1
|
Feng C, Yan J, Luo T, Zhang H, Zhang H, Yuan Y, Chen Y, Chen H. Vitamin B12 ameliorates gut epithelial injury via modulating the HIF-1 pathway and gut microbiota. Cell Mol Life Sci 2024; 81:397. [PMID: 39261351 PMCID: PMC11391010 DOI: 10.1007/s00018-024-05435-5] [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: 04/30/2024] [Revised: 08/27/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
Inflammatory bowel diseases (IBDs) are immune chronic diseases characterized by recurrent episodes, resulting in continuous intestinal barrier damage and intestinal microbiota dysbiosis. Safe strategies aimed at stabilizing and reducing IBDs recurrence have been vigorously pursued. Here, we constructed a recurrent intestinal injury Drosophila model and found that vitamin B12 (VB12), an essential co-factor for organism physiological functions, could effectively protect the intestine and reduce dextran sulfate sodium-induced intestinal barrier disruption. VB12 also alleviated microbial dysbiosis in the Drosophila model and inhibited the growth of gram-negative bacteria. We demonstrated that VB12 could mitigate intestinal damage by activating the hypoxia-inducible factor-1 signaling pathway in injured conditions, which was achieved by regulating the intestinal oxidation. In addition, we also validated the protective effect of VB12 in a murine acute colitis model. In summary, we offer new insights and implications for the potential supportive role of VB12 in the management of recurrent IBDs flare-ups.
Collapse
Affiliation(s)
- Chenxi Feng
- Division of Gastrointestinal Surgery, Laboratory of Stem Cell and Anti-Aging Research, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jinhua Yan
- Center of Gerontology and Geriatrics, Laboratory of Stem Cell and Anti-Aging Research, National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ting Luo
- Center of Gerontology and Geriatrics, Laboratory of Stem Cell and Anti-Aging Research, National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Hong Zhang
- Department of Gastroenterology and Hepatology and Laboratory of Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Hu Zhang
- Department of Gastroenterology and Hepatology and Laboratory of Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yu Yuan
- Division of Gastrointestinal Surgery, Laboratory of Stem Cell and Anti-Aging Research, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi Chen
- Division of Gastrointestinal Surgery, Laboratory of Stem Cell and Anti-Aging Research, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Haiyang Chen
- Division of Gastrointestinal Surgery, Laboratory of Stem Cell and Anti-Aging Research, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Center of Gerontology and Geriatrics, Laboratory of Stem Cell and Anti-Aging Research, National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
2
|
Huang Z, Teng W, Yao L, Xie K, Hang S, He R, Li Y. mTOR signaling pathway regulation HIF-1 α effects on LPS induced intestinal mucosal epithelial model damage. BMC Mol Cell Biol 2024; 25:13. [PMID: 38654163 PMCID: PMC11036631 DOI: 10.1186/s12860-024-00509-5] [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: 04/14/2023] [Accepted: 04/05/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Sepsis-induced small-intestinal injury is associated with increased morbidity and mortality. Our previous study and other papers have shown that HIF-1α has a protective effect on intestinal mucosal injury in septic rats. The purpose of this study is to further verify the protective effect of HIF-1α on intestinal mucosa and its molecular mechanism in vitro experiments. METHODS Caco-2 cells were selected and experiment was divided into 2 parts. Part I: HIF-1α activator and inhibitor were used to treat lipopolysacchrides (LPS)-stimulated Caco-2 cells respectively, to explore the effect of HIF-1α on LPS induced Caco-2 cell epithelial model; Part II: mTOR activator or inhibitor combined with or without HIF-1α activator, inhibitor to treat LPS-stimulated Caco-2 cells respectively, and then the molecular mechanism of HIF-1α reducing LPS induced Caco-2 cell epithelial model damage was detected. RESULTS The results showed that HIF-1α activator decreased the permeability and up regulated tight junction (TJ) expression, while HIF-1α inhibitor had the opposite effect with the HIF-1α activator. mTOR activation increased, while mTOR inhibition decreased HIF-1α protein and expression of its downstream target molecules, which can be attenuated by HIF-1α activator or inhibitor. CONCLUSION This study once again confirmed that HIF-1α alleviates LPS-induced mucosal epithelial model damage through P70S6K signalling pathway. It is of great value to explore whether HIF-2α plays crucial roles in the regulation of mucosal epithelial model functions in the future.
Collapse
Affiliation(s)
- Zeyong Huang
- Department of Anesthesiology, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren College, 310015, Hangzhou, China
| | - Wenbin Teng
- Department of Anesthesiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, 310001, Hangzhou, China
| | - Liuxu Yao
- Rehabilitation Medicine Center, Department of Anesthesiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, China
| | - Kai Xie
- Department of Anesthesiology, Shaoxing People's Hospital, Zhejiang University, 312000, Shaoxing, China
| | - Suqin Hang
- Department of Anesthesiology, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren College, 310015, Hangzhou, China
| | - Rui He
- Department of Anesthesiology, Shaoxing People's Hospital, Zhejiang University, 312000, Shaoxing, China.
| | - Yuhong Li
- Department of Anesthesiology, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren College, 310015, Hangzhou, China.
- Department of Anesthesiology, Shulan (Hangzhou) Hospital, Shulan International Medical College, Shuren University, 848 Dongxin Road, Xiacheng District, 310004, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Chen TC, Chang SW. Non-lethal exposure to short-wavelength light-emitting diodes modulates tight-junction structure in human corneal epithelial cells via cAMP-dependent signaling. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 252:112869. [PMID: 38368634 DOI: 10.1016/j.jphotobiol.2024.112869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/30/2024] [Accepted: 02/13/2024] [Indexed: 02/20/2024]
Abstract
Light-emitting diodes (LED)-derived lights have been widely used as a medical treatment in photobiomodulation (PBM). However, the PBM effects in ophthalmology are less well investigated. Herein, we explored the effect of LED-generated light on the tight-junction (TJ) formation in human corneal epithelial cells (HCEs). The HCEs were separately exposed to monochromatic LEDs at wavelengths of 365 nm (UVA), 420 nm (violet), 470 nm (blue), 530 nm (green), 590 nm (amber), 660 nm (deep red), and 740 nm (far red) at 10 J/cm2/day for 1 and 2 days. Long-term cultivation of HCEs without LED exposure for up to 14 days was established as a control. The effects of both LED wavelength and culture duration on cell morphology, cAMP-regulated proteins, TJ-associated proteins, and cell growth-associated proteins were also analyzed. Together with the increase in cell number during prolonged cultivation, cAMP, ZO-1, ZO-2, CLDN1, and CLDN4 all increased significantly during long-term cultivation without LED exposure. There was no difference in HCE viability after exposure to all monochromatic LEDs at an accumulated dose of 20 J/cm2. As determined by immunoblotting, UVA, violet, and blue light increased intracellular cAMP, ZO-1, ZO-2, CLDN1, and CLDN4 expression, respectively. UVA and violet, but not blue, light increased PKAreg-pS77 expression. However, none of the other treatments changed the expression of PKAcat-pT197, VASP-pS157, Bax, Bcl-2, or Bcl-xL. Immunofluorescence staining confirmed the formation of TJ structures. The expressions of ZO-1, ZO-2, CLDN1, and CLDN4 as well as TJ structures 2 days following UVA, violet, and blue exposure were similar to those of control cells after 9 days of cultivation. We conclude that short-wavelength LEDs at non-lethal exposure intensities accelerated the formation of TJ structure in HCEs via a cAMP-dependent regulatory cascade.
Collapse
Affiliation(s)
- Tsan-Chi Chen
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan; Department of Medical Research, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Shu-Wen Chang
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan; College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
4
|
Liu W, Xie J, Jiang H, Zhou J, Lu X, Zuo D, Dong L, Chen Q. Mannose attenuates intestinal epithelial tight junction damage in experimental colitis mice by activating the AXIN-AMPK pathway. Int Immunopharmacol 2024; 127:111319. [PMID: 38064812 DOI: 10.1016/j.intimp.2023.111319] [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: 09/20/2023] [Revised: 11/20/2023] [Accepted: 11/27/2023] [Indexed: 01/18/2024]
Abstract
Mannose is a unique natural sugar that can be found in a variety of fruits and vegetables. During the past decades, mannose has been reported to be effective in promoting immune tolerance and suppressing inflammatory diseases. Metabolic dysfunction and altered inflammation have clear implications for the development and progression of inflammatory diseases. Herein, we intended to reveal the molecular mechanism of mannose in protecting against intestinal epithelial damage in experimental colitis. We showed that mannose treatment significantly attenuated dextran sodium sulfate (DSS)-induced intestinal barrier damage. The AMPK pathway was responsible for the mannose-mediated protective effect in DSS-induced intestinal epithelial damage. Mechanistically, mannose promoted the axis inhibition protein (AXIN)-based AMPK activation, thereby preventing mitochondrial dysfunction and tight junction disruption in response to the DSS challenge. Cumulatively, the results indicate the use of mannose as a novel approach to treat IBD and other diseases involving tight junction dysfunction. The therapeutic effect of mannose is related to its regulatory function in AMPK pathway activation.
Collapse
Affiliation(s)
- Wenxin Liu
- Clinical Research Institute of Zhanjiang, Central People's Hospital of Zhanjiang, Guangdong Medical University, Zhanjiang, Guangdong 524045, China
| | - Jingwen Xie
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Honglian Jiang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, Guangzhou, Guangdong 510030, China
| | - Jia Zhou
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xiao Lu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Daming Zuo
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, China; Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Lijun Dong
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Qingyun Chen
- Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou 510080, Guangdong 519041, China.
| |
Collapse
|
5
|
Morello S, Cicala C. Ecto-5'-nucleotidase (CD73): an emerging role as prognostic factor in allergic sensitization. Inflamm Res 2024; 73:111-115. [PMID: 38087076 DOI: 10.1007/s00011-023-01820-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 01/10/2024] Open
Abstract
Over the years, the importance of the epithelium in the assessment of allergic sensitization and development of allergic diseases has increased. Sensitization to allergens appears to be influenced by genetic and external environmental factors. However, not all subjects exposed to environmental factors that damage epithelial cells suffer from allergic diseases. On this basis, identifying the signaling pathways that characterize the different phenotypes and endotypes of allergy is of high priority for a successful personalized therapy. Ecto-5'-nucleotidase/CD73 is a membrane-bound enzyme responsible for extracellular adenosine accumulation from AMP derived, in turn, from the hydrolysis of extracellular ATP. Current knowledge suggests that CD73 expression and enzymatic activity at epithelial barriers would be of fundamental importance to control the first defense against allergens, by preserving both physical and immunological epithelial barrier functions. Here, we highlight evidence for a crucial role of CD73 in features of allergic sensitization and the potential of this enzyme as prognostic marker and target of therapeutic intervention.
Collapse
Affiliation(s)
- Silvana Morello
- Department of Pharmacy, University of Salerno, 84084, Fisciano, Italy.
| | - Carla Cicala
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131, Naples, Italy.
| |
Collapse
|
6
|
Frusciante MR, Signori MF, Parmeggiani B, Grings M, Pramio J, Cecatto C, de Andrade Silveira J, Aubin MR, Santos LA, Paz AH, Wajner M, Leipnitz G. Disruption of Bioenergetics in the Intestine of Wistar Rats Caused by Hydrogen Sulfide and Thiosulfate: A Potential Mechanism of Chronic Hemorrhagic Diarrhea in Ethylmalonic Encephalopathy. Cell Biochem Biophys 2023; 81:683-695. [PMID: 37589888 DOI: 10.1007/s12013-023-01161-0] [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] [Accepted: 08/05/2023] [Indexed: 08/18/2023]
Abstract
Ethylmalonic encephalopathy (EE) is a severe inherited metabolic disorder that causes tissue accumulation of hydrogen sulfide (sulfide) and thiosulfate in patients. Although symptoms are predominantly neurological, chronic hemorrhagic diarrhea associated with intestinal mucosa abnormalities is also commonly observed. Considering that the pathophysiology of intestinal alterations in EE is virtually unknown and that sulfide and thiosulfate are highly reactive molecules, the effects of these metabolites were investigated on bioenergetic production and transfer in the intestine of rats. We observed that sulfide reduced NADH- and FADH2-linked mitochondrial respiration in the intestine, which was avoided by reduced glutathione (GSH) but not by melatonin. Thiosulfate did not change respiration. Moreover, both metabolites markedly reduced the activity of total, cytosolic and mitochondrial isoforms of creatine kinase (CK) in rat intestine. Noteworthy, the addition of GSH but not melatonin, apocynin, and Trolox (hydrosoluble vitamin E) prevented the change in the activities of total CK and its isoforms caused by sulfide and thiosulfate, suggesting a direct protein modification on CK structure by these metabolites. Sulfide further increased thiol content in the intestine, suggesting a modulation in the redox state of these groups. Finally, sulfide and thiosulfate decreased the viability of Caco-2 intestinal cells. Our data suggest that bioenergetic impairment caused by sulfide and thiosulfate is a mechanism involved in the gastrointestinal abnormalities found in EE.
Collapse
Affiliation(s)
- Marina Rocha Frusciante
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 2600 Ramiro Barcelos Street-Attached, Porto Alegre, RS, 90035-003, Brazil
| | - Marian Flores Signori
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 2600 Ramiro Barcelos Street-Attached, Porto Alegre, RS, 90035-003, Brazil
| | - Belisa Parmeggiani
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 2600 Ramiro Barcelos Street-Attached, Porto Alegre, RS, 90035-003, Brazil
| | - Mateus Grings
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 2600 Ramiro Barcelos Street-Attached, Porto Alegre, RS, 90035-003, Brazil
| | - Julia Pramio
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 2600 Ramiro Barcelos Street-Attached, Porto Alegre, RS, 90035-003, Brazil
| | - Cristiane Cecatto
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 2600 Ramiro Barcelos Street-Attached, Porto Alegre, RS, 90035-003, Brazil
| | - Josyane de Andrade Silveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 2600 Ramiro Barcelos Street-Attached, Porto Alegre, RS, 90035-003, Brazil
| | - Mariana Rauback Aubin
- Programa de Pós-Graduação em Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 500 Sarmento Leite Street, Porto Alegre, RS, 90035-190, Brazil
- Laboratório de Células, Tecidos e Genes - Centro de Pesquisa Experimental, HCPA, Porto Alegre, RS, Brazil
| | - Larissa Aguiar Santos
- Programa de Pós-Graduação em Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 500 Sarmento Leite Street, Porto Alegre, RS, 90035-190, Brazil
- Laboratório de Células, Tecidos e Genes - Centro de Pesquisa Experimental, HCPA, Porto Alegre, RS, Brazil
| | - Ana Helena Paz
- Programa de Pós-Graduação em Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 500 Sarmento Leite Street, Porto Alegre, RS, 90035-190, Brazil
- Laboratório de Células, Tecidos e Genes - Centro de Pesquisa Experimental, HCPA, Porto Alegre, RS, Brazil
| | - Moacir Wajner
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 2600 Ramiro Barcelos Street-Attached, Porto Alegre, RS, 90035-003, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 2600 Ramiro Barcelos Street-Attached, Porto Alegre, RS, 90035-003, Brazil
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, 2350 Ramiro Barcelos Street, Porto Alegre, RS, 90035-903, Brazil
| | - Guilhian Leipnitz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 2600 Ramiro Barcelos Street-Attached, Porto Alegre, RS, 90035-003, Brazil.
- Programa de Pós-Graduação em Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 500 Sarmento Leite Street, Porto Alegre, RS, 90035-190, Brazil.
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 2600 Ramiro Barcelos Street-Attached, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
7
|
Chaukimath P, Frankel G, Visweswariah SS. The metabolic impact of bacterial infection in the gut. FEBS J 2023; 290:3928-3945. [PMID: 35731686 DOI: 10.1111/febs.16562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 06/02/2022] [Accepted: 06/21/2022] [Indexed: 08/17/2023]
Abstract
Bacterial infections of the gut are one of the major causes of morbidity and mortality worldwide. The interplay between the pathogen and the host is finely balanced, with the bacteria evolving to proliferate and establish infection. In contrast, the host mounts a response to first restrict and then eliminate the infection. The intestine is a rapidly proliferating tissue, and metabolism is tuned to cater to the demands of proliferation and differentiation along the crypt-villus axis (CVA) in the gut. As bacterial pathogens encounter the intestinal epithelium, they elicit changes in the host cell, and core metabolic pathways such as the tricarboxylic acid (TCA) cycle, lipid metabolism and glycolysis are affected. This review highlights the mechanisms utilized by diverse gut bacterial pathogens to subvert host metabolism and describes host responses to the infection.
Collapse
Affiliation(s)
- Pooja Chaukimath
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Gad Frankel
- Centre for Molecular Bacteriology and Infection and Department of Life Sciences, Imperial College, London, UK
| | - Sandhya S Visweswariah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| |
Collapse
|
8
|
Wiącek J, Szurkowska J, Kryściak J, Galecka M, Karolkiewicz J. No changes in the abundance of selected fecal bacteria during increased carbohydrates consumption period associated with the racing season in amateur road cyclists. PeerJ 2023; 11:e14594. [PMID: 36700000 PMCID: PMC9869777 DOI: 10.7717/peerj.14594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/28/2022] [Indexed: 01/22/2023] Open
Abstract
Background Cyclists often use high-carbohydrate, low-fiber diets to optimize the glycogen stores and to avoid the gastrointestinal distress during both, the trainings and the competitions. The impact of such dietary changes on gut microbiota is not fully known. Methods We assessed the abundances of Faecalibacterium prausnitzii, Akkermansia muciniphila, Bifidobacterium spp., and Bacteroides spp. and the fecal pH in 14 amateur cyclists during the racing season. Eleven healthy men formed the control group. Results Despite significant differences in the diet composition and physical endurance levels of amateur cyclists before the competition season (1st term) and control group (carbohydrates: 52.2% ± 4.9% vs 41.9% ± 6.6%; VO2max: 56.1 ± 6.0 vs 39.7 ± 7.7; p < 0.01; respectively), we did not observe any significant differences in studied gut bacteria abundances or fecal pH between the groups. Although the cyclists' carbohydrates consumption (2nd term) have increased throughout the season (4.48 g/kg b.w. ± 1.56 vs 5.18 g/kg b.w. ± 1.99; p < 0.05), the studied gut bacteria counts and fecal pH remained unchanged. It seems that the amateur cyclists' diet with increased carbohydrates intake does not alter the gut microbiota, but further research is needed to assess the potential impact of even higher carbohydrates consumption (over 6 g/kg b.w.).
Collapse
Affiliation(s)
- Jakub Wiącek
- Department of Food and Nutrition, Poznan University of Physical Education, Poznań, Greater Poland, Poland
| | - Joanna Szurkowska
- Department of Food and Nutrition, Poznan University of Physical Education, Poznań, Greater Poland, Poland
| | - Jakub Kryściak
- Department of Physiology and Biochemistry, Poznan University of Physical Education, Poznań, Greater Poland, Poland
| | | | - Joanna Karolkiewicz
- Department of Food and Nutrition, Poznan University of Physical Education, Poznań, Greater Poland, Poland
| |
Collapse
|
9
|
Mannose ameliorates experimental colitis by protecting intestinal barrier integrity. Nat Commun 2022; 13:4804. [PMID: 35974017 PMCID: PMC9381535 DOI: 10.1038/s41467-022-32505-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 08/02/2022] [Indexed: 12/23/2022] Open
Abstract
Metabolite alteration has been associated with the pathogenesis of inflammatory bowel disease (IBD), including colitis. Mannose, a natural bioactive monosaccharide that is involved in metabolism and synthesis of glycoproteins, exhibits anti-inflammatory and anti-oxidative activities. We show here that the circulating level of mannose is increased in patients with IBD and mice with experimental colitis. Mannose treatment attenuates intestinal barrier damage in two mouse colitis models, dextran sodium sulfate (DSS)-induced colitis and spontaneous colitis in IL-10-deficient mice. We demonstrate that mannose treatment enhanced lysosomal integrity and limited the release of cathepsin B, preventing mitochondrial dysfunction and myosin light chain kinase (MLCK)-induced tight junction disruption in the context of intestinal epithelial damage. Mannose exerts a synergistic therapeutic effect with mesalamine on mouse colitis. Cumulatively, the results indicate that mannose supplementation may be an optional approach to the treatment of colitis and other diseases associated with intestinal barrier dysfunction.
Collapse
|
10
|
Wen J, Lyu P, Stolzer I, Xu J, Gießl A, Lin Z, Andreev D, Kachler K, Song R, Meng X, Cao S, Guggino G, Ciccia F, Günther C, Schett G, Bozec A. Epithelial HIF2α expression induces intestinal barrier dysfunction and exacerbation of arthritis. Ann Rheum Dis 2022; 81:annrheumdis-2021-222035. [PMID: 35710307 DOI: 10.1136/annrheumdis-2021-222035] [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: 12/20/2021] [Accepted: 05/14/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To investigate how the mucosal barrier in the intestine influences the development of arthritis, considering that metabolic changes in the intestinal epithelium influence its barrier function. METHODS Intestinal hypoxia inducible factor (HIF)-2α expression was assessed before, at onset and during experimental arthritis and human rheumatoid arthritis (RA). Intestinal epithelial cell-specific HIF2α conditional knock-out mice were generated (HIF2α∆IEC) and subjected to collagen-induced arthritis. Clinical and histological courses of arthritis were recorded; T-cell and B-cell subsets were analysed in the gut and secondary lymphatic organs; and intestinal epithelial cells were subjected to molecular mRNA sequencing in HIF2α∆IEC and littermate control mice. The gut intestinal HIF2α target genes were delineated by chromatin immunoprecipitation and luciferase experiments. Furthermore, pharmacological HIF2α inhibitor PT2977 was used for inhibition of arthritis. RESULTS Intestinal HIF2α expression peaked at onset of experimental arthritis and RA. Conditionally, deletion of HIF2α in gut epithelial cells inhibited arthritis and was associated with improved intestinal barrier function and less intestinal and lymphatic Th1 and Th17 activation. Mechanistically, HIF2α induced the transcription of the pore-forming claudin (CLDN)-15, which inhibits intestinal barrier integrity. Furthermore, treatment with HIF2α inhibitor decreased claudin-15 expression in epithelial cells and inhibited arthritis. CONCLUSION These findings show that the HIF2α-CLDN15 axis is critical for the breakdown of intestinal barrier function at onset of arthritis, highlighting the functional link between intestinal homeostasis and arthritis.
Collapse
Affiliation(s)
- Jinming Wen
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Pang Lyu
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Iris Stolzer
- Department of Internal Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jin Xu
- Department of Orthopaedic Surgery, Sun Yat-Sen University, Guangzhou, China
| | - Andreas Gießl
- Department of Animal Physiology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Zhen Lin
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Darja Andreev
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Katerina Kachler
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Rui Song
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Xianyi Meng
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Shan Cao
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Giuliana Guggino
- Rheumatology Section, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University Hospital P. Giaccone, Palermo, Italy
| | - Francesco Ciccia
- Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Claudia Günther
- Department of Internal Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
11
|
Ardizzone A, Lanza M, Casili G, Campolo M, Paterniti I, Cuzzocrea S, Esposito E. Efficacy of a Novel Therapeutic, Based on Natural Ingredients and Probiotics, in a Murine Model of Multiple Food Intolerance and Maldigestion. Nutrients 2022; 14:nu14112251. [PMID: 35684051 PMCID: PMC9182885 DOI: 10.3390/nu14112251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 02/07/2023] Open
Abstract
Patients with hypersensitive gut mucosa often suffer from food intolerances (FIs) associated with an inadequate gastrointestinal function that affects 15-20% of the population. Current treatments involve elimination diets, but require careful control, are difficult to maintain long-term, and diagnosis remains challenging. This study aims to evaluate the beneficial effects of a novel therapeutic of natural (NTN) origin containing food-grade polysaccharides, proteins, and grape seed extract to restore intestinal function in a murine model of fructose, carbohydrate, and fat intolerances. All experiments were conducted in four-week-old male CD1 mice. To induce FIs, mice were fed with either a high-carbohydrate diet (HCD), high-fat diet (HFD), or high-fructose diet (HFrD), respectively. After two weeks of treatment, several parameters and endpoints were evaluated such as food and water intake, body weight, histological score in several organs, gut permeability, intestinal epithelial integrity, and biochemical endpoints. Our results demonstrated that the therapeutic agent significantly restored gut barrier integrity and permeability compromised by every FIs induction. Restoration of intestinal function by NTN treatment has consequently improved tissue damage in several functional organs involved in the diagnostic of each intolerance such as the pancreas for HCD and liver for HFD and HFrD. Taken together, our results support NTN as a promising natural option in the non-pharmacological strategy for the recovery of intestinal dysregulation, supporting the well-being of the gastrointestinal tract.
Collapse
|
12
|
Joung EJ, Cao L, Gwon WG, Kwon MS, Lim KT, Kim HR. Meroterpenoid-Rich Ethanoic Extract of Sargassum macrocarpum Ameliorates Dextran Sulfate Sodium-Induced Colitis in Mice. Foods 2022; 11:foods11030329. [PMID: 35159480 PMCID: PMC8834051 DOI: 10.3390/foods11030329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 11/28/2022] Open
Abstract
Colitis is a colon mucosal disorder characterized by intestinal damage and inflammation. This current study aimed to evaluate the effect of meroterpenoid-rich ethanoic extract of a brown algae, Sargassum macrocarpum (MES) on dextran sulfate sodium (DSS)-induced colitis in mice and explore the possible mechanisms. Mice were given 4% DSS in drinking water for 7 days to induce colitis, followed by 3 days of regular water. MES (12 mg/kg body weight) or celecoxib (10 mg/kg body weight) was administrated orally to mice on a daily basis during these 10 days. Both MES and celecoxib supplementations significantly attenuated DSS-induced weight loss, shortening of colon length, elevated myeloperoxidase activity as well as histomorphological changes of colon. MES and celecoxib reduced the inflammation level of colon tissue, as indicated by its suppression on a panel of pro-inflammatory cytokines, including interleukin (IL)-1β, IL-17, tumor necrosis factor α, and interferon γ, and a group of inflammatory proteins, including intracellular adhesion molecule 1, vascular adhesion molecule 1, matrix metalloproteinase (MMP)-2, MMP-9, MMP-13, and inducible nitric oxidase. In addition, their administration down-regulated pro-inflammatory cytokines in serum. Moreover, the supplementation of MES suppressed the DSS-induced hyperactivation of Akt, JNK, and NF-κB signaling pathways. Taken together, our results demonstrate that MES ameliorates DSS-induced colitis in mice, suggesting that MES may have therapeutic implications for the treatment of colitis.
Collapse
Affiliation(s)
- Eun-Ji Joung
- Department of Food Science and Nutrition, Pukyong National University, Busan 608737, Korea; (E.-J.J.); (W.-G.G.); (M.-S.K.)
| | - Lei Cao
- Institute of Marine Sciences, Pukyong National University, Busan 608737, Korea;
| | - Wi-Gyeong Gwon
- Department of Food Science and Nutrition, Pukyong National University, Busan 608737, Korea; (E.-J.J.); (W.-G.G.); (M.-S.K.)
| | - Mi-Sung Kwon
- Department of Food Science and Nutrition, Pukyong National University, Busan 608737, Korea; (E.-J.J.); (W.-G.G.); (M.-S.K.)
| | - Kwon Taek Lim
- Department of Display Engineering, Pukyong National University, Busan 608737, Korea;
- Department of Smart Green Technology Engineering, Pukyong National University, Busan 608737, Korea
| | - Hyeung-Rak Kim
- Department of Food Science and Nutrition, Pukyong National University, Busan 608737, Korea; (E.-J.J.); (W.-G.G.); (M.-S.K.)
- Correspondence: ; Tel.: +82-051-629-5847
| |
Collapse
|
13
|
Starr AE, Deeke SA, Ning Z, de Nanassy J, Singleton R, Benchimol EI, Mack DR, Stintzi A, Figeys D. Associations between Cellular Energy and Pediatric Inflammatory Bowel Disease Patient Response to Treatment. J Proteome Res 2021; 20:4393-4404. [PMID: 34424714 DOI: 10.1021/acs.jproteome.1c00341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Inflammatory bowel diseases (IBDs), including Crohn's disease (CD) and ulcerative colitis, are chronic diseases of the gastrointestinal tract, with an unknown etiology, that affect over 6.8 million people worldwide. To characterize disease pathogenesis, proteomic and bioinformatic analyses were performed on colon biopsies collected during diagnostic endoscopy from 119 treatment-naïve pediatric patients, including from 78 IBD patients and 41 non-IBD patients who served as controls. Due to the presence of noninflamed and/or inflamed regions in IBD patients, up to two biopsies were obtained from IBD patients as compared to a single noninflamed biopsy from non-IBD pediatric control patients. Additional biopsies were obtained and analyzed from 33 of the IBD patients after IBD-directed therapeutic intervention for comparison of pre- and post-treatment proteomes. SuperSILAC was utilized to perform quantitative analysis of homogenized tissues, which were processed by filter-aided sample preparation. Hierarchical clustering and principal component analyses revealed proteomic patterns that distinguished inflamed from noninflamed tissues independent of therapy. Gene ontology revealed that proteins downregulated in inflammation are associated with metabolism, whereas upregulated proteins contribute to protein processing. A comparison of pre- and post-treatment proteomes from CD patients identified over 100 proteins that are significantly different between patients who responded and those who did not respond to therapy, including creatine kinase B and basigin.
Collapse
|
14
|
Peach JT, Wilson SM, Gunderson LD, Frothingham L, Tran T, Walk ST, Yeoman CJ, Bothner B, Miles MP. Temporal metabolic response yields a dynamic biosignature of inflammation. iScience 2021; 24:102817. [PMID: 34355150 PMCID: PMC8319798 DOI: 10.1016/j.isci.2021.102817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/20/2021] [Accepted: 07/02/2021] [Indexed: 12/15/2022] Open
Abstract
Chronic low-grade inflammation is a subclinical condition directly and indirectly linked to the development of a wide range of diseases responsible for the vast majority of morbidity. To examine mechanisms coupled to chronic disease, a group of overweight and obese human subjects without known inflammatory diseases participated in a high-fat meal challenge as an acute inflammation stimulus. Analysis of serum metabolites grouped by baseline cytokine levels revealed that single samples had little power in differentiating groups. However, an analysis that incorporated temporal response separated inflammatory response phenotypes and allowed us to create a metabolic signature of inflammation which revealed metabolic components that are crucial to a cytokine-mediated inflammation response. The use of temporal response, rather than a single time point, improved metabolomic prediction of high postprandial inflammation responses and led to the development of a dynamic biosignature as a potential tool for stratifying risk to a wide range of diseases. Dynamic responses often provide insight into disease pathology Temporal metabolic responses to acute inflammation were explored in obese people Temporal metabolite levels differentiated low, mid, and high inflammation groups Inflammation-linked metabolites were shown to be predictors of cytokine responses
Collapse
Affiliation(s)
- Jesse T Peach
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59718, USA
| | - Stephanie M Wilson
- Department of Health and Human Development, Montana State University, Bozeman, MT 59718, USA
| | - Logan D Gunderson
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59718, USA
| | - Lizzi Frothingham
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59718, USA
| | - Tan Tran
- Department of Math, Montana State University, Bozeman, MT 59718, USA
| | - Seth T Walk
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59718, USA
| | - Carl J Yeoman
- Department of Range and Animal Sciences, Montana State University, Bozeman, MT 59718, USA
| | - Brian Bothner
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59718, USA
| | - Mary P Miles
- Department of Health and Human Development, Montana State University, Bozeman, MT 59718, USA
| |
Collapse
|
15
|
Creatine Supplementation for Patients with Inflammatory Bowel Diseases: A Scientific Rationale for a Clinical Trial. Nutrients 2021; 13:nu13051429. [PMID: 33922654 PMCID: PMC8145094 DOI: 10.3390/nu13051429] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/18/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022] Open
Abstract
Based on theoretical considerations, experimental data with cells in vitro, animal studies in vivo, as well as a single case pilot study with one colitis patient, a consolidated hypothesis can be put forward, stating that “oral supplementation with creatine monohydrate (Cr), a pleiotropic cellular energy precursor, is likely to be effective in inducing a favorable response and/or remission in patients with inflammatory bowel diseases (IBD), like ulcerative colitis and/or Crohn’s disease”. A current pilot clinical trial that incorporates the use of oral Cr at a dose of 2 × 7 g per day, over an initial period of 2 months in conjunction with ongoing therapies (NCT02463305) will be informative for the proposed larger, more long-term Cr supplementation study of 2 × 3–5 g of Cr per day for a time of 3–6 months. This strategy should be insightful to the potential for Cr in reducing or alleviating the symptoms of IBD. Supplementation with chemically pure Cr, a natural nutritional supplement, is well tolerated not only by healthy subjects, but also by patients with diverse neuromuscular diseases. If the outcome of such a clinical pilot study with Cr as monotherapy or in conjunction with metformin were positive, oral Cr supplementation could then be used in the future as potentially useful adjuvant therapeutic intervention for patients with IBD, preferably together with standard medication used for treating patients with chronic ulcerative colitis and/or Crohn’s disease.
Collapse
|
16
|
Abstract
SARS-CoV2 infection or COVID-19 has created panic around the world since its first origin in December 2019 in Wuhan city, China. The COVID-19 pandemic has infected more than 46.4 million people, with 1,199,727 deaths. The immune system plays a crucial role in the severity of COVID-19 and the development of pneumonia-induced acute lung injury (ALI) or acute respiratory distress syndrome (ARDS). Along with providing protection, both innate and T cell-based adaptive immune response dysregulate during severe SARS-CoV2 infection. This dysregulation is more pronounced in older population and patients with comorbidities (Diabetes, hypertension, obesity, other pulmonary and autoimmune diseases). However, COVID-19 patients develop protective antibodies (Abs) against SARS-CoV2, but they do not long for last. The induction of the immune response against the pathogen also requires metabolic energy that generates through the process of immunometabolism. The change in the metabolic stage of immune cells from homeostasis to an inflammatory or infectious environment is called immunometabolic reprogramming. The article describes the cellular immunology (macrophages, T cells, B cells, dendritic cells, NK cells and pulmonary epithelial cells (PEC) and vascular endothelial cells) and the associated immune response during COVID-19. Immunometabolism may serve as a cell-specific therapeutic approach to target COVID-19.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, Brisbane, Queensland, Australia.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
17
|
Khailova L, Robison J, Jaggers J, Ing R, Lawson S, Treece A, Soranno D, Osorio Lujan S, Davidson JA. Tissue alkaline phosphatase activity and expression in an experimental infant swine model of cardiopulmonary bypass with deep hypothermic circulatory arrest. JOURNAL OF INFLAMMATION-LONDON 2020; 17:27. [PMID: 32817746 PMCID: PMC7422466 DOI: 10.1186/s12950-020-00256-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/30/2020] [Indexed: 01/12/2023]
Abstract
Background Infant cardiac surgery with cardiopulmonary bypass results in decreased circulating alkaline phosphatase that is associated with poor postoperative outcomes. Bovine intestinal alkaline phosphatase infusion represents a novel therapy for post-cardiac surgery organ injury. However, the effects of cardiopulmonary bypass and bovine-intestinal alkaline phosphatase infusion on tissue-level alkaline phosphatase activity/expression are unknown. Methods Infant pigs (n = 20) underwent cardiopulmonary bypass with deep hypothermic circulatory arrest followed by four hours of intensive care. Seven control animals underwent mechanical ventilation only. Cardiopulmonary bypass/deep hypothermic circulatory arrest animals were given escalating doses of bovine intestinal alkaline phosphatase infusion (0-25 U/kg/hr.; n = 5/dose). Kidney, liver, ileum, jejunum, colon, heart and lung were collected for measurement of tissue alkaline phosphatase activity and mRNA. Results Tissue alkaline phosphatase activity varied significantly across organs with the highest levels found in the kidney and small intestine. Cardiopulmonary bypass with deep hypothermic circulatory arrest resulted in decreased kidney alkaline phosphatase activity and increased lung alkaline phosphatase activity, with no significant changes in the other organs. Alkaline phosphatase mRNA expression was increased in both the lung and the ileum. The highest dose of bovine intestinal alkaline phosphatase resulted in increased kidney and liver tissue alkaline phosphatase activity. Conclusions Changes in alkaline phosphatase activity after cardiopulmonary bypass with deep hypothermic circulatory arrest and bovine intestinal alkaline phosphatase delivery are tissue specific. Kidneys, lung, and ileal alkaline phosphatase appear most affected by cardiopulmonary bypass with deep hypothermic circulatory arrest and further research is warranted to determine the mechanism and biologic importance of these changes.
Collapse
Affiliation(s)
- Ludmila Khailova
- Department of Pediatrics, University of Colorado, 13123 East 16th Ave, Box 100, Aurora, CO 80045 USA
| | - Justin Robison
- Department of Pediatrics, University of Colorado, 13123 East 16th Ave, Box 100, Aurora, CO 80045 USA
| | - James Jaggers
- Department of Surgery, University of Colorado, Aurora, CO USA
| | - Richard Ing
- Department of Anesthesiology, University of Colorado, Aurora, CO USA
| | - Scott Lawson
- Children's Hospital Colorado, Heart Institute, Aurora, CO USA
| | - Amy Treece
- Department of Pathology, University of Colorado, Aurora, CO USA
| | - Danielle Soranno
- Department of Pediatrics, University of Colorado, 13123 East 16th Ave, Box 100, Aurora, CO 80045 USA
| | - Suzanne Osorio Lujan
- Department of Pediatrics, University of Colorado, 13123 East 16th Ave, Box 100, Aurora, CO 80045 USA
| | - Jesse A Davidson
- Department of Pediatrics, University of Colorado, 13123 East 16th Ave, Box 100, Aurora, CO 80045 USA
| |
Collapse
|
18
|
Li Y, Guo R, Zhang M, Chen P, Li J, Sun Y. Protective effect of emodin on intestinal epithelial tight junction barrier integrity in rats with sepsis induced by cecal ligation and puncture. Exp Ther Med 2020; 19:3521-3530. [PMID: 32346413 PMCID: PMC7185184 DOI: 10.3892/etm.2020.8625] [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] [Received: 06/04/2019] [Accepted: 01/17/2020] [Indexed: 12/14/2022] Open
Abstract
The present study investigated the protective effects of emodin on intestinal epithelial tight junction (TJ) barrier integrity in cecal ligation and puncture (CLP)-induced septic rats and its possible mechanisms of action. Healthy male Sprague-Dawley rats were randomly divided into three groups (n=20 per group): Sham group, CLP group and CLP + emodin group. Animals were sacrificed at 12 and 24 h after the model was established. Abdominal aortic blood and specimens of the ileum were harvested for analysis. The histopathological changes in intestinal mucosa and the ultrastructures of intestinal epithelial cells were investigated using light microscopy and transmission electron microscopy. The integrity of the intestinal barrier was assessed by examining plasma diamine oxidase (DAO) levels and the ratio of urine lactulose to mannitol (L/M). The levels of the intestinal TJ proteins claudin-3, zonula occludens (ZO)-1 and occludin were detected using immunohistochemistry, western blotting and reverse transcription-quantitative PCR. The results showed that the pathological damage to intestinal mucosa and the intestinal tissue injury score in the CLP + emodin group were significantly reduced compared to those of the CLP group, and the differences were more obvious at 24 h compared with 12 h. DAO activity and the L/M ratio in the emodin pre-treatment group decreased significantly at 24 h compared with the CLP groups. The protein and mRNA levels of the TJ proteins claudin-3, ZO-1 and occludin in the emodin pre-treatment groups at 12 and 24 h were increased, while occludin mRNA level was found to be decreased compared with the CLP groups. The present study suggested that emodin may significantly reduce the damage to the intestinal epithelial barrier in sepsis, inhibit intestinal barrier permeability and protect intestinal barrier integrity. Emodin may protect intestinal barrier integrity by elevating expression levels of the TJ proteins claudin-3, ZO-1 and occludin in CLP rats.
Collapse
Affiliation(s)
- Yanjun Li
- Department of Emergency, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Ruimin Guo
- Department of Emergency, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Mengying Zhang
- Department of Emergency, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Peng Chen
- Department of Emergency, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Jingxin Li
- Department of Emergency, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Yanni Sun
- Department of Emergency, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China.,Department of Emergency, Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Shanghai 200062, P.R. China
| |
Collapse
|
19
|
Renga G, Oikonomou V, Moretti S, Stincardini C, Bellet MM, Pariano M, Bartoli A, Brancorsini S, Mosci P, Finocchi A, Rossi P, Costantini C, Garaci E, Goldstein AL, Romani L. Thymosin β4 promotes autophagy and repair via HIF-1α stabilization in chronic granulomatous disease. Life Sci Alliance 2019; 2:2/6/e201900432. [PMID: 31719116 PMCID: PMC6851533 DOI: 10.26508/lsa.201900432] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023] Open
Abstract
This study demonstrates that thymosin β4 stabilizes HIF-1a to promote autophagy and up-regulate genes involved in tissue and mucosal barrier protection in chronic granulomatous disease. Chronic granulomatous disease (CGD) is a genetic disorder of the NADPH oxidase characterized by increased susceptibility to infections and hyperinflammation associated with defective autophagy and increased inflammasome activation. Herein, we demonstrate that thymosin β4 (Tβ4), a g-actin sequestering peptide with multiple and diverse intracellular and extracellular activities affecting inflammation, wound healing, fibrosis, and tissue regeneration, promoted in human and murine cells noncanonical autophagy, a form of autophagy associated with phagocytosis and limited inflammation via the death-associated protein kinase 1. We further show that the hypoxia inducible factor-1 (HIF-1)α was underexpressed in CGD but normalized by Tβ4 to promote autophagy and up-regulate genes involved in mucosal barrier protection. Accordingly, inflammation and granuloma formation were impaired and survival increased in CGD mice with colitis or aspergillosis upon Tβ4 treatment or HIF-1α stabilization. Thus, the promotion of endogenous pathways of inflammation resolution through HIF-1α stabilization is druggable in CGD by Tβ4.
Collapse
Affiliation(s)
- Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Vasilis Oikonomou
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Silvia Moretti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Andrea Bartoli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Paolo Mosci
- Internal Medicine, Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Andrea Finocchi
- Department of Pediatrics, Unit of Immune and Infectious Diseases, Children's Hospital Bambino Gesù, Rome, Italy
| | - Paolo Rossi
- Department of Pediatrics, Unit of Immune and Infectious Diseases, Children's Hospital Bambino Gesù, Rome, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Enrico Garaci
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Rome, Italy
| | - Allan L Goldstein
- Department of Biochemistry and Molecular Medicine, the George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
20
|
Nedeljkovic N. Complex regulation of ecto-5'-nucleotidase/CD73 and A 2AR-mediated adenosine signaling at neurovascular unit: A link between acute and chronic neuroinflammation. Pharmacol Res 2019; 144:99-115. [PMID: 30954629 DOI: 10.1016/j.phrs.2019.04.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/01/2019] [Accepted: 04/03/2019] [Indexed: 12/20/2022]
Abstract
The review summarizes available data regarding the complex regulation of CD73 at the neurovascular unit (NVU) during neuroinflammation. Based on available data we propose the biphasic pattern of CD73 regulation at NVU, with an early attenuation and a postponed up-regulation of CD73 activity. Transient attenuation of CD73 activity on leukocyte/vascular endothelium and leukocyte/astrocyte surface, required for the initiation of a neuroinflammatory response, may be effectuated either by catalytic inhibition of CD73 and/or by shedding of the CD73 molecule from the cell surface, while postponed induction of CD73 is effectuated by transcriptional up-regulation of Nt5e and posttranslational modifications. Neuroinflammatory conditions are also associated with significant enhancement and gain-of-function of A2AR-mediated adenosine signaling. However, in contrast to the temporary prevalence of A2AR over A1R signaling during an acute inflammatory response, prolonged induction of A2AR and resulting perpetual CD73/A2AR coupling may be a contributing factors in the transition between acute and chronic neuroinflammation. Thus, pharmacological targeting of the CD73/A2AR axis may attenuate inflammatory response and ameliorate neurological deficits in chronic neuroinflammatory conditions.
Collapse
Affiliation(s)
- Nadezda Nedeljkovic
- Department of General Physiology and Biophysics, Faculty of Biology University of Belgrade, Studentski trg 3, Belgrade 11001, Serbia.
| |
Collapse
|
21
|
Saroha V, Josephson CD, Patel RM. Epidemiology of Necrotizing Enterocolitis: New Considerations Regarding the Influence of Red Blood Cell Transfusions and Anemia. Clin Perinatol 2019; 46:101-117. [PMID: 30771812 PMCID: PMC6383803 DOI: 10.1016/j.clp.2018.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
This article summarizes available evidence on the relationship between red blood cell transfusion and anemia, and necrotizing enterocolitis (NEC). We review recent studies that highlight the uncertainty of the effect of red blood cell transfusion on NEC and the potential role of anemia. We also discuss potential pathophysiologic effects of both red blood cell transfusion and anemia and highlight strategies to prevent anemia and red blood cell transfusion. We also discuss ongoing randomized trials that are likely to provide important new evidence to guide red blood cell transfusion practices.
Collapse
Affiliation(s)
- Vivek Saroha
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 2015 Uppergate Dr. NE, 3 floor, Atlanta, GA 30322. Tel 404-727-3236.
| | - Cassandra D. Josephson
- Center for Transfusion and Cellular Therapies, Departments of Pathology and Laboratory Medicine and Pediatrics, Emory University School of Medicine, 101 Woodruff Cir, Atlanta, GA 30322. Tel 404-785-4553.
| | - Ravi Mangal Patel
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 2015 Uppergate Dr. NE, 3 floor, Atlanta, GA 30322. Tel 404-727-3236.
| |
Collapse
|
22
|
Jiang L, Lv J, Liu J, Hao X, Ren F, Guo H. Donkey milk lysozyme ameliorates dextran sulfate sodium-induced colitis by improving intestinal barrier function and gut microbiota composition. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.07.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
23
|
Abstract
Mucosal tissues represent surfaces that are exposed to the outside world and provide a conduit for internal and external communication. Tissues such as the intestine and the lung are lined by layer(s) of epithelial cells that, when organized in three dimensions, provide a critical barrier to the flux of luminal contents. This selective barrier is provided through the regulated expression of junctional proteins and mucins. Tissue oxygen metabolism is central to the maintenance of homeostasis in the mucosa. In some organs (e.g., the colon), low baseline Po2 determines tissue metabolism and results in basal expression of the transcription factor, hypoxia-inducible factor (HIF), which is enhanced after ischemia/inflammation. Recent studies have indicated that HIF contributes fundamentally to the expression of barrier-related genes and in the regulation of barrier-adaptive responses within the mucosa. Here, we briefly review recent literature on the topic of hypoxia and HIF regulation of barrier in mucosal health and during disease.
Collapse
|
24
|
Wang RX, Colgan SP. Special pro-resolving mediator (SPM) actions in regulating gastro-intestinal inflammation and gut mucosal immune responses. Mol Aspects Med 2017; 58:93-101. [PMID: 28232096 PMCID: PMC5797700 DOI: 10.1016/j.mam.2017.02.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 02/16/2017] [Indexed: 12/19/2022]
Abstract
Surfaces covered by epithelial cells, termed mucosal surfaces, serve special functions as selectively permeable barriers that partition the host and the outside world. Given its close association to microbial antigens, the intestinal mucosa has evolved creative mechanisms to maintain homeostasis, to prevent excessive inflammatory responses, and to promote rapid and full inflammatory resolution. In recent years, an active role for the epithelium has been attributed to the local generation of specialized pro-resolving mediators (SPMs) in the maintenance of immunological homeostasis. In this brief review, we highlight evidence that the epithelium actively contributes to coordination and resolution of inflammation, principally through the generation of SPMs. These autacoids are derived from omega-6 and omega-3 polyunsaturated fatty acids. Acting through widely expressed G-protein coupled receptors, SPMs are implicated in the resolution of acute inflammation that manifests specific, epithelial-directed actions focused on mucosal-homeostasis, including regulation of leukocyte trafficking, the generation of antimicrobial peptides, the dampening of endotoxin signaling, and the attenuation of mucosal cytokine responses.
Collapse
Affiliation(s)
- Ruth X Wang
- Departments of Medicine and Immunology and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Sean P Colgan
- Departments of Medicine and Immunology and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
25
|
Colgan SP, Campbell EL, Kominsky DJ. Hypoxia and Mucosal Inflammation. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 11:77-100. [PMID: 27193451 DOI: 10.1146/annurev-pathol-012615-044231] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sites of inflammation are defined by significant changes in metabolic activity. Recent studies have suggested that O2 metabolism and hypoxia play a prominent role in inflammation so-called "inflammatory hypoxia," which results from a combination of recruited inflammatory cells (e.g., neutrophils and monocytes), the local proliferation of multiple cell types, and the activation of multiple O2-consuming enzymes during inflammation. These shifts in energy supply and demand result in localized regions of hypoxia and have revealed the important function off the transcription factor HIF (hypoxia-inducible factor) in the regulation of key target genes that promote inflammatory resolution. Analysis of these pathways has provided multiple opportunities for understanding basic mechanisms of inflammation and has defined new targets for intervention. Here, we review recent work addressing tissue hypoxia and metabolic control of inflammation and immunity.
Collapse
Affiliation(s)
- Sean P Colgan
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045; .,Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Eric L Campbell
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045; .,Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Douglas J Kominsky
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045.,Department of Microbiology and Immunology, Montana State University, Bozeman, Montana 59717
| |
Collapse
|
26
|
Colgan SP, Campbell EL. Oxygen metabolism and innate immune responses in the gut. J Appl Physiol (1985) 2017; 123:1321-1327. [PMID: 28705991 DOI: 10.1152/japplphysiol.00113.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 06/27/2017] [Accepted: 07/06/2017] [Indexed: 01/02/2023] Open
Abstract
Epithelial cells of the mucosa provide a first line of defense to prevent the inappropriate translocation of luminal antigens, and therefore contribute significantly to nonspecific innate immunity. In the gastrointestinal (GI) tract, barrier is provided by multiple components of the mucosa, including mucus production, epithelial junctional complexes, and the production of antimicrobial molecules. In recent years, it is better appreciated that tissue oxygen metabolism is key to homeostasis in the mucosa. The intestine, for example, maintains a low baseline Po2 level due to high rates of metabolism, countercurrent blood flow, and the presence of a steep oxygen gradient across the luminal aspect of tissue surface. As a result, hypoxia and hypoxia-inducible factor (HIF)-dependent signaling exists even in the healthy, unperturbed intestinal mucosa. In a number of examples, HIF has been demonstrated both to promote barrier function during homeostasis and to promote resolution of active inflammation. Hypoxia-elicited factors that contribute to innate responses in the mucosa include the transcriptional regulation of mucin genes, junction proteins, and autophagic flux. Here, we review current literature related to hypoxia and innate immunity in health and during mucosal inflammation.
Collapse
Affiliation(s)
- Sean P Colgan
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado; and
| | - Eric L Campbell
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| |
Collapse
|
27
|
Chun C, Zheng L, Colgan SP. Tissue metabolism and host-microbial interactions in the intestinal mucosa. Free Radic Biol Med 2017; 105:86-92. [PMID: 27687211 PMCID: PMC5797701 DOI: 10.1016/j.freeradbiomed.2016.09.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/23/2016] [Accepted: 09/26/2016] [Indexed: 01/03/2023]
Abstract
In recent years, studies in the gastrointestinal (GI) mucosa have taught us a number of important lessons related to tissue oxygenation and metabolism in health and disease. The highly vascularized mucosa lies immediately adjacent to an anaerobic lumen containing trillions of metabolically active microbes (i.e. the microbiome) that results in one of the more austere tissue microenvironments in the body. These studies have also implicated a prominent role for oxygen metabolism and hypoxia in inflammation, so called "inflammatory hypoxia", that results from the activation of multiple oxygen consuming enzymes. Inflammation-associated shifts in the composition of the microbiome and microbial-derived metabolites have revealed a prominent role for the transcription factor hypoxia-inducible factor (HIF) in the regulation of key target genes that promote inflammatory resolution. Analyses of these pathways have provided a multitude of opportunities for understanding basic mechanisms of both homeostasis and disease and have defined new targets for intervention. Here, we review recent advances in our understanding of metabolic influences on host-microbe interactions in the GI mucosa.
Collapse
Affiliation(s)
- Carlene Chun
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, United States
| | - Leon Zheng
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, United States
| | - Sean P Colgan
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, United States.
| |
Collapse
|
28
|
Hypoxia and inflammatory bowel disease. Microbes Infect 2017; 19:210-221. [DOI: 10.1016/j.micinf.2016.09.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/08/2016] [Accepted: 09/13/2016] [Indexed: 12/17/2022]
|
29
|
Zhdanov AV, Okkelman IA, Golubeva AV, Doerr B, Hyland NP, Melgar S, Shanahan F, Cryan JF, Papkovsky DB. Quantitative analysis of mucosal oxygenation using ex vivo imaging of healthy and inflamed mammalian colon tissue. Cell Mol Life Sci 2017; 74:141-151. [PMID: 27510419 PMCID: PMC11107550 DOI: 10.1007/s00018-016-2323-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/14/2016] [Accepted: 08/02/2016] [Indexed: 12/18/2022]
Abstract
Colonic inflammation is associated with decreased tissue oxygenation, significantly affecting gut homeostasis. However, the crosstalk between O2 consumption and supply in the inflamed tissue are not fully understood. Using a murine model of colitis, we analysed O2 in freshly prepared samples of healthy and inflamed colon tissue. We developed protocols for efficient ex vivo staining of mouse distal colon mucosa with a cell-penetrating O2 sensitive probe Pt-Glc and high-resolution imaging of O2 concentration in live tissue by confocal phosphorescence lifetime-imaging microscopy (PLIM). Microscopy analysis revealed that Pt-Glc stained mostly the top 50-60 μm layer of the mucosa, with high phosphorescence intensity in epithelial cells. Measured O2 values in normal mouse tissue ranged between 5 and 35 μM (4-28 Torr), tending to decrease in the deeper tissue areas. Four-day treatment with dextran sulphate sodium (DSS) triggered colon inflammation, as evidenced by an increase in local IL6 and mKC mRNA levels, but did not affect the gross architecture of colonic epithelium. We further observed an increase in oxygenation, partial activation of hypoxia inducible factor (HIF) 1 signalling, and negative trends in pyruvate dehydrogenase activity and O2 consumption rate in the colitis mucosa, suggesting a decrease in mitochondrial respiration, which is known to be regulated via HIF-1 signalling and pyruvate oxidation rate. These results along with efficient staining with Pt-Glc of rat and human colonic mucosa reveal high potential of PLIM platform as a powerful tool for the high-resolution analysis of the intestinal tissue oxygenation in patients with inflammatory bowel disease and other pathologies, affecting tissue respiration.
Collapse
Affiliation(s)
- Alexander V Zhdanov
- School of Biochemistry and Cell Biology, University College Cork, Cavanagh Pharmacy Building, College Road, Cork, Ireland.
| | - Irina A Okkelman
- School of Biochemistry and Cell Biology, University College Cork, Cavanagh Pharmacy Building, College Road, Cork, Ireland
| | - Anna V Golubeva
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Barbara Doerr
- School of Biochemistry and Cell Biology, University College Cork, Cavanagh Pharmacy Building, College Road, Cork, Ireland
| | - Niall P Hyland
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Silvia Melgar
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Fergus Shanahan
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Dmitri B Papkovsky
- School of Biochemistry and Cell Biology, University College Cork, Cavanagh Pharmacy Building, College Road, Cork, Ireland
| |
Collapse
|
30
|
Bowser JL, Broaddus RR. CD73s protection of epithelial integrity: Thinking beyond the barrier. Tissue Barriers 2016; 4:e1224963. [PMID: 28123924 DOI: 10.1080/21688370.2016.1224963] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/09/2016] [Accepted: 08/10/2016] [Indexed: 01/05/2023] Open
Abstract
The prevailing view of CD73 in cancer is that it is overexpressed in tumors and promotes cancer progression by dampening local T cell-mediated immune responses. We recently found that CD73 is down-regulated in poorly-differentiated and advanced stage endometrial carcinoma compared to normal endometrium and well-differentiated, early stage tumors. We revealed that CD73-generated adenosine induces a physiological response to protect epithelial integrity in well-differentiated, early stage endometrial carcinoma. The ability of CD73-generated adenosine to protect the barrier is not so different from its ability to induce immunosuppression and other physiological responses in cancerous tissues. In this commentary we examine the complexity of CD73 in cancer and suggest that a "one size fits all" approach to the role of CD73/adenosine in cancer is no longer warranted. Given that tumors often hijack normal cellular responses, we also provide consideration on how CD73s known role to protect barrier function may have implications in promoting tumor progression.
Collapse
Affiliation(s)
- Jessica L Bowser
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Russell R Broaddus
- Department of Pathology, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| |
Collapse
|
31
|
Wu C, Wang X, Jiang T, Li C, Zhang L, Gao X, Tian F, Li N, Li J. Partial Enteral Nutrition Mitigated Ischemia/Reperfusion-Induced Damage of Rat Small Intestinal Barrier. Nutrients 2016; 8:nu8080502. [PMID: 27548209 PMCID: PMC4997415 DOI: 10.3390/nu8080502] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/07/2016] [Accepted: 08/11/2016] [Indexed: 01/03/2023] Open
Abstract
Background and Aims: This study was designed to investigate a relatively optimum dose of partial enteral nutrition (PEN) which effectively attenuates intestinal barrier dysfunction initiated by ischemia/reperfusion injury (IRI). Methods: In experiment 1, 60 male Sprague-Dawley (SD) rats were subjected to intestinal IRI and assigned to six groups according to the different proportion of EN administrations: namely total parenteral nutrition (TPN or 0%EN), 10%EN, 20%EN, 40%EN, 60%EN, and total enteral nutrition (TEN or 100%) groups, the deficits of intraluminal calorie were supplemented by PN. In experiment 2, 50 male SD rats were subjected to intestinal IRI and divided into five groups based on the results of experiment 1: TPN, TEN, 20%EN, TPN plus pretreatment with NF-κB antagonist 30 min before IRI (TPN+PDTC), and TPN plus pretreatment with HIF-1α antagonist 30 min before IRI (TPN+YC-1) groups. Results: In experiment 1, previous IRI combined with subsequent EN shortage disrupted the structure of intestinal epithelial cell and tight junctions (TJs). While 20% dose of EN had an obviously protective effect on these detrimental consequences. In experiment 2, compared with TPN only, 20%EN exerted a significant protection of barrier function of intestinal epithelium. Analogous results were observed when TPN combined with specific NF-κB/HIF-1α inhibitors (PDTC and YC-1). Meanwhile, the expression of NF-κB/HIF-1α had a similar trend among the groups. Conclusions: Our findings indicate that 20%EN is the minimally effective dosage of EN which promotes the recovery of intestinal barrier function after IRI in a rat model. Furthermore, we discreetly speculate that this benefit is, at least partly, related to NF-κB/HIF-1α pathway expression.
Collapse
Affiliation(s)
- Chao Wu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Xinying Wang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Tingting Jiang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Chaojun Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University and Model Animal Research Center, National Resource Center for Mutant Mice, Nanjing 210093, China.
| | - Li Zhang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Xuejin Gao
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Feng Tian
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Ning Li
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Jieshou Li
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| |
Collapse
|
32
|
Tong LC, Wang Y, Wang ZB, Liu WY, Sun S, Li L, Su DF, Zhang LC. Propionate Ameliorates Dextran Sodium Sulfate-Induced Colitis by Improving Intestinal Barrier Function and Reducing Inflammation and Oxidative Stress. Front Pharmacol 2016; 7:253. [PMID: 27574508 PMCID: PMC4983549 DOI: 10.3389/fphar.2016.00253] [Citation(s) in RCA: 200] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/02/2016] [Indexed: 12/15/2022] Open
Abstract
Propionate is a short chain fatty acid that is abundant as butyrate in the gut and blood. However, propionate has not been studied as extensively as butyrate in the treatment of colitis. The present study was to investigate the effects of sodium propionate on intestinal barrier function, inflammation and oxidative stress in dextran sulfate sodium (DSS)-induced colitis mice. Animals in DSS group received drinking water from 1 to 6 days and DSS [3% (w/v) dissolved in double distilled water] instead of drinking water from 7 to 14 days. Animals in DSS+propionate (DSS+Prop) group were given 1% sodium propionate for 14 consecutive days and supplemented with 3% DSS solution on day 7–14. Intestinal barrier function, proinflammatory factors, oxidative stress, and signal transducer and activator of transcription 3 (STAT3) signaling pathway in the colon were determined. It was found that sodium propionate ameliorated body weight loss, colon-length shortening and colonic damage in colitis mice. Sodium propionate significantly inhibited the increase of FITC-dextran in serum and the decrease of zonula occludens-1 (ZO-1), occludin, and E-cadherin expression in the colonic tissue. It also inhibited the expression of interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α) mRNA and phosphorylation of STAT3 in colitis mice markedly, reduced the myeloperoxidase (MPO) level, and increased the superoxide dismutase and catalase level in colon and serum compared with DSS group. Sodium propionate inhibited macrophages with CD68 marker infiltration into the colonic mucosa of colitis mice. These results suggest that oral administration of sodium propionate could ameliorate DSS-induced colitis mainly by improving intestinal barrier function and reducing inflammation and oxidative stress via the STAT3 signaling pathway.
Collapse
Affiliation(s)
- Ling-Chang Tong
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese MedicineShanghai, China; Department of Pharmacology, College of Pharmacy, Second Military Medical UniversityShanghai, China
| | - Yue Wang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese MedicineShanghai, China; Department of Pharmacy, Ningxia Medical UniversityYinchuan, China
| | - Zhi-Bin Wang
- Department of Pharmacology, College of Pharmacy, Second Military Medical University Shanghai, China
| | - Wei-Ye Liu
- Department of Pharmacology, College of Pharmacy, Second Military Medical University Shanghai, China
| | - Sheng Sun
- Department of Pharmacology, College of Pharmacy, Second Military Medical University Shanghai, China
| | - Ling Li
- Department of Pharmacology, College of Pharmacy, Second Military Medical University Shanghai, China
| | - Ding-Feng Su
- Department of Pharmacology, College of Pharmacy, Second Military Medical University Shanghai, China
| | - Li-Chao Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine Shanghai, China
| |
Collapse
|
33
|
Glover LE, Lee JS, Colgan SP. Oxygen metabolism and barrier regulation in the intestinal mucosa. J Clin Invest 2016; 126:3680-3688. [PMID: 27500494 DOI: 10.1172/jci84429] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mucosal surfaces are lined by epithelial cells and provide an important barrier to the flux of antigens from the outside. This barrier is provided at a number of levels, including epithelial junctional complexes, mucus production, and mucosa-derived antimicrobials. Tissue metabolism is central to the maintenance of homeostasis in the mucosa. In the intestine, for example, baseline pO2 levels are uniquely low due to counter-current blood flow and the presence of large numbers of bacteria. As such, hypoxia and HIF signaling predominates normal intestinal metabolism and barrier regulation during both homeostasis and active inflammation. Contributing factors that elicit important adaptive responses within the mucosa include the transcriptional regulation of tight junction proteins, metabolic regulation of barrier components, and changes in autophagic flux. Here, we review recent literature around the topic of hypoxia and barrier function in health and during disease.
Collapse
|
34
|
Experimental Colitis Is Attenuated by Cardioprotective Diet Supplementation That Reduces Oxidative Stress, Inflammation, and Mucosal Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8473242. [PMID: 26881044 PMCID: PMC4736595 DOI: 10.1155/2016/8473242] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 12/01/2015] [Indexed: 12/19/2022]
Abstract
Inflammatory bowel diseases (IBD) such as ulcerative colitis (UC) and Crohn's disease (CD) are multifactorial, relapsing disorders of the gastrointestinal tract. However, the etiology is still poorly understood but involves altered immune responses, epithelial dysfunction, environmental factors, and nutrition. Recently, we have shown that the diet supplement corabion has cardioprotective effects due to reduction of oxidative stress and inflammation. Since oxidative stress and inflammation are also prominent risk factors in IBD, we speculated that corabion also has beneficial effects on experimental colitis. Colitis was induced in male mice by administration of 3.5% (w/v) dextran sulfate sodium (DSS) in drinking water for a period of 3 or 7 days with or without daily gavage feeding of corabion consisting of vitamin C, vitamin E, L-arginine, and eicosapentaenoic and docosahexaenoic acid. We found that corabion administration attenuated DSS-induced colon shortening, tissue damage, and disease activity index during the onset of colitis. Mechanistically, these effects could be explained by reduced neutrophil recruitment, oxidative stress, production of proinflammatory cytokines, and internalization of the junctional proteins ZO-1 and E-cadherin leading to less edema formation. Thus, corabion may be a useful diet supplement for the management of chronic inflammatory intestinal disorders such as IBD.
Collapse
|
35
|
Luscinskas FW, Leick M, Newton G, Nusrat A. Introduction for the special issue on "Tissue Barriers in Inflammation". Tissue Barriers 2015; 3:e1015825. [PMID: 25927017 PMCID: PMC4389789 DOI: 10.1080/21688370.2015.1015825] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
This issue of Tissue Barriers contains the inaugural special issue devoted to recent advances in barrier function of endothelial and epithelial cells. We used this opportunity to invite experts in vascular endothelial cell biology and epithelial cell biology to comment on critical questions and problems in permeability of organ and tissue barriers, and to provide insight into common areas in these fields, namely how these cells maintain homeostasis and response to injury and infection. To complement these reviews, this issue also contains four research articles that explore specific questions related respiratory and intestinal epithelial cell function.
Collapse
Affiliation(s)
- Francis W Luscinskas
- Center for Excellence in Vascular Biology; Department
of Pathology; Brigham and Women's Hospital; Harvard Medical
School; Boston, MA USA
| | - Marion Leick
- Center for Excellence in Vascular Biology; Department
of Pathology; Brigham and Women's Hospital; Harvard Medical
School; Boston, MA USA
| | - Gail Newton
- Center for Excellence in Vascular Biology; Department
of Pathology; Brigham and Women's Hospital; Harvard Medical
School; Boston, MA USA
| | - Asma Nusrat
- Center for Excellence in Vascular Biology; Department
of Pathology; Brigham and Women's Hospital; Harvard Medical
School; Boston, MA USA
- Present affiliation: Pathology and Laboratory
Medicine; Emory University School of Medicine; Atlanta, GA
USA
| |
Collapse
|