RESULTS The data showed heterogeneity of lipid metabolic processes in liver and epididymal white adipose tissue due to inflammation mediated by macrophage infiltration. Meanwhile, we found that the macrophage SCAP was abnormally increased in the adipose and liver tissues of PD-fed mice. Intriguingly, the SCAPΔMϕ mice attenuated PD-induced metaflammation and ectopic lipid deposition by reducing hepatic stimulator of interferon gene (STING)-nuclear factor kappa B (NF-κB) pathway activation. In-depth molecular analysis revealed that SCAP specifically recruits the STING and tank-binding kinase 1 onto the Golgi to activate the NF-κB in macrophages, thereby promoting the release of inflammatory factors. This process ultimately led to an increased lipolysis in adipocytes and lipid uptake and synthesis in hepatocytes. CONCLUSIONS Our findings suggest that SCAP acts as a novel regulator of the macrophage inflammatory response and the pathogenesis of lean NAFLD by activating the STING-NF-κB signaling pathway. Inhibition of macrophage SCAP may represent a new therapeutic strategy for the treatment of lean NAFLD.
Collapse
Affiliation(s)
- Xinyu Huang
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yingcheng Yao
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xiaoli Hou
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Li Wei
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yuhan Rao
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yu Su
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Guo Zheng
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xiong Z. Ruan
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China,John Moorhead Research Laboratory, Centre for Nephrology, University College London Medical School, Royal Free Campus, University College London, London, United Kingdom
| | - Danyang Li
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China,Correspondence Address correspondence to: Danyang Li, PhD or Yaxi Chen, PhD, Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, 400016 Chongqing, China. fax: +86-23-68486780.
| | - Yaxi Chen
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China,Correspondence Address correspondence to: Danyang Li, PhD or Yaxi Chen, PhD, Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, 400016 Chongqing, China. fax: +86-23-68486780.
| |
Collapse
|
25
|
Giallongo S, Řeháková D, Biagini T, Lo Re O, Raina P, Lochmanová G, Zdráhal Z, Resnick I, Pata P, Pata I, Mistrík M, de Magalhães JP, Mazza T, Koutná I, Vinciguerra M. Histone Variant macroH2A1.1 Enhances Nonhomologous End Joining-dependent DNA Double-strand-break Repair and Reprogramming Efficiency of Human iPSCs. Stem Cells 2022; 40:35-48. [PMID: 35511867 PMCID: PMC9199840 DOI: 10.1093/stmcls/sxab004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/02/2021] [Indexed: 01/21/2023]
Abstract
DNA damage repair (DDR) is a safeguard for genome integrity maintenance. Increasing DDR efficiency could increase the yield of induced pluripotent stem cells (iPSC) upon reprogramming from somatic cells. The epigenetic mechanisms governing DDR during iPSC reprogramming are not completely understood. Our goal was to evaluate the splicing isoforms of histone variant macroH2A1, macroH2A1.1, and macroH2A1.2, as potential regulators of DDR during iPSC reprogramming. GFP-Trap one-step isolation of mtagGFP-macroH2A1.1 or mtagGFP-macroH2A1.2 fusion proteins from overexpressing human cell lines, followed by liquid chromatography-tandem mass spectrometry analysis, uncovered macroH2A1.1 exclusive interaction with Poly-ADP Ribose Polymerase 1 (PARP1) and X-ray cross-complementing protein 1 (XRCC1). MacroH2A1.1 overexpression in U2OS-GFP reporter cells enhanced specifically nonhomologous end joining (NHEJ) repair pathway, while macroH2A1.1 knock-out (KO) mice showed an impaired DDR capacity. The exclusive interaction of macroH2A1.1, but not macroH2A1.2, with PARP1/XRCC1, was confirmed in human umbilical vein endothelial cells (HUVEC) undergoing reprogramming into iPSC through episomal vectors. In HUVEC, macroH2A1.1 overexpression activated transcriptional programs that enhanced DDR and reprogramming. Consistently, macroH2A1.1 but not macroH2A1.2 overexpression improved iPSC reprogramming. We propose the macroH2A1 splicing isoform macroH2A1.1 as a promising epigenetic target to improve iPSC genome stability and therapeutic potential.
Collapse
Affiliation(s)
- Sebastiano Giallongo
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Daniela Řeháková
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
| | - Tommaso Biagini
- Laboratory of Bioinformatics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Oriana Lo Re
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
- Department of Translational Stem Cell Biology, Research Institute of the Medical University of Varna (RIMUV), Varna, Bulgaria
| | - Priyanka Raina
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Gabriela Lochmanová
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Zbyněk Zdráhal
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- National Centre for Biomolecular Research, Masaryk University, Brno, Czech Republic
| | - Igor Resnick
- Department of Translational Stem Cell Biology, Research Institute of the Medical University of Varna (RIMUV), Varna, Bulgaria
- Program for Hematology, Immunology, BMT and Cell therapy, St. Marina University Hospital, Varna, Bulgaria
- Department of Medical Genetics, Medical University of Varna, Varna, Bulgaria
| | - Pille Pata
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
- IVEX Lab, Akadeemia 15, Tallinn, Estonia
| | - Illar Pata
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Martin Mistrík
- Laboratory of Genome Integrity, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Tommaso Mazza
- Department of Translational Stem Cell Biology, Research Institute of the Medical University of Varna (RIMUV), Varna, Bulgaria
| | - Irena Koutná
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
- Department of Translational Stem Cell Biology, Research Institute of the Medical University of Varna (RIMUV), Varna, Bulgaria
| |
Collapse
|
26
|
Shi Y, Zou Y, Xiong Y, Zhang S, Song M, An X, Liu C, Zhang W, Chen S. Host Gasdermin D restrains systemic endotoxemia by capturing Proteobacteria in the colon of high-fat diet-feeding mice. Gut Microbes 2021; 13:1946369. [PMID: 34275417 PMCID: PMC8288038 DOI: 10.1080/19490976.2021.1946369] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Gasdermin D (GSDMD) functions as a key pyroptotic executor through its secreted N-terminal domain (GSDMD-N). However, the functional relevance and mechanistic basis of the precise roles of host colonic GSDMD in high-fat diet (HFD)-induced gut dysbiosis and systemic endotoxemia remain elusive. In this study, we demonstrate that HFD feeding triggers GSDMD-N secretion of both T-lymphocytes and enterocytes in mouse colons. GSDMD deficiency aggravates HFD-induced systemic endotoxemia, gut barrier impairment, and colonic inflammation. More importantly, active GSDMD-N kills the Proteobacteria phylum via directly interacting with Cardiolipin. Mechanistically, we identify that the Glu236 (a known residue for GSDMD protein cleavage) is a bona fide important site for the bacterial recognition of GSDMD. Collectively, our findings explain the mechanism by which colonic GSDMD-N maintains low levels of HFD-induced metabolic endotoxemia. A GSDMD-N mimetic containing an exposed Glu236 site could be an attractive strategy for the treatment of HFD-induced metabolic endotoxemia.
Collapse
Affiliation(s)
- Yujie Shi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yixin Zou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yonghong Xiong
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shiyao Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China,Wenxiang Zhang State Key Laboratory of Natural Medicines, China Pharmaceutical University, #639 Longmian Avenue, Nanjing211198, China
| | - Mingming Song
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiaofei An
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenxiang Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China,Wenxiang Zhang State Key Laboratory of Natural Medicines, China Pharmaceutical University, #639 Longmian Avenue, Nanjing211198, China
| | - Siyu Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China,CONTACT Siyu Chen
| |
Collapse
|
27
|
Morais AHDA, Passos TS, de Lima Vale SH, da Silva Maia JK, Maciel BLL. Obesity and the increased risk for COVID-19: mechanisms and nutritional management. Nutr Res Rev 2021; 34:209-221. [PMID: 33183383 PMCID: PMC7737140 DOI: 10.1017/s095442242000027x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 10/23/2020] [Accepted: 11/10/2020] [Indexed: 01/07/2023]
Abstract
The global COVID-19 (coronavirus disease 2019) pandemic has become a complex problem that overlaps with a growing public health problem, obesity. Obesity alters different components of the innate and adaptive immune responses, creating a chronic and low-grade state of inflammation. Nutritional status is closely related to a better or worse prognosis of viral infections. Excess weight has been recognised as a risk factor for COVID-19 complications. In addition to the direct risk, obesity triggers other diseases such as diabetes and hypertension, increasing the risk of severe COVID-19. The present review explains the diets that induce obesity and the importance of different foods in this process. We also review tissue disruption in obesity, leading to impaired immune responses and the possible mechanisms by which obesity and its co-morbidities increase COVID-19 morbidity and mortality. Nutritional strategies that support the immune system in patients with obesity and with COVID-19 are also discussed in light of the available data, considering the severity of the infection. The discussions held may contribute to combating this global emergency and planning specific public health policy.
Collapse
Affiliation(s)
- Ana Heloneida de Araújo Morais
- Nutrition Postgraduate Program, Centre for Health Sciences, Federal University of Rio Grande do Norte, Natal, RN59078-970, Brazil
- Biochemistry Postgraduate Program, Biosciences Centre, Federal University of Rio Grande do Norte, Natal, RN59078-970, Brazil
- Department of Nutrition, Centre for Health Sciences, Federal University of Rio Grande do Norte, Natal, RN59078-970, Brazil
| | - Thais Sousa Passos
- Department of Nutrition, Centre for Health Sciences, Federal University of Rio Grande do Norte, Natal, RN59078-970, Brazil
| | - Sancha Helena de Lima Vale
- Department of Nutrition, Centre for Health Sciences, Federal University of Rio Grande do Norte, Natal, RN59078-970, Brazil
| | - Juliana Kelly da Silva Maia
- Nutrition Postgraduate Program, Centre for Health Sciences, Federal University of Rio Grande do Norte, Natal, RN59078-970, Brazil
- Department of Nutrition, Centre for Health Sciences, Federal University of Rio Grande do Norte, Natal, RN59078-970, Brazil
| | - Bruna Leal Lima Maciel
- Nutrition Postgraduate Program, Centre for Health Sciences, Federal University of Rio Grande do Norte, Natal, RN59078-970, Brazil
- Department of Nutrition, Centre for Health Sciences, Federal University of Rio Grande do Norte, Natal, RN59078-970, Brazil
| |
Collapse
|
28
|
Vasilyeva LE, Drapkina OM. Impact of Gut Microbiota on the Risk of Cardiometabolic Diseases Development. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2021. [DOI: 10.20996/1819-6446-2021-10-14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Obesity is a multifactorial disease that leads to excessive adipose tissue accumulation, mainly visceral fat. Importance and prevalence of obesity has increased significantly in recent decades all over the world. Until now, the pandemic of obesity has been associated more to lifestyle changes: excessive eating and low physical activity. In recent years, special attention has been paid to studying of composition and functions of intestinal microbiota as major factor in development of obesity and related comorbidities, such as hypertension, cardiac ischemia, heart failure and others. It is proved that gut microbiota affects extraction, accumulation and consumption of energy derived from food, lipid metabolism and immune response. It is also revealed that composition of the microbiota is different in thin and obese people. Thus, study of the relationship between intestinal microbiota composition and risk factors for cardiovascular diseases, in particular obesity, is an actual task. The purpose of this review is analyzing of literature about assessment of relationship between composition and functions of intestinal microbiota in the diagnostics, prevention and treatment of obesity and cardiovascular diseases.
Collapse
Affiliation(s)
- L. E. Vasilyeva
- National Medical Research Center for Therapy and Preventive Medicine
| | - O. M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine
| |
Collapse
|
29
|
Tian J, Bai B, Gao Z, Yang Y, Wu H, Wang X, Wang J, Li M, Tong X. Alleviation Effects of GQD, a Traditional Chinese Medicine Formula, on Diabetes Rats Linked to Modulation of the Gut Microbiome. Front Cell Infect Microbiol 2021; 11:740236. [PMID: 34692563 PMCID: PMC8531589 DOI: 10.3389/fcimb.2021.740236] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/22/2021] [Indexed: 01/14/2023] Open
Abstract
Gegen Qinlian Decoction (GQD) is a Chinese herbal medicine that has been reported to significantly decrease blood glucose levels, which is suggested to be related to interactions with the gut microbiota. However, the protective effect of GQD on intestinal barrier function with regard to its influence on the gut microbiota has not been explored to date. In this study, we investigated the role of the gut microbiota in mediating the hypoglycemic mechanism of GQD in type 2 diabetes mellitus (T2DM) rats induced by a single intraperitoneal injection of streptozotocin after 4 weeks of high-fat diet feeding. The T2DM rats were randomly allocated to receive GQD, metformin (Met), or saline for 12 consecutive weeks, and changes in metabolic parameters, intestinal barrier function, and inflammation were investigated. Gut microbiota was analyzed using 16S rRNA gene sequencing from fecal samples, and statistical analyses were performed to correlate microbiota composition with phenotypes of the T2DM rats. GQD administration decreased the levels of blood glucose and inflammatory cytokines, and increased the levels of tight junction proteins. Besides, GQD had a protective effect on islet function, restoring intestinal permeability, and inhibiting inflammation, as evidenced by increases in the levels of serum C-peptide, occludin, and claudin-1 in the colon, and also improved the expression of serum inflammatory factors. In addition, GQD regulated the structure of the gut microbiota by increasing the proportions of short-chain fatty acids-producing and anti-inflammatory bacteria, and decreasing the proportions of conditioned pathogenic bacteria associated with the diabetic phenotype. Overall, these findings suggest that GQD could ameliorate hyperglycemia and protect islet function by regulating the structure of the gut microbiota, thereby restoring intestinal permeability and inhibiting inflammation in T2DM rats. Our study thus suggests that the hypoglycemic mechanism of GQD is mediated by its modulation of the gut microbiota.
Collapse
Affiliation(s)
- Jiaxing Tian
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bingbing Bai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Science, Beijing, China
| | - Zezheng Gao
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yingying Yang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haoran Wu
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinmiao Wang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jun Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Science, Beijing, China
| | - Min Li
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaolin Tong
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
30
|
Zeng Q, Yang Z, Wang F, Li D, Liu Y, Wang D, Zhao X, Li Y, Wang Y, Feng X, Chen J, Li Y, Zheng Y, Kenney T, Gu H, Feng S, Li S, He Y, Xu X, Dai W. Association between metabolic status and gut microbiome in obese populations. Microb Genom 2021; 7. [PMID: 34356001 PMCID: PMC8549370 DOI: 10.1099/mgen.0.000639] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Despite that obesity is associated with many metabolic diseases, a significant proportion (10–30 %) of obese individuals is recognized as ‘metabolically healthy obeses’ (MHOs). The aim of the current study is to characterize the gut microbiome for MHOs as compared to ‘metabolically unhealthy obeses’ (MUOs). We compared the gut microbiome of 172 MHO and 138 MUO individuals from Chongqing (China) (inclined to eat red meat and food with a spicy taste), and performed validation with selected biomarkers in 40 MHOs and 33 MUOs from Quanzhou (China) (inclined to eat seafood and food with a light/bland taste). The genera Alistipes, Faecalibacterium and Odoribacter had increased abundance in both Chongqing and Quanzhou MHOs. We also observed different microbial functions in MUOs compared to MHOs, including an increased abundance of genes associated with glycan biosynthesis and metabolism. In addition, the microbial gene markers identified from the Chongqing cohort bear a moderate accuracy [AUC (area under the operating characteristic curve)=0.69] for classifying MHOs distinct from MUOs in the Quanzhou cohort. These findings indicate that gut microbiome is significantly distinct between MHOs and MUOs, implicating the potential of the gut microbiome in stratification and refined management of obesity.
Collapse
Affiliation(s)
- Qiang Zeng
- Health Management Institute, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, PR China
| | - Zhenyu Yang
- School of Statistics and Data Science, Nankai University, Tianjin 300000, PR China.,Key Laboratory for Medical Data Analysis and Statistical Research of Tianjin, Tianjin 300000, PR China
| | - Fei Wang
- Health Management Institute, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, PR China
| | - Dongfang Li
- Department of Microbial Research, WeHealthGene Institute, Shenzhen 518000, PR China
| | - Yanhong Liu
- Department of Microbial Research, WeHealthGene Institute, Shenzhen 518000, PR China
| | - Daxi Wang
- Melbourne Veterinary School, Department of Veterinary Biosciences, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Xiaolan Zhao
- Southwest Hospital, Third Military Medical University, Chongqing 400000, PR China
| | - Yinhu Li
- Department of Computer Science, College of Science and Engineering, City University of Hong Kong, Hong Kong 999077, PR China
| | - Yu Wang
- Health Management Center, 91st Hospital of the People's Liberation Army, Quanzhou 362000, PR China
| | - Xin Feng
- Department of Microbial Research, WeHealthGene Institute, Shenzhen 518000, PR China
| | - Jiaxing Chen
- Department of Computer Science, College of Science and Engineering, City University of Hong Kong, Hong Kong 999077, PR China
| | - Yongli Li
- Department of Health Management, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Yuejie Zheng
- Department of Respiratory Medicine, Shenzhen Children's Hospital, Shenzhen 518000, PR China
| | - Toby Kenney
- Department of Mathematics and Statistics, Dalhousie University, Halifax 15000, Canada
| | - Hong Gu
- Department of Mathematics and Statistics, Dalhousie University, Halifax 15000, Canada
| | - Su Feng
- School of Statistics and Data Science, Nankai University, Tianjin 300000, PR China
| | - Shuangcheng Li
- Department of Computer Science, College of Science and Engineering, City University of Hong Kong, Hong Kong 999077, PR China
| | - Yuan He
- Health Management Institute, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, PR China.,National Research Institute for Health, Beijing 100000, PR China
| | - Ximing Xu
- School of Statistics and Data Science, Nankai University, Tianjin 300000, PR China.,Key Laboratory for Medical Data Analysis and Statistical Research of Tianjin, Tianjin 300000, PR China
| | - Wenkui Dai
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen 518000, PR China
| |
Collapse
|
31
|
Tamarind (Tamarindus indica L.) Seed a Candidate Protein Source with Potential for Combating SARS-CoV-2 Infection in Obesity. Drug Target Insights 2021; 15:5-12. [PMID: 33840996 PMCID: PMC8025844 DOI: 10.33393/dti.2021.2192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 03/11/2021] [Indexed: 12/17/2022] Open
Abstract
Introduction: Obesity and coronavirus disease (COVID)-19 are overlapping pandemics, and one might worsen the other. Methods: This narrative review discusses one of the primary mechanisms to initiate acute respiratory distress syndrome, uncontrolled systemic inflammation in COVID-19, and presents a potential candidate for adjuvant treatment. Blocking the S protein binding to angiotensin-converting enzyme 2 (ACE-2) and the 3C-like protease (3CL pro) is an effective strategy against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Results: Host proteases such as FURIN, trypsin, and transmembrane serine protease 2 (TMPRSS) act in S protein activation. Tamarind trypsin inhibitor (TTI) shows several beneficial effects on the reduction of inflammatory markers (tumor necrosis factor α [TNF-α], leptin) and biochemical parameters (fasting glycemia, triglycerides, and very low-density lipoprotein [VLDL]), in addition to improving pancreatic function and mucosal integrity in an obesity model. TTI may inhibit the action of proteases that collaborate with SARS-CoV-2 infection and the neutrophil activity characteristic of lung injury promoted by the virus. Conclusion: Thus, TTI may contribute to combating two severe overlapping problems with high cost and social complex implications, obesity and COVID-19.
Collapse
|
32
|
Chinniah R, Sevak V, Pandi S, Ravi PM, Vijayan M, Kannan A, Karuppiah B. HLA-DRB1 genes and the expression dynamics of HLA CIITA determine the susceptibility to T2DM. Immunogenetics 2021; 73:291-305. [PMID: 33754173 DOI: 10.1007/s00251-021-01212-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/01/2021] [Indexed: 12/18/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a disease with polygenic inheritance. The expression of major histocompatibility complex class II genes are regulated by several trans-activators. We have studied the expression of HLA-DRB1, RFX, CIITA-P1, PIV transactivators, immunophenotyping of cells, SNPs in CIITA-168 (A/G) and IFN-γ + 874 (T/A) in T2DM patients and controls (n = 201 each). We observed increased frequencies of DRB1*03, DRB1*04 and DRB1*07 and decreased frequencies of DRB1*10, DRB1*14, and DRB1*15 alleles among patients. Significant up-regulations of HLA-DRB1 genes were observed in patients (p < 0.0001). Down-regulated expressions were documented in DRB1*03-homo (p < 0.002) and DRB1*04-homo (p < 0.009) patients. No significant differences were observed for CIITA-P1 expression except DRB1*04-pooled (p < 0.0113). The CIITA-PIV was up-regulated in overall (p < 0.0001), DRB1*03-pooled (p < 0.0006), DRB1*03-hetero (p < 0.0006) and DRB1*03-homo (p < 0.001) T2DM patients. However, significant down-regulations were documented for DRB1*04-pooled (p < 0.040), DRB1*04-hetero (p < 0.060), and DRB1*04-homo (p < 0.027) combinations. Further, significant down-regulations of RFX5 were observed in overall (p < 0.0006), DRB1*04-pooled (p < 0.0022), and DRB1*04-hetero (p < 0.0004) combinations. Immunophenotyping studies revealed significant increase of CD45+ CD14-, CD19+, CD14- and CD8 cells and elevated level of expression of IFN-γ (p < 0.0001) in patients. A significant increase of TT (p < 3.35 × 10-6) and decrease of TA (p < 4.57 × 10-4) genotypes of IFN-γ + 874 (T/A) and an increase of GG (p < 0.001) and decrease of AG (p < 8.24 × 10-5) genotypes of CIITA-168 A/G SNPs were observed. The combinatorial analysis revealed susceptible associations for DRB1*03 + AA, *03 + AG, *03 + GG and *04 + GG and protective associations for DRB1*10 + AG, *10 + GG, *15 + AG, and *14 + GG combinations. Thus, the present study corroborated the effect of differential expressions of promoters of risk alleles in the pathogenesis of T2DM.
Collapse
Affiliation(s)
- Rathika Chinniah
- Department of Immunology, School of Biological Sciences, Madurai, Tamil Nadu, 625021, India
| | - Vandit Sevak
- Department of Immunology, School of Biological Sciences, Madurai, Tamil Nadu, 625021, India
| | - Sasiharan Pandi
- Department of Immunology, School of Biological Sciences, Madurai, Tamil Nadu, 625021, India
| | - Padma Malini Ravi
- Department of Immunology, School of Biological Sciences, Madurai, Tamil Nadu, 625021, India
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Arun Kannan
- The Madurai Institute of Diabetes and Endocrine Practice Research, Madurai, Tamil Nadu, 625 001, India
| | - Balakrishnan Karuppiah
- Department of Immunology, School of Biological Sciences, Madurai, Tamil Nadu, 625021, India.
| |
Collapse
|
33
|
McNamara MP, Singleton JM, Cadney MD, Ruegger PM, Borneman J, Garland T. Early-life effects of juvenile Western diet and exercise on adult gut microbiome composition in mice. J Exp Biol 2021; 224:jeb239699. [PMID: 33431595 PMCID: PMC7929929 DOI: 10.1242/jeb.239699] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/06/2021] [Indexed: 02/06/2023]
Abstract
Alterations to the gut microbiome caused by changes in diet, consumption of antibiotics, etc., can affect host function. Moreover, perturbation of the microbiome during critical developmental periods potentially has long-lasting impacts on hosts. Using four selectively bred high runner and four non-selected control lines of mice, we examined the effects of early-life diet and exercise manipulations on the adult microbiome by sequencing the hypervariable internal transcribed spacer region of the bacterial gut community. Mice from high runner lines run ∼3-fold more on wheels than do controls, and have several other phenotypic differences (e.g. higher food consumption and body temperature) that could alter the microbiome, either acutely or in terms of coevolution. Males from generation 76 were given wheels and/or a Western diet from weaning until sexual maturity at 6 weeks of age, then housed individually without wheels on standard diet until 14 weeks of age, when fecal samples were taken. Juvenile Western diet reduced bacterial richness and diversity after the 8-week washout period (equivalent to ∼6 human years). We also found interactive effects of genetic line type, juvenile diet and/or juvenile exercise on microbiome composition and diversity. Microbial community structure clustered significantly in relation to both line type and diet. Western diet also reduced the relative abundance of Muribaculum intestinale These results constitute one of the first reports of juvenile diet having long-lasting effects on the adult microbiome after a substantial washout period. Moreover, we found interactive effects of diet with early-life exercise exposure, and a dependence of these effects on genetic background.
Collapse
Affiliation(s)
- Monica P McNamara
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, Riverside, CA 91521, USA
| | - Jennifer M Singleton
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, Riverside, CA 91521, USA
| | - Marcell D Cadney
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, Riverside, CA 91521, USA
| | - Paul M Ruegger
- Department of Microbiology and Plant Pathology, University of California, Riverside, Riverside, CA 91521, USA
| | - James Borneman
- Department of Microbiology and Plant Pathology, University of California, Riverside, Riverside, CA 91521, USA
| | - Theodore Garland
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, Riverside, CA 91521, USA
| |
Collapse
|
34
|
Li N, Li J, Zhang Q, Gao S, Quan X, Liu P, Xu C. Effects of endocrine disrupting chemicals in host health: Three-way interactions between environmental exposure, host phenotypic responses, and gut microbiota. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 271:116387. [PMID: 33401209 DOI: 10.1016/j.envpol.2020.116387] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/20/2020] [Accepted: 12/23/2020] [Indexed: 06/12/2023]
Abstract
Endocrine disrupting chemicals (EDCs) have gradually become a global health hazard in recent decades. Gut microbiota (GM) provides a crucial interface between the environment and the human body. A triad relationship may exist between EDCs exposure, host phenotypic background, and GM effects. In this review, we attempted to parse out the contribution of GM on the alteration of host phenotypic responses induced by EDCs, suggesting that GM intervention may be used as a therapeutic strategy to limit the expansion of pathogen. These studies can increase the understanding of pathogenic mechanisms, and help to identify the modifiable environmental factors and microbiota characteristics in people with underlying disease susceptibility for prevention and remediation.
Collapse
Affiliation(s)
- Na Li
- Pediatric Department, Ruijin Hospital, Shanghai Jiaotong University. School of Medicine, Shanghai, China; Institute of Tropical Medicine, Hainan Medical University, HaiKou, China
| | - Jinhua Li
- School of Public Health, Jilin University, Changchun, China
| | - Qingqing Zhang
- Pediatric Department, Ruijin Hospital, Shanghai Jiaotong University. School of Medicine, Shanghai, China
| | - Shenshen Gao
- Pediatric Department, Ruijin Hospital, Shanghai Jiaotong University. School of Medicine, Shanghai, China
| | - Xu Quan
- Pediatric Department, Ruijin Hospital, Shanghai Jiaotong University. School of Medicine, Shanghai, China
| | - Ping Liu
- Pediatric Department, Ruijin Hospital, Shanghai Jiaotong University. School of Medicine, Shanghai, China
| | - Chundi Xu
- Pediatric Department, Ruijin Hospital, Shanghai Jiaotong University. School of Medicine, Shanghai, China.
| |
Collapse
|
35
|
Qu H, Song L, Zhang Y, Gao ZY, Shi DZ. The Effect of Prebiotic Products on Decreasing Adiposity Parameters in Overweight and Obese Individuals: A Systematic Review and Meta- Analysis. Curr Med Chem 2021; 28:419-431. [PMID: 31886746 DOI: 10.2174/0929867327666191230110128] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/09/2019] [Accepted: 11/10/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Prebiotics are substrates selectively utilized by host microorganisms to confer health benefits. The potential of prebiotics to decrease body weight in overweight/obese individuals was suggested by some clinical and animal studies. However, these studies were based on relatively small sample sizes and the precise effects of prebiotic products have not yet been evaluated. Therefore, the present meta-analysis of Randomized Controlled Trials (RCTs) was designed to comprehensively assess the effects of prebiotic products on overweight and obese individuals. METHODS PubMed, EMBASE and Cochrane Library were searched to identify RCT investigating the effects of prebiotic products on overweight and obese individuals. We calculated the pooled weighted mean difference (WMD) to assess the effects of prebiotic products on Body Mass Index (BMI), body weight, fat mass and inflammatory biomarkers. RESULTS Twelve RCTs with a total of 535 overweight and obese individuals were enrolled. Compared with placebo, prebiotic products decreased C reactive protein (WMD, -1.06 mg/L; 95%CI, -1.72 to - 0.40; p=0.002), tumour necrosis factor-α(WMD, -0.64 pg/mL; 95%CI, -1.11 to -0.18; p=0.006) and other inflammatory markers, such as interleukin-1β,lipopolysaccharide (p<0.05); whereas no reductions in BMI (WMD, -0.20 kg/m2; 95%CI, -0.58 to 0.19; p=0.32), body weight (WMD, -0.51 kg; 95%CI, -1.18 to 0.16; p=0.14) and fat mass (WMD, 0.11 kg; 95%CI, -0.04 to 0.25; p=0.15) were observed. CONCLUSION In the present analysis, comprehensive evidence suggested that prebiotic products did not decrease adiposity parameters (BMI, body weight and body fat mass), but they could decrease the levels of systemic inflammatory biomarkers, implying adherence to prebiotic products might be a promising complementary approach to managing inflammatory states in overweight and obese individuals.
Collapse
Affiliation(s)
- Hua Qu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lei Song
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhu-Ye Gao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Da-Zhuo Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
36
|
Inhibition of pyruvate dehydrogenase kinase influence microbiota and metabolomic profile in pancreatic cancer xenograft mice. BMC Res Notes 2020; 13:540. [PMID: 33208188 PMCID: PMC7672971 DOI: 10.1186/s13104-020-05384-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/10/2020] [Indexed: 11/17/2022] Open
Abstract
Objective Despite recent advances in treatment options, pancreatic cancer remains the most deadly major cancer. Targeting metabolism represents an emerging anti-cancer strategy. Results Metagenomic 16S analysis was employed to explore the effect of Dichloroacetate (DCA) on the composition of the fecal microbiota and metabolomic profile was assessed on in vivo pancreatic cancer mouse xenograft model. Pancreatic cancer xenograft mice displayed a shift of microbiota’ profile as compared to control mice without DCA treatment and a significant decrease of the purine bases inosine xanthine together with their metabolically-related compound hypoxanthine were observed in the DCA treated group as compared to the control group. Two aminoacids methionine and aspartic acid resulted decreased and increased respectively. DCA affects tumor environment and studies are needed in order to understand whether DCA supplementation could be supportive as synergistic approach to enhance the efficacy of existing cancer treatments in pancreatic cancer patients.
Collapse
|
37
|
Lin YC, Chen YT, Li KY, Chen MJ. Investigating the Mechanistic Differences of Obesity-Inducing Lactobacillus kefiranofaciens M1 and Anti-obesity Lactobacillus mali APS1 by Microbolomics and Metabolomics. Front Microbiol 2020; 11:1454. [PMID: 32733406 PMCID: PMC7360855 DOI: 10.3389/fmicb.2020.01454] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 06/04/2020] [Indexed: 12/27/2022] Open
Abstract
Many studies have investigated the anti-obesity effects of probiotics in animal models and humans. However, few studies have focused on the mechanisms of obesity-inducing probiotics. In a previous study, we demonstrated that specific bacterial strains isolated from kefir, Lactobacillus kefirnofaciens M1 and Lactobacillus mali APS1, possess obesity and anti-obesity effects, respectively, in high-fat diet (HFD)-induced obese mice. Thus, in the present study, we systematically investigated whether APS1 and M1 affect energy homeostasis and lipid metabolism in HFD-induced obese mice and how this might be achieved. We observed that the M1/APS1 intervention influenced fat accumulation by regulating adipogenesis and inflammation-related marker expression both in vitro and in a HFD induced C57BL/6J mice model. We also observed putative links between key taxa and possible metabolic processes of the gut microbiota. Notably, families Christensenellaceae and S24_7 were negatively correlated with body weight gain through increase in the essential esterized carnitine for energy expenditure. These results suggest the importance of specific probiotic interventions affecting leanness and obesity of subjects under a HFD, which are operated by modulating the tripartite relationship among the host, microbiota, and metabolites.
Collapse
Affiliation(s)
- Yu-Chun Lin
- Livestock Research Institute, Council of Agriculture, Executive Yuan, Tainan, Taiwan.,Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Yung-Tsung Chen
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Kuan-Yi Li
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Ming-Ju Chen
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
38
|
Dong Z, Liu Y, Pan H, Wang H, Wang X, Xu X, Xiao K, Liu M, Xu Z, Li L, Zhang Y. The Effects of High-Salt Gastric Intake on the Composition of the Intestinal Microbiota in Wistar Rats. Med Sci Monit 2020; 26:e922160. [PMID: 32504527 PMCID: PMC7297027 DOI: 10.12659/msm.922160] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background A high-salt diet may result in chronic disease and changes in the intestinal microbiota. This pilot study aimed to investigate the microbial composition of the intestine in Wistar rats given intragastric high-salt infusions for four weeks. Material/Methods Six 4-week-old male Wistar rats were fed standard chow and divided into the high-salt group (n=3) and the control study group (n=3). Rats in the high-salt group were given 1 ml of 10% NaCl solution intragastrically three times per week for four weeks. The fecal pellets were collected, and the microbiota was characterized using 16S rRNA gene sequencing that targeted the V4 region. The relative abundance of microbial populations was compared using linear discriminant analysis effect size (LEfSe) statistical analysis for the identification of biomarkers between two or more groups, principal component analysis (PCA), and linear discriminant analysis (LDA). Microbial genome prediction was performed using the phylogenetic investigation of communities by reconstructing the unobserved states (PICRUSt) bioinformatics software. Results There was no significant difference in the alpha diversity of the fecal microbiota between the high-salt group and the control group. However, PCA showed structural segregation between the two groups. Further analysis using LEfSe showed that the intestinal contents in the high-salt group had significantly reduced populations of Lactobacillus and Prevotella NK3B31, and a significant increase in Alloprevotella and Prevotella 9, without physiological or pathological changes. Conclusions A pilot study in Wistar rats showed that high-salt intake was associated with a change in the composition of the intestinal microbiota.
Collapse
Affiliation(s)
- Zhaogang Dong
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Yuanbin Liu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Hongwei Pan
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Hongchun Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Xin Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Xiaofei Xu
- Infertility Center, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Ke Xiao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Min Liu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Zhiyun Xu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Lanbo Li
- Department of Animal Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Yi Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
39
|
Yang X, Liang S, Guo F, Ren Z, Yang X, Long F. Gut microbiota mediates the protective role of Lactobacillus plantarum in ameliorating deoxynivalenol-induced apoptosis and intestinal inflammation of broiler chickens. Poult Sci 2020; 99:2395-2406. [PMID: 32359574 PMCID: PMC7597391 DOI: 10.1016/j.psj.2019.10.034] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/07/2019] [Accepted: 10/11/2019] [Indexed: 12/02/2022] Open
Abstract
The protection of Lactobacillus plantarum JM113 against deoxynivalenol (DON)-induced apoptosis and intestinal inflammation on the jejunum of broiler chickens and the potential roles of gut microbiota were determined. A total of 144 one-day-old male broilers (Arbor Acres) were randomly divided into 3 treatment groups consisting of 6 replicates with 8 birds per replicate, including the CON (basal diet), the DON (basal diet + 10 mg/kg DON), and the DL (basal diet + 10 mg/kg DON + 1 × 109 CFU/kg L. plantarum JM113). The DON-diet decreased (P < 0.05) the mRNA expression of mucosal defense proteins and mechanistic target of rapamycin pathway genes. Meanwhile, DON challenge significantly increased Bcl-2-associated X gene/B-cell lymphoma 2 gene (Bcl-2) in the jejunum (P < 0.05) and demonstrated proapoptosis status. In contrast, the DL group showed normal immunity-related gene expression of jejunal mucosa and manifested a superior antiapoptosis status. Adding L. plantarum JM113 significantly raised (P < 0.05) propionic acid, n-butyric acid, and total short-chain fatty acids concentrations in cecal contents of birds fed with DON diet. In addition, DON exposure altered bacterial community structure and disturbed the abundance of several bacterial phyla, families, and genera, leading to dysbiosis. Supplementation with JM113 shifted the gut microbiota composition to that of the CON group. Finally, Spearman correlation analysis suggested that most positive correlations with the mRNA expression of immunity-related and apoptosis-regulatory gene were observed within the phylum Bacteroidetes, and most negative correlations with the indicators were observed within the phylum Firmicutes. The mRNA expression of Bcl-2, TLR2, mTOR, Raptor, and RPS6KB1 (P < 0.05), which are regarded as important cell proliferation and antiapoptosis parameters, were significantly negatively associated with the relative abundances of norank_f__Erysipelotrichaceae, Subdoligranulum, and Anaeroplasma, whereas they had a strong positive correlation with Ruminococcaceae_UCG-004, Alistipes, and Ruminococcaceae_NK4A214_group. These results implied that L. plantarum JM113 supplementation could ameliorate DON-induced apoptosis and intestinal inflammation via manipulating the bacterial community composition and could be used as a potential candidate to attenuate intestinal impairments.
Collapse
Affiliation(s)
- Xin Yang
- College of Animal Science and Technology, Northwest A & F University, Yangling, Shaanxi 712100, P.R. China
| | - Saisai Liang
- College of Animal Science and Technology, Northwest A & F University, Yangling, Shaanxi 712100, P.R. China
| | - Fangshen Guo
- College of Animal Science and Technology, Northwest A & F University, Yangling, Shaanxi 712100, P.R. China
| | - Zhouzheng Ren
- College of Animal Science and Technology, Northwest A & F University, Yangling, Shaanxi 712100, P.R. China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A & F University, Yangling, Shaanxi 712100, P.R. China.
| | - Fangyu Long
- College of Food Science and Engineering, Northwest A & F University, Yangling, Shaanxi 712100, P.R. China.
| |
Collapse
|
40
|
Chait A, den Hartigh LJ. Adipose Tissue Distribution, Inflammation and Its Metabolic Consequences, Including Diabetes and Cardiovascular Disease. Front Cardiovasc Med 2020; 7:22. [PMID: 32158768 PMCID: PMC7052117 DOI: 10.3389/fcvm.2020.00022] [Citation(s) in RCA: 716] [Impact Index Per Article: 143.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/10/2020] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue plays essential roles in maintaining lipid and glucose homeostasis. To date several types of adipose tissue have been identified, namely white, brown, and beige, that reside in various specific anatomical locations throughout the body. The cellular composition, secretome, and location of these adipose depots define their function in health and metabolic disease. In obesity, adipose tissue becomes dysfunctional, promoting a pro-inflammatory, hyperlipidemic and insulin resistant environment that contributes to type 2 diabetes mellitus (T2DM). Concurrently, similar features that result from adipose tissue dysfunction also promote cardiovascular disease (CVD) by mechanisms that can be augmented by T2DM. The mechanisms by which dysfunctional adipose tissue simultaneously promote T2DM and CVD, focusing on adipose tissue depot-specific adipokines, inflammatory profiles, and metabolism, will be the focus of this review. The impact that various T2DM and CVD treatment strategies have on adipose tissue function and body weight also will be discussed.
Collapse
Affiliation(s)
- Alan Chait
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Laura J den Hartigh
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
41
|
Han X, Shao H, Wang Y, Hu A, Chen R, Chen Q. Composition of the bacterial community in the gastrointestinal tract of Kunming mice. ELECTRON J BIOTECHN 2020. [DOI: 10.1016/j.ejbt.2019.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
42
|
Lo Re O, Mazza T, Giallongo S, Sanna P, Rappa F, Vinh Luong T, Li Volti G, Drovakova A, Roskams T, Van Haele M, Tsochatzis E, Vinciguerra M. Loss of histone macroH2A1 in hepatocellular carcinoma cells promotes paracrine-mediated chemoresistance and CD4 +CD25 +FoxP3 + regulatory T cells activation. Am J Cancer Res 2020; 10:910-924. [PMID: 31903159 PMCID: PMC6929991 DOI: 10.7150/thno.35045] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 10/06/2019] [Indexed: 12/11/2022] Open
Abstract
Rationale: Loss of histone macroH2A1 induces appearance of cancer stem cells (CSCs)-like cells in hepatocellular carcinoma (HCC). How CSCs interact with the tumor microenvironment and the adaptive immune system is unclear. Methods: We screened aggressive human HCC for macroH2A1 and CD44 CSC marker expression. We also knocked down (KD) macroH2A1 in HCC cells, and performed integrated transcriptomic and secretomic analyses. Results: Human HCC showed low macroH2A1 and high CD44 expression compared to control tissues. MacroH2A1 KD CSC-like cells transferred paracrinally their chemoresistant properties to parental HCC cells. MacroH2A1 KD conditioned media transcriptionally reprogrammed parental HCC cells activated regulatory CD4+/CD25+/FoxP3+ T cells (Tregs). Conclusions: Loss of macroH2A1 in HCC cells drives cancer stem-cell propagation and evasion from immune surveillance.
Collapse
|
43
|
Ahmad AF, Dwivedi G, O'Gara F, Caparros-Martin J, Ward NC. The gut microbiome and cardiovascular disease: current knowledge and clinical potential. Am J Physiol Heart Circ Physiol 2019; 317:H923-H938. [PMID: 31469291 DOI: 10.1152/ajpheart.00376.2019] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. The human body is populated by a diverse community of microbes, dominated by bacteria, but also including viruses and fungi. The largest and most complex of these communities is located in the gastrointestinal system and, with its associated genome, is known as the gut microbiome. Gut microbiome perturbations and related dysbiosis have been implicated in the progression and pathogenesis of CVD, including atherosclerosis, hypertension, and heart failure. Although there have been advances in the characterization and analysis of the gut microbiota and associated bacterial metabolites, the exact mechanisms through which they exert their action are not well understood. This review will focus on the role of the gut microbiome and associated functional components in the development and progression of atherosclerosis. Potential treatments to alter the gut microbiome to prevent or treat atherosclerosis and CVD are also discussed.
Collapse
Affiliation(s)
- Adilah F Ahmad
- Medical School, University of Western Australia, Perth, Western Australia, Australia.,Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
| | - Girish Dwivedi
- Medical School, University of Western Australia, Perth, Western Australia, Australia.,Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia.,Department of Cardiology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Fergal O'Gara
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Western Australia, Australia.,Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.,BIOMERIT Research Centre, School of Microbiology, University College Cork, National University of Ireland, Cork, Ireland.,Telethon Kids Institute, Children's Hospital, Perth, Western Australia, Australia
| | - Jose Caparros-Martin
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Western Australia, Australia.,Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Natalie C Ward
- Medical School, University of Western Australia, Perth, Western Australia, Australia.,School of Public Health, Curtin University, Perth Western Australia, Australia
| |
Collapse
|
44
|
Panebianco C, Potenza A, Andriulli A, Pazienza V. Exploring the microbiota to better understand gastrointestinal cancers physiology. Clin Chem Lab Med 2019; 56:1400-1412. [PMID: 29630505 DOI: 10.1515/cclm-2017-1163] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 02/26/2018] [Indexed: 02/07/2023]
Abstract
Gastrointestinal cancers account for around 40% of cancer-related deaths worldwide, representing a global health burden. There is a growing body of evidence highlighting the link between microbiota and gastrointestinal tumorigenesis and/or resistance to therapy. In the present manuscript, we reviewed the published studies on the relationship between the microbiota and the different gastrointestinal tumors, namely, gastric, colorectal and esophageal, including also the cancer of accessory organs such as liver and pancreas. There is an emergent interest in the manipulation of gastrointestinal microflora in order to understand the gastrointestinal tumorigenesis' processes and the establishment of chemoresistance mechanisms.
Collapse
Affiliation(s)
- Concetta Panebianco
- Gastroenterology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo (FG), Italy
| | - Adele Potenza
- Dietetic and Clinical Nutrition Unit IRCCS "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo (FG), Italy
| | - Angelo Andriulli
- Gastroenterology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo (FG), Italy
| | - Valerio Pazienza
- Gastroenterology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, Viale dei Cappuccini, 1, 71013 San Giovanni Rotondo (FG), Italy, Phone: +39-0882.416281, Fax: +39-0882.410271
| |
Collapse
|
45
|
Wang T, Fan C, Yao A, Xu X, Zheng G, You Y, Jiang C, Zhao X, Hou Y, Hung MC, Lin X. The Adaptor Protein CARD9 Protects against Colon Cancer by Restricting Mycobiota-Mediated Expansion of Myeloid-Derived Suppressor Cells. Immunity 2019; 49:504-514.e4. [PMID: 30231984 DOI: 10.1016/j.immuni.2018.08.018] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 03/29/2018] [Accepted: 07/06/2018] [Indexed: 01/19/2023]
Abstract
The adaptor protein CARD9 links detection of fungi by surface receptors to the activation of the NF-κB pathway. Mice deficient in CARD9 exhibit dysbiosis and are more susceptible to colitis. Here we examined the impact of Card9 deficiency in the development of colitis-associated colon cancer (CAC). Treatment of Card9-/- mice with AOM-DSS resulted in increased tumor loads as compared to WT mice and in the accumulation of myeloid-derived suppressor cells (MDSCs) in tumor tissue. The impaired fungicidal functions of Card9-/- macrophages led to increased fungal loads and variation in the overall composition of the intestinal mycobiota, with a notable increase in C. tropicalis. Bone marrow cells incubated with C. tropicalis exhibited MDSC features and suppressive functions. Fluconazole treatment suppressed CAC in Card9-/- mice and was associated with decreased MDSC accumulation. The frequency of MDSCs in tumor tissues of colon cancer patients correlated positively with fungal burden, pointing to the relevance of this regulatory axis in human disease.
Collapse
Affiliation(s)
- Tingting Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Department of Molecular and Cellular Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Chaogang Fan
- General Surgery, Jinling Hospital affiliated Medical School, Nanjing University, Nanjing 210093, China
| | - Anran Yao
- The State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Xingwei Xu
- General Surgery, Jinling Hospital affiliated Medical School, Nanjing University, Nanjing 210093, China
| | - Guoxing Zheng
- Institute for Immunology, Tsinghua University School of Medicine, Tsinghua, Beijing 100084, China; Tsinghua University-Peking University Joint Center for Life Sciences, Beijing 100084, China
| | - Yun You
- Department of Molecular and Cellular Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Changying Jiang
- Department of Molecular and Cellular Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xueqiang Zhao
- Institute for Immunology, Tsinghua University School of Medicine, Tsinghua, Beijing 100084, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xin Lin
- Institute for Immunology, Tsinghua University School of Medicine, Tsinghua, Beijing 100084, China; Tsinghua University-Peking University Joint Center for Life Sciences, Beijing 100084, China.
| |
Collapse
|
46
|
Chen DM, Zhang ML, Shi ZQ, Li CQ, Wang Q, Song JP, Xu Q, Li H, Zeng QP. Anti-inflammatory and Anti-infectious Dietary Paradigms May Be Crucial for Visceral Weight Reduction. Front Immunol 2019; 10:422. [PMID: 30906298 PMCID: PMC6418406 DOI: 10.3389/fimmu.2019.00422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 02/18/2019] [Indexed: 01/01/2023] Open
Affiliation(s)
- Dong-Mei Chen
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meng-Le Zhang
- School of Basic Course, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhu-Qing Shi
- Science and Technology Industrial Park, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chang-Qing Li
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jian-Ping Song
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qin Xu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - He Li
- School of Basic Course, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qing-Ping Zeng
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
47
|
Cerdó T, García-Santos JA, G Bermúdez M, Campoy C. The Role of Probiotics and Prebiotics in the Prevention and Treatment of Obesity. Nutrients 2019; 11:E635. [PMID: 30875987 PMCID: PMC6470608 DOI: 10.3390/nu11030635] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 03/06/2019] [Accepted: 03/11/2019] [Indexed: 02/06/2023] Open
Abstract
Obesity is a global pandemic complex to treat due to its multifactorial pathogenesis-an unhealthy lifestyle, neuronal and hormonal mechanisms, and genetic and epigenetic factors are involved. Scientific evidence supports the idea that obesity and metabolic consequences are strongly related to changes in both the function and composition of gut microbiota, which exert an essential role in modulating energy metabolism. Modifications of gut microbiota composition have been associated with variations in body weight and body mass index. Lifestyle modifications remain as primary therapy for obesity and related metabolic disorders. New therapeutic strategies to treat/prevent obesity have been proposed, based on pre- and/or probiotic modulation of gut microbiota to mimic that found in healthy non-obese subjects. Based on human and animal studies, this review aimed to discuss mechanisms through which gut microbiota could act as a key modifier of obesity and related metabolic complications. Evidence from animal studies and human clinical trials suggesting potential beneficial effects of prebiotic and various probiotic strains on those physical, biochemical, and metabolic parameters related to obesity is presented. As a conclusion, a deeper knowledge about pre-/probiotic mechanisms of action, in combination with adequately powered, randomized controlled follow-up studies, will facilitate the clinical application and development of personalized healthcare strategies.
Collapse
Affiliation(s)
- Tomás Cerdó
- Department of Pediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain.
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
- Instituto de Investigación Biosanitaria ibs, GRANADA, Health Sciences Technological Park, 18012 Granada, Spain.
| | - José Antonio García-Santos
- Department of Pediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain.
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
- Instituto de Investigación Biosanitaria ibs, GRANADA, Health Sciences Technological Park, 18012 Granada, Spain.
| | - Mercedes G Bermúdez
- Department of Pediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain.
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
- Instituto de Investigación Biosanitaria ibs, GRANADA, Health Sciences Technological Park, 18012 Granada, Spain.
| | - Cristina Campoy
- Department of Pediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain.
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
- Instituto de Investigación Biosanitaria ibs, GRANADA, Health Sciences Technological Park, 18012 Granada, Spain.
- Spanish Network of Biomedical Research in Epidemiology and Public Health (CIBERESP), Granada's node, Carlos III Health Institute of Health Carlos III, 28029 Madrid, Spain.
| |
Collapse
|
48
|
|
49
|
Porras D, Nistal E, Martínez-Flórez S, González-Gallego J, García-Mediavilla MV, Sánchez-Campos S. Intestinal Microbiota Modulation in Obesity-Related Non-alcoholic Fatty Liver Disease. Front Physiol 2018; 9:1813. [PMID: 30618824 PMCID: PMC6305464 DOI: 10.3389/fphys.2018.01813] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 12/05/2018] [Indexed: 12/12/2022] Open
Abstract
Obesity and associated comorbidities, including non-alcoholic fatty liver disease (NAFLD), are a major concern to public well-being worldwide due to their high prevalence among the population, and its tendency on the rise point to as important threats in the future. Therapeutic approaches for obesity-associated disorders have been circumscribed to lifestyle modifications and pharmacological therapies have demonstrated limited efficacy. Over the last few years, different studies have shown a significant role of intestinal microbiota (IM) on obesity establishment and NAFLD development. Therefore, modulation of IM emerges as a promising therapeutic strategy for obesity-associated diseases. Administration of prebiotic and probiotic compounds, fecal microbiota transplantation (FMT) and exercise protocols have shown a modulatory action over the IM. In this review we provide an overview of current approaches targeting IM which have shown their capacity to counteract NAFLD and metabolic syndrome features in human patients and animal models.
Collapse
Affiliation(s)
- David Porras
- Institute of Biomedicine, University of León, León, Spain
| | - Esther Nistal
- Institute of Biomedicine, University of León, León, Spain.,Department of Gastroenterology, Complejo Asistencial Universitario de León, León, Spain
| | | | - Javier González-Gallego
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain
| | - María Victoria García-Mediavilla
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain
| | - Sonia Sánchez-Campos
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain
| |
Collapse
|
50
|
Pretorius L, Kell DB, Pretorius E. Iron Dysregulation and Dormant Microbes as Causative Agents for Impaired Blood Rheology and Pathological Clotting in Alzheimer's Type Dementia. Front Neurosci 2018; 12:851. [PMID: 30519157 PMCID: PMC6251002 DOI: 10.3389/fnins.2018.00851] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/30/2018] [Indexed: 12/13/2022] Open
Abstract
Alzheimer’s disease and other similar dementias are debilitating neurodegenerative disorders whose etiology and pathogenesis remain largely unknown, even after decades of research. With the anticipated increase in prevalence of Alzheimer’s type dementias among the more susceptible aging population, the need for disease-modifying treatments is urgent. While various hypotheses have been put forward over the last few decades, we suggest that Alzheimer’s type dementias are triggered by external environmental factors, co-expressing in individuals with specific genetic susceptibilities. These external stressors are defined in the Iron Dysregulation and Dormant Microbes (IDDM) hypothesis, previously put forward. This hypothesis is consistent with current literature in which serum ferritin levels of individuals diagnosed with Alzheimer’s disease are significantly higher compared those of age- and gender-matched controls. While iron dysregulation contributes to oxidative stress, it also causes microbial reactivation and virulence of the so-called dormant blood (and tissue) microbiome. Dysbiosis (changes in the microbiome) or previous infections can contribute to the dormant blood microbiome (atopobiosis1), and also directly promotes systemic inflammation via the amyloidogenic formation and shedding of potent inflammagens such as lipopolysaccharides. The simultaneous iron dysregulation and microbial aberrations affect the hematological system, promoting fibrin amylodiogenesis, and pathological clotting. Systemic inflammation and oxidative stress can contribute to blood brain barrier permeability and the ensuing neuro-inflammation, characteristic of Alzheimer’s type dementias. While large inter-individual variability exists, especially concerning disease pathogenesis, the IDDM hypothesis acknowledges primary causative factors which can be targeted for early diagnosis and/or for prevention of disease progression.
Collapse
Affiliation(s)
- Lesha Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Douglas B Kell
- School of Chemistry, The University of Manchester, Manchester, United Kingdom.,The Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| | - Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|