1
|
AlMarzooqi SK, Almarzooqi F, Sadida HQ, Jerobin J, Ahmed I, Abou-Samra AB, Fakhro KA, Dhawan P, Bhat AA, Al-Shabeeb Akil AS. Deciphering the complex interplay of obesity, epithelial barrier dysfunction, and tight junction remodeling: Unraveling potential therapeutic avenues. Obes Rev 2024; 25:e13766. [PMID: 38745386 DOI: 10.1111/obr.13766] [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: 08/09/2023] [Revised: 03/11/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
Obesity stands as a formidable global health challenge, predisposing individuals to a plethora of chronic illnesses such as cardiovascular disease, diabetes, and cancer. A confluence of genetic polymorphisms, suboptimal dietary choices, and sedentary lifestyles significantly contribute to the elevated incidence of obesity. This multifaceted health issue profoundly disrupts homeostatic equilibrium at both organismal and cellular levels, with marked alterations in gut permeability as a salient consequence. The intricate mechanisms underlying these alterations have yet to be fully elucidated. Still, evidence suggests that heightened inflammatory cytokine levels and the remodeling of tight junction (TJ) proteins, particularly claudins, play a pivotal role in the manifestation of epithelial barrier dysfunction in obesity. Strategic targeting of proteins implicated in these pathways and metabolites such as short-chain fatty acids presents a promising intervention for restoring barrier functionality among individuals with obesity. Nonetheless, recognizing the heterogeneity among affected individuals is paramount; personalized medical interventions or dietary regimens tailored to specific genetic backgrounds and allergy profiles may prove indispensable. This comprehensive review delves into the nexus of obesity, tight junction remodeling, and barrier dysfunction, offering a critical appraisal of potential therapeutic interventions.
Collapse
Affiliation(s)
- Sara K AlMarzooqi
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Fajr Almarzooqi
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Hana Q Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Jayakumar Jerobin
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ikhlak Ahmed
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Abdul-Badi Abou-Samra
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Khalid A Fakhro
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| |
Collapse
|
2
|
Altyar AE, Afzal M, Ghaboura N, Alharbi KS, Alenezi SK, Sayyed N, Kazmi I. Barbaloin Protects Pentylenetetrazol-Induced Cognitive Deficits in Rodents via Modulation of Neurotransmitters and Inhibition of Oxidative-Free-Radicals-Led Inflammation. Pharmaceuticals (Basel) 2024; 17:699. [PMID: 38931365 PMCID: PMC11206990 DOI: 10.3390/ph17060699] [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: 04/22/2024] [Revised: 05/09/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Epilepsy is defined by an excessive level of activity in the neurons and coordinated bursts of electrical activity, resulting in the occurrence of seizure episodes. The precise cause of epileptogenesis remains uncertain; nevertheless, the etiology of epilepsy may involve neuroinflammation, oxidative stress, and malfunction of the neurotransmitter system. OBJECTIVE The goal of this investigation was to assess barbaloin's protective properties with respect to pentylenetetrazol (PTZ)-)-induced cognitive deficits in rats via antioxidative, anti-inflammatory, and neurotransmitter-modulating effects. METHODS Wistar rats were subjected to PTZ [40 mg/kg (i.p.)], which induced cognitive decline. Behavior assessment using a kindling score, open-field test (OFT), novel object recognition test (NORT), and assays for superoxide dismutase (SOD), reduced glutathione (GSH), catalase (CAT), malondialdehyde (MDA), acetylcholinesterase (AChE), caspase-3, nitric oxide (NO), interleukins-1β (IL-1β), tumor necrosis factor-α (TNF-α), IL-6, nuclear factor kappa-B (NF-κB), Bcl-2 and Bax, and neurotransmitter levels [GABA, DA, NE, and serotonin (5-HT)] were performed. RESULTS The treatment of rats with barbaloin resulted in behavior improvement and significant changes in the levels of GSH, SOD, CAT, MDA, AChE, NO, IL-6, IL-1β, TNF-α, NF-κB, caspase-3, Bcl-2, and Bax compared to the PTZ control group. Barbaloin treatment resulted in notable changes in neurotransmitter levels (GABA, NE, 5-HT, DA) compared to the PTZ group. CONCLUSIONS The ongoing study has gathered evidence indicating that the injection of barbaloin has resulted in significant improvements in cognitive performance in rats. This is achieved by inhibiting oxidative stress, enhancing the activity of natural antioxidant enzymes, reducing cytokine levels, and increasing the levels of neurotransmitters in the brain. These results were detected in comparison to a PTZ control and can be attributed to the potent anti-inflammatory and antioxidant capabilities of barbaloin, which could be linked to its neuroprotective properties. Barbaloin may potentially increase cognitive decline and boost neuronal survival by altering the expression of Bax, caspase-3, Bcl-2.
Collapse
Affiliation(s)
- Ahmad Essam Altyar
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, P.O. Box 80260, Jeddah 21589, Saudi Arabia
- Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeedah 21442, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeedah 21442, Saudi Arabia
| | - Nehmat Ghaboura
- Department of Pharmacy Practice, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeedah 21442, Saudi Arabia;
| | - Khalid Saad Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim 51452, Saudi Arabia; (K.S.A.); (S.K.A.)
| | - Sattam Khulaif Alenezi
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim 51452, Saudi Arabia; (K.S.A.); (S.K.A.)
| | - Nadeem Sayyed
- Glocal School of Pharmacy, Glocal University, Mirzapur-Pole, Saharanpur 247121, India;
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, P.O. Box. 80200, Jeddah 21589, Saudi Arabia;
| |
Collapse
|
3
|
Drolia R, Bryant DB, Tenguria S, Jules-Culver ZA, Thind J, Amelunke B, Liu D, Gallina NLF, Mishra KK, Samaddar M, Sawale MR, Mishra DK, Cox AD, Bhunia AK. Listeria adhesion protein orchestrates caveolae-mediated apical junctional remodeling of epithelial barrier for Listeria monocytogenes translocation. mBio 2024; 15:e0282123. [PMID: 38376160 PMCID: PMC10936185 DOI: 10.1128/mbio.02821-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/09/2024] [Indexed: 02/21/2024] Open
Abstract
The cellular junctional architecture remodeling by Listeria adhesion protein-heat shock protein 60 (LAP-Hsp60) interaction for Listeria monocytogenes (Lm) passage through the epithelial barrier is incompletely understood. Here, using the gerbil model, permissive to internalin (Inl) A/B-mediated pathways like in humans, we demonstrate that Lm crosses the intestinal villi at 48 h post-infection. In contrast, the single isogenic (lap- or ΔinlA) or double (lap-ΔinlA) mutant strains show significant defects. LAP promotes Lm translocation via endocytosis of cell-cell junctional complex in enterocytes that do not display luminal E-cadherin. In comparison, InlA facilitates Lm translocation at cells displaying apical E-cadherin during cell extrusion and mucus expulsion from goblet cells. LAP hijacks caveolar endocytosis to traffic integral junctional proteins to the early and recycling endosomes. Pharmacological inhibition in a cell line and genetic knockout of caveolin-1 in mice prevents LAP-induced intestinal permeability, junctional endocytosis, and Lm translocation. Furthermore, LAP-Hsp60-dependent tight junction remodeling is also necessary for InlA access to E-cadherin for Lm intestinal barrier crossing in InlA-permissive hosts. IMPORTANCE Listeria monocytogenes (Lm) is a foodborne pathogen with high mortality (20%-30%) and hospitalization rates (94%), particularly affecting vulnerable groups such as pregnant women, fetuses, newborns, seniors, and immunocompromised individuals. Invasive listeriosis involves Lm's internalin (InlA) protein binding to E-cadherin to breach the intestinal barrier. However, non-functional InlA variants have been identified in Lm isolates, suggesting InlA-independent pathways for translocation. Our study reveals that Listeria adhesion protein (LAP) and InlA cooperatively assist Lm entry into the gut lamina propria in a gerbil model, mimicking human listeriosis in early infection stages. LAP triggers caveolin-1-mediated endocytosis of critical junctional proteins, transporting them to early and recycling endosomes, facilitating Lm passage through enterocytes. Furthermore, LAP-Hsp60-mediated junctional protein endocytosis precedes InlA's interaction with basolateral E-cadherin, emphasizing LAP and InlA's cooperation in enhancing Lm intestinal translocation. This understanding is vital in combating the severe consequences of Lm infection, including sepsis, meningitis, encephalitis, and brain abscess.
Collapse
Affiliation(s)
- Rishi Drolia
- Department of Food Science, Molecular Food Microbiology Laboratory, Purdue University, West Lafayette, Indiana, USA
- Department of Biological Science, Old Dominion University, Norfolk, Virginia, USA
- Department of Biological Science, Eastern Kentucky University, Richmond, Kentucky, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, Indiana, USA
| | - Donald B. Bryant
- Department of Biological Science, Eastern Kentucky University, Richmond, Kentucky, USA
| | - Shivendra Tenguria
- Department of Food Science, Molecular Food Microbiology Laboratory, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, Indiana, USA
| | - Zuri A. Jules-Culver
- Department of Biological Science, Old Dominion University, Norfolk, Virginia, USA
| | - Jessie Thind
- Department of Biological Science, Eastern Kentucky University, Richmond, Kentucky, USA
| | - Breanna Amelunke
- Department of Biological Science, Eastern Kentucky University, Richmond, Kentucky, USA
| | - Dongqi Liu
- Department of Food Science, Molecular Food Microbiology Laboratory, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, Indiana, USA
| | - Nicholas L. F. Gallina
- Department of Food Science, Molecular Food Microbiology Laboratory, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, Indiana, USA
| | - Krishna K. Mishra
- Department of Food Science, Molecular Food Microbiology Laboratory, Purdue University, West Lafayette, Indiana, USA
| | - Manalee Samaddar
- Department of Food Science, Molecular Food Microbiology Laboratory, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, Indiana, USA
| | - Manoj R. Sawale
- Department of Food Science, Molecular Food Microbiology Laboratory, Purdue University, West Lafayette, Indiana, USA
| | - Dharmendra K. Mishra
- Department of Food Science, Molecular Food Microbiology Laboratory, Purdue University, West Lafayette, Indiana, USA
| | - Abigail D. Cox
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - Arun K. Bhunia
- Department of Food Science, Molecular Food Microbiology Laboratory, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, Indiana, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
4
|
Segui-Perez C, Stapels DAC, Ma Z, Su J, Passchier E, Westendorp B, Wubbolts RW, Wu W, van Putten JPM, Strijbis K. MUC13 negatively regulates tight junction proteins and intestinal epithelial barrier integrity via protein kinase C. J Cell Sci 2024; 137:jcs261468. [PMID: 38345099 PMCID: PMC10984281 DOI: 10.1242/jcs.261468] [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: 07/07/2023] [Accepted: 01/29/2024] [Indexed: 03/14/2024] Open
Abstract
Glycosylated mucin proteins contribute to the essential barrier function of the intestinal epithelium. The transmembrane mucin MUC13 is an abundant intestinal glycoprotein with important functions for mucosal maintenance that are not yet completely understood. We demonstrate that in human intestinal epithelial monolayers, MUC13 localized to both the apical surface and the tight junction (TJ) region on the lateral membrane. MUC13 deletion resulted in increased transepithelial resistance (TEER) and reduced translocation of small solutes. TEER buildup in ΔMUC13 cells could be prevented by addition of MLCK, ROCK or protein kinase C (PKC) inhibitors. The levels of TJ proteins including claudins and occludin were highly increased in membrane fractions of MUC13 knockout cells. Removal of the MUC13 cytoplasmic tail (CT) also altered TJ composition but did not affect TEER. The increased buildup of TJ complexes in ΔMUC13 and MUC13-ΔCT cells was dependent on PKC. The responsible PKC member might be PKCδ (or PRKCD) based on elevated protein levels in the absence of full-length MUC13. Our results demonstrate for the first time that a mucin protein can negatively regulate TJ function and stimulate intestinal barrier permeability.
Collapse
Affiliation(s)
- Celia Segui-Perez
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Daphne A. C. Stapels
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Ziliang Ma
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A*STAR), 138648 Singapore, Singapore
- Department of Pharmacy, National University of Singapore, 117543 Singapore, Singapore
| | - Jinyi Su
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Elsemieke Passchier
- UMAB, Department of Laboratory Pharmacy and Biomedical Genetics, Center for Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Bart Westendorp
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism and Cancer, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Richard W. Wubbolts
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism and Cancer, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Wei Wu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A*STAR), 138648 Singapore, Singapore
- Department of Pharmacy, National University of Singapore, 117543 Singapore, Singapore
| | - Jos P. M. van Putten
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Karin Strijbis
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| |
Collapse
|
5
|
Matheus VA, Oliveira RB, Maschio DA, Tada SFS, Soares GM, Mousovich-Neto F, Costa RG, Mori MA, Barbosa HCL, Collares-Buzato CB. Butyrate restores the fat/lean mass ratio balance and energy metabolism and reinforces the tight junction-mediated intestinal epithelial barrier in prediabetic mice independently of its anti-inflammatory and epigenetic actions. J Nutr Biochem 2023; 120:109409. [PMID: 37364792 DOI: 10.1016/j.jnutbio.2023.109409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/24/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023]
Abstract
Tissue/cellular actions of butyrate on energy metabolism and intestinal barrier in normal metabolic conditions or prediabetes are still unclear. In this work, we investigated the beneficial effect of dietary supplementation with sodium butyrate on energy metabolism, body mass composition, and intestinal epithelial barrier mediated by tight junction (TJ) in chow diet-fed normal and high-fat diet (HF)-fed prediabetic mice, considering the well-known butyrate action as an epigenetic and inflammatory regulator. Butyrate significantly reduced the fat/lean mass ratio, slightly ameliorated dyslipidemia, restored oral glucose tolerance, and increased basal energy expenditure in prediabetic HF-fed mice but had no effect on control animals. Such effects were observed in the absence of significant alterations in the hypothalamic expression of orexigenic and anorexigenic genes and motor activity. Also, butyrate suppressed the whitening effect of HF on brown adipose tissue but did not affect cell bioenergetics in immortalized UCP1-positive adipocytes in vitro. Butyrate reinforced the intestinal epithelial barrier in HF-fed mice and in Caco-2 monolayers, which involved higher trafficking of TJ proteins to the cell-cell contact region of the intestinal epithelia, without affecting TJ gene expression or the acetylation level of histones H3 and H4 in vivo. All metabolic and intestinal effects of butyrate in prediabetic mice occurred in the absence of detectable changes in systemic or local inflammation, or alterations in endotoxemia markers. Butyrate has no effect on chow diet-fed mice but, in the context of HF-induced prediabetes, it prevents metabolic and intestinal dysfunctions independently of its anti-inflammatory and epigenetic actions.
Collapse
Affiliation(s)
- Valquiria A Matheus
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| | - Ricardo B Oliveira
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Daniela A Maschio
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil; Obesity and Comorbidities Research Center, Institute of Biology, UNICAMP, Campinas, Brazil
| | - Susely F S Tada
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Gabriela M Soares
- Obesity and Comorbidities Research Center, Institute of Biology, UNICAMP, Campinas, Brazil
| | - Felippe Mousovich-Neto
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Raul G Costa
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Marcelo A Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil; Obesity and Comorbidities Research Center, Institute of Biology, UNICAMP, Campinas, Brazil
| | - Helena C L Barbosa
- Obesity and Comorbidities Research Center, Institute of Biology, UNICAMP, Campinas, Brazil
| | - Carla B Collares-Buzato
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| |
Collapse
|
6
|
Tokumon R, Sebastián I, Humbel BM, Okura N, Yamanaka H, Yamashiro T, Toma C. Degradation of p0071 and p120-catenin during adherens junction disassembly by Leptospira interrogans. Front Cell Infect Microbiol 2023; 13:1228051. [PMID: 37795382 PMCID: PMC10545952 DOI: 10.3389/fcimb.2023.1228051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/23/2023] [Indexed: 10/06/2023] Open
Abstract
Leptospira interrogans disseminates hematogenously to reach the target organs by disrupting epithelial adherens junctions (AJs), thus causing leptospirosis, which is a globally neglected zoonotic disease. L. interrogans induces E-cadherin (E-cad) endocytosis and cytoskeletal rearrangement during AJ disassembly, but the detailed mechanism remains unknown. Elucidation of AJ disassembly mechanisms will guide new approaches to developing vaccines and diagnostic methods. In this study, we combine proteomic and imaging analysis with chemical inhibition studies to demonstrate that disrupting the AJs of renal proximal tubule epithelial cells involves the degradation of two armadillo repeat-containing proteins, p0071 and p120-catenin, that stabilize E-cad at the plasma membrane. Combining proteasomal and lysosomal inhibitors substantially prevented p120-catenin degradation, and monolayer integrity destruction without preventing p0071 proteolysis. In contrast, the pan-caspase inhibitor Z-VAD-FMK inhibited p0071 proteolysis and displacement of both armadillo repeat-containing proteins from the cell-cell junctions. Our results show that L. interrogans induces p120-catenin and p0071 degradation, which mutually regulates E-cad stability by co-opting multiple cellular degradation pathways. This strategy may allow L. interrogans to disassemble AJs and disseminate through the body efficiently.
Collapse
Affiliation(s)
- Romina Tokumon
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Isabel Sebastián
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Bruno M. Humbel
- Provost Office, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- Microscopy Center, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobuhiko Okura
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Hidenori Yamanaka
- Environmental Technology Department, Chemicals Evaluation and Research Institute, Saitama, Japan
| | - Tetsu Yamashiro
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Claudia Toma
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| |
Collapse
|
7
|
Rajeev V, Chai YL, Poh L, Selvaraji S, Fann DY, Jo DG, De Silva TM, Drummond GR, Sobey CG, Arumugam TV, Chen CP, Lai MKP. Chronic cerebral hypoperfusion: a critical feature in unravelling the etiology of vascular cognitive impairment. Acta Neuropathol Commun 2023; 11:93. [PMID: 37309012 PMCID: PMC10259064 DOI: 10.1186/s40478-023-01590-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/14/2023] Open
Abstract
Vascular cognitive impairment (VCI) describes a wide spectrum of cognitive deficits related to cerebrovascular diseases. Although the loss of blood flow to cortical regions critically involved in cognitive processes must feature as the main driver of VCI, the underlying mechanisms and interactions with related disease processes remain to be fully elucidated. Recent clinical studies of cerebral blood flow measurements have supported the role of chronic cerebral hypoperfusion (CCH) as a major driver of the vascular pathology and clinical manifestations of VCI. Here we review the pathophysiological mechanisms as well as neuropathological changes of CCH. Potential interventional strategies for VCI are also reviewed. A deeper understanding of how CCH can lead to accumulation of VCI-associated pathology could potentially pave the way for early detection and development of disease-modifying therapies, thus allowing preventive interventions instead of symptomatic treatments.
Collapse
Affiliation(s)
- Vismitha Rajeev
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Luting Poh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Sharmelee Selvaraji
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, Singapore
| | - David Y Fann
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - T Michael De Silva
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore.
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
8
|
Javaid S, Alqahtani F, Ashraf W, Anjum SMM, Rasool MF, Ahmad T, Alasmari F, Alasmari AF, Alqarni SA, Imran I. Tiagabine suppresses pentylenetetrazole-induced seizures in mice and improves behavioral and cognitive parameters by modulating BDNF/TrkB expression and neuroinflammatory markers. Biomed Pharmacother 2023; 160:114406. [PMID: 36791567 DOI: 10.1016/j.biopha.2023.114406] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/10/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Tiagabine (Tia), a new-generation antiseizure drug that mimics the GABAergic signaling by inhibiting GABA transporter type-1, is the least studied molecule in chronic epilepsy models with comorbid neurobehavioral and neuroinflammatory parameters. Therefore, the current study investigated the effects of Tia in a real-time manner on electroencephalographic (EEG) activity, behavioral manifestations and mRNA expression in pentylenetetrazole (PTZ)-kindled mice. Male BALB/c mice were treated with tiagabine (0.5, 1 and 2 mg/kg) for 21 days with simultaneous PTZ (40 mg/kg) injection every other day for a total of 11 injections and monitored for seizure progression with synchronized validation through EEG recordings from cortical electrodes. The post-kindling protection from anxiety and memory deficit was verified by a battery of behavioral experiments. Isolated brains were evaluated for oxidative alterations and real-time changes in mRNA expression for BDNF/TrkB, GAT-1 and GAT-3 as well as neuroinflammatory markers. Experimental results revealed that Tia at the dose of 2 mg/kg maximally inhibited the development of full bloom seizure and reduced epileptic spike discharges from the cortex. Furthermore, Tia dose-dependently exerted the anxiolytic effects and protected from PTZ-evoked cognitive impairment. Tia reduced lipid peroxidation and increased superoxide dismutase and glutathione levels in the brain via augmentation of GABAergic modulation. PTZ-induced upregulated BDNF/TrkB signaling and pro-inflammatory cytokines were mitigated by Tia with upregulation of GAT-1 and GAT-3 transporters in whole brains. In conclusion, the observed effects of Tia might have resulted from reduced oxidative stress, BDNF/TrkB modulation and mitigated neuroinflammatory markers expression leading to reduced epileptogenesis and improved epilepsy-related neuropsychiatric effects.
Collapse
Affiliation(s)
- Sana Javaid
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan; Department of Pharmacy, The Women University, Multan 60000, Pakistan
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Waseem Ashraf
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Syed Muhammad Muneeb Anjum
- The Institute of Pharmaceutical Sciences, University of Veterinary & Animal Sciences, Lahore 75270, Pakistan
| | - Muhammad Fawad Rasool
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Tanveer Ahmad
- Institut pour l'Avancée des Biosciences, Centre de Recherche UGA / INSERM U1209 / CNRS 5309, Université Grenoble Alpes, France
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh Abdullah Alqarni
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Imran Imran
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan.
| |
Collapse
|
9
|
He J, Hu L, Deng Q, Sun L, Zhao Y, Fang Z, Wang C, Zhao J. Carboxymethyl pachymaran attenuates short-term stress induced depressive behaviours and over-expression of occludin and claudin-2 in the blood–brain-barrier by regulating inflammatory cytokines- JNK/ERK/p38 pathway. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023] Open
|
10
|
Yang X, Chatterjee V, Zheng E, Reynolds A, Ma Y, Villalba N, Tran T, Jung M, Smith DJ, Wu MH, Yuan SY. Burn Injury-Induced Extracellular Vesicle Production and Characteristics. Shock 2022; 57:228-242. [PMID: 35613455 PMCID: PMC9246995 DOI: 10.1097/shk.0000000000001938] [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] [Indexed: 11/26/2022]
Abstract
ABSTRACT Extracellular vesicles (EVs) are nano-sized membrane-bound particles containing biologically active cargo molecules. The production and molecular composition of EVs reflect the physiological state of parent cells, and once released into the circulation, they exert pleiotropic functions via transferring cargo contents. Thus, circulating EVs not only serve as biomarkers, but also mediators in disease processes or injury responses. In the present study, we performed a comprehensive analysis of plasma EVs from burn patients and healthy subjects, characterizing their size distribution, concentration, temporal changes, cell origins, and cargo protein contents. Our results indicated that burn injury induced a significant increase in circulating EVs, the response peaked at the time of admission and declined over the course of recovery. Importantly, EV production correlated with injury severity, as indicated by the total body surface area and depth of burn, requirement for critical care/ICU stay, hospitalization length, wound infection, and concurrence of sepsis. Burn patients with inhalation injury showed a higher level of EVs than those without inhalation injury. We also evaluated patient demographics (age and sex) and pre-existing conditions (hypertension, obesity, and smoking) and found no significant correlation between these conditions and overall EV production. At the molecular level, flow cytometric analysis showed that the burn-induced EVs were largely derived from leukocytes and endothelial cells (ECs), which are known to be activated postburn. Additionally, a high level of zona-occludens-1 (ZO-1), a major constituent of tight junctions, was identified in burn EV cargos, indicative of injury in tissues that form barriers via tight junctions. Moreover, when applied to endothelial cell monolayers, burn EVs caused significant barrier dysfunction, characterized by decreased transcellular barrier resistance and disrupted cell-cell junction continuity. Taken together, these data suggest that burn injury promotes the production of EVs containing unique cargo proteins in a time-dependent manner; the response correlates with injury severity and worsened clinical outcomes. Functionally, burn EVs serve as a potent mediator capable of reducing endothelial barrier resistance and impairing junction integrity, a pathophysiological process underlying burn-associated tissue dysfunction. Thus, further in-depth characterization of circulating EVs will contribute to the development of new prognostic tools or therapeutic targets for advanced burn care.
Collapse
Affiliation(s)
- Xiaoyuan Yang
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Victor Chatterjee
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Ethan Zheng
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Amanda Reynolds
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Yonggang Ma
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Nuria Villalba
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Thanh Tran
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Michelle Jung
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - David J. Smith
- Department of Plastic Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Mack H. Wu
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Sarah Y. Yuan
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| |
Collapse
|
11
|
Kang J, Zhou Y, Zhu C, Ren T, Zhang Y, Xiao L, Fang B. Ginsenoside Rg1 Mitigates Porcine Intestinal Tight Junction Disruptions Induced by LPS through the p38 MAPK/NLRP3 Inflammasome Pathway. TOXICS 2022; 10:toxics10060285. [PMID: 35736894 PMCID: PMC9228030 DOI: 10.3390/toxics10060285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 12/24/2022]
Abstract
Inflammation leads to porcine tight junction disruption of small intestinal epithelial cells, resulting in intestinal dysfunction. Herein, we established lipopolysaccharide (LPS)-induced in-vivo and in-vitro inflammatory models. The results revealed that LPS induced tight junction disruption in IPEC-J2 cells by downregulating tight-junction-related protein zonula occludens-1 (ZO-1), occludin and claudin-1 expression, while ginsenoside Rg1 rescued such inhibition and abrogated the upregulated expression of phosphorylation p38 MAPK. The p38 MAPK inhibitor (SB203580) showed a similar effect with Rg1 and attenuated the LPS-induced inhibition of ZO-1, occludin and claudin-1 expression, which is consistent with the reduced expression of NLRP3 inflammasome and IL-1β. Furthermore, the specific inhibitors of NLRP3 and IL-1β result in increased expression of tight-junction-related protein, demonstrating that p38 MAPK signaling was associated with Rg1 suppression of tight junction disruption. Besides, LPS treatment decreased the expression of ZO-1, occludin and claudin-1 through p38 MAPK signaling, and caused abnormal morphological changes in murine ileum. Meanwhile, Rg1 attenuated the decreased expression of ZO-1, occludin and claudin-1 and partially alleviated LPS-induced morphological changes in murine ileum. In summary, these findings characterized a novel mechanism by which Rg1 alleviates LPS-induced intestinal tight junction disruption by inhibiting the p38 MAPK-mediated NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Jian Kang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510630, China; (J.K.); (Y.Z.); (C.Z.); (T.R.)
| | - Yanhong Zhou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510630, China; (J.K.); (Y.Z.); (C.Z.); (T.R.)
| | - Chunyang Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510630, China; (J.K.); (Y.Z.); (C.Z.); (T.R.)
| | - Tian Ren
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510630, China; (J.K.); (Y.Z.); (C.Z.); (T.R.)
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China;
| | - Longfei Xiao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 100096, China
- Correspondence: (L.X.); (B.F.)
| | - Binghu Fang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510630, China; (J.K.); (Y.Z.); (C.Z.); (T.R.)
- Correspondence: (L.X.); (B.F.)
| |
Collapse
|
12
|
Puerta-Guardo H, Biering SB, de Sousa FTG, Shu J, Glasner DR, Li J, Blanc SF, Beatty PR, Harris E. Flavivirus NS1 Triggers Tissue-Specific Disassembly of Intercellular Junctions Leading to Barrier Dysfunction and Vascular Leak in a GSK-3β-Dependent Manner. Pathogens 2022; 11:615. [PMID: 35745469 PMCID: PMC9228372 DOI: 10.3390/pathogens11060615] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/13/2022] [Accepted: 05/19/2022] [Indexed: 02/05/2023] Open
Abstract
The flavivirus nonstructural protein 1 (NS1) is secreted from infected cells and contributes to endothelial barrier dysfunction and vascular leak in a tissue-dependent manner. This phenomenon occurs in part via disruption of the endothelial glycocalyx layer (EGL) lining the endothelium. Additionally, we and others have shown that soluble DENV NS1 induces disassembly of intercellular junctions (IJCs), a group of cellular proteins critical for maintaining endothelial homeostasis and regulating vascular permeability; however, the specific mechanisms by which NS1 mediates IJC disruption remain unclear. Here, we investigated the relative contribution of five flavivirus NS1 proteins, from dengue (DENV), Zika (ZIKV), West Nile (WNV), Japanese encephalitis (JEV), and yellow fever (YFV) viruses, to the expression and localization of the intercellular junction proteins β-catenin and VE-cadherin in endothelial cells from human umbilical vein and brain tissues. We found that flavivirus NS1 induced the mislocalization of β-catenin and VE-cadherin in a tissue-dependent manner, reflecting flavivirus disease tropism. Mechanistically, we observed that NS1 treatment of cells triggered internalization of VE-cadherin, likely via clathrin-mediated endocytosis, and phosphorylation of β-catenin, part of a canonical IJC remodeling pathway during breakdown of endothelial barriers that activates glycogen synthase kinase-3β (GSK-3β). Supporting this model, we found that a chemical inhibitor of GSK-3β reduced both NS1-induced permeability of human umbilical vein and brain microvascular endothelial cell monolayers in vitro and vascular leakage in a mouse dorsal intradermal model. These findings provide insight into the molecular mechanisms regulating NS1-mediated endothelial dysfunction and identify GSK-3β as a potential therapeutic target for treatment of vascular leakage during severe dengue disease.
Collapse
Affiliation(s)
- Henry Puerta-Guardo
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
- Laboratorio de Virologia, CIR-Biomedicas y Unidad Colaborativa de Bioensayos Entomologicos (UCBE), Universidad Autonoma de Yucatan, Merida 97000, Mexico
| | - Scott B. Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| | - Francielle Tramontini Gomes de Sousa
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| | - Jeffrey Shu
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| | - Dustin R. Glasner
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| | - Jeffrey Li
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| | - Sophie F. Blanc
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| | - P. Robert Beatty
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| |
Collapse
|
13
|
Collares-Buzato CB, Carvalho CP. Is type 2 diabetes mellitus another intercellular junction-related disorder? Exp Biol Med (Maywood) 2022; 247:743-755. [PMID: 35466731 DOI: 10.1177/15353702221090464] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Type 2 diabetes mellitus (T2D) is nowadays a worldwide epidemic and has become a major challenge for health systems around the world. It is a multifactorial disorder, characterized by a chronic state of hyperglycemia caused by defects in the production as well as in the peripheral action of insulin. This minireview highlights the experimental and clinical evidence that supports the novel idea that intercellular junctions (IJs)-mediated cell-cell contacts play a role in the pathogenesis of T2D. It focuses on IJs repercussion for endocrine pancreas, intestinal barrier, and kidney dysfunctions that contribute to the onset and evolution of this metabolic disorder.
Collapse
Affiliation(s)
- Carla B Collares-Buzato
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, CEP 13083-970, Brazil
| | - Carolina Pf Carvalho
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Santos, SP, CEP 11015-020, Brazil
| |
Collapse
|
14
|
Deoxycholic Acid Modulates Cell-Junction Gene Expression and Increases Intestinal Barrier Dysfunction. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030723. [PMID: 35163990 PMCID: PMC8839472 DOI: 10.3390/molecules27030723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 12/11/2022]
Abstract
Diet-related obesity is associated with increased intestinal hyperpermeability. High dietary fat intake causes an increase in colonic bile acids (BAs), particularly deoxycholic acid (DCA). We hypothesize that DCA modulates the gene expression of multiple cell junction pathways and increases intestinal permeability. With a human Caco-2 cell intestinal model, we used cell proliferation, PCR array, biochemical, and immunofluorescent assays to examine the impact of DCA on the integrity of the intestinal barrier and gene expression. The Caco-2 cells were grown in monolayers and challenged with DCA at physiological, sub-mM, concentrations. DCA increased transcellular and paracellular permeability (>20%). Similarly, DCA increased intracellular reactive oxidative species production (>100%) and accompanied a decrease (>40%) in extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathways. Moreover, the mRNA levels of 23 genes related to the epithelial barrier (tight junction, focal adhesion, gap junction, and adherens junction pathways) were decreased (>40%) in (0.25 mM) DCA-treated Caco-2 cells compared to untreated cells. Finally, we demonstrated that DCA decreased (>58%) the protein content of occludin present at the cellular tight junctions and the nucleus of epithelial cells. Collectively, DCA decreases the gene expression of multiple pathways related to cell junctions and increases permeability in a human intestinal barrier model.
Collapse
|
15
|
Cervero P, Vrenken K, Klose M, Rehm K, Linder S. Nectin stabilization at adherens junctions is counteracted by Rab5a-dependent endocytosis. Eur J Cell Biol 2021; 100:151184. [PMID: 34826799 DOI: 10.1016/j.ejcb.2021.151184] [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: 06/27/2017] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 11/19/2022] Open
Abstract
Cell-cell junctions undergo constant remodeling, which is crucial for the control of vascular integrity. Indeed, transport of junctional components such as cadherins is understood in increasing depth. However, little is known about the respective pathways regulating localization of nectin at cell-cell junctions. Here, we performed an siRNA-based screen of vesicle regulators of the RabGTPase family, leading to the identification of a novel role for Rab5a in the endocytosis nectin-2 at adherens junctions of primary human endothelial cells (HUVEC). Confocal microscopy experiments revealed disordered nectin-2 localization at adherens junctions upon Rab5a depletion. In addition, internalized nectin-2 was shown to prominently localize to Rab5a-positive vesicles in both fixed and living cells. As shown previously, nectin-2 stabilization at junctions is achieved via drebrin-dependent coupling to the subcortical actin cytoskeleton. Consistently, depletion of drebrin in this study leads to enhanced internalization of nectin-2 from junctions. Strikingly, simultaneous silencing of Rab5a and drebrin restored the junctional localization of nectin-2, pointing to Rab5a as counteracting the drebrin-dependent stabilization of nectin-2 at adherens junctions. This mechanism could be further validated by transendothelial resistance measurements. Collectively, our results identify Rab5a as a key player in the endocytosis of nectin-2 and thus in the regulation of adherens junction integrity in primary human endothelial cells.
Collapse
Affiliation(s)
- Pasquale Cervero
- Institut für medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Kirsten Vrenken
- Institut für medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Eppendorf, Martinistr. 52, 20246 Hamburg, Germany; Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, P.O.Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Matthias Klose
- Institut für medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Kerstin Rehm
- Institut für medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - Stefan Linder
- Institut für medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| |
Collapse
|
16
|
Rehman S, Gora AH, Siriyappagouder P, Brugman S, Fernandes JMO, Dias J, Kiron V. Zebrafish intestinal transcriptome highlights subdued inflammatory responses to dietary soya bean and efficacy of yeast β-glucan. JOURNAL OF FISH DISEASES 2021; 44:1619-1637. [PMID: 34237181 DOI: 10.1111/jfd.13484] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 06/13/2023]
Abstract
Anti-nutritional factors in dietary components can have a negative impact on the intestinal barrier. Here, we present soya bean-induced changes in the intestine of juvenile zebrafish and the effect of yeast β-glucan through a transcriptomic approach. The inclusion of soya bean meal affected the expression of several intestinal barrier function-related genes like arl4ca, rab25b, rhoub, muc5ac, muc5d, clcn2c and cltb in zebrafish. Several metabolic genes like cyp2x10.2, cyp2aa2, aldh3a2b, crata, elovl4, elovl6, slc51a, gpat2 and ATP-dependent peptidase activity (lonrf, clpxb) were altered in the intestinal tissue. The expression of immune-related genes like nlrc3, nlrp12, gimap8, prdm1 and tph1a, and genes related to cell cycle, DNA damage and DNA repair (e.g. spo11, rad21l1, nabp1b, spata22, tdrd9) were also affected in the soya bean fed group. Furthermore, our study suggests the plausible effect of yeast β-glucan through the modulation of several genes that regulate immune responses and barrier integrity. Our findings indicate a subdued inflammation in juvenile zebrafish fed soya bean meal and the efficacy of β-glucan to counter these subtle inflammatory responses.
Collapse
Affiliation(s)
- Saima Rehman
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Adnan H Gora
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | | | - Sylvia Brugman
- Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands
| | | | | | - Viswanath Kiron
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| |
Collapse
|
17
|
High-Fat Diet Induces Disruption of the Tight Junction-Mediated Paracellular Barrier in the Proximal Small Intestine Before the Onset of Type 2 Diabetes and Endotoxemia. Dig Dis Sci 2021; 66:3359-3374. [PMID: 33104937 DOI: 10.1007/s10620-020-06664-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 10/06/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND/AIM A link between an impaired intestinal barrier, endotoxemia, and the pathogenesis of metabolic diseases, such as type 2 diabetes mellitus (T2DM), has been proposed. In previous work, we have demonstrated that the tight junction (TJ)-mediated intestinal barrier in ileum/colon was marginally changed in prediabetic mice; therefore, it does not seem to mainly contribute to the T2DM onset. In this study, the TJ-mediated epithelial barrier in the duodenum and jejunum was evaluated in mice during the development of type 2 prediabetes. METHODS/RESULTS HF diet induced prediabetes after 60 days associated with a significant rise in intestinal permeability to the small-sized marker Lucifer yellow in these mice, with no histological signs of mucosal inflammation or rupture of the proximal intestine epithelium. As revealed by immunofluorescence, TJ proteins, such as claudins-1, -2, -3, and ZO-1, showed a significant decrease in junctional content in duodenum and jejunum epithelia, already after 15 days of treatment, suggesting a rearrangement of the TJ structure. However, no significant change in total cell content of these proteins was observed in intestinal epithelium homogenates, as assessed by immunoblotting. Despite the changes in intestinal permeability and TJ structure, the prediabetic mice showed similar LPS, zonulin, and TNF-α levels in plasma or adipose tissue, and in intestinal segments as compared to the controls. CONCLUSION Disruption of the TJ-mediated paracellular barrier in the duodenum and jejunum is an early event in prediabetes development, which occurs in the absence of detectable endotoxemia/inflammation and may contribute to the HF diet-induced increase in intestinal permeability.
Collapse
|
18
|
Bartosova M, Zhang C, Schaefer B, Herzog R, Ridinger D, Damgov I, Levai E, Marinovic I, Eckert C, Romero P, Sallay P, Ujszaszi A, Unterwurzacher M, Wagner A, Hildenbrand G, Warady BA, Schaefer F, Zarogiannis SG, Kratochwill K, Schmitt CP. Glucose Derivative Induced Vasculopathy in Children on Chronic Peritoneal Dialysis. Circ Res 2021; 129:e102-e118. [PMID: 34233458 DOI: 10.1161/circresaha.121.319310] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Maria Bartosova
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
| | - Conghui Zhang
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
| | - Betti Schaefer
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
| | - Rebecca Herzog
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria (R.H., M.U., A.W., K.K.)
| | - David Ridinger
- Kirchhoff Institute for Physics (D.R., G.H.), University of Heidelberg, Heidelberg, Germany
| | - Ivan Damgov
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
| | - Eszter Levai
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
- ELKH-SE, Pediatrics and Nephrology Research Group, Budapest, Hungary (E.L.)
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary (E.L., P.S.)
| | - Iva Marinovic
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
| | - Christoph Eckert
- Institute of Pathology (C.E.), University of Heidelberg, Heidelberg, Germany
| | - Philipp Romero
- Division of Pediatric Surgery, Department of General, Visceral and Transplantation Surgery (P.R.), University of Heidelberg, Heidelberg, Germany
| | - Peter Sallay
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary (E.L., P.S.)
| | - Akos Ujszaszi
- Division of Nephrology, Heidelberg University Hospital, Heidelberg, Germany (A.U.)
| | - Markus Unterwurzacher
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria (R.H., M.U., A.W., K.K.)
| | - Anja Wagner
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria (R.H., M.U., A.W., K.K.)
| | - Georg Hildenbrand
- Kirchhoff Institute for Physics (D.R., G.H.), University of Heidelberg, Heidelberg, Germany
| | | | - Franz Schaefer
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
| | - Sotirios G Zarogiannis
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
- Department of Physiology, Faculty of Medicine, University of Thessaly, Larissa, Greece (S.G.Z.)
| | - Klaus Kratochwill
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria (R.H., M.U., A.W., K.K.)
| | - Claus Peter Schmitt
- Center for Pediatric and Adolescent Medicine (M.B., C.Z., B.S., I.D., E.L., I.M., F.S., S.G.Z., C.P.S.), University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
19
|
Chánez-Paredes S, Montoya-García A, Castro-Ochoa KF, García-Cordero J, Cedillo-Barrón L, Shibayama M, Nava P, Flemming S, Schlegel N, Gautreau AM, Vargas-Robles H, Mondragón-Flores R, Schnoor M. The Arp2/3 Inhibitory Protein Arpin Is Required for Intestinal Epithelial Barrier Integrity. Front Cell Dev Biol 2021; 9:625719. [PMID: 34012961 PMCID: PMC8128147 DOI: 10.3389/fcell.2021.625719] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/17/2021] [Indexed: 12/23/2022] Open
Abstract
The intestinal epithelial barrier (IEB) depends on stable interepithelial protein complexes such as tight junctions (TJ), adherens junctions (AJ), and the actin cytoskeleton. During inflammation, the IEB is compromised due to TJ protein internalization and actin remodeling. An important actin regulator is the actin-related protein 2/3 (Arp2/3) complex, which induces actin branching. Activation of Arp2/3 by nucleation-promoting factors is required for the formation of epithelial monolayers, but little is known about the relevance of Arp2/3 inhibition and endogenous Arp2/3 inhibitory proteins for IEB regulation. We found that the recently identified Arp2/3 inhibitory protein arpin was strongly expressed in intestinal epithelial cells. Arpin expression decreased in response to tumor necrosis factor (TNF)α and interferon (IFN)γ treatment, whereas the expression of gadkin and protein interacting with protein C-kinase α-subunit 1 (PICK1), other Arp2/3 inhibitors, remained unchanged. Of note, arpin coprecipitated with the TJ proteins occludin and claudin-1 and the AJ protein E-cadherin. Arpin depletion altered the architecture of both AJ and TJ, increased actin filament content and actomyosin contractility, and significantly increased epithelial permeability, demonstrating that arpin is indeed required for maintaining IEB integrity. During experimental colitis in mice, arpin expression was also decreased. Analyzing colon tissues from ulcerative colitis patients by Western blot, we found different arpin levels with overall no significant changes. However, in acutely inflamed areas, arpin was significantly reduced compared to non-inflamed areas. Importantly, patients receiving mesalazine had significantly higher arpin levels than untreated patients. As arpin depletion (theoretically meaning more active Arp2/3) increased permeability, we wanted to know whether Arp2/3 inhibition would show the opposite. Indeed, the specific Arp2/3 inhibitor CK666 ameliorated TNFα/IFNγ-induced permeability in established Caco-2 monolayers by preventing TJ disruption. CK666 treatment also attenuated colitis development, colon tissue damage, TJ disruption, and permeability in dextran sulphate sodium (DSS)-treated mice. Our results demonstrate that loss of arpin triggers IEB dysfunction during inflammation and that low arpin levels can be considered a novel hallmark of acute inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | - Mineko Shibayama
- Department of Infectomics and Molecular Pathogenesis, CINVESTAV-IPN, Mexico City, Mexico
| | - Porfirio Nava
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, Mexico City, Mexico
| | - Sven Flemming
- Department of Surgery I, University Hospital Würzburg, Würzburg, Germany
| | - Nicolas Schlegel
- Department of Surgery I, University Hospital Würzburg, Würzburg, Germany
| | | | | | | | - Michael Schnoor
- Department of Molecular Biomedicine, CINVESTAV-IPN, Mexico City, Mexico
| |
Collapse
|
20
|
Ji Y, Fan X, Zhang Y, Li J, Dai Z, Wu Z. Glycine regulates mucosal immunity and the intestinal microbial composition in weaned piglets. Amino Acids 2021; 54:385-398. [PMID: 33839961 DOI: 10.1007/s00726-021-02976-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023]
Abstract
Glycine is an amino acid with a diverse array of health benefits regarding metabolism, immunity, and development. The aim of this study was to test the hypothesis that glycine supplementation alters the intestinal microbial composition and improves the intestinal mucosal immunity of weaned piglets. One hundred and twenty-eight weaned piglets divided into 4 groups were fed with a corn- and soybean meal-based diet supplemented with 0 (control), 0.5, 1, or 2% glycine for 7 days. The intestinal microbiota and tissue samples from the control and the 2% glycine-supplemented piglets were collected for determination of the composition of microbial community and the intestinal mucosal barrier function. Piglets fed with diet containing 2% glycine, instead of 0.5% or 1% glycine, presented elevated average daily gain and feed conversion ratio, as compared with the control. 2% glycine enhanced the abundance of mucins in the jejunum and ileum and mRNA level of porcine β-defensin (pBD) 2 and pBD-3, as well as the protein level of secretory immunoglobulin A (sIgA) in the jejunum. The mRNA expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6, and the protein level of phosphorylated p38 mitogen-activated protein kinase (MAPK), signal transducer and activator of transcription 3 (STAT3), nuclear factor (NF)-κB p65, and claudin-2 in the jejunum were lower in the 2% glycine group than that in the control. In addition, an elevated ratio of CD4+/CD8+ T lymphocytes was observed in the jejunum of piglets receiving diet supplemented with 2% glycine. The colon content of piglets fed with 2% glycine exhibited a reduction in abundance of pathogenic bacteria (Escherichia-Shigella, Clostridium, and Burkholderiales) and an increase in short-chain fatty acid-producing bacteria (Blautia, Lachnospiraceae, Anaerostipes, and Prevotella) in comparison with the control. We conclude that dietary supplementation with 2% glycine improves the intestinal immunological barrier function and the microbial composition, therefore, contributing to the growth performance of weaned piglets.
Collapse
Affiliation(s)
- Yun Ji
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China
| | - Xiaoxiao Fan
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China
| | - Yunchang Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China
| | - Ju Li
- Henan Yinfa Animal Husbandry Co., Xinzheng, 451100, Henan, China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
21
|
Gao Y, Bao X, Meng L, Liu H, Wang J, Zheng N. Aflatoxin B1 and Aflatoxin M1 Induce Compromised Intestinal Integrity through Clathrin-Mediated Endocytosis. Toxins (Basel) 2021; 13:184. [PMID: 33801329 PMCID: PMC8002210 DOI: 10.3390/toxins13030184] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 02/08/2023] Open
Abstract
With the growing diversity and complexity of diet, humans are at risk of simultaneous exposure to aflatoxin B1 (AFB1) and aflatoxin M1 (AFM1), which are well-known contaminants in dairy and other agricultural products worldwide. The intestine represents the first barrier against external contaminants; however, evidence about the combined effect of AFB1 and AFM1 on intestinal integrity is lacking. In vivo, the serum biochemical parameters related to intestinal barrier function, ratio of villus height/crypt depth, and distribution pattern of claudin-1 and zonula occluden-1 were significantly affected in mice exposed to 0.3 mg/kg b.w. AFB1 and 3.0 mg/kg b.w. AFM1. In vitro results on differentiated Caco-2 cells showed that individual and combined AFB1 (0.5 and 4 μg/mL) and AFM1 (0.5 and 4 μg/mL) decreased cell viability and trans-epithelial electrical resistance values as well as increased paracellular permeability of fluorescein isothiocyanate-dextran in a dose-dependent manner. Furthermore, AFM1 aggravated AFB1-induced compromised intestinal barrier, as demonstrated by the down-regulation of tight junction proteins and their redistribution, particularly internalization. Adding the inhibitor chlorpromazine illustrated that clathrin-mediated endocytosis partially contributed to the compromised intestinal integrity. Synergistic and additive effects were the predominant interactions, suggesting that these toxins are likely to have negative effects on human health.
Collapse
Affiliation(s)
- Yanan Gao
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.G.); (X.B.); (L.M.); (H.L.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection, Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xiaoyu Bao
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.G.); (X.B.); (L.M.); (H.L.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection, Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lu Meng
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.G.); (X.B.); (L.M.); (H.L.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection, Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Huimin Liu
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.G.); (X.B.); (L.M.); (H.L.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection, Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jiaqi Wang
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.G.); (X.B.); (L.M.); (H.L.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection, Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.G.); (X.B.); (L.M.); (H.L.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection, Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
22
|
Liu D, Qian T, Sun S, Jiang JJ. Laryngopharyngeal Reflux and Inflammatory Responses in Mucosal Barrier Dysfunction of the Upper Aerodigestive Tract. J Inflamm Res 2021; 13:1291-1304. [PMID: 33447069 PMCID: PMC7801919 DOI: 10.2147/jir.s282809] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/16/2020] [Indexed: 12/29/2022] Open
Abstract
The upper aerodigestive tract (UAT) is the first line of defense against environmental stresses such as antigens, microbes, inhalants, foods, etc., and mucins, intracellular junctions, epithelial cells, and immune cells are the major constituents of this defensive mucosal barrier. Laryngopharyngeal reflux (LPR) is recognized as an independent risk factor for UAT mucosal disorders, and in this review, we describe the components and functions of the mucosal barrier and the results of LPR-induced mucosal inflammation in the UAT. We discuss the interactions between the refluxate and the mucosal components and the mechanisms through which these damaging events disrupt and alter the mucosal barriers. In addition, we discuss the dynamic alterations in the mucosal barrier that might be potential therapeutic targets for LPR-induced disorders.
Collapse
Affiliation(s)
- Danling Liu
- Otorhinolaryngology Department, ENT Institute, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai 200032, People's Republic of China
| | - Tingting Qian
- Otorhinolaryngology Department, ENT Institute, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai 200032, People's Republic of China
| | - Shan Sun
- Otorhinolaryngology Department, ENT Institute, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai 200032, People's Republic of China
| | - Jack J Jiang
- Otorhinolaryngology Department, ENT Institute, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai 200032, People's Republic of China.,Department of Surgery, Division of Otolaryngology Head and Neck Surgery, University of Wisconsin Medical School, Madison, WI 53792-7375, USA
| |
Collapse
|
23
|
Hall CHT, Lee JS, Murphy EM, Gerich ME, Dran R, Glover LE, Abdulla ZI, Skelton MR, Colgan SP. Creatine Transporter, Reduced in Colon Tissues From Patients With Inflammatory Bowel Diseases, Regulates Energy Balance in Intestinal Epithelial Cells, Epithelial Integrity, and Barrier Function. Gastroenterology 2020; 159:984-998.e1. [PMID: 32433978 PMCID: PMC7891846 DOI: 10.1053/j.gastro.2020.05.033] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 05/04/2020] [Accepted: 05/11/2020] [Indexed: 01/03/2023]
Abstract
BACKGROUND & AIMS Patients with inflammatory bowel diseases (IBDs) have intestinal barrier dysfunction. Creatine regulates energy distribution within cells and reduces the severity of colitis in mice. We studied the functions of the creatine transporter solute carrier family 6 member 8 (SLC6A8, also called CRT) in intestinal epithelial cells (IECs) and mice, and we measured levels in mucosal biopsies from patients with IBD. METHODS Colon biopsy specimens from patients with IBD (30 with Crohn's disease and 27 with ulcerative colitis) and 30 patients without IBD (control individuals) and colon tissues from mice (with and without disruption of Crt) were analyzed by immunofluorescence, immunoblots, and/or quantitative reverse-transcription polymerase chain reaction (qRT-PCR). CRT was knocked down or overexpressed in T84 cells, which were analyzed by immunofluorescence, immunoblots, high-performance liquid chromatography (to measure creatine levels), qRT-PCR, transepithelial electrical resistance, barrier function, actin localization, wound healing, mitochondrial oxygen consumption, and glycolysis extracellular acidification rate assays. Organoids from colon cells of CRT-knockout mice and control mice were analyzed by qRT-PCR, immunoblot, and transepithelial electrical resistance. RESULTS CRT localized around tight junctions (TJs) of T84 IECs. In analyses of IECs with CRT knockdown or overexpression, we found that CRT regulates intracellular creatine, barrier formation, and wound healing. CRT-knockout organoids also had diminished barrier formation. In the absence of adequate creatine, IECs transition toward a stressed, glycolysis-predominant form of metabolism; this resulted in leaky TJs and mislocalization of actin and TJ proteins. Colon tissues from patients with IBD had reduced levels of CRT messenger RNA compared with those from control individuals. CONCLUSIONS In an analysis of IEC cell lines and colonoids derived from CRT-knockout mice, we found that CRT regulates energy balance in IECs and thereby epithelial integrity and barrier function. Mucosal biopsy specimens from patients with ulcerative colitis and inactive Crohn's disease have lower levels of CRT, which might contribute to the reduced barrier function observed in patients with IBD.
Collapse
Affiliation(s)
- Caroline H T Hall
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital Colorado, Aurora, Colorado; Mucosal Inflammation Program, University of Colorado, Aurora, Colorado
| | - J Scott Lee
- Mucosal Inflammation Program, University of Colorado, Aurora, Colorado; Division of Gastroenterology and Hepatology, University of Colorado, Aurora, Colorado
| | - Emily M Murphy
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital Colorado, Aurora, Colorado; Mucosal Inflammation Program, University of Colorado, Aurora, Colorado; Division of Gastroenterology and Hepatology, University of Colorado, Aurora, Colorado
| | - Mark E Gerich
- Mucosal Inflammation Program, University of Colorado, Aurora, Colorado; Division of Gastroenterology and Hepatology, University of Colorado, Aurora, Colorado
| | - Rachael Dran
- Mucosal Inflammation Program, University of Colorado, Aurora, Colorado; Division of Gastroenterology and Hepatology, University of Colorado, Aurora, Colorado
| | - Louis E Glover
- Mucosal Inflammation Program, University of Colorado, Aurora, Colorado; Division of Gastroenterology and Hepatology, University of Colorado, Aurora, Colorado; School of Biochemistry and Immunology, Trinity College Dublin, Ireland
| | - Zuhair I Abdulla
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, Ohio
| | - Matthew R Skelton
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, Ohio
| | - Sean P Colgan
- Mucosal Inflammation Program, University of Colorado, Aurora, Colorado; Division of Gastroenterology and Hepatology, University of Colorado, Aurora, Colorado.
| |
Collapse
|
24
|
Slifer ZM, Blikslager AT. The Integral Role of Tight Junction Proteins in the Repair of Injured Intestinal Epithelium. Int J Mol Sci 2020; 21:ijms21030972. [PMID: 32024112 PMCID: PMC7036844 DOI: 10.3390/ijms21030972] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/10/2019] [Accepted: 10/14/2019] [Indexed: 12/29/2022] Open
Abstract
The intestinal epithelial monolayer forms a transcellular and paracellular barrier that separates luminal contents from the interstitium. The paracellular barrier consists of a highly organized complex of intercellular junctions that is primarily regulated by apical tight junction proteins and tight junction-associated proteins. This homeostatic barrier can be lost through a multitude of injurious events that cause the disruption of the tight junction complex. Acute repair after injury leading to the reestablishment of the tight junction barrier is crucial for the return of both barrier function as well as other cellular functions, including water regulation and nutrient absorption. This review provides an overview of the tight junction complex components and how they link to other plasmalemmal proteins, such as ion channels and transporters, to induce tight junction closure during repair of acute injury. Understanding the components of interepithelial tight junctions and the mechanisms of tight junction regulation after injury is crucial for developing future therapeutic targets for patients experiencing dysregulated intestinal permeability.
Collapse
|
25
|
de Oliveira RB, Matheus VA, Canuto LP, De Sant'ana A, Collares-Buzato CB. Time-dependent alteration to the tight junction structure of distal intestinal epithelia in type 2 prediabetic mice. Life Sci 2019; 238:116971. [PMID: 31634462 DOI: 10.1016/j.lfs.2019.116971] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/13/2019] [Accepted: 10/14/2019] [Indexed: 01/18/2023]
Abstract
AIM High-fat diet (HFD) intake has been associated with changes in intestinal microbiota composition, increased intestinal permeability, and onset of type 2 diabetes mellitus (T2DM). The aim of this work was twofold: 1) to investigate the structural and functional alterations of the tight junction (TJ)-mediated intestinal epithelial barrier of ileum and colon, that concentrate most of the microbiota, after exposure to a HFD for 15, 30 and 60 days, and 2) to assess the effect of in vitro exposure to free fatty acids (FFAs), one of the components of HFD, on paracellular barrier of colon-derived Caco-2 cells. METHODS/KEY FINDINGS HFD exposure induced progressive metabolic changes in male mice that culminated in prediabetes after 60d. Morphological analysis of ileum and colon mucosa showed no signs of epithelial rupture or local inflammation but changes in the junctional content/distribution and/or cellular content of TJ-associated proteins (claudins-1, -2, -3, and occludin) in intestinal epithelia were seen mainly after a prediabetes state has been established. This impairment in TJ structure was not associated with significant changes in intestinal permeability to FITC-dextran. Exposure of Caco-2 monolayers to palmitic or linoleic acids seems to induce a reinforcement of TJ structure while treatment with oleic acid had a more diverse effect on TJ protein distribution. SIGNIFICANCE TJ structure in distal intestinal epithelia can be specifically impaired by HFD intake at early stage of T2DM, but not by FFAs in vitro. Since the TJ change in ileum/colon was marginal, probably it does not contribute to the disease onset.
Collapse
Affiliation(s)
- Ricardo Beltrame de Oliveira
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Valquiria Aparecida Matheus
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Leandro Pereira Canuto
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Ariane De Sant'ana
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Carla Beatriz Collares-Buzato
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
26
|
Ma Q, Zhang Z, Shim JK, Venkatraman TN, Lascola CD, Quinones QJ, Mathew JP, Terrando N, Podgoreanu MV. Annexin A1 Bioactive Peptide Promotes Resolution of Neuroinflammation in a Rat Model of Exsanguinating Cardiac Arrest Treated by Emergency Preservation and Resuscitation. Front Neurosci 2019; 13:608. [PMID: 31258464 PMCID: PMC6587399 DOI: 10.3389/fnins.2019.00608] [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: 03/01/2019] [Accepted: 05/28/2019] [Indexed: 12/19/2022] Open
Abstract
Neuroinflammation initiated by damage-associated molecular patterns, including high mobility group box 1 protein (HMGB1), has been implicated in adverse neurological outcomes following lethal hemorrhagic shock and polytrauma. Emergency preservation and resuscitation (EPR) is a novel method of resuscitation for victims of exsanguinating cardiac arrest, shown in preclinical studies to improve survival with acceptable neurological recovery. Sirtuin 3 (SIRT3), the primary mitochondrial deacetylase, has emerged as a key regulator of metabolic and energy stress response pathways in the brain and a pharmacological target to induce a neuronal pro-survival phenotype. This study aims to examine whether systemic administration of an Annexin-A1 bioactive peptide (ANXA1sp) could resolve neuroinflammation and induce sirtuin-3 regulated cytoprotective pathways in a novel rat model of exsanguinating cardiac arrest and EPR. Adult male rats underwent hemorrhagic shock and ventricular fibrillation, induction of profound hypothermia, followed by resuscitation and rewarming using cardiopulmonary bypass (EPR). Animals randomly received ANXA1sp (3 mg/kg, in divided doses) or vehicle. Neuroinflammation (HMGB1, TNFα, IL-6, and IL-10 levels), cerebral cell death (TUNEL, caspase-3, pro and antiapoptotic protein levels), and neurologic scores were assessed to evaluate the inflammation resolving effects of ANXA1sp following EPR. Furthermore, western blot analysis and immunohistochemistry were used to interrogate the mechanisms involved. Compared to vehicle controls, ANXA1sp effectively reduced expression of cerebral HMGB1, IL-6, and TNFα and increased IL-10 expression, which were associated with improved neurological scores. ANXA1sp reversed EPR-induced increases in expression of proapoptotic protein Bax and reduction in antiapoptotic protein Bcl-2, with a corresponding decrease in cerebral levels of cleaved caspase-3. Furthermore, ANXA1sp induced autophagic flux (increased LC3II and reduced p62 expression) in the brain. Mechanistically, these findings were accompanied by upregulation of the mitochondrial protein deacetylase Sirtuin-3, and its downstream targets FOXO3a and MnSOD in ANXA1sp-treated animals. Our data provide new evidence that engaging pro-resolving pharmacological strategies such as Annexin-A1 biomimetic peptides can effectively attenuate neuroinflammation and enhance the neuroprotective effects of EPR after exsanguinating cardiac arrest.
Collapse
Affiliation(s)
- Qing Ma
- Systems Modeling of Perioperative Organ Injury Laboratory, Department of Anesthesiology, Duke University, Durham, NC, United States
| | - Zhiquan Zhang
- Neuroinflammation and Cognitive Outcomes Laboratory, Department of Anesthesiology, Duke University, Durham, NC, United States.,Center for Translational Pain Medicine, Duke University, Durham, NC, United States
| | - Jae-Kwang Shim
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Christopher D Lascola
- Departments of Radiology and Neurobiology, Duke University, Durham, NC, United States.,Duke-UNC Brain Imaging and Analysis Center, Duke University, Durham, NC, United States
| | - Quintin J Quinones
- Systems Modeling of Perioperative Organ Injury Laboratory, Department of Anesthesiology, Duke University, Durham, NC, United States
| | - Joseph P Mathew
- Department of Anesthesiology, Duke University, Durham, NC, United States
| | - Niccolò Terrando
- Neuroinflammation and Cognitive Outcomes Laboratory, Department of Anesthesiology, Duke University, Durham, NC, United States.,Center for Translational Pain Medicine, Duke University, Durham, NC, United States
| | - Mihai V Podgoreanu
- Systems Modeling of Perioperative Organ Injury Laboratory, Department of Anesthesiology, Duke University, Durham, NC, United States
| |
Collapse
|
27
|
Maes M, Sirivichayakul S, Kanchanatawan B, Vodjani A. Breakdown of the Paracellular Tight and Adherens Junctions in the Gut and Blood Brain Barrier and Damage to the Vascular Barrier in Patients with Deficit Schizophrenia. Neurotox Res 2019; 36:306-322. [PMID: 31077000 DOI: 10.1007/s12640-019-00054-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/29/2019] [Accepted: 04/23/2019] [Indexed: 12/15/2022]
Abstract
Deficit schizophrenia is characterized by leaky intestinal tight and adherens junctions and bacterial translocation. Here we examine whether (deficit) schizophrenia is accompanied by leaky paracellular, transcellular, and vascular barriers in the gut and blood-brain barriers. We measured IgA responses to occludin, claudin-5, E-cadherin, and β-catenin (paracellular pathway, PARA); talin, actin, vinculin, and epithelial intermediate filament (transcellular pathway, TRANS); and plasmalemma vesicle-associated protein (PLVAP, vascular pathway) in 78 schizophrenia patients and 40 controls. IgA responses to claudin-5, E-cadherin, and β-catenin, the sum of the four PARA proteins, and the ratio PARA/TRANS were significantly higher in deficit schizophrenia patients than in nondeficit schizophrenia patients and controls. A large part of the variance in PHEMN (psychosis, hostility, excitation, mannerism, and negative) symptoms, psychomotor retardation, formal thought disorders, verbal fluency, word list memory, word list recall, and executive functions was explained by the PARA/TRANS ratio coupled with plasma IgA responses to Gram-negative bacteria, IgM to malondialdehyde, CCL-11 (eotaxin), IgA levels of the ratio of noxious to more protective tryptophan catabolites (NOX/PRO TRYCATs), and a plasma immune activation index. Moreover, IgA levels to Gram-negative bacteria were significantly associated with IgA to E-cadherin, β-catenin, and PLVAP, while IgA levels to claudin-5 were significantly predicted by IgA to E-cadherin, NOX/PRO TRYCAT ratio, Gram-negative bacteria, and CCL11. The phenomenology of the deficit syndrome is to a large extent explained by the cumulative effects of lowered natural IgM, breakdown of the paracellular and vascular pathways, increased bacterial translocation, peripheral immune-inflammatory responses, and indices of BBB breakdown.
Collapse
Affiliation(s)
- Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand. .,Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria. .,IMPACT Strategic Research Center, Barwon Health, Deakin University, Geelong, VIC, Australia.
| | | | - Buranee Kanchanatawan
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Aristo Vodjani
- Immunosciences Lab., Inc, Los Angeles, CA, USA.,Cyrex Labs, LLC, Phoenix, AZ, USA.,Department of Preventive Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
28
|
Maes M, Sirivichayakul S, Kanchanatawan B, Vodjani A. Upregulation of the Intestinal Paracellular Pathway with Breakdown of Tight and Adherens Junctions in Deficit Schizophrenia. Mol Neurobiol 2019; 56:7056-7073. [PMID: 30972627 DOI: 10.1007/s12035-019-1578-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 03/20/2019] [Indexed: 12/12/2022]
Abstract
In 2001, the first author of this paper reported that schizophrenia is associated with an increased frequency of the haptoglobin (Hp)-2 gene. The precursor of Hp-2 is zonulin, a molecule that affects intercellular tight junction integrity. Recently, we reported increased plasma IgA/IgM responses to Gram-negative bacteria in deficit schizophrenia indicating leaky gut and gut dysbiosis. The current study was performed to examine the integrity of the paracellular (tight and adherens junctions) and transcellular (cytoskeletal proteins) pathways in deficit versus non-deficit schizophrenia. We measured IgM responses to zonulin, occludin, E-cadherin, talin, actin, and vinculin in association with IgA responses to Gram-negative bacteria, CCL-11, IgA responses to tryptophan catabolites (TRYCATs), immune activation and IgM to malondialdehyde (MDA), and NO-cysteinyl in 78 schizophrenia patients and 40 controls. We found that the ratio of IgM to zonulin + occludin/talin + actin + viculin (PARA/TRANS) was significantly greater in deficit than those in non-deficit schizophrenia and higher in schizophrenia than those in controls and was significantly associated with increased IgA responses to Gram-negative bacteria. IgM responses to zonulin were positively associated with schizophrenia (versus controls), while IgM to occludin was significantly associated with deficit schizophrenia (versus non-deficit schizophrenia and controls). A large part of the variance (90.8%) in negative and PHEM (psychosis, hostility, excitation, and mannerism) symptoms was explained by PARA/TRANS ratio, IgA to Gram-negative bacteria, IgM to E-cadherin and MDA, and memory dysfunctions, while 53.3% of the variance in the latter was explained by PARA/TRANS ratio, IgA to Gram-negative bacteria, CCL-11, TRYCATs, and immune activation. The results show an upregulated paracellular pathway with breakdown of the tight and adherens junctions and increased bacterial translocation in deficit schizophrenia. These dysfunctions in the intestinal paracellular route together with lowered natural IgM, immune activation, and production of CCL-11 and TRYCATs contribute to the phenomenology of deficit schizophrenia.
Collapse
Affiliation(s)
- Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand. .,Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria. .,IMPACT Strategic Research Center, Barwon Health, Deakin University, Geelong, Vic, Australia.
| | | | - Buranee Kanchanatawan
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Aristo Vodjani
- Immunosciences Laboratory, Inc., Los Angeles, CA, USA.,Cyrex Laboratories, LLC, Phoenix, AZ, USA.,Department of Preventive Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
29
|
Zeisel MB, Dhawan P, Baumert TF. Tight junction proteins in gastrointestinal and liver disease. Gut 2019; 68:547-561. [PMID: 30297438 PMCID: PMC6453741 DOI: 10.1136/gutjnl-2018-316906] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/16/2018] [Accepted: 08/19/2018] [Indexed: 12/11/2022]
Abstract
Over the past two decades a growing body of evidence has demonstrated an important role of tight junction (TJ) proteins in the physiology and disease biology of GI and liver disease. On one side, TJ proteins exert their functional role as integral proteins of TJs in forming barriers in the gut and the liver. Furthermore, TJ proteins can also be expressed outside TJs where they play important functional roles in signalling, trafficking and regulation of gene expression. A hallmark of TJ proteins in disease biology is their functional role in epithelial-to-mesenchymal transition. A causative role of TJ proteins has been established in the pathogenesis of colorectal cancer and gastric cancer. Among the best characterised roles of TJ proteins in liver disease biology is their function as cell entry receptors for HCV-one of the most common causes of hepatocellular carcinoma. At the same time TJ proteins are emerging as targets for novel therapeutic approaches for GI and liver disease. Here we review our current knowledge of the role of TJ proteins in the pathogenesis of GI and liver disease biology and discuss their potential as therapeutic targets.
Collapse
Affiliation(s)
- Mirjam B. Zeisel
- Inserm U1052, CNRS UMR 5286, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL), Lyon, France
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE
- Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE
- VA Nebraska-Western Iowa Health Care System, Omaha, NE
| | - Thomas F. Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Institut Hospitalo-Universitaire, Pôle hépato-digestif, Nouvel Hôpital Civil, Strasbourg, France
| |
Collapse
|
30
|
Groeger S, Meyle J. Oral Mucosal Epithelial Cells. Front Immunol 2019; 10:208. [PMID: 30837987 PMCID: PMC6383680 DOI: 10.3389/fimmu.2019.00208] [Citation(s) in RCA: 204] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 01/23/2019] [Indexed: 12/14/2022] Open
Abstract
Cellular Phenotype and Apoptosis: The function of epithelial tissues is the protection of the organism from chemical, microbial, and physical challenges which is indispensable for viability. To fulfill this task, oral epithelial cells follow a strongly regulated scheme of differentiation that results in the formation of structural proteins that manage the integrity of epithelial tissues and operate as a barrier. Oral epithelial cells are connected by various transmembrane proteins with specialized structures and functions. Keratin filaments adhere to the plasma membrane by desmosomes building a three-dimensional matrix. Cell-Cell Contacts and Bacterial Influence: It is known that pathogenic oral bacteria are able to affect the expression and configuration of cell-cell junctions. Human keratinocytes up-regulate immune-modulatory receptors upon stimulation with bacterial components. Periodontal pathogens including P. gingivalis are able to inhibit oral epithelial innate immune responses through various mechanisms and to escape from host immune reaction, which supports the persistence of periodontitis and furthermore is able to affect the epithelial barrier function by altering expression and distribution of cell-cell interactions including tight junctions (TJs) and adherens junctions (AJs). In the pathogenesis of periodontitis a highly organized biofilm community shifts from symbiosis to dysbiosis which results in destructive local inflammatory reactions. Cellular Receptors: Cell-surface located toll like receptors (TLRs) and cytoplasmatic nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) belong to the pattern recognition receptors (PRRs). PRRs recognize microbial parts that represent pathogen-associated molecular patterns (PAMPs). A multimeric complex of proteins known as inflammasome, which is a subset of NLRs, assembles after activation and proceeds to pro-inflammatory cytokine release. Cytokine Production and Release: Cytokines and bacterial products may lead to host cell mediated tissue destruction. Keratinocytes are able to produce diverse pro-inflammatory cytokines and chemokines, including interleukin (IL)-1, IL-6, IL-8 and tumor necrosis factor (TNF)-α. Infection by pathogenic bacteria such as Porphyromonas gingivalis (P. gingivalis) and Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans) can induce a differentiated production of these cytokines. Immuno-modulation, Bacterial Infection, and Cancer Cells: There is a known association between bacterial infection and cancer. Bacterial components are able to up-regulate immune-modulatory receptors on cancer cells. Interactions of bacteria with tumor cells could support malignant transformation an environment with deficient immune regulation. The aim of this review is to present a set of molecular mechanisms of oral epithelial cells and their reactions to a number of toxic influences.
Collapse
Affiliation(s)
- Sabine Groeger
- Department of Periodontology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Joerg Meyle
- Department of Periodontology, Justus-Liebig-University of Giessen, Giessen, Germany
| |
Collapse
|
31
|
Paudel YN, Shaikh MF, Chakraborti A, Kumari Y, Aledo-Serrano Á, Aleksovska K, Alvim MKM, Othman I. HMGB1: A Common Biomarker and Potential Target for TBI, Neuroinflammation, Epilepsy, and Cognitive Dysfunction. Front Neurosci 2018; 12:628. [PMID: 30271319 PMCID: PMC6142787 DOI: 10.3389/fnins.2018.00628] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/21/2018] [Indexed: 12/13/2022] Open
Abstract
High mobility group box protein 1 (HMGB1) is a ubiquitous nuclear protein released by glia and neurons upon inflammasome activation and activates receptor for advanced glycation end products (RAGE) and toll-like receptor (TLR) 4 on the target cells. HMGB1/TLR4 axis is a key initiator of neuroinflammation. In recent days, more attention has been paid to HMGB1 due to its contribution in traumatic brain injury (TBI), neuroinflammatory conditions, epileptogenesis, and cognitive impairments and has emerged as a novel target for those conditions. Nevertheless, HMGB1 has not been portrayed as a common prognostic biomarker for these HMGB1 mediated pathologies. The current review discusses the contribution of HMGB1/TLR4/RAGE signaling in several brain injury, neuroinflammation mediated disorders, epileptogenesis and cognitive dysfunctions and in the light of available evidence, argued the possibilities of HMGB1 as a common viable biomarker of the above mentioned neurological dysfunctions. Furthermore, the review also addresses the result of preclinical studies focused on HMGB1 targeted therapy by the HMGB1 antagonist in several ranges of HMGB1 mediated conditions and noted an encouraging result. These findings suggest HMGB1 as a potential candidate to be a common biomarker of TBI, neuroinflammation, epileptogenesis, and cognitive dysfunctions which can be used for early prediction and progression of those neurological diseases. Future study should explore toward the translational implication of HMGB1 which can open the windows of opportunities for the development of innovative therapeutics that could prevent several associated HMGB1 mediated pathologies discussed herein.
Collapse
Affiliation(s)
- Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Ayanabha Chakraborti
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yatinesh Kumari
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Ángel Aledo-Serrano
- Department of Neurology, Epilepsy Program, Hospital Ruber Internacional, Madrid, Spain
| | - Katina Aleksovska
- Medical Faculty, Department of Neurology, "Saints Cyril and Methodius" University, Skopje, Macedonia
| | | | - Iekhsan Othman
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
32
|
Blaschke M, Koepp R, Lenz C, Kruppa J, Jung K, Siggelkow H. Crohn's disease patient serum changes protein expression in a human mesenchymal stem cell model in a linear relationship to patients' disease stage and to bone mineral density. J Clin Transl Endocrinol 2018; 13:26-38. [PMID: 30003044 PMCID: PMC6039964 DOI: 10.1016/j.jcte.2018.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/07/2018] [Accepted: 06/18/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Crohn's disease (CD) is associated with a higher prevalence of osteoporosis, a complication that is recognized as a significant cause of morbidity. Its pathogenesis is controversial, but the activity of CD is one contributing factor. METHODS We stimulated SCP-1 cells (mesenchymal stem cell line) under osteogenic conditions with serum from adult patients with CD in the symptomatic phase (SP) and in remission (R) and with control sera. Concentrations of IL-6, IL-1 beta, and TNF alpha in the sera were measured. Patients were classified as normal or osteopenic/osteoporotic based on bone mineral density (BMD) T-score measurements. After 14 days in culture, protein expression and gene ontology (GO) annotation analysis was performed. RESULTS Cytokine concentrations (IL-6, IL-1 beta, TNF alpha) varied within sera groups. None of the cytokines were significantly increased in the symptomatic phase compared to remission. Protein analysis revealed 17 proteins regulated by the SP versus R phase sera of disease. A linear relationship between CDAI (Crohn's disease activity index) and normalized protein expression of APOA1 and 2, TTR, CDKAL1 and TUBB6 could be determined. Eleven proteins were found to be differentially regulated comparing osteoporosis-positive and osteoporosis-negative sera. Gene annotation and further analysis identified these genes as part of heme and erythrocyte metabolism, as well as involved in hypoxia and in endocytosis. A significant linear relationship between bone mineral density and normalized protein expression could be determined for proteins FABP3 and TTR. CONCLUSION Our explorative results confirm our hypothesis that factors in serum from patients with CD change the protein expression pattern of human immortalized osteoblast like cells. We suggest, that these short time changes indeed influence factors of bone metabolism.
Collapse
Affiliation(s)
- Martina Blaschke
- Clinic of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Regine Koepp
- Clinic of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Christof Lenz
- Bioanalytical Mass Spectrometry, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
- Institute of Clinical Chemistry, University Medical Center, 37075 Göttingen, Germany
| | - Jochen Kruppa
- Department of Medical Statistics, University Medical Center Göttingen, Humboldtallee 32, 37073 Göttingen, Germany
- Genomics and Bioinformatics of Infectious Diseases, University of Veterinary Medicine Hannover, Institute of animal breeding and Genetics, 30559 Hannover, Germany
| | - Klaus Jung
- Department of Medical Statistics, University Medical Center Göttingen, Humboldtallee 32, 37073 Göttingen, Germany
- Institute of Biometry and Clinical Epidemiology, Charite, University Medical Center Berlin, Chariteplatz 1, 10117 Berlin, Germany
| | - Heide Siggelkow
- Clinic of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, 37075 Göttingen, Germany
- MVZ Endokrinologikum Göttingen, von Siebold-Str. 3, 37075 Göttingen, Germany
| |
Collapse
|
33
|
Jia Z, Wang X, Wei X, Zhao G, Foster KW, Qiu F, Gao Y, Yuan F, Yu F, Thiele GM, Bronich TK, O’Dell JR, Wang D. Micelle-Forming Dexamethasone Prodrug Attenuates Nephritis in Lupus-Prone Mice without Apparent Glucocorticoid Side Effects. ACS NANO 2018; 12:7663-7681. [PMID: 29965725 PMCID: PMC6117746 DOI: 10.1021/acsnano.8b01249] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 07/02/2018] [Indexed: 05/27/2023]
Abstract
Nephritis is one of the major complications of systemic lupus erythematosus. While glucocorticoids (GCs) are frequently used as the first-line treatment for lupus nephritis (LN), long-term GC usage is often complicated by severe adverse effects. To address this challenge, we have developed a polyethylene glycol-based macromolecular prodrug (ZSJ-0228) of dexamethasone, which self-assembles into micelles in aqueous media. When compared to the dose equivalent daily dexamethasone 21-phosphate disodium (Dex) treatment, monthly intravenous administration of ZSJ-0228 for two months significantly improved the survival of lupus-prone NZB/W F1 mice and was much more effective in normalizing proteinuria, with clear histological evidence of nephritis resolution. Different from the dose equivalent daily Dex treatment, monthly ZSJ-0228 administration has no impact on the serum anti-double-stranded DNA (anti-dsDNA) antibody level but can significantly reduce renal immune complex deposition. No significant systemic toxicities of GCs ( e. g., total IgG reduction, adrenal gland atrophy, and osteopenia) were found to be associated with ZSJ-0228 treatment. In vivo imaging and flow cytometry studies revealed that the fluorescent-labeled ZSJ-0228 primarily distributed to the inflamed kidney after systemic administration, with renal myeloid cells and proximal tubular epithelial cells mainly responsible for its kidney retention. Collectively, these data suggest that the ZSJ-0228's potent local anti-inflammatory/immunosuppressive effects and improved safety may be attributed to its nephrotropicity and cellular sequestration at the inflamed kidney tissues. Pending further optimization, it may be developed into an effective and safe therapy for improved clinical management of LN.
Collapse
Affiliation(s)
- Zhenshan Jia
- Department
of Pharmaceutical Sciences, College of Pharmacy, Department of Pathology
and Microbiology, College of Medicine, Department of Biostatistics, College
of Public Health, and Division of Rheumatology, Department of Internal
Medicine, College of Medicine, University
of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Xiaobei Wang
- Department
of Pharmaceutical Sciences, College of Pharmacy, Department of Pathology
and Microbiology, College of Medicine, Department of Biostatistics, College
of Public Health, and Division of Rheumatology, Department of Internal
Medicine, College of Medicine, University
of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Xin Wei
- Department
of Pharmaceutical Sciences, College of Pharmacy, Department of Pathology
and Microbiology, College of Medicine, Department of Biostatistics, College
of Public Health, and Division of Rheumatology, Department of Internal
Medicine, College of Medicine, University
of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Gang Zhao
- Department
of Pharmaceutical Sciences, College of Pharmacy, Department of Pathology
and Microbiology, College of Medicine, Department of Biostatistics, College
of Public Health, and Division of Rheumatology, Department of Internal
Medicine, College of Medicine, University
of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Kirk W. Foster
- Department
of Pharmaceutical Sciences, College of Pharmacy, Department of Pathology
and Microbiology, College of Medicine, Department of Biostatistics, College
of Public Health, and Division of Rheumatology, Department of Internal
Medicine, College of Medicine, University
of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Fang Qiu
- Department
of Pharmaceutical Sciences, College of Pharmacy, Department of Pathology
and Microbiology, College of Medicine, Department of Biostatistics, College
of Public Health, and Division of Rheumatology, Department of Internal
Medicine, College of Medicine, University
of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Yangyang Gao
- Department
of Pharmaceutical Sciences, College of Pharmacy, Department of Pathology
and Microbiology, College of Medicine, Department of Biostatistics, College
of Public Health, and Division of Rheumatology, Department of Internal
Medicine, College of Medicine, University
of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Fang Yuan
- Department
of Pharmaceutical Sciences, College of Pharmacy, Department of Pathology
and Microbiology, College of Medicine, Department of Biostatistics, College
of Public Health, and Division of Rheumatology, Department of Internal
Medicine, College of Medicine, University
of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Fang Yu
- Department
of Pharmaceutical Sciences, College of Pharmacy, Department of Pathology
and Microbiology, College of Medicine, Department of Biostatistics, College
of Public Health, and Division of Rheumatology, Department of Internal
Medicine, College of Medicine, University
of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Geoffrey M. Thiele
- Department
of Pharmaceutical Sciences, College of Pharmacy, Department of Pathology
and Microbiology, College of Medicine, Department of Biostatistics, College
of Public Health, and Division of Rheumatology, Department of Internal
Medicine, College of Medicine, University
of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Tatiana K. Bronich
- Department
of Pharmaceutical Sciences, College of Pharmacy, Department of Pathology
and Microbiology, College of Medicine, Department of Biostatistics, College
of Public Health, and Division of Rheumatology, Department of Internal
Medicine, College of Medicine, University
of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - James R. O’Dell
- Department
of Pharmaceutical Sciences, College of Pharmacy, Department of Pathology
and Microbiology, College of Medicine, Department of Biostatistics, College
of Public Health, and Division of Rheumatology, Department of Internal
Medicine, College of Medicine, University
of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Dong Wang
- Department
of Pharmaceutical Sciences, College of Pharmacy, Department of Pathology
and Microbiology, College of Medicine, Department of Biostatistics, College
of Public Health, and Division of Rheumatology, Department of Internal
Medicine, College of Medicine, University
of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| |
Collapse
|
34
|
Gu M, Jia Q, Zhang Z, Bai N, Xu X, Xu B. Soya-saponins induce intestinal inflammation and barrier dysfunction in juvenile turbot (Scophthalmus maximus). FISH & SHELLFISH IMMUNOLOGY 2018; 77:264-272. [PMID: 29625242 DOI: 10.1016/j.fsi.2018.04.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 02/25/2018] [Accepted: 04/02/2018] [Indexed: 06/08/2023]
Abstract
Soybean meal-induced enteritis (SBMIE) is a well-described condition in the distal intestine (DI) of several cultured fish species, but the exact cause is still unclear. The work on Atlantic salmon and zebrafish suggested soya-saponins, as heat-stable anti-nutritional factors in soybean meal, are the major causal agents. However, this conclusion was not supported by the research on some other fish, such as gilthead sea bream and European sea bass. Our previous work proved that soybean could induce SBMIE on turbot and the present work aimed to investigate whether soya-saponins alone could cause SBMIE and the effects of soya-saponins on the intestinal barrier function in juvenile turbot. Turbots with initial weight 11.4 ± 0.02 g were fed one of four fishmeal-based diets containing graded levels of soya-saponins (0, 2.5, 7.5, 15 g kg-1) for 8 weeks. At the end of the trial, all fish were weighed and plasma was obtained for diamine oxidase (DAO) activity and d-lactate level analysis and DI was sampled for histological evaluation and quantification of antioxidant parameters and inflammatory marker genes. The activities of superoxide dismutase, catalase, glutathione peroxidase, glutathione reductase and intestinal glutathione level were selected to evaluated intestinal antioxidant system. The distal intestinal epithelial cell (IEC) proliferation and apoptosis were investigated by proliferating cell nuclear antigen (PCNA) labelling and TdT-mediated dUTP nick end labeling (TUNEL), respectively. The results showed that soya-saponins caused significantly dose-dependent decrease in the growth performance and nutrient utilization (p < 0.05). Enteritis developed in DI of the fish fed diet containing soya-saponins. Significantly dose-dependent increases in severity of the inflammation concomitant with up-regulated expression of il-1β, il-8, and tnf-α, increased IEC proliferation and apoptosis, and decreases in selected antioxidant parameters were detected (p < 0.05). The epithelial permeability (evaluated by the plasma DAO activity and d-lactate level) was significantly increased with the increasing of dietary level of soya-saponins (p < 0.05), which was concomitant with the destroyed the intracellular junctions. In conclusion, the present work proved that soya-saponins induced enteritis and compromised the intestinal barrier functions. Based on the present work, strategies focus on regulation of cell apoptosis, epithelial permeability, intracellular junctions and redox homeostasis worth further investigating to develop new and efficient ways for SBMIE alleviation.
Collapse
Affiliation(s)
- Min Gu
- Marine College, Shandong University at Weihai, 180 Wenhua West Road, Weihai, 264209, PR China
| | - Qian Jia
- Marine College, Shandong University at Weihai, 180 Wenhua West Road, Weihai, 264209, PR China
| | - Zhiyu Zhang
- Marine College, Shandong University at Weihai, 180 Wenhua West Road, Weihai, 264209, PR China
| | - Nan Bai
- Marine College, Shandong University at Weihai, 180 Wenhua West Road, Weihai, 264209, PR China.
| | - Xiaojie Xu
- Marine College, Shandong University at Weihai, 180 Wenhua West Road, Weihai, 264209, PR China
| | - Bingying Xu
- Marine College, Shandong University at Weihai, 180 Wenhua West Road, Weihai, 264209, PR China
| |
Collapse
|
35
|
Rios-Arce ND, Collins FL, Schepper JD, Steury MD, Raehtz S, Mallin H, Schoenherr DT, Parameswaran N, McCabe LR. Epithelial Barrier Function in Gut-Bone Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1033:151-183. [PMID: 29101655 DOI: 10.1007/978-3-319-66653-2_8] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The intestinal epithelial barrier plays an essential role in maintaining host homeostasis. The barrier regulates nutrient absorption as well as prevents the invasion of pathogenic bacteria in the host. It is composed of epithelial cells, tight junctions, and a mucus layer. Several factors, such as cytokines, diet, and diseases, can affect this barrier. These factors have been shown to increase intestinal permeability, inflammation, and translocation of pathogenic bacteria. In addition, dysregulation of the epithelial barrier can result in inflammatory diseases such as inflammatory bowel disease. Our lab and others have also shown that barrier disruption can have systemic effects including bone loss. In this chapter, we will discuss the current literature to understand the link between intestinal barrier and bone. We will discuss how inflammation, aging, dysbiosis, and metabolic diseases can affect intestinal barrier-bone link. In addition, we will highlight the current suggested mechanism between intestinal barrier and bone.
Collapse
Affiliation(s)
- Naiomy Deliz Rios-Arce
- Comparative Medicine and Integrative Biology Program, East Lansing, MI, USA.,Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Fraser L Collins
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | | | - Michael D Steury
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Sandi Raehtz
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Heather Mallin
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Danny T Schoenherr
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Narayanan Parameswaran
- Comparative Medicine and Integrative Biology Program, East Lansing, MI, USA. .,Department of Physiology, Michigan State University, East Lansing, MI, USA.
| | - Laura R McCabe
- Department of Physiology and Department of Radiology, Biomedical Imaging Research Centre, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
36
|
Schneeberger K, Roth S, Nieuwenhuis EES, Middendorp S. Intestinal epithelial cell polarity defects in disease: lessons from microvillus inclusion disease. Dis Model Mech 2018; 11:11/2/dmm031088. [PMID: 29590640 PMCID: PMC5894939 DOI: 10.1242/dmm.031088] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The intestinal epithelium is a highly organized tissue. The establishment of epithelial cell polarity, with distinct apical and basolateral plasma membrane domains, is pivotal for both barrier formation and for the uptake and vectorial transport of nutrients. The establishment of cell polarity requires a specialized subcellular machinery to transport and recycle proteins to their appropriate location. In order to understand and treat polarity-associated diseases, it is necessary to understand epithelial cell-specific trafficking mechanisms. In this Review, we focus on cell polarity in the adult mammalian intestine. We discuss how intestinal epithelial polarity is established and maintained, and how disturbances in the trafficking machinery can lead to a polarity-associated disorder, microvillus inclusion disease (MVID). Furthermore, we discuss the recent developments in studying MVID, including the creation of genetically manipulated cell lines, mouse models and intestinal organoids, and their uses in basic and applied research. Summary: Microvillus inclusion disease serves as a useful model to enhance our understanding of the intestinal trafficking and polarity machinery in health and disease.
Collapse
Affiliation(s)
- Kerstin Schneeberger
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands
| | - Sabrina Roth
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands
| | - Edward E S Nieuwenhuis
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands
| | - Sabine Middendorp
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands .,Regenerative Medicine Center Utrecht, University Medical Centre (UMC) Utrecht, 3584 CT, Utrecht, The Netherlands
| |
Collapse
|
37
|
Jiang M, Taghizadeh F, Steyger PS. Potential Mechanisms Underlying Inflammation-Enhanced Aminoglycoside-Induced Cochleotoxicity. Front Cell Neurosci 2017; 11:362. [PMID: 29209174 PMCID: PMC5702304 DOI: 10.3389/fncel.2017.00362] [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: 09/20/2017] [Accepted: 11/03/2017] [Indexed: 12/20/2022] Open
Abstract
Aminoglycoside antibiotics remain widely used for urgent clinical treatment of life-threatening infections, despite the well-recognized risk of permanent hearing loss, i.e., cochleotoxicity. Recent studies show that aminoglycoside-induced cochleotoxicity is exacerbated by bacteriogenic-induced inflammation. This implies that those with severe bacterial infections (that induce systemic inflammation), and are treated with bactericidal aminoglycosides are at greater risk of drug-induced hearing loss than previously recognized. Incorporating this novel comorbid factor into cochleotoxicity risk prediction models will better predict which individuals are more predisposed to drug-induced hearing loss. Here, we review the cellular and/or signaling mechanisms by which host-mediated inflammatory responses to infection could enhance the trafficking of systemically administered aminoglycosides into the cochlea to enhance the degree of cochleotoxicity over that in healthy preclinical models. Once verified, these mechanisms will be potential targets for novel pharmacotherapeutics that reduce the risk of drug-induced hearing loss (and acute kidney damage) without compromising the life-saving bactericidal efficacy of aminoglycosides.
Collapse
Affiliation(s)
- Meiyan Jiang
- Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Farshid Taghizadeh
- Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Peter S Steyger
- Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR, United States.,National Center for Rehabilitative Auditory Research, VA Portland Health Care System, Portland, OR, United States
| |
Collapse
|
38
|
Cai J, Culley MK, Zhao Y, Zhao J. The role of ubiquitination and deubiquitination in the regulation of cell junctions. Protein Cell 2017; 9:754-769. [PMID: 29080116 PMCID: PMC6107491 DOI: 10.1007/s13238-017-0486-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Accepted: 10/09/2017] [Indexed: 12/11/2022] Open
Abstract
Maintenance of cell junctions plays a crucial role in the regulation of cellular functions including cell proliferation, permeability, and cell death. Disruption of cell junctions is implicated in a variety of human disorders, such as inflammatory diseases and cancers. Understanding molecular regulation of cell junctions is important for development of therapeutic strategies for intervention of human diseases. Ubiquitination is an important type of post-translational modification that primarily regulates endogenous protein stability, receptor internalization, enzyme activity, and protein-protein interactions. Ubiquitination is tightly regulated by ubiquitin E3 ligases and can be reversed by deubiquitinating enzymes. Recent studies have been focusing on investigating the effect of protein stability in the regulation of cell-cell junctions. Ubiquitination and degradation of cadherins, claudins, and their interacting proteins are implicated in epithelial and endothelial barrier disruption. Recent studies have revealed that ubiquitination is involved in regulation of Rho GTPases’ biological activities. Taken together these studies, ubiquitination plays a critical role in modulating cell junctions and motility. In this review, we will discuss the effects of ubiquitination and deubiquitination on protein stability and expression of key proteins in the cell-cell junctions, including junction proteins, their interacting proteins, and small Rho GTPases. We provide an overview of protein stability in modulation of epithelial and endothelial barrier integrity and introduce potential future search directions to better understand the effects of ubiquitination on human disorders caused by dysfunction of cell junctions.
Collapse
Affiliation(s)
- Junting Cai
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Asthma, and Critical Care Medicine, Department of Medicine, The University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Miranda K Culley
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Asthma, and Critical Care Medicine, Department of Medicine, The University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Yutong Zhao
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Asthma, and Critical Care Medicine, Department of Medicine, The University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jing Zhao
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Asthma, and Critical Care Medicine, Department of Medicine, The University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
39
|
Yang T, Mei H, Xu D, Zhou W, Zhu X, Sun L, Huang X, Wang X, Shu T, Liu J, Ding J, Hassan HM, Zhang L, Jiang Z. Early indications of ANIT-induced cholestatic liver injury: Alteration of hepatocyte polarization and bile acid homeostasis. Food Chem Toxicol 2017; 110:1-12. [PMID: 28986171 DOI: 10.1016/j.fct.2017.09.051] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 09/04/2017] [Accepted: 09/29/2017] [Indexed: 02/08/2023]
Abstract
Hepatocyte polarization is essential for biliary secretion, and loss of polarity causes bile secretory failure and hepatotoxicity. Here, we showed that alpha-naphthyl isothiocyanate (ANIT)-induced liver injury was accompanied by the dynamic interruption of bile acid homeostasis in rat plasma, liver and bile, which was characterized by the redistribution of bile acids in plasma and bile and a small range of fluctuations in the liver. Molecular mechanism studies indicated that these factors are dynamically mediated by the disruption of bile acid transporters and hepatic tight junctions. Dynamic changes in tight junction (TJ) permeability were observed by hepatobiliary barrier function assessment. Hepatocyte polarization was disrupted by ANIT before the development of cholestatic hepatotoxicity and alteration of bile acid metabolic profiles, which were assayed by high-performance liquid chromatography-tandem mass spectrometry, further verifying TJ deficiency. S1PR1 activation with SEW2871 reduced ANIT-induced liver injury by reducing the total serum bile acid concentration, liver functional enzyme activity and inflammation. Our data suggest that hepatocyte polarization plays an important role in maintaining bile acid homeostasis before the development of cholestatic hepatotoxicity and that TJs were more prominent in the early stage of cholestasis. S1PR1 may be a potential target for the prevention of drug-induced cholestatic liver injury.
Collapse
Affiliation(s)
- Tingting Yang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Huifang Mei
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Dengqiu Xu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Wang Zhou
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoyu Zhu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Lixin Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Xin Huang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Xue Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Ting Shu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Jia Liu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Jiaxin Ding
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - H M Hassan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Screening and Pharmacodynamics Evaluation, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing 210009, China.
| |
Collapse
|
40
|
Miyazaki K, Hashimoto K, Sato M, Watanabe M, Tomikawa N, Kanno S, Kawasaki Y, Momoi N, Hosoya M. Establishment of a method for evaluating endothelial cell injury by TNF-α in vitro for clarifying the pathophysiology of virus-associated acute encephalopathy. Pediatr Res 2017; 81:942-947. [PMID: 28157839 DOI: 10.1038/pr.2017.28] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 12/15/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Virus-associated acute encephalopathy (VAE) is a severe central nervous system complication caused by common viral infections in children. The pathophysiology of VAE is thought to be endothelial injury. This study was designed to establish an in vitro VAE model for evaluating endothelial injury caused by the proinflammatory cytokine TNF-α. METHODS Transwell-grown human umbilical vein endothelial cells (HUVECs) monolayers were incubated with serially diluted TNF-α. Transendothelial electrical resistance (TER) was measured using impedance spectroscopy. Permeability changes of HUVECs after TNF-α treatment were determined by fluorescein isothiocyanate (FITC)-conjugated dextran. Moreover, TNF-α-induced morphological changes in claudin-5 and apoptosis were observed by immunofluorescent staining. RESULTS The decrease in TER, time of TER recovery to baseline, and increase in permeability were all dependent on TNF-α concentration. Immunofluorescent staining showed that claudin-5 was delocalized after TNF-α treatment in a dose-dependent manner. In addition, some apoptotic cells were observed at high TNF-α concentrations. CONCLUSION TER measurement combined with a permeability assay could be useful for evaluating vascular endothelial cell permeability in an in vitro model. These evaluation methods will contribute to both the development of specific treatments focusing on vascular permeability, and the search for a novel therapeutic strategy in VAE treatment.
Collapse
Affiliation(s)
- Kyohei Miyazaki
- Department of Pediatrics, Fukushima Medical University, Fukushima, Japan
| | - Koichi Hashimoto
- Department of Pediatrics, Fukushima Medical University, Fukushima, Japan
| | - Masatoki Sato
- Department of Pediatrics, Fukushima Medical University, Fukushima, Japan
| | - Masahiro Watanabe
- Department of Pediatrics, Fukushima Medical University, Fukushima, Japan
| | - Naoki Tomikawa
- Department of Basic Pathology, Fukushima Medical University, Fukushima, Japan
| | - Shuto Kanno
- Department of Pediatrics, Fukushima Medical University, Fukushima, Japan
| | - Yukihiko Kawasaki
- Department of Pediatrics, Fukushima Medical University, Fukushima, Japan
| | - Nobuo Momoi
- Department of Pediatrics, Fukushima Medical University, Fukushima, Japan
| | - Mitsuaki Hosoya
- Department of Pediatrics, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
41
|
Buzza MS, Johnson TA, Conway GD, Martin EW, Mukhopadhyay S, Shea-Donohue T, Antalis TM. Inflammatory cytokines down-regulate the barrier-protective prostasin-matriptase proteolytic cascade early in experimental colitis. J Biol Chem 2017; 292:10801-10812. [PMID: 28490634 DOI: 10.1074/jbc.m116.771469] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 05/02/2017] [Indexed: 12/17/2022] Open
Abstract
Compromised gastrointestinal barrier function is strongly associated with the progressive and destructive pathologies of the two main forms of irritable bowel disease (IBD), ulcerative colitis (UC), and Crohn's disease (CD). Matriptase is a membrane-anchored serine protease encoded by suppression of tumorigenicity-14 (ST14) gene, which is critical for epithelial barrier development and homeostasis. Matriptase barrier-protective activity is linked with the glycosylphosphatidylinositol (GPI)-anchored serine protease prostasin, which is a co-factor for matriptase zymogen activation. Here we show that mRNA and protein expression of both matriptase and prostasin are rapidly down-regulated in the initiating inflammatory phases of dextran sulfate sodium (DSS)-induced experimental colitis in mice, and, significantly, the loss of these proteases precedes the appearance of clinical symptoms, suggesting their loss may contribute to disease susceptibility. We used heterozygous St14 hypomorphic mice expressing a promoter-linked β-gal reporter to show that inflammatory colitis suppresses the activity of the St14 gene promoter. Studies in colonic T84 cell monolayers revealed that barrier disruption by the colitis-associated Th2-type cytokines, IL-4 and IL-13, down-regulates matriptase as well as prostasin through phosphorylation of the transcriptional regulator STAT6 and that inhibition of STAT6 with suberoylanilide hydroxamic acid (SAHA) restores protease expression and reverses cytokine-induced barrier dysfunction. Both matriptase and prostasin are significantly down-regulated in colonic tissues from human subjects with active ulcerative colitis or Crohn's disease, implicating the loss of this barrier-protective protease pathway in the pathogenesis of irritable bowel disease.
Collapse
Affiliation(s)
- Marguerite S Buzza
- From the Center for Vascular and Inflammatory Diseases and Department of Physiology and
| | - Tierra A Johnson
- From the Center for Vascular and Inflammatory Diseases and Department of Physiology and
| | - Gregory D Conway
- From the Center for Vascular and Inflammatory Diseases and Department of Physiology and
| | - Erik W Martin
- From the Center for Vascular and Inflammatory Diseases and Department of Physiology and
| | | | - Terez Shea-Donohue
- the Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Toni M Antalis
- From the Center for Vascular and Inflammatory Diseases and Department of Physiology and
| |
Collapse
|
42
|
Garcia-Hernandez V, Quiros M, Nusrat A. Intestinal epithelial claudins: expression and regulation in homeostasis and inflammation. Ann N Y Acad Sci 2017; 1397:66-79. [PMID: 28493289 DOI: 10.1111/nyas.13360] [Citation(s) in RCA: 273] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/17/2017] [Accepted: 03/23/2017] [Indexed: 12/31/2022]
Abstract
The intestinal epithelium forms a highly dynamic and selective barrier that controls absorption of fluid and solutes while restricting pathogen access to underlying tissues. Barrier properties are achieved by intercellular junctions that include an apical tight junction (TJ) and subjacent adherens junctions and desmosomes. The TJ tetraspan claudin proteins form pores between epithelial cells to control paracellular fluid and ion movement. In addition to regulation of barrier function, claudin family members control epithelial homeostasis and are expressed in a spatiotemporal manner in the intestinal crypt-luminal axis. This delicate balance of physiologic differential claudin protein expression is altered during mucosal inflammation. Inflammatory mediators influence transcriptional regulation, as well as endocytic trafficking, targeting, and retention of claudins in the TJ. Increased expression of intestinal epithelial claudin-1, -2, and -18 with downregulation of claudin-3, -4, -5, -7, -8, and -12 has been observed in intestinal inflammatory disorders. Such changes in claudin proteins modify the epithelial barrier function in addition to influencing epithelial and mucosal homeostasis. An improved understanding of the regulatory mechanisms that control epithelial claudin proteins will provide strategies to strengthen the epithelial barrier function and restore mucosal homeostasis in inflammatory disorders.
Collapse
Affiliation(s)
| | - Miguel Quiros
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
43
|
Stamatovic SM, Johnson AM, Sladojevic N, Keep RF, Andjelkovic AV. Endocytosis of tight junction proteins and the regulation of degradation and recycling. Ann N Y Acad Sci 2017; 1397:54-65. [PMID: 28415156 DOI: 10.1111/nyas.13346] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/08/2017] [Accepted: 03/09/2017] [Indexed: 12/31/2022]
Abstract
Internalization of tight junction (TJ) proteins from the plasma membrane is a pivotal mechanism regulating TJ plasticity and function in both epithelial and endothelial barrier tissues. Once internalized, the TJ proteins enter complex vesicular machinery, where further trafficking is directly dependent on the initiating stimulus and downstream signaling pathways that regulate the sorting and destiny of TJ proteins, as well as on cell and barrier responses. The destiny of internalized TJ proteins is recycling to the plasma membrane or sorting to late endosomes and degradation. This review highlights recent advances in our knowledge of endocytosis and vesicular trafficking of TJ proteins in both epithelial and endothelial cells. A greater understanding of these processes may allow for the development of methods to modulate barrier permeability for drug delivery or prevent barrier dysfunction in disease states.
Collapse
Affiliation(s)
| | | | | | - Richard F Keep
- Neurosurgery.,Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| | | |
Collapse
|
44
|
Salvo Romero E, Alonso Cotoner C, Pardo Camacho C, Casado Bedmar M, Vicario M. The intestinal barrier function and its involvement in digestive disease. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2017; 107:686-96. [PMID: 26541659 DOI: 10.17235/reed.2015.3846/2015] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The gastrointestinal mucosal surface is lined with epithelial cells representing an effective barrier made up with intercellular junctions that separate the inner and the outer environments, and block the passage of potentially harmful substances. However, epithelial cells are also responsible for the absorption of nutrients and electrolytes, hence a semipermeable barrier is required that selectively allows a number of substances in while keeping others out. To this end, the intestine developed the "intestinal barrier function", a defensive system involving various elements, both intra- and extracellular, that work in a coordinated way to impede the passage of antigens, toxins, and microbial byproducts, and simultaneously preserves the correct development of the epithelial barrier, the immune system, and the acquisition of tolerance against dietary antigens and the intestinal microbiota. Disturbances in the mechanisms of the barrier function favor the development of exaggerated immune responses; while exact implications remain unknown, changes in intestinal barrier function have been associated with the development of inflammatory conditions in the gastrointestinal tract. This review details de various elements of the intestinal barrier function, and the key molecular and cellular changes described for gastrointestinal diseases associated with dysfunction in this defensive mechanism.
Collapse
Affiliation(s)
| | | | | | | | - María Vicario
- Gastroenteroogia, Vall d'Hebron Institut de REcerca, España
| |
Collapse
|
45
|
Luissint AC, Parkos CA, Nusrat A. Inflammation and the Intestinal Barrier: Leukocyte-Epithelial Cell Interactions, Cell Junction Remodeling, and Mucosal Repair. Gastroenterology 2016; 151:616-32. [PMID: 27436072 PMCID: PMC5317033 DOI: 10.1053/j.gastro.2016.07.008] [Citation(s) in RCA: 354] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/13/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023]
Abstract
The intestinal tract is lined by a single layer of columnar epithelial cells that forms a dynamic, permeable barrier allowing for selective absorption of nutrients, while restricting access to pathogens and food-borne antigens. Precise regulation of epithelial barrier function is therefore required for maintaining mucosal homeostasis and depends, in part, on barrier-forming elements within the epithelium and a balance between pro- and anti-inflammatory factors in the mucosa. Pathologic states, such as inflammatory bowel disease, are associated with a leaky epithelial barrier, resulting in excessive exposure to microbial antigens, recruitment of leukocytes, release of soluble mediators, and ultimately mucosal damage. An inflammatory microenvironment affects epithelial barrier properties and mucosal homeostasis by altering the structure and function of epithelial intercellular junctions through direct and indirect mechanisms. We review our current understanding of complex interactions between the intestinal epithelium and immune cells, with a focus on pathologic mucosal inflammation and mechanisms of epithelial repair. We discuss leukocyte-epithelial interactions, as well as inflammatory mediators that affect the epithelial barrier and mucosal repair. Increased knowledge of communication networks between the epithelium and immune system will lead to tissue-specific strategies for treating pathologic intestinal inflammation.
Collapse
Affiliation(s)
- Anny-Claude Luissint
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Asma Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan.
| |
Collapse
|
46
|
García-Ponce A, Chánez Paredes S, Castro Ochoa KF, Schnoor M. Regulation of endothelial and epithelial barrier functions by peptide hormones of the adrenomedullin family. Tissue Barriers 2016; 4:e1228439. [PMID: 28123925 DOI: 10.1080/21688370.2016.1228439] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 08/15/2016] [Accepted: 08/17/2016] [Indexed: 01/16/2023] Open
Abstract
The correct regulation of tissue barriers is of utmost importance for health. Barrier dysfunction accompanies inflammatory disorders and, if not controlled properly, can contribute to the development of chronic diseases. Tissue barriers are formed by monolayers of epithelial cells that separate organs from their environment, and endothelial cells that cover the vasculature, thus separating the blood stream from underlying tissues. Cells within the monolayers are connected by intercellular junctions that are linked by adaptor molecules to the cytoskeleton, and the regulation of these interactions is critical for the maintenance of tissue barriers. Many endogenous and exogenous molecules are known to regulate barrier functions in both ways. Proinflammatory cytokines weaken the barrier, whereas anti-inflammatory mediators stabilize barriers. Adrenomedullin (ADM) and intermedin (IMD) are endogenous peptide hormones of the same family that are produced and secreted by many cell types during physiologic and pathologic conditions. They activate certain G-protein-coupled receptor complexes to regulate many cellular processes such as cytokine production, actin dynamics and junction stability. In this review, we summarize current knowledge about the barrier-stabilizing effects of ADM and IMD in health and disease.
Collapse
Affiliation(s)
- Alexander García-Ponce
- Department of Molecular Biomedicine, Center for Investigation and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN) , Mexico City, Mexico
| | - Sandra Chánez Paredes
- Department of Molecular Biomedicine, Center for Investigation and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN) , Mexico City, Mexico
| | - Karla Fabiola Castro Ochoa
- Department of Molecular Biomedicine, Center for Investigation and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN) , Mexico City, Mexico
| | - Michael Schnoor
- Department of Molecular Biomedicine, Center for Investigation and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN) , Mexico City, Mexico
| |
Collapse
|
47
|
Hegan PS, Chandhoke SK, Barone C, Egan M, Bähler M, Mooseker MS. Mice lacking myosin IXb, an inflammatory bowel disease susceptibility gene, have impaired intestinal barrier function and superficial ulceration in the ileum. Cytoskeleton (Hoboken) 2016; 73:163-79. [PMID: 26972322 DOI: 10.1002/cm.21292] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 03/09/2016] [Accepted: 03/10/2016] [Indexed: 12/16/2022]
Abstract
Genetic studies have implicated MYO9B, which encodes myosin IXb (Myo9b), a motor protein with a Rho GTPase activating domain (RhoGAP), as a susceptibility gene for inflammatory bowel disease (IBD). Moreover, we have recently shown that knockdown of Myo9b in an intestinal epithelial cell line impairs wound healing and barrier function. Here, we investigated whether mice lacking Myo9b have impaired intestinal barrier function and features of IBD. Myo9b knock out (KO) mice exhibit impaired weight gain and fecal occult blood (indicator of gastrointestinal bleeding), and increased intestinal epithelial cell apoptosis could be detected along the entire intestinal axis. Histologic analysis revealed intestinal mucosal damage, most consistently observed in the ileum, which included superficial ulceration and neutrophil infiltration. Focal lesions contained neutrophils and ultrastructural examination confirmed epithelial discontinuity and the deposition of extracellular matrix. We also observed impaired mucosal barrier function in KO mice. Transepithelial electrical resistance of KO ileum is >3 fold less than WT ileum. The intestinal mucosa is also permeable to high molecular weight dextran, presumably due to the presence of mucosal surface ulcerations. There is loss of tight junction-associated ZO-1, decreased lateral membrane associated E-cadherin, and loss of terminal web associated cytokeratin filaments. Consistent with increased Rho activity in the KO, there is increased subapical expression of activated myosin II (Myo2) based on localization of phosphorylated Myo2 regulatory light chain. Except for a delay in disease onset in the KO, no difference in dextran sulfate sodium-induced colitis and lethality was observed between wild-type and Myo9b KO mice.
Collapse
Affiliation(s)
- Peter S Hegan
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut
| | - Surjit K Chandhoke
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut
| | - Christina Barone
- Department of Respiratory Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Marie Egan
- Department of Respiratory Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Martin Bähler
- Institute of Molecular Cell Biology, Westfalian Wilhelms University, Münster, Germany
| | - Mark S Mooseker
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut.,Departments of Cell Biology and Pathology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
48
|
Zhang JM, Zhu YB, Li HG, Luan SM, Song CY, Deng X, Chen YX. Protection of Sophocarpine on Colonic Barrier in DSS-induced Acute Colitis in Mice by Increasing Expression of HNF4α. CHINESE HERBAL MEDICINES 2015. [DOI: 10.1016/s1674-6384(15)60048-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
49
|
Abstract
Intestinal mucosal barrier function is the capacity of the intestine to provide adequate containment of luminal microorganisms and molecules while preserving the ability to absorb nutrients. The central element is the epithelial layer, which physically separates the lumen and the internal milieu and is in charge of vectorial transport of ions, nutrients, and other substances. The secretion of mucus-forming mucins, sIgA, and antimicrobial peptides reinforces the mucosal barrier on the extraepithelial side, while a variety of immune cells contributes to mucosal defense in the inner side. Thus, the mucosal barrier is of physical, biochemical, and immune nature. In addition, the microbiota may be viewed as part of this system because of the mutual influence occurring between the host and the luminal microorganisms. Alteration of the mucosal barrier function with accompanying increased permeability and/or bacterial translocation has been linked with a variety of conditions, including inflammatory bowel disease. Genetic and environmental factors may converge to evoke a defective function of the barrier, which in turn may lead to overt inflammation of the intestine as a result of an exacerbated immune reaction toward the microbiota. According to this hypothesis, inflammatory bowel disease may be both precipitated and treated by either stimulation or downregulation of the different elements of the mucosal barrier, with the outcome depending on timing, the cell type affected, and other factors. In this review, we cover briefly the elements of the barrier and their involvement in functional defects and the resulting phenotype.
Collapse
|
50
|
Wang H, Li X, Wang C, Zhu D, Xu Y. Abnormal ultrastructure of intestinal epithelial barrier in mice with alcoholic steatohepatitis. Alcohol 2014; 48:787-93. [PMID: 25454538 DOI: 10.1016/j.alcohol.2014.08.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 08/08/2014] [Accepted: 08/26/2014] [Indexed: 12/12/2022]
Abstract
Intestinal barrier dysfunction caused by chronic alcohol consumption is closely associated with disruption of the intestinal epithelial apical junction complex. The present study was undertaken to directly display by transmission electron microscopy the abnormal ultrastructure of the intestinal epithelial barrier in mice with alcoholic steatohepatitis. The results showed that chronic alcohol consumption could induce obvious liver injury, with diffuse lipid accumulation and focal inflammatory cell infiltration in the liver, assessed by hematoxylin and eosin staining. The indicators of intestinal barrier dysfunction, d-lactic acid and lipopolysaccharide, were significantly higher in alcohol-fed mice than in control mice. Alcohol exposure obviously caused high permeability in the ileum to fluorescein isothiocyanate-dextran (FD-4; molecular weight 4000). Transmission electron microscopy demonstrated that tight junctions and adherens junctions expanded noticeably in alcohol-fed mice. Although the tight junction (TJ) length of alcohol-fed mice had no significant difference compared with control mice, the adherens junction (AJ) length of alcohol-fed mice significantly decreased compared with control mice. Additionally, the ratios of both TJmax/TJmin and AJmax/AJmin were significantly larger in alcohol-fed mice than in control liquid-fed mice. In conclusion, high intestinal permeability caused by alcohol attributes to the irregular ultrastructure of the intestinal epithelial barrier.
Collapse
Affiliation(s)
- Hongyan Wang
- Department of Gastroenterology, Beijing Tian Tan Hospital, Capital Medical University, 6 Xili, Tiantan, Dongcheng District, Beijing 100050 PR China
| | - Xin Li
- Department of Gastroenterology, Beijing Tian Tan Hospital, Capital Medical University, 6 Xili, Tiantan, Dongcheng District, Beijing 100050 PR China
| | - Chen Wang
- Department of Gastroenterology, Beijing Tian Tan Hospital, Capital Medical University, 6 Xili, Tiantan, Dongcheng District, Beijing 100050 PR China
| | - Dong Zhu
- Department of Gastroenterology, Beijing Tian Tan Hospital, Capital Medical University, 6 Xili, Tiantan, Dongcheng District, Beijing 100050 PR China.
| | - Youqing Xu
- Department of Gastroenterology, Beijing Tian Tan Hospital, Capital Medical University, 6 Xili, Tiantan, Dongcheng District, Beijing 100050 PR China.
| |
Collapse
|