1
|
Dugbartey GJ, Sener A. Organ Toxicity by Immunosuppressive Drugs in Solid Organ Transplantation. RECENT ADVANCES IN THERAPEUTIC DRUG MONITORING AND CLINICAL TOXICOLOGY 2022:255-271. [DOI: 10.1007/978-3-031-12398-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
2
|
Han Y, Wu L, Ling Q, Wu P, Zhang C, Jia L, Weng H, Wang B. Intestinal Dysbiosis Correlates With Sirolimus-induced Metabolic Disorders in Mice. Transplantation 2021; 105:1017-1029. [PMID: 33116044 DOI: 10.1097/tp.0000000000003494] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Long-time use of pharmacological immunosuppressive agents frequently leads to metabolic disorders. Most studies have focused on islet toxicity leading to posttransplantation diabetes mellitus. In contrast, the link between intestinal dysbiosis and immunosuppressive drug-induced metabolic disorders remains unclear. METHODS We established a mouse model of metabolic abnormality via sirolimus treatment. Fecal microbiota was examined using 16S rRNA gene MiSeq sequencing. Intestinal barrier function was assessed using fluorescein isothiocyanate-dextran assay and mucus immunostaining. Systemic inflammation was determined using a multiplexed fluorescent bead-based immunoassay. RESULTS Sirolimus induced dyslipidemia and glucose intolerance in mice in a dose-dependent manner. Interestingly, the clinical-mimicking dose of sirolimus altered the intestinal microbiota community, which was characterized by the enrichment of Proteobacteria, depletion of Akkermansia, and potential function shifts to those involved in lipid metabolism and the immune system. In addition, the clinical-mimicking dose of sirolimus reduced the thickness of the intestinal mucosal layer, increased the intestinal permeability, and enriched the circulating pro-inflammatory factors, including interleukin (IL)-12, IL-6, monocyte chemotactic protein 1, granulocyte-macrophage colony stimulating factor, and IL-1β. Our results showed a close association between intestinal dysbiosis, intestinal barrier failure, systemic inflammation, and metabolic disorders. Furthermore, we demonstrated that oral intervention in the gut microbiota by Lactobacillus rhamnosus HN001 protected against intestinal dysbiosis, especially by depleting the lipopolysaccharide-producing Proteobacteria, and attenuated the sirolimus-induced systemic inflammation, dyslipidemia, and insulin resistance. CONCLUSIONS Our study demonstrated a potentially causative role of intestinal dysbiosis in sirolimus-induced metabolic disorders, which will provide a novel therapeutic target for transplant recipients.
Collapse
Affiliation(s)
- Yuqiu Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Ling
- Department of Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, Zhejiang, China
| | - Pin Wu
- Division of Throat Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chenzhi Zhang
- Department of Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, Zhejiang, China
| | - Longfei Jia
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Honglei Weng
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Baohong Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Hangzhou, China
| |
Collapse
|
3
|
Carron C, Pais de Barros JP, Gaiffe E, Deckert V, Adda-Rezig H, Roubiou C, Laheurte C, Masson D, Simula-Faivre D, Louvat P, Moulin B, Frimat L, Rieu P, Mousson C, Durrbach A, Heng AE, Saas P, Ducloux D, Lagrost L, Bamoulid J. End-Stage Renal Disease-Associated Gut Bacterial Translocation: Evolution and Impact on Chronic Inflammation and Acute Rejection After Renal Transplantation. Front Immunol 2019; 10:1630. [PMID: 31474974 PMCID: PMC6706794 DOI: 10.3389/fimmu.2019.01630] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/01/2019] [Indexed: 01/04/2023] Open
Abstract
Chronic inflammation in end-stage renal disease (ESRD) is partly attributed to gut bacterial translocation (GBT) due to loss of intestinal epithelium integrity. Increased levels of circulating lipopolysaccharide (LPS) –a surrogate marker of GBT– contribute to maintain a chronic inflammatory state. However, circulating LPS can be neutralized by lipoproteins and transported to the liver for elimination. While ESRD-associated GBT has been widely described, less is known about its changes and impact on clinical outcome after kidney transplantation (KT). One hundred and forty-six renal transplant recipients with serum samples obtained immediately before and 1 year after transplantation (1-Year post KT) were included. Intestinal epithelium integrity (iFABP), total LPS (by measuring 3-hydroxymyristate), LPS activity (biologically active LPS measured by the LAL assay), inflammatory biomarkers (sCD14 and cytokines), lipoproteins and LPS-binding proteins (LBP and phospholipid transfer protein [PLTP] activity) were simultaneously measured. At 1-Year post KT, iFABP decreased but remained higher than in normal volunteers. Total LPS concentration remained stable while LPS activity decreased. Inflammation biomarkers decreased 1-Year post KT. We concomitantly observed an increase in lipoproteins. Higher sCD14 levels before transplantation was associated with lower incidence of acute rejection. Although GBT remained stable after KT, the contemporary increase in lipoproteins could bind circulating LPS and contribute concomitantly to neutralization of LPS activity, as well as improvement in ESRD-associated chronic inflammation. Chronic exposure to LPS in ESRD could promote endotoxin tolerance and explain why patients with higher pre-transplant sCD14 are less prompt to develop acute rejection after transplantation.
Collapse
Affiliation(s)
- Clémence Carron
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Besançon, France
| | | | - Emilie Gaiffe
- FHU INCREASE, Besançon, France.,INSERM CIC-1431, LabEx LipSTIC, Clinical Investigation Center in Biotherapy, University Hospital of Besançon, Fédération Hospitalo-Universitaire INCREASE, Besançon, France
| | - Valérie Deckert
- INSERM, LabEx LipSTIC, Univ. Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
| | - Hanane Adda-Rezig
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Besançon, France
| | - Caroline Roubiou
- Department of Nephrology, Dialysis, and Renal Transplantation, University Hospital of Besançon, Besançon, France
| | - Caroline Laheurte
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Besançon, France.,Plateforme de BioMonitoring, EFS Bourgogne Franche-Comté, Besançon, France
| | - David Masson
- INSERM, LabEx LipSTIC, Univ. Bourgogne Franche-Comté, LNC UMR1231, Dijon, France.,CHU Dijon, Biochimie et Service de la Recherche, Dijon, France
| | - Dominique Simula-Faivre
- Department of Nephrology, Dialysis, and Renal Transplantation, University Hospital of Besançon, Besançon, France
| | - Pascale Louvat
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Besançon, France.,Plateforme de BioMonitoring, EFS Bourgogne Franche-Comté, Besançon, France
| | - Bruno Moulin
- Department of Nephrology, CHU Strasbourg, Dialysis, and Renal Transplantation, Strasbourg, France
| | - Luc Frimat
- Department of Nephrology, CHU Nancy, Dialysis, and Renal Transplantation, Nancy, France
| | - Philippe Rieu
- Department of Nephrology, CHU Reims, Dialysis, and Renal Transplantation, Reims, France
| | - Christiane Mousson
- Department of Nephrology, CHU Dijon, Dialysis, and Renal Transplantation, Dijon, France
| | - Antoine Durrbach
- Department of Nephrology, CHU Kremlin-Bicêtre, Dialysis, and Renal Transplantation, Le Kremlin-Bicêtre, France
| | - Anne-Elisabeth Heng
- Department of Nephrology, CHU Clermont-Ferrand, Dialysis, and Renal Transplantation, Clermont-Ferrand, France
| | - Philippe Saas
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Besançon, France.,FHU INCREASE, Besançon, France.,INSERM CIC-1431, LabEx LipSTIC, Clinical Investigation Center in Biotherapy, University Hospital of Besançon, Fédération Hospitalo-Universitaire INCREASE, Besançon, France.,Plateforme de BioMonitoring, EFS Bourgogne Franche-Comté, Besançon, France
| | - Didier Ducloux
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Besançon, France.,FHU INCREASE, Besançon, France.,INSERM CIC-1431, LabEx LipSTIC, Clinical Investigation Center in Biotherapy, University Hospital of Besançon, Fédération Hospitalo-Universitaire INCREASE, Besançon, France.,Department of Nephrology, Dialysis, and Renal Transplantation, University Hospital of Besançon, Besançon, France
| | - Laurent Lagrost
- INSERM, LabEx LipSTIC, Univ. Bourgogne Franche-Comté, LNC UMR1231, Dijon, France.,CHU Dijon, Biochimie et Service de la Recherche, Dijon, France
| | - Jamal Bamoulid
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Besançon, France.,FHU INCREASE, Besançon, France.,Department of Nephrology, Dialysis, and Renal Transplantation, University Hospital of Besançon, Besançon, France
| |
Collapse
|
4
|
Jia J, Tian X, Jiang J, Ren Z, Lu H, He N, Xie H, Zhou L, Zheng S. Structural shifts in the intestinal microbiota of rats treated with cyclosporine A after orthotropic liver transplantation. Front Med 2019; 13:451-460. [PMID: 31020543 DOI: 10.1007/s11684-018-0675-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 10/27/2018] [Indexed: 02/07/2023]
Abstract
Understanding the effect of immunosuppressive agents on intestinal microbiota is important to reduce the mortality and morbidity from orthotopic liver transplantation (OLT). We investigated the relationship between the commonly used immunosuppressive agent cyclosporine A (CSA) and the intestinal microbial variation in an OLT model. The rat samples were divided as follows: (1) N group (normal control); (2) I group (isograft LT, Brown Norway [BN] rat to BN); (3) R group (allograft LT, Lewis to BN rat); and (4) CSA group (R group treated with CSA). The intestinal microbiota was assayed by denaturing gradient gel electrophoresis profiles and by using real-time polymerase chain reaction. The liver histopathology and the alanine/aspartate aminotransferase ratio after LT were both ameliorated by CSA. In the CSA group, the numbers of rDNA gene copies of Clostridium cluster I, Clostridium cluster XIV, and Enterobacteriaceae decreased, whereas those of Faecalibacterium prausnitzii increased compared with the R group. Cluster analysis indicated that the samples from the N, I, and CSA groups were clustered, whereas the other clusters contained the samples from the R group. Hence, CSA ameliorates hepatic graft injury and partially restores gut microbiota following LT, and these may benefit hepatic graft rejection.
Collapse
Affiliation(s)
- Junjun Jia
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xinyao Tian
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jianwen Jiang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhigang Ren
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Haifeng Lu
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, 310003, China.,State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Ning He
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Haiyang Xie
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shusen Zheng
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China. .,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, 310003, China. .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
5
|
Immunosuppressive drugs and the gastrointestinal tract in renal transplant patients. Transplant Rev (Orlando) 2018; 33:55-63. [PMID: 30473173 DOI: 10.1016/j.trre.2018.11.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/07/2018] [Accepted: 11/10/2018] [Indexed: 12/26/2022]
Abstract
Gastrointestinal (GI) discomfort is common after renal transplantation and can be caused by the use of various immunosuppressive drugs. GI symptoms affect the quality of life, lead to an impaired graft survival and an increased mortality. Moreover, diseases and disturbances of the GI tract also affect the pharmacokinetics of immunosuppressive drugs. This review addresses the interaction between immunosuppressive agents and GI disorders. The GI tract is involved in the metabolism of several immunosuppressive drugs. Calcineurin inhibitors, mTor inhibitors, and corticosteroids are subjected to metabolism by the intestinal cytochrome P450 (CYP3A) and by the drug efflux pump ABCB1. Mycophenolate is partly metabolized in the stomach and intestine and undergoes enterohepatic recirculation. Gastrointestinal disturbances can lead to a modified exposure to immunosuppressive drugs. In the first and second part of this review, we focus on the role of the GI tract in the pharmacokinetics of the immunosuppressive drugs and how to adjust immunosuppressive therapy in patients with vomiting, need for tube feeding, delayed gastric emptying, intestinal resection, and diarrhea. In the third part, we review the GI adverse effects of the various immunosuppressive drugs, with special attention for diarrhea and dyspepsia. Finally, we discuss the effects of drugs used for relief of GI complaints on the exposure to immunosuppressive agents.
Collapse
|
6
|
Van Skike CE, Jahrling JB, Olson AB, Sayre NL, Hussong SA, Ungvari Z, Lechleiter JD, Galvan V. Inhibition of mTOR protects the blood-brain barrier in models of Alzheimer's disease and vascular cognitive impairment. Am J Physiol Heart Circ Physiol 2018; 314:H693-H703. [PMID: 29351469 PMCID: PMC5966773 DOI: 10.1152/ajpheart.00570.2017] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/29/2017] [Accepted: 12/13/2017] [Indexed: 01/05/2023]
Abstract
An intact blood-brain barrier (BBB) limits entry of proinflammatory and neurotoxic blood-derived factors into the brain parenchyma. The BBB is damaged in Alzheimer's disease (AD), which contributes significantly to the progression of AD pathologies and cognitive decline. However, the mechanisms underlying BBB breakdown in AD remain elusive, and no interventions are available for treatment or prevention. We and others recently established that inhibition of the mammalian/mechanistic target of rapamycin (mTOR) pathway with rapamycin yields significant neuroprotective effects, improving cerebrovascular and cognitive function in mouse models of AD. To test whether mTOR inhibition protects the BBB in neurological diseases of aging, we treated hAPP(J20) mice modeling AD and low-density lipoprotein receptor-null (LDLR-/-) mice modeling vascular cognitive impairment with rapamycin. We found that inhibition of mTOR abrogates BBB breakdown in hAPP(J20) and LDLR-/- mice. Experiments using an in vitro BBB model indicated that mTOR attenuation preserves BBB integrity through upregulation of specific tight junction proteins and downregulation of matrix metalloproteinase-9 activity. Together, our data establish mTOR activity as a critical mediator of BBB breakdown in AD and, potentially, vascular cognitive impairment and suggest that rapamycin and/or rapalogs could be used for the restoration of BBB integrity. NEW & NOTEWORTHY This report establishes mammalian/mechanistic target of rapamycin as a critical mediator of blood-brain barrier breakdown in models of Alzheimer's disease and vascular cognitive impairment and suggests that drugs targeting the target of rapamycin pathway could be used for the restoration of blood-brain barrier integrity in disease states.
Collapse
MESH Headings
- Alzheimer Disease/drug therapy
- Alzheimer Disease/enzymology
- Alzheimer Disease/pathology
- Alzheimer Disease/psychology
- Animals
- Behavior, Animal
- Blood-Brain Barrier/drug effects
- Blood-Brain Barrier/enzymology
- Blood-Brain Barrier/pathology
- Cell Line
- Cognition
- Dementia, Vascular/drug therapy
- Dementia, Vascular/enzymology
- Dementia, Vascular/pathology
- Dementia, Vascular/psychology
- Disease Models, Animal
- Female
- Male
- Matrix Metalloproteinase 9/metabolism
- Mechanistic Target of Rapamycin Complex 1/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Protein Kinase Inhibitors/pharmacology
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Sirolimus/pharmacology
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- TOR Serine-Threonine Kinases/metabolism
- Tight Junction Proteins/metabolism
- Tight Junctions/drug effects
- Tight Junctions/enzymology
- Tight Junctions/pathology
Collapse
Affiliation(s)
- Candice E Van Skike
- Department of Cellular and Integrative Physiology and Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio , San Antonio, Texas
| | - Jordan B Jahrling
- Department of Cellular and Integrative Physiology and Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio , San Antonio, Texas
| | - Angela B Olson
- Department of Cellular and Integrative Physiology and Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio , San Antonio, Texas
| | - Naomi L Sayre
- Department of Neurosurgery, University of Texas Health San Antonio , San Antonio, Texas
- Department of Veterans Affairs, South Texas Veterans Health Care System, San Antonio, Texas
| | - Stacy A Hussong
- Department of Cellular and Integrative Physiology and Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio , San Antonio, Texas
- Department of Veterans Affairs, South Texas Veterans Health Care System, San Antonio, Texas
| | - Zoltan Ungvari
- Department of Geriatric Medicine and Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| | - James D Lechleiter
- Department of Cellular and Structural Biology, South Texas Research Facility Neuroscience Center, University of Texas Health San Antonio, San Antonio, Texas
| | - Veronica Galvan
- Department of Cellular and Integrative Physiology and Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio , San Antonio, Texas
- Department of Veterans Affairs, South Texas Veterans Health Care System, San Antonio, Texas
| |
Collapse
|
7
|
Dall'Agnol DJR, Corá LA, Teixeira MDCB, de Lima MB, Gama LA, Miranda JRDA, Américo MF. Gastrointestinal disorders after immunosuppression: an experimental model to evaluate the influence of monotherapy on motility parameters. Exp Physiol 2017; 102:924-933. [PMID: 28556421 DOI: 10.1113/ep086267] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/19/2017] [Indexed: 12/19/2022]
Abstract
NEW FINDINGS What is the central question of this study? The aim was to propose an animal model for investigating the effects of immunosuppressive monotherapy on gastrointestinal motility using a non-invasive biomagnetic technique. What is main finding and its importance? In our experimental study, immunosuppressive drugs currently in use accelerated gastric emptying whilst increasing the frequency and amplitude of gastric contractions after treatment, except for Mycophenolate and azathioprine. Alternating current biosusceptometry is a useful tool to evaluate side-effects of drugs on the gastrointestinal tract, which will help in understanding the symptoms and improving clinical management of patients. The aim was to propose an animal model for investigating the effects of immunosuppressive monotherapy on gastrointestinal motility using a non-invasive biomagnetic technique. Male Wistar rats were randomly distributed into the following treatment groups: ciclosporin, tacrolimus, prednisone, sirolimus, mycophenolate mofetil, everolimus, azathioprine and control. Each animal was treated for 14 days by gavage with dosages ranging from 1 to 20 mg kg-1 day-1 considering the area-to-volume ratio and hepatic metabolism. Gastrointestinal transit and gastric contractility measurements were evaluated by alternating current biosusceptometry before and after treatment. Gastric emptying was faster in animals treated with tacrolimus, prednisone, sirolimus and everolimus compared with control animals (126.7 ± 12.7 min). There was a significant increase in the frequency of contractions after ciclosporin, tacrolimus, azathioprine and sirolimus treatment compared with control animals (4.6 ± 0.3 cycles min-1 ). Increases in the amplitude of contraction were observed after treatment with tacrolimus, sirolimus and everolimus compared with control rats (34.9 ± 6.0 dB). The results showed that our animal model was suitable for demonstrating that most immunosuppressive drugs currently in use impaired at least one gastrointestinal motility parameter. As a non-invasive technique, alternating current biosusceptometry is a potentially useful tool for evaluation of side-effects of drugs in gastrointestinal tract, helping us to understand the symptoms to improve clinical management of patients.
Collapse
Affiliation(s)
- Denize Jussara Rupolo Dall'Agnol
- Postgraduate Program in Pharmacology and Biotechnology - São Paulo State University - UNESP, Institute of Biosciences, Botucatu-SP, Brazil
| | | | | | - Maysa Bruno de Lima
- Institute of Biological Sciences and Health - Federal University of Mato Grosso - UFMT, Barra do Garças, Mato Grosso, Brazil
| | - Loyane Almeida Gama
- Postgraduate Program in Pharmacology and Biotechnology - São Paulo State University - UNESP, Institute of Biosciences, Botucatu-SP, Brazil
| | - José Ricardo de Arruda Miranda
- Department of Physics and Biophysics - São Paulo State University - UNESP, Institute of Biosciences - Botucatu/SP, Brazil
| | - Madileine Francely Américo
- Institute of Biological Sciences and Health - Federal University of Mato Grosso - UFMT, Barra do Garças, Mato Grosso, Brazil
| |
Collapse
|
8
|
Khan N, Binder L, Pantakani DVK, Asif AR. MPA Modulates Tight Junctions' Permeability via Midkine/PI3K Pathway in Caco-2 Cells: A Possible Mechanism of Leak-Flux Diarrhea in Organ Transplanted Patients. Front Physiol 2017; 8:438. [PMID: 28694783 PMCID: PMC5483464 DOI: 10.3389/fphys.2017.00438] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/09/2017] [Indexed: 12/16/2022] Open
Abstract
Mycophenolic acid (MPA) is prescribed to prevent allograft rejection in organ transplanted patients. However, its use is sporadically linked to leak flux diarrhea and other gastrointestinal (GI) disturbances in around 75% of patients through yet unknown mechanisms. Recently, we identified Midkine as a modulator of tight junctions (TJs) permeability in MPA treated Caco-2 monolayer. In the present study, we investigated the possible involvement of Midkine dependent PI3K pathway in alteration of TJs under MPA treatment. Caco-2 cells were grown as monolayer to develop TJs and were treated for 72 h with DMSO (control) or MPA in presence and absence of Midkine inhibitor (iMDK) or PI3K inhibitors (LY/AMG). Caco-2 monolayer integrity was assessed by transepithelial electrical resistance (TEER) and FITC-dextran assays. Our functional assays showed that PI3K inhibitors (LY/AMG) can significantly inhibit the compromised TJs integrity of MPA-treated Caco-2 cells monolayer. Chromatin immunoprecipitation analyses showed a significant epigenetic activation of Midkine, PI3K, Cdx-2, and Cldn-2 genes and epigenetic repression of Cldn-1 gene after MPA treatment. The MPA-induced epigenetic alterations were further confirmed by mRNA and protein expression analysis. Collectively, our data shows that PI3K pathway as the downstream target of Midkine which in turn modulates p38MAPK and pAKT signaling to alter TJs permeability in Caco-2 cell monolayers treated with MPA. These results highlight the possible use of either Midkine or PI3K inhibitors as therapeutic agents to prevent MPA induced GI disturbances.
Collapse
Affiliation(s)
- Niamat Khan
- Institute for Clinical Chemistry/UMG-Laboratories, University Medical CenterGoettingen, Germany.,Department of Biotechnology and Genetic Engineering, Kohat University of Science and TechnologyKohat, Pakistan
| | - Lutz Binder
- Institute for Clinical Chemistry/UMG-Laboratories, University Medical CenterGoettingen, Germany.,German Center for Cardiovascular Research, Partner Site GoettingenGoettingen, Germany
| | - D V Krishna Pantakani
- Institute for Clinical Chemistry/UMG-Laboratories, University Medical CenterGoettingen, Germany.,German Center for Cardiovascular Research, Partner Site GoettingenGoettingen, Germany
| | - Abdul R Asif
- Institute for Clinical Chemistry/UMG-Laboratories, University Medical CenterGoettingen, Germany.,German Center for Cardiovascular Research, Partner Site GoettingenGoettingen, Germany
| |
Collapse
|
9
|
Reiner J, Hsieh CJ, Straarup C, Bodammer P, Schäffler H, Graepler F, Stüker D, Kratt T, Linnebacher M, Nadalin S, Witte M, Königsrainer A, Lamprecht G. After Intestinal Transplantation Kidney Function Is Impaired by Downregulation of Epithelial Ion Transporters in the Ileum. Transplant Proc 2016; 48:499-506. [DOI: 10.1016/j.transproceed.2015.12.068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 12/29/2015] [Indexed: 12/18/2022]
|
10
|
Khan N, Pantakani DVK, Binder L, Qasim M, Asif AR. Immunosuppressant MPA Modulates Tight Junction through Epigenetic Activation of MLCK/MLC-2 Pathway via p38MAPK. Front Physiol 2015; 6:381. [PMID: 26733876 PMCID: PMC4687409 DOI: 10.3389/fphys.2015.00381] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 11/24/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Mycophenolic acid (MPA) is an important immunosuppressive drug (ISD) prescribed to prevent graft rejection in the organ transplanted patients, however, its use is also associated with adverse side effects like sporadic gastrointestinal (GI) disturbances. Recently, we reported the MPA induced tight junctions (TJs) deregulation which involves MLCK/MLC-2 pathway. Here, we investigated the global histone acetylation as well as gene-specific chromatin signature of several genes associated with TJs regulation in Caco-2 cells after MPA treatment. RESULTS The epigenetic analysis shows that MPA treatment increases the global histone acetylation levels as well as the enrichment for transcriptional active histone modification mark (H3K4me3) at promoter regions of p38MAPK, ATF-2, MLCK, and MLC-2. In contrast, the promoter region of occludin was enriched for transcriptional repressive histone modification mark (H3K27me3) after MPA treatment. In line with the chromatin status, MPA treatment increased the expression of p38MAPK, ATF-2, MLCK, and MLC-2 both at transcriptional and translational level, while occludin expression was negatively influenced. Interestingly, the MPA induced gene expression changes and functional properties of Caco-2 cells could be blocked by the inhibition of p38MAPK using a chemical inhibitor (SB203580). CONCLUSIONS Collectively, our results highlight that MPA disrupts the structure of TJs via p38MAPK-dependent activation of MLCK/MLC-2 pathway that results in decreased integrity of Caco-2 monolayer. These results led us to suggest that p38MAPK-mediated lose integrity of epithelial monolayer could be the possible cause of GI disturbance (barrier dysfunction) in the intestine, leading to leaky style diarrhea observed in the organ-transplanted patients treated with MPA.
Collapse
Affiliation(s)
- Niamat Khan
- Proteomics Group, Institute for Clinical Chemistry/UMG-Laboratories, University Medical CentreGoettingen, Germany; Department of Biotechnology and Genetic Engineering, Kohat University of Science and TechnologyKohat, Pakistan
| | - D V Krishna Pantakani
- Proteomics Group, Institute for Clinical Chemistry/UMG-Laboratories, University Medical Centre Goettingen, Germany
| | - Lutz Binder
- Proteomics Group, Institute for Clinical Chemistry/UMG-Laboratories, University Medical Centre Goettingen, Germany
| | - Muhammad Qasim
- Proteomics Group, Institute for Clinical Chemistry/UMG-Laboratories, University Medical CentreGoettingen, Germany; Department of Microbiology, Kohat University of Science and TechnologyKohat, Pakistan
| | - Abdul R Asif
- Proteomics Group, Institute for Clinical Chemistry/UMG-Laboratories, University Medical Centre Goettingen, Germany
| |
Collapse
|
11
|
Li Z, Sun F, Zhang Y, Chen H, He N, Chen H, Song P, Wang Y, Yan S, Zheng S. Tacrolimus Induces Insulin Resistance and Increases the Glucose Absorption in the Jejunum: A Potential Mechanism of the Diabetogenic Effects. PLoS One 2015; 10:e0143405. [PMID: 26599323 PMCID: PMC4657894 DOI: 10.1371/journal.pone.0143405] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/07/2015] [Indexed: 12/16/2022] Open
Abstract
Background The use of the immunosuppressive drug tacrolimus (TAC) is related to new onset diabetes after transplantation. Herein, we examined the effect of intraperitoneal administered TAC on intestinal glucose absorption in mice. Methods Animals received low, medium, or high dose TAC (0.5, 1, or 5 mg/kg/d, respectively), or 0.9% saline solution (control) for 14 days. Oral glucose tolerance test (OGTT), insulin concentration test, and serum TAC concentration measurements was performed after 14 days of TAC exposure. Plasma insulin was assessed and electrogenic glucose absorption were measured by the sodium-dependent increase of the short-circuit current. Expression levels of the glucose transporters sodium glucose co-transporter (SGLT) 1, glucose transporter (GLUT) 2, and GLUT5 were also determined. Results Oral glucose absorption assessed by OGTT was significantly enhanced in the low, medium, and high groups. Serum insulin was elevated in the medium and high group compared with the control. Moreover, glucose-induced Isc was significantly higher in TAC administrated groups, which indicates that SGLT1 activity increased. Transcription levels and protein abundance of SGLT1 in the experimental groups also increased compared with the control. Conclusions TAC induced insulin resistance and strengthened intestinal glucose absorption by increasing the activity and expression of the glucose transporter, SGLT1.
Collapse
Affiliation(s)
- Zhiwei Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fei Sun
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yaohui Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hao Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ningning He
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hui Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Penghong Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yan Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Yan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
- * E-mail:
| |
Collapse
|
12
|
Dall'Agnol DJR, Hauschildt AT, Lima MB, Corá LA, Teixeira MCB, Américo MF. Effects of immunosuppressive drugs on gastrointestinal transit of rats: effects of tacrolimus, cyclosporine, and prednisone. Transplant Proc 2015; 46:1872-4. [PMID: 25131057 DOI: 10.1016/j.transproceed.2014.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Triple immunosuppressive therapy after organ transplantation may cause several gastrointestinal disturbances. It is difficult to identify which drug causes more complications, requiring an appropriate animal model. The aim was to compare the gastrointestinal transit in immunosuppressed rats under triple immunosuppressive therapy. Male rats were immunosuppressed by gavage during 14 days with tacrolimus (n = 10), cyclosporine (n = 12), and prednisone (n = 9). Animals received a magnetic pellet before (control) and after treatment that was monitored at predetermined intervals by AC biosusceptometry, a noninvasive and radiation-free technique. The following parameters were measured: creatinine serum, mean time of gastric emptying (MGET), mean time to reach cecum (MCAT), and mean transit time through small bowel (MSBTT). The differences were analyzed by ANOVA (Tukey). Our results showed that MGET of animals treated with prednisone, cyclosporine, and tacrolimus were reduced compared with control subjects (P < .03, P < .009, and P < .002, respectively). There was no difference in MCAT, whereas MSBTT was longer for tacrolimus and prednisone compared with control subjects (P < .004 and P < .004, respectively). Also, prednisone and tacrolimus presented a reduced MGET (P < .05 and P < .01, respectively) compared with cyclosporine. Our data showed a low serum creatinine level and no difference among groups regarding renal function. In summary, cyclosporine has less effect on the gastrointestinal transit; however, all of these drugs should be carefully prescribed to prevent gastrointestinal symptoms and improve quality of life after transplantation.
Collapse
Affiliation(s)
- D J R Dall'Agnol
- Universidade Federal de Mato Grosso-UFMT, Barra do Garças/MT, Brazil
| | - A T Hauschildt
- Universidade Federal de Mato Grosso-UFMT, Barra do Garças/MT, Brazil
| | - M B Lima
- Universidade Federal de Mato Grosso-UFMT, Barra do Garças/MT, Brazil
| | - L A Corá
- Universidade Estadual de Ciências da Saúde de Alagoas-UNCISAL, Maceió/AL, Brazil.
| | - M C B Teixeira
- Universidade Estadual de Ciências da Saúde de Alagoas-UNCISAL, Maceió/AL, Brazil
| | - M F Américo
- Universidade Federal de Mato Grosso-UFMT, Barra do Garças/MT, Brazil
| |
Collapse
|
13
|
Shen B, Yu H, Yu T, Shen J, Meng N, Cai X. A CD52 antibody impairs mouse-transplanted intestinal tight junctions. J Surg Res 2015; 196:278-84. [PMID: 25796109 DOI: 10.1016/j.jss.2015.02.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 01/28/2015] [Accepted: 02/20/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Induction immunosuppression strategies using a CD52 monoclonal antibody (mAb) have been introduced for small bowel transplantation, resulting in improved outcomes. However, little information is known about the effects of the antibody on the microstructure of the intestinal barrier, which functions to prevent bacterial translocation. In this study, we used a murine orthotopic small bowel transplantation model to investigate the impact of a CD52 mAb on tight junctions (TJs), which are considered a central part of the intestinal barrier, of the transplanted intestine. MATERIALS AND METHODS C57BL/6 mice were used as recipients. The grafts were harvested from BALB/c mice in allogeneic groups and C57BL/6 mice in syngeneic groups. The anti-mouse CD52 mAb was applied as a surrogate antibody. Transmission electron microscopy was used to evaluate the TJ ultrastructure. The expression of the TJ proteins occludin and ZO-1 was analyzed by Western blot. The distribution of TJ proteins was observed by immunofluorescence, and the permeability of the transplanted intestine was assessed in vivo using FITC-dextran. RESULTS After CD52 mAb application, a compromised TJ ultrastructure was observed. In addition, TJ protein expression (occludin and ZO-1) decreased and the intestinal permeability increased. CONCLUSIONS The anti-mouse CD52 mAb impaired the transplanted intestinal TJ and barrier.
Collapse
Affiliation(s)
- Bo Shen
- Department of General Surgery, Institute of Micro-Invasive Surgery of Zhejiang University, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Yu
- Department of General Surgery, Institute of Micro-Invasive Surgery of Zhejiang University, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tunan Yu
- Department of General Surgery, Institute of Micro-Invasive Surgery of Zhejiang University, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiliang Shen
- Zhejiang University School of Medicine, Hangzhou, China
| | - Ning Meng
- Zhejiang University School of Medicine, Hangzhou, China
| | - Xiujun Cai
- Department of General Surgery, Institute of Micro-Invasive Surgery of Zhejiang University, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
14
|
Qasim M, Rahman H, Ahmed R, Oellerich M, Asif AR. Mycophenolic acid mediated disruption of the intestinal epithelial tight junctions. Exp Cell Res 2014; 322:277-89. [PMID: 24509232 DOI: 10.1016/j.yexcr.2014.01.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 12/21/2013] [Accepted: 01/21/2014] [Indexed: 12/12/2022]
Abstract
Gastrointestinal toxicity is a common adverse effect of mycophenolic acid (MPA) treatment in organ transplant patients, through poorly understood mechanisms. Phosphorylation of myosin light chain 2 (MLC2) is associated with epithelial tight junction (TJ) modulation which leads to defective epithelial barrier function, and has been implicated in GI diseases. The aim of this study was to investigate whether MPA could induce epithelial barrier permeability via MLC2 regulation. Caco-2 monolayers were exposed to therapeutic concentrations of MPA, and MLC2 and myosin light chain kinase (MLCK) expression were analyzed using PCR and immunoblotting. Epithelial cell permeability was assessed by measuring transepithelial resistance (TER) and the flux of paracellular permeability marker FITC-dextran across the epithelial monolayers. MPA increased the expression of MLC2 and MLCK at both the transcriptional and translational levels. In addition, the amount of phosphorylated MLC2 was increased after MPA treatment. Confocal immunofluorescence analysis showed redistribution of TJ proteins (ZO-1 and occludin) after MPA treatment. This MPA mediated TJ disruption was not due to apoptosis or cell death. Additionally ML-7, a specific inhibitor of MLCK was able to reverse both the MPA mediated decrease in TER and the increase in FITC-dextran influx, suggesting a modulating role of MPA on epithelial barrier permeability via MLCK activity. These results suggest that MPA induced alterations in MLC2 phosphorylation and may have a role in the patho-physiology of intestinal epithelial barrier disruption and may be responsible for the adverse effects (GI toxicity) of MPA on the intestine.
Collapse
Affiliation(s)
- Muhammad Qasim
- Institute of Clinical Chemistry/UMG-Laboratories, University Medical Centre, Robert Koch Strasse 40, 37075 Goettingen, Germany; Department of Microbiology, Kohat University of Science and Technology, 26000 Kohat, Pakistan
| | - Hazir Rahman
- Institute of Clinical Chemistry/UMG-Laboratories, University Medical Centre, Robert Koch Strasse 40, 37075 Goettingen, Germany; Department of Microbiology, Kohat University of Science and Technology, 26000 Kohat, Pakistan
| | - Raees Ahmed
- Institute for Applied Science and Clinical Trials GmbH - IFS, Georg-August University, 37075 Goettingen, Germany
| | - Michael Oellerich
- Institute of Clinical Chemistry/UMG-Laboratories, University Medical Centre, Robert Koch Strasse 40, 37075 Goettingen, Germany
| | - Abdul R Asif
- Institute of Clinical Chemistry/UMG-Laboratories, University Medical Centre, Robert Koch Strasse 40, 37075 Goettingen, Germany.
| |
Collapse
|
15
|
Pazetti R, Pêgo-Fernandes PM, Jatene FB. Adverse effects of immunosuppressant drugs upon airway epithelial cell and mucociliary clearance: implications for lung transplant recipients. Drugs 2014; 73:1157-69. [PMID: 23842748 DOI: 10.1007/s40265-013-0089-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Optimal post-transplantation immunosuppression is critical to the survival of the graft and the patient after lung transplantation. Immunosuppressant agents target various aspects of the immune system to maximize graft tolerance while minimizing medication toxicities and side effects. The vast majority of patients receive maintenance immunosuppressive therapy consisting of a triple-drug regimen including a calcineurin inhibitor, a cell cycle inhibitor and a corticosteroid. Although these immunosuppressant drugs are frequently used after transplantation and to control inflammatory processes, limited data are available with regard to their effects on cells other than those from the immunological system. Notably, the airway epithelial cell is of interest because it may contribute to development of bronchiolitis obliterans through production of pro-inflammatory cytokines. This review focuses the current armamentarium of immunosuppressant drugs used after lung transplantation and their main side effects upon airway epithelial cells and mucociliary clearance.
Collapse
Affiliation(s)
- Rogerio Pazetti
- Laboratory of Thoracic Surgery Research-LIM61, Department of Cardiopneumology, Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Avenida Doutor Arnaldo, 455, 1o. Andar, Sala 1220, Pacaembu, São Paulo, SP, 01246-000, Brazil.
| | | | | |
Collapse
|
16
|
Fölster-Holst R, Dähnhardt-Pfeiffer S, Dähnhardt D, Proksch E. The role of skin barrier function in atopic dermatitis: an update. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/edm.12.17] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|