1
|
Alqadi R, Alqumia A, Alhomoud IS, Alhowail A, Aldubayan M, Mohammed HA, Alhmoud H, Khan RA. Cyclosporine: Immunosuppressive effects, entwined toxicity, and clinical modulations of an organ transplant drug. Transpl Immunol 2025; 88:102147. [PMID: 39549927 DOI: 10.1016/j.trim.2024.102147] [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: 07/21/2024] [Revised: 11/10/2024] [Accepted: 11/10/2024] [Indexed: 11/18/2024]
Abstract
The discovery and use of cyclosporine since its inception into the clinics in the '70s and up have played a crucial role in advancing transplant therapy, and containment of the immune-based rejections. The drug has improved the high rates of acute rejections and has supported early graft survival. However, the long-term survival of renal allografts is still less prevalent, and an in-depth analysis, as well as reported findings led us to believe that there is a chronic irreversible component to the drug, that is tackled through its metabolites, and that causes toxicity, which has led to new therapies, including monoclonal antibody-based medications. A recap of the immunosuppressive effects, and entwined toxicity of the drug, now relegated primarily to bone marrow early transplants, is being overviewed for the past protocols that were used to minimize, and avoid, or use this calcineurin inhibitor class of drug, cyclosporine, in combination with other drugs. The current review circumvents the cyclosporine's mechanism of action, pathophysiology, cytochrome roles, and other factors associated with acute and chronic toxicity developments. The review also attempts to find conclusive strategies reported in the recent studies to avoid its toxic side effects, and develop a safe-use strategy for the drug. Gastrointestinal decontamination, supporting the airway, monitoring for signs of respiratory insufficiency, monitoring for severe reactions, such as seizures, need for administration of oxygen, and avoiding the administration of drugs, that increase the blood levels of the cyclosporine, are beneficial interventions, when encountering cyclosporine toxicity cases. The constrained therapeutic outcomes have also led to redesign, and making use of combined formulations to reassess the pharmacokinetics of the drug.
Collapse
Affiliation(s)
- Razan Alqadi
- Department of Pharmacy, King Saud Hospital, Unaizah, Qassim 56249, Saudi Arabia
| | - Amal Alqumia
- Department of Pharmacy, King Fahd Specialist Hospital, Buraydah, Qassim 52719, Saudi Arabia
| | - Ibrahim S Alhomoud
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Ahmad Alhowail
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Maha Aldubayan
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Hamdoon A Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Hussam Alhmoud
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Riaz A Khan
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia.
| |
Collapse
|
2
|
Abd-Eldayem AM, Makram SM, Messiha BAS, Abd-Elhafeez HH, Abdel-Reheim MA. Cyclosporine-induced kidney damage was halted by sitagliptin and hesperidin via increasing Nrf2 and suppressing TNF-α, NF-κB, and Bax. Sci Rep 2024; 14:7434. [PMID: 38548778 PMCID: PMC10978894 DOI: 10.1038/s41598-024-57300-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/16/2024] [Indexed: 04/01/2024] Open
Abstract
Cyclosporine A (CsA) is employed for organ transplantation and autoimmune disorders. Nephrotoxicity is a serious side effect that hampers the therapeutic use of CsA. Hesperidin and sitagliptin were investigated for their antioxidant, anti-inflammatory, and tissue-protective properties. We aimed to investigate and compare the possible nephroprotective effects of hesperidin and sitagliptin. Male Wistar rats were utilized for induction of CsA nephrotoxicity (20 mg/kg/day, intraperitoneally for 7 days). Animals were treated with sitagliptin (10 mg/kg/day, orally for 14 days) or hesperidin (200 mg/kg/day, orally for 14 days). Blood urea, serum creatinine, albumin, cystatin-C (CYS-C), myeloperoxidase (MPO), and glucose were measured. The renal malondialdehyde (MDA), glutathione (GSH), catalase, and SOD were estimated. Renal TNF-α protein expression was evaluated. Histopathological examination and immunostaining study of Bax, Nrf-2, and NF-κB were performed. Sitagliptin or hesperidin attenuated CsA-mediated elevations of blood urea, serum creatinine, CYS-C, glucose, renal MDA, and MPO, and preserved the serum albumin, renal catalase, SOD, and GSH. They reduced the expressions of TNF-α, Bax, NF-κB, and pathological kidney damage. Nrf2 expression in the kidney was raised. Hesperidin or sitagliptin could protect the kidney against CsA through the mitigation of oxidative stress, apoptosis, and inflammation. Sitagliptin proved to be more beneficial than hesperidin.
Collapse
Affiliation(s)
- Ahmed M Abd-Eldayem
- Department of Medical Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt.
- Department of Pharmacology, Faculty of Medicine, Merit University, Sohâg, Egypt.
| | | | | | - Hanan H Abd-Elhafeez
- Department of Cell and Tissue, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, Egypt
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra, Saudi Arabia
| |
Collapse
|
3
|
2-Methoxyestradiol TPGS Micelles Attenuate Cyclosporine A-Induced Nephrotoxicity in Rats through Inhibition of TGF-β1 and p-ERK1/2 Axis. Antioxidants (Basel) 2022; 11:antiox11081499. [PMID: 36009218 PMCID: PMC9405159 DOI: 10.3390/antiox11081499] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 01/25/2023] Open
Abstract
The immunosuppressant cyclosporine A (CSA) has been linked to serious renal toxic effects. Although 2-methoxyestradiol (2ME) possesses a wide range of pharmacological abilities, it suffers poor bioavailability after oral administration. The purpose of this study was to evaluate the potential of 2ME loaded D-ɑ-tocopheryl polyethylene glycol succinate (TPGS) micelles to prevent CSA-induced nephrotoxicity in rats. A 2ME-TPGS was prepared and showed particle size of 44.3 ± 3.5 nm with good entrapment efficiency and spherical structures. Male Wistar rats were divided into 5 groups, namely: Control, Vehicle, CSA, CSA + 2ME-Raw, and CSA + 2ME-Nano. CSA was injected daily at a SC dose of 20 mg/kg. Both 2ME-Raw and 2ME-Nano were given daily at oral doses of 5 mg/kg. Treatments continued for three successive weeks. 2ME-TPGS exerted significant protective effects against CSA nephrotoxicity. This was evidenced in ameliorating deterioration of renal functions, attenuation of pathological changes in kidney tissues, exerting significant anti-fibrotic, antioxidant, and anti-inflammatory effects together with significant anti-apoptotic effects. Western blot analyses showed both 2ME-Raw and 2ME-Nano significantly inhibited protein expression of TGF-β1 and phospho-ERK (p-ERK). It was observed that 2ME-TPGS, in almost all experiments, exerted superior protective effects as compared with 2ME-Raw. In conclusion, 2ME loaded in a TPGS nanocarrier possesses significant protective activities against CSA-induced kidney injury in rats. This is attributable to 2ME anti-fibrotic, antioxidant, anti-inflammatory, and anti-apoptotic activities which are mediated at least partly by inhibition of TGF-β1/p-ERK axis.
Collapse
|
4
|
Abstract
INTRODUCTION Liver transplantation is a life-changing event for patients and survival following transplantation has improved significantly since the first transplantation in 1967. Following liver transplantation, patients face a unique set of healthcare management decisions including transplantation-specific complications, recurrence of primary liver disease, as well as metabolic and malignancy concerns related to immunosuppression. As more patients with liver disease receive transplantation and live longer, understanding and managing these patients will require not only transplant specialist but also local subspecialist and primary care physicians. AREAS COVERED This review covers common issues related to the management of patients following liver transplantation including immunosuppression, liver allograft dysfunction, metabolic complications, as well as routine health maintenance such as immunizations and cancer screening. EXPERT OPINION Optimizing medical care for patients following liver transplant will benefit from ensuring all providers, not just transplant specialist, have a basic understanding of the common issues encountered in the post-transplant patient. This review provides an overview of common healthcare concerns and management options for patients following liver transplantation.
Collapse
Affiliation(s)
- Nicholas Hoppmann
- Division of Gastroenterology and Hepatology, University of Alabama at Birmingham , Birmingham, Alabama, USA
| | - Omar Massoud
- Division of Gastroenterology and Hepatology, University of Alabama at Birmingham , Birmingham, Alabama, USA
| |
Collapse
|
5
|
Lacquaniti A, Campo S, Casuscelli Di Tocco T, Rovito S, Bucca M, Ragusa A, Monardo P. Acute and chronic kidney disease after pediatric liver transplantation: An underestimated problem. Clin Transplant 2020; 34:e14082. [PMID: 32949054 DOI: 10.1111/ctr.14082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 08/02/2020] [Accepted: 08/15/2020] [Indexed: 12/13/2022]
Abstract
Acute and chronic kidney injuries represent critical issues after liver transplantation (LTx), but whereas renal dysfunction in adult transplant patients is well documented, little is known about its prevalence in childhood. It is a challenge to accurately evaluate renal function in patients with liver disease, due to several confounding factors. Creatinine-based equations estimating glomerular filtration rate, validated in nephropathic patients without hepatic issues, are frequently inaccurate in end-stage liver disease, underestimating the real impact of renal disease. Moreover, whereas renal issues observed within 1 year from LTx were often related to acute injuries, kidney damage observed after 5-7 years from LTx, is due to chronic, irreversible mechanisms. Most immunosuppression protocols are based on calcineurin inhibitors (CNIs) and corticosteroids, but mycophenolate mofetil or sirolimus could play significant roles, also in children. Early diagnosis and personalized treatment represent the bases of kidney disease management, in order to minimize its close relation with increased mortality. This review analyzed acute and chronic kidney damage after pediatric LTx, also discussing the impact of pre-existent renal disease. The main immunosuppressant strategies have been reviewed, highlighting their impact on kidney function. Different methods assessing renal function were reported, with the potential application of new renal biomarkers.
Collapse
Affiliation(s)
- Antonio Lacquaniti
- Department of Internal Medicine, Nephrology and Dialysis Unit, Papardo Hospital of Messina, Messina, Italy
| | - Susanna Campo
- Department of Internal Medicine, Nephrology and Dialysis Unit, Papardo Hospital of Messina, Messina, Italy
| | - Teresa Casuscelli Di Tocco
- Department of Internal Medicine, Nephrology and Dialysis Unit, Papardo Hospital of Messina, Messina, Italy
| | - Stefania Rovito
- Department of Internal Medicine, Nephrology and Dialysis Unit, Papardo Hospital of Messina, Messina, Italy
| | - Maurizio Bucca
- Department of Internal Medicine, Nephrology and Dialysis Unit, Papardo Hospital of Messina, Messina, Italy
| | - Antonino Ragusa
- Department of Internal Medicine, Nephrology and Dialysis Unit, Papardo Hospital of Messina, Messina, Italy
| | - Paolo Monardo
- Department of Internal Medicine, Nephrology and Dialysis Unit, Papardo Hospital of Messina, Messina, Italy
| |
Collapse
|
6
|
Zhou X, Qu Y, Gan G, Zhu S, Huang Y, Liu Y, Zhu J, Xie B, Tan Z. Cyclosporine A Plus Ischemic Postconditioning Improves Neurological Function in Rats After Cardiac Resuscitation. Neurocrit Care 2020; 32:812-821. [PMID: 31576483 DOI: 10.1007/s12028-019-00849-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND OBJECTIVE Attenuation of neuronal apoptosis helps maintain neurological function in patients after cardiac arrest. After ischemia-reperfusion, both cyclosporin A (CsA) and ischemic postconditioning independently protect mitochondria and thus reduce nerve injury. This study employed a rat model to evaluate the neuroprotective effect of combining ischemic postconditioning with CsA after cardiopulmonary resuscitation (CPR). METHODS Rats were apportioned equally to model control, postconditioned, CsA-treated, or CsA + postconditioned groups. Asphyxial cardiac arrest was imposed using modified Utstein-style guidelines. In the appropriate groups, postconditioning was implemented by ischemia and reperfusion (clamping and loosening the left femoral artery); CsA treatment was delivered with a single intravenous dose. Neurological deficits were scored at different times after CPR. Histological evaluation and electron microscopy were used to evaluate tissue damage, and TUNEL and flow cytometry were used to measure the apoptotic rate of hippocampal neurons and size of the mitochondrial permeability transition pore (mPTP) opening. RESULTS The apoptotic rate was significantly lower in the postconditioned and CsA-treated groups compared with the model control and lowest in the CsA + postconditioned group. By histological evaluation and electron microscopy, the least damage was observed in the CsA + postconditioned group. The neurological deficit score of the CsA + postconditioned group was significantly higher than that of the CsA-treated group, but the size of the mPTP openings of these two groups was comparable. CONCLUSION Ischemic postconditioning combined with CsA exerted a better neuroprotective effect after CPR than did either postconditioning or CsA alone. Inhibiting the opening of the mPTP is not the only neuroprotective mechanism.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Anesthesiology, General Hospital of Central Theater Command of People's Liberation Army of China, Wuhan, China
- Southern Medical University, Guangzhou, China
| | - YanLiang Qu
- Department of Anesthesiology, No. 971 Hospital of the PLA Navy, Qingdao, China
| | - GuoShen Gan
- Southern Medical University, Guangzhou, China
| | - ShuiBo Zhu
- Department of Thoracic Cardiovascular Surgery, General Hospital of Central Theater Command of People's Liberation Army of China, 627 Wuluo Road, Wuhan, 430070, Hubei, China.
- Southern Medical University, Guangzhou, China.
| | - Yang Huang
- Southern Medical University, Guangzhou, China
| | - Yong Liu
- Department of Thoracic Cardiovascular Surgery, General Hospital of Central Theater Command of People's Liberation Army of China, 627 Wuluo Road, Wuhan, 430070, Hubei, China
| | - Jian Zhu
- Department of Thoracic Cardiovascular Surgery, General Hospital of Central Theater Command of People's Liberation Army of China, 627 Wuluo Road, Wuhan, 430070, Hubei, China
| | - Biao Xie
- Southern Medical University, Guangzhou, China
| | - ZhiTian Tan
- Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Abstract
Atopic dermatitis (AD) is a chronic, inflammatory skin disease characterized by pruritus, inflammatory erythematous skin lesions, and skin-barrier defect. Current mainstay treatments of emollients, steroids, calcineurin inhibitors, and immunosuppressants have limited efficacy and potentially serious side effects. Recent advances and understanding of the pathogenesis of AD have resulted in new therapies that target specific pathways with increased efficacy and the potential for less systemic side effects. New FDA-approved therapies for AD are crisaborole and dupilumab. The JAK-STAT inhibitors (baricitinib, upadacitinib, PF-04965842, ASN002, tofacitinib, ruxolitinib, and delgocitinib) have the most promising results of the emerging therapies. Other drugs with potential include the aryl hydrocarbon receptor modulating agent tapinarof, the IL-4/IL-13 antagonists lebrikizumab and tralokinumab, and the IL-31Rα antagonist nemolizumab. In this review, new and emerging AD therapies will be discussed along with their mechanisms of action and their potential based on clinical study data.
Collapse
Affiliation(s)
- Henry L Nguyen
- Department of Dermatology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55902, USA
| | - Katelyn R Anderson
- Department of Dermatology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55902, USA
| | - Megha M Tollefson
- Department of Dermatology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55902, USA.
| |
Collapse
|
8
|
Fries JWU. MicroRNAs as markers to monitor endothelin-1 signalling and potential treatment in renal disease: Carcinoma - proteinuric damage - toxicity. Biol Cell 2019; 111:169-186. [PMID: 30866090 DOI: 10.1111/boc.201800059] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/01/2019] [Accepted: 02/25/2019] [Indexed: 12/11/2022]
Abstract
This review highlights new developments in miRNA as diagnostic and surveillance tools in diseases damaging the renal proximal tubule mediated by endothelin in the field of renal carcinoma, proteinuric kidney disease and tubulotoxicity. A new mechanism in the miRNA regulation of proteins leads to the binding of the miRNA directly to the DNA with premature transcriptional termination and hence the formation of truncated protein isoforms (Mxi2, Vim3). These isoforms are mediated through miRNA15a or miRNA 498, respectively. ET-1 can activate a cytoplasmic complex consisting of NF-κB p65, MAPK p38α, and PKCα. Consequently, PKCα does not transmigrate into the nucleus, which leads to the loss of suppression of a primiRNA15a, maturation of this miRNA in the cytoplasm, tubular secretion and detectability in the urine. This mechanism has been shown in renal cell carcinoma and in proteinuric disease as a biomarker for the activation of the signalling pathway. Similarly, ET-1 induced miRNA 498 transmigrates into the nucleus to form the truncated protein Vim3, which is a biomarker for the benign renal cell tumour, oncocytoma. In tubulotoxicity, ET-1 induced miRNa133a down-regulating multiple-drug-resistant related protein-2, relevant for proteinuric and cisplatin/cyclosporine A toxicity. Current advantages and limitations of miRNAs as urinary biomarkers are discussed.
Collapse
Affiliation(s)
- Jochen W U Fries
- Department of Pathology, University Hospital of Koeln, 50931, Koeln, Germany
| |
Collapse
|
9
|
Marrow Mesenchymal Stem Cells Effectively Reduce Histologic Changes in a Rat Model of Chronic Renal Allograft Rejection. Transplant Proc 2018; 49:2194-2203. [PMID: 29149982 DOI: 10.1016/j.transproceed.2017.09.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 09/06/2017] [Accepted: 09/22/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Chronic allograft rejection remains as the leading cause of the chronic renal grafts loss post-transplantation. No therapy has been found really effective to prevent and treat chronic allograft rejection. Mesenchymal stem cells (MSCs) have characteristics of immunomodulation and are expected to be used for inducing graft immune tolerance in organ transplantation. We investigated the efficacy and safety of early infusion of donor-derived marrow MSCs in a rat model of chronic renal allograft rejection. METHODS Orthotopic kidney transplantations were performed in a rat strain combination of Sprague-Dawley (SD) → Wistar. The native right kidneys of recipient rats were kept intact as internal control of each graft. Twenty-three successfully transplanted recipient rats were divided into 3 groups: group 1 (the MSCs therapy group) (n = 8) and group 2 (the control group) (n = 8) both received a 10-day course of cyclosporine (CsA) (2 mg/kg intraperitoneally) to prevent initial acute rejection. MSCs (1 × 107) of first dosage and an additional dosage were injected into group 1 postoperative days (PODs) 0, 3, and 7. Group 2 received 0.9% saline solution in addition to CsA as the control group. Group 3 consisted of recipients (n = 7) receiving neither immunosuppression nor MSCs. Renal histopathology and immunohistochemistry of transforming growth factor β1 (TGF-β1) was examined at week 12. Safety of MSC infusion was determined by observing symptoms and signs after infusion and performing gross anatomy at week 12. RESULTS All the grafts of group 3 developed acute rejection and were rejected within 4 weeks. Bone marrow MSCs significantly decreased the severity of mononuclear cell interstitial inflammation, tubular atrophy, interstitial fibrosis, and vascular fibrous intimal thickening in renal grafts (P < .001). MSCs also greatly reduced the glomerulosclerosis rate of the transplanted kidneys of group 1 (P < .001). The TGF-ß1 expression of group 1 was weaker than that of group 2 (P = .043). There were no symptoms or signs of severe adverse side effects observed. CONCLUSIONS Early bone marrow MSCs infusion on PODs 0, 3, and 7 are effective and safe for chronic renal allograft rejection in rats. MSCs hold significant promise for clinical transplantation to treat chronic renal allograft rejection and prolong the renal graft survival.
Collapse
|
10
|
Leal R, Tsapepas D, Crew RJ, Dube GK, Ratner L, Batal I. Pathology of Calcineurin and Mammalian Target of Rapamycin Inhibitors in Kidney Transplantation. Kidney Int Rep 2018; 3:281-290. [PMID: 30276344 PMCID: PMC6161639 DOI: 10.1016/j.ekir.2017.10.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/15/2017] [Accepted: 10/23/2017] [Indexed: 12/26/2022] Open
Abstract
The recent evolution in immunosuppression therapy has led to significant improvement in short-term kidney allograft outcomes; however, this progress did not translate into similar improvement in long-term graft survival. The latter, at least in part, is likely to be attributed to immunosuppressant side effects. In this review, we focus on the histologic manifestations of calcineurin inhibitor and mammalian target of rapamycin inhibitor toxicity. We discuss the pathologic features attributed to such toxicity and allude to the lack of highly specific pathognomonic lesions. Finally, we highlight the importance of clinicopathologic correlation to achieve a meaningful pathologic interpretation.
Collapse
Affiliation(s)
- Rita Leal
- Department of Nephrology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Department of Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| | - Demetra Tsapepas
- Department of Pharmacy, NewYork-Presbyterian Hospital, New York, New York, USA
| | - Russell J. Crew
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| | - Geoffrey K. Dube
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| | - Lloyd Ratner
- Department of Surgery, Division of Transplantation, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| | - Ibrahim Batal
- Department of Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
11
|
Everolimus immunosuppression for renal protection, reduction of allograft vasculopathy and prevention of allograft rejection in de-novo heart transplant recipients: could we have it all? Curr Opin Organ Transplant 2017; 22:198-206. [PMID: 28463861 DOI: 10.1097/mot.0000000000000409] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW De-novo introduction of everolimus (Eve) in heart transplant recipients opens for early reduction of calcineurin inhibitors (CNI) and potential of preserving renal function, attenuate progression of coronary allograft vasculopathy (CAV) and maintain rejection efficacy. RECENT FINDINGS The first trials demonstrated adequate rejection prophylaxis and favorable outcomes on CAV, but observed enhanced nephrotoxicity because of insufficient CNI reduction. The SCHEDULE trial compared de-novo Eve with significantly reduced CNI exposure and conversion to CNI-free treatment week 7-11 postheart transplant, with standard CNI immunosuppression. Improved renal function and attenuation of CAV was found among Eve patients, with higher numbers of treated acute rejections observed. With sustained superior renal and CAV related data also after 36 months with the Eve protocol, cardiac function was equally well preserved in both groups. According to the International Society of Heart and Lunge Transplantation registry, mammalian target of rapamycin inhibitor treatment is uncommon during the first postoperative year, with a prevalence of 20% in patients after 5 years. SUMMARY Current evidence suggests a greater benefit from these immunosuppressives if introduced at an earlier timepoint. Immunosuppressive protocols based on Eve treatment in de-novo patients should be further investigated and developed, enabling CNI avoidance before accelerating side-effects lead to irreversible damage.
Collapse
|
12
|
Melilli E, Manonelles A, Montero N, Grinyo J, Martinez-Castelao A, Bestard O, Cruzado J. Impact of immunosuppressive therapy on arterial stiffness in kidney transplantation: are all treatments the same? Clin Kidney J 2017; 11:413-421. [PMID: 29988241 PMCID: PMC6007381 DOI: 10.1093/ckj/sfx120] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 09/06/2017] [Indexed: 02/07/2023] Open
Abstract
Arterial stiffness is a biologic process related to ageing and its relationship with cardiovascular risk is well established. Several methods are currently available for non-invasive measurement of arterial stiffness that provide valuable information to further assess patients’ vascular status in real time. In kidney transplantation recipients, several factors could accelerate the stiffness process, such as the use of calcineurin inhibitors (CNIs), the presence of chronic kidney disease and other classical cardiovascular factors, which would explain, at least in part, the high cardiovascular mortality and morbidity. Despite the importance of arterial stiffness as a biomarker of cardiovascular risk, and unlike other cardiovascular risk factors (e.g. left ventricular hypertrophy), only a few clinical trials or retrospective studies of kidney recipients have evaluated its impact. In this review we describe the clinical impact of arterial stiffness as a prognostic marker of cardiovascular disease and the effects of different immunosuppressive regimens on its progression, focusing on the potential benefits of CNI-sparing protocols and supporting the rationale for individualization of immunosuppression in patients with lower arterial elasticity. Among the immunosuppressive drugs, a belatacept-based regimen seems to offer better vascular protection compared with CNIs, although further studies are needed to confirm the preliminary positive results.
Collapse
Affiliation(s)
- Edoardo Melilli
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
- Correspondence and offprint requests to: Edoardo Melilli; E-mail:
| | - Anna Manonelles
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
| | - Nuria Montero
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
| | - Josep Grinyo
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
| | | | - Oriol Bestard
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
| | - Josep Cruzado
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
| |
Collapse
|
13
|
Lemos SVD, Vianna IG, Castiglia YMM, Golim MDA, Souza AVGD, Carvalho LRD, Deffune E, Nascimento PD, Módolo NSP, Vianna PTG. Cyclosporine A attenuates apoptosis and necrosis after ischemia-reperfusion-induced renal injury in transiently hyperglycemic rats. Acta Cir Bras 2017; 32:203-210. [PMID: 28403344 DOI: 10.1590/s0102-865020170030000004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 02/21/2017] [Indexed: 11/22/2022] Open
Abstract
Purpose: To investigate the effects of cyclosporine A on renal ischemia-reperfusion injury during transient hyperglycemia in rats. Methods: In a model of ischemia-reperfusion-induced renal injury and transiently induced hyperglycemia by intraperitoneal injection of glucose, 2.5 g.kg-1, Wistar rats were anesthetized with either isoflurane or propofol and received intravenous cyclosporine A, 5 mg.kg-1, five minutes before reperfusion. Comparison groups were isoflurane and propofol sham groups and isoflurane and propofol ischemia-reperfusion-induced renal injury. Renal tubular cell viability was quantitatively assessed by flow cytometry after cell culture and classified as early apoptosis, necrotic cells, and intact cells. Results: Early apoptosis was significantly higher in isoflurane and propofol anesthetized animals subjected to renal ischemia-reperfusion injury when compared to both cyclosporine A treated and sham groups. Necrosis percentage was significantly higher in propofol-anesthetized animals subjected to renal ischemia-reperfusion injury. The percentage of intact cells was lower in both, isoflurane and propofol anesthetized animals subjected to renal ischemia-reperfusion injury. Conclusion: In a model of ischemia-reperfusion-induced renal injury, cyclosporine A, 5 m.kg-1, administered five minutes before renal reperfusion in rats with acute-induced hyperglycemia under either isoflurano or propofol anesthesia, attenuated early apoptosis and preserved viability in renal tubular cells, regardless of the anesthetic used.
Collapse
Affiliation(s)
- Sylvio Valença de Lemos
- Fellow PhD degree, Postgraduate Program in Anesthesiology, Department of Anesthesiology, Botucatu Medical School, Universidade Estadual de São Paulo (UNESP), Botucatu-SP, Brazil. Conception and design of the study, acquisition of data, manuscript writing, critical revision
| | - Isabela Galvão Vianna
- Fellow, Experimental Surgery, Department of Anesthesiology, Botucatu Medical School, UNESP, Botucatu-SP, Brazil. Manuscript writing
| | - Yara Marcondes Machado Castiglia
- PhD, Full Professor, Department of Anesthesiology, Botucatu Medical School, UNESP, Botucatu-SP, Brazil. Critical revision, final approval of the version to be published
| | - Marjorie de Assis Golim
- PhD, Biologist, Department of Internal Medicine, Botucatu Medical School, UNESP, Botucatu-SP, Brazil. Technical procedures, histopathological examinations
| | - Aparecida Vitória Gonçalves de Souza
- Fellow PhD degree, Postgraduate Program in Anesthesiology, Department of Anesthesiology, Botucatu Medical School, UNESP, Botucatu-SP, Brazil. Acquisition of data
| | - Lídia Raquel de Carvalho
- PhD, Assistant Professor, Department of Biostatistics, Bioscience Institute of Botucatu, UNESP, Botucatu-SP, Brazil. Statistical analysis
| | - Elenice Deffune
- PhD, Assistant Professor, Department of Internal Medicine, Botucatu Medical School, UNESP, Botucatu-SP, Brazil. Technical procedures, histopathological examinations
| | - Paulo do Nascimento
- PhD, Associate Professor, Department of Anesthesiology, Botucatu Medical School, UNESP, Botucatu-SP, Brazil. Manuscript preparation and writing, critical revision
| | - Norma Sueli Pinheiro Módolo
- PhD, Full Professor, Department of Anesthesiology, Botucatu Medical School, UNESP, Botucatu-SP, Brazil. Manuscript preparation and writing, critical revision
| | - Pedro Thadeu Galvão Vianna
- PhD, Full Professor, Department of Anesthesiology, Botucatu Medical School, UNESP, Botucatu-SP, Brazil. Conception and design of the study, analysis and interpretation of data, critical revision, final approval of the version to be published
| |
Collapse
|
14
|
Abdel-Hafez MA, Abou-El-Hana NM, Erfan AA, El-Gamasy M, Abdel-Nabi H. Predictive risk factors of steroid dependent nephrotic syndrome in children. J Nephropathol 2017; 6:180-186. [PMID: 28975099 PMCID: PMC5607981 DOI: 10.15171/jnp.2017.31] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 01/20/2017] [Indexed: 12/23/2022] Open
Abstract
Background:
Development of steroid dependency is one of the difficult problems in the
management of children with idiopathic nephrotic syndrome, leading to increased
morbidity, complications and cost of treatment. Thus, predicting early in the disease
course will be useful in counseling parents and may improve treatment strategy.
Objectives:
To determine the clinical characteristics that can predict the development of
steroid dependency early in the initial episodes of steroid sensitive nephrotic syndrome
(SSNS).
Patients and Methods:
The study included 52 children with SSNS. Their ages ranged from
3 to 16 years. Patients were divided into two groups. Group A consisted of 24 patients
with steroid dependency or frequent relapses nephrotic syndrome and group B consisted
of 28 patients with complete remission or recurrent nephrotic syndrome. Data obtained
retrospectively from patients’ files.
Results:
Children who require a cumulative steroid dose equal or more than 140 mg/kg
to maintain remission during the first 6 months of the disease are at high risk to require
steroid sparing agents (SSA) for disease control, and who did not achieve remission by
day 20 of the initial prednisone course became steroid dependent with 96% specificity but
with low sensitivity (50%). All steroid dependent children in this study showed relapses
associated significantly with upper respiratory tract infections.
Conclusions:
Cumulative steroid dose in the first 6 months of treatment and the need of
more than 20 days to achieve initial remission can predict steroid dependency in children
with nephrotic syndrome.
Collapse
Affiliation(s)
| | | | - Adel Ali Erfan
- Pediatric Department, Faculty of Medicine, Tanta University, Egypt
| | | | - Hend Abdel-Nabi
- Pediatric Department, Faculty of Medicine, Tanta University, Egypt
| |
Collapse
|
15
|
Bennett J, Cassidy H, Slattery C, Ryan MP, McMorrow T. Tacrolimus Modulates TGF-β Signaling to Induce Epithelial-Mesenchymal Transition in Human Renal Proximal Tubule Epithelial Cells. J Clin Med 2016; 5:jcm5050050. [PMID: 27128949 PMCID: PMC4882479 DOI: 10.3390/jcm5050050] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 04/16/2016] [Accepted: 04/19/2016] [Indexed: 01/05/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT), a process which describes the trans-differentiation of epithelial cells into motile mesenchymal cells, is pivotal in stem cell behavior, development and wound healing, as well as contributing to disease processes including fibrosis and cancer progression. Maintenance immunosuppression with calcineurin inhibitors (CNIs) has become routine management for renal transplant patient, but unfortunately the nephrotoxicity of these drugs has been well documented. HK-2 cells were exposed to Tacrolimus (FK506) and EMT markers were assessed by RT PCR and western blot. FK506 effects on TGF-β mRNA were assessed by RT PCR and TGF-β secretion was measured by ELISA. The impact of increased TGF-β secretion on Smad signaling pathways was investigated. The impact of inhibition of TGF-β signaling on EMT processes was assessed by scratch-wound assay. The results presented in this study suggest that FK506 initiates EMT processes in the HK-2 cell line, with altered expression of epithelial and myofibroblast markers evident. Additionally, the study demonstrates that FK506 activation of the TGF-β/ SMAD pathways is an essential step in the EMT process. Overall the results demonstrate that EMT is heavily involved in renal fibrosis associated with CNI nephrotoxicity.
Collapse
Affiliation(s)
- Jason Bennett
- Centre for Cell Signaling and Inflammation, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK.
| | - Hilary Cassidy
- Renal Disease Research Group, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland.
| | - Craig Slattery
- Renal Disease Research Group, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland.
| | - Michael P Ryan
- Renal Disease Research Group, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland.
| | - Tara McMorrow
- Renal Disease Research Group, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland.
| |
Collapse
|
16
|
Rosner MH, Perazella MA, Choi MJ. American Society of Nephrology Quiz and Questionnaire 2015: Electrolytes and Acid-Base Disorders. Clin J Am Soc Nephrol 2016; 11:735-44. [PMID: 26825098 DOI: 10.2215/cjn.12801215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The Nephrology Quiz and Questionnaire remains an extremely popular session for attendees of the annual Kidney Week meeting of the American Society of Nephrology. During the 2015 meeting the conference hall was once again overflowing with eager quiz participants. Topics covered by the experts included electrolyte and acid-base disorders, glomerular disease, end-stage renal disease and dialysis, and kidney transplantation. Complex cases representing each of these categories together with single-best-answer questions were prepared and submitted by the panel of experts. Before the meeting, training program directors of nephrology fellowship programs and nephrology fellows in the United States answered the questions through an internet-based questionnaire. During the live session members of the audience tested their knowledge and judgment on the same series of case-oriented questions in a quiz. The audience compared their answers in real time using a cell-phone app containing the answers of the nephrology fellows and training program directors. The results of the online questionnaire were displayed, and then the quiz answers were discussed. As always, the audience, lecturers, and moderators enjoyed this highly educational session. This article recapitulates the session and reproduces selected content of educational value for theClinical Journal of the American Society of Nephrologyreaders. Enjoy the clinical cases and expert discussions.
Collapse
Affiliation(s)
- Mitchell H Rosner
- Division of Nephrology, University of Virginia Health System, Charlottesville, Virginia;
| | - Mark A Perazella
- Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut; and
| | - Michael J Choi
- Division of Nephrology, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
17
|
Manito N, Delgado JF, Crespo-Leiro MG, Arizón JM, Segovia J, González-Vílchez F, Mirabet S, Lage E, Pascual-Figal D, Díaz B, Palomo J, Rábago G, Sanz M, Blasco T, Roig E. Twelve-month efficacy and safety of the conversion to everolimus in maintenance heart transplant recipients. World J Transplant 2015; 5:310-319. [PMID: 26722659 PMCID: PMC4689942 DOI: 10.5500/wjt.v5.i4.310] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/31/2015] [Accepted: 10/13/2015] [Indexed: 02/05/2023] Open
Abstract
AIM: To determine the clinical reasons for conversion to everolimus (EVL) and long-term outcomes in heart transplant (HT) recipients.
METHODS: A retrospective 12-mo study has been carried out in 14 Spanish centres to assess the efficacy and safety of conversion to EVL in maintenance HT recipients.
RESULTS: Two hundred and twenty-two patients were included (mean age: 53 ± 10.5 years; mean time from HT: 8.1 ± 4.5 years). The most common reasons for conversion were nephrotoxicity (30%), chronic allograft vasculopathy (20%) and neoplasms (17%). The doses and mean levels of EVL at baseline (conversion to EVL) and after one year were 1.3 ± 0.3 and 1.2 ± 0.6 mg/d and 6.4 ± 3.4 and 5.6 ± 2.5 ng/mL, respectively. The percentage of patients receiving calcineurin inhibitors (CNIs) at baseline and on the final visit was 95% and 65%, respectively. The doses and mean levels of CNIs decreased between baseline and month 12 from 142.2 ± 51.6 to 98.0 ± 39.4 mg/d (P < 0.001) and from 126.1 ± 50.9 to 89.2 ± 47.7 ng/mL (P < 0.001), respectively, for cyclosporine, and from 2.9 ± 1.8 to 2.6 ± 1.9 mg/d and from 8.3 ± 4.0 to 6.5 ± 2.7 ng/mL (P = 0.011) for tacrolimus. In the subgroup of patients converted because of nephrotoxicity, creatinine clearance increased from 34.9 ± 10.1 to 40.4 ± 14.4 mL/min (P < 0.001). There were 37 episodes of acute rejection in 24 patients (11%). The most frequent adverse events were oedemas (12%), infections (9%) and gastrointestinal problems (6%). EVL was suspended in 44 patients (20%). Since the database was closed at the end of the study, no further follow-up data is available.
CONCLUSION: Conversion to EVL in maintenance HT recipients allowed minimisation or suspension of the CNIs, with improved kidney function in the patients with nephrotoxicity, after 12 mo.
Collapse
|
18
|
Yang X, Sherwin CMT, Yu T, Yellepeddi VK, Brunner HI, Vinks AA. Pharmacokinetic modeling of therapies for systemic lupus erythematosus. Expert Rev Clin Pharmacol 2015; 8:587-603. [PMID: 26143647 DOI: 10.1586/17512433.2015.1059751] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
With the increasing use of different types of therapies in treating autoimmune diseases such as systemic lupus erythematosus (SLE), there is a need to utilize pharmacokinetic (PK) strategies to optimize the clinical outcome of these treatments. Various PK analysis approaches, including population PK modeling and physiologically based PK modeling, have been used to evaluate drug PK characteristics and population variability or to predict drug PK profiles in a mechanistic manner. This review outlines the PK modeling of major SLE therapies including immunosuppressants (methotrexate, azathioprine, mycophenolate and cyclophosphamide, among others) and immunomodulators (intravenous immunoglobulin). It summarizes the population PK modeling, physiologically based PK modeling and model-based individualized dosing strategies to improve the therapeutic outcomes in SLE patients.
Collapse
Affiliation(s)
- Xiaoyan Yang
- a 1 Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | | | | | | | | | | |
Collapse
|
19
|
|
20
|
Janus N, Launay-Vacher V, Sebbag L, Despins P, Epailly E, Pavie A, Obadia JF, Pattier S, Varnous S, Pezzella V, Trillaud L, Deray G, Guillemain R. Renal insufficiency, mortality, and drug management in heart transplant. Results of the CARIN study. Transpl Int 2014; 27:931-8. [DOI: 10.1111/tri.12359] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 12/12/2013] [Accepted: 05/12/2014] [Indexed: 11/28/2022]
Affiliation(s)
- Nicolas Janus
- Service ICAR; Nephrology Department; Pitie-Salpetriere Hospital; Paris France
| | | | - Laurent Sebbag
- Heart Transplant Department; Louis Pradel Cardiology Hospital; Lyon France
| | - Philippe Despins
- Thoracic Transplant Department; North Hospital CHU Nantes; Nantes France
| | - Eric Epailly
- Cardiac Surgery Department; Nouvel Hôpital Civil; Strasbourg France
| | - Alain Pavie
- Thorac and Cardiovasculars Surgery Department; Pitie-Salpetriere Hospital; Paris France
| | | | - Sabine Pattier
- Thoracic Transplant Department; North Hospital CHU Nantes; Nantes France
| | - Shaïda Varnous
- Thorac and Cardiovasculars Surgery Department; Pitie-Salpetriere Hospital; Paris France
| | - Veronica Pezzella
- Thoracic Transplant Department; Georges Pompidou European Hospital; Paris France
| | - Laurence Trillaud
- Service ICAR; Nephrology Department; Pitie-Salpetriere Hospital; Paris France
| | - Gilbert Deray
- Nephrology Department; Pitie-Salpetriere Hospital; Paris France
| | - Romain Guillemain
- Thoracic Transplant Department; Georges Pompidou European Hospital; Paris France
| |
Collapse
|
21
|
Sereno J, Rodrigues-Santos P, Vala H, Rocha-Pereira P, Alves R, Fernandes J, Santos-Silva A, Carvalho E, Teixeira F, Reis F. Transition from cyclosporine-induced renal dysfunction to nephrotoxicity in an in vivo rat model. Int J Mol Sci 2014; 15:8979-97. [PMID: 24853130 PMCID: PMC4057770 DOI: 10.3390/ijms15058979] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 05/05/2014] [Accepted: 05/13/2014] [Indexed: 01/16/2023] Open
Abstract
Cyclosporin A (CsA), a calcineurin inhibitor, remain the cornerstone of immunosuppressive regimens, regardless of nephrotoxicity, which depends on the duration of drug exposure. The mechanisms and biomarkers underlying the transition from CsA-induced renal dysfunction to nephrotoxicity deserve better elucidation, and would help clinical decisions. This study aimed to clarify these issues, using a rat model of short- and long-term CsA (5 mg/kg bw/day) treatments (3 and 9 weeks, respectively). Renal function was assessed on serum and urine; kidney tissue was used for histopathological characterization and gene and/or protein expression of markers of proliferation, fibrosis and inflammation. In the short-term, creatinine and blood urea nitrogen (BUN) levels increased and clearances decreased, accompanied by glomerular filtration rate (GFR) reduction, but without kidney lesions; at that stage, CsA exposure induced proliferating cell nuclear antigen (PCNA), transforming growth factor beta 1 (TGF-β1), factor nuclear kappa B (NF-κβ) and Tumor Protein P53 (TP53) kidney mRNA up-regulation. In the long-term treatment, renal dysfunction data was accompanied by glomerular and tubulointerstitial lesions, with remarkable kidney mRNA up-regulation of the mammalian target of rapamycin (mTOR) and the antigen identified by monoclonal antibody Ki-67 (Mki67), accompanied by mTOR protein overexpression. Transition from CsA-induced renal dysfunction to nephrotoxicity is accompanied by modification of molecular mechanisms and biomarkers, being mTOR one of the key players for kidney lesion evolution, thus suggesting, by mean of molecular evidences, that early CsA replacement by mTOR inhibitors is indeed the better therapeutic choice to prevent chronic allograft nephropathy.
Collapse
Affiliation(s)
- José Sereno
- Laboratory of Pharmacology & Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal.
| | - Paulo Rodrigues-Santos
- Institute of Immunology, Faculty of Medicine, University of Coimbra, Coimbra 3004-504, Portugal.
| | - Helena Vala
- Agrarian School of Viseu (ESAV), Polytechnic Institute of Viseu, Viseu 3500-606, Portugal.
| | | | - Rui Alves
- University Nephrology Unit, Faculty of Medicine, University of Coimbra, Coimbra 3004-504, Portugal.
| | - João Fernandes
- Laboratory of Pharmacology & Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal.
| | - Alice Santos-Silva
- Biochemistry Department, Pharmacy Faculty, Porto University, Porto 4050-313, Portugal.
| | - Eugénia Carvalho
- Laboratory of Pharmacology & Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal.
| | - Frederico Teixeira
- Laboratory of Pharmacology & Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal.
| | - Flávio Reis
- Laboratory of Pharmacology & Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal.
| |
Collapse
|
22
|
Conversion to sirolimus ameliorates cyclosporine-induced nephropathy in the rat: focus on serum, urine, gene, and protein renal expression biomarkers. BIOMED RESEARCH INTERNATIONAL 2014; 2014:576929. [PMID: 24971338 PMCID: PMC4055143 DOI: 10.1155/2014/576929] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 04/17/2014] [Indexed: 02/06/2023]
Abstract
Protocols of conversion from cyclosporin A (CsA) to sirolimus (SRL) have been widely used in immunotherapy after transplantation to prevent CsA-induced nephropathy, but the molecular mechanisms underlying these protocols remain nuclear. This study aimed to identify the molecular pathways and putative biomarkers of CsA-to-SRL conversion in a rat model. Four animal groups (n = 6) were tested during 9 weeks: control, CsA, SRL, and conversion (CsA for 3 weeks followed by SRL for 6 weeks). Classical and emergent serum, urinary, and kidney tissue (gene and protein expression) markers were assessed. Renal lesions were analyzed in hematoxylin and eosin, periodic acid-Schiff, and Masson's trichrome stains. SRL-treated rats presented proteinuria and NGAL (serum and urinary) as the best markers of renal impairment. Short CsA treatment presented slight or even absent kidney lesions and TGF-β, NF-κβ, mTOR, PCNA, TP53, KIM-1, and CTGF as relevant gene and protein changes. Prolonged CsA exposure aggravated renal damage, without clear changes on the traditional markers, but with changes in serums TGF-β and IL-7, TBARs clearance, and kidney TGF-β and mTOR. Conversion to SRL prevented CsA-induced renal damage evolution (absent/mild grade lesions), while NGAL (serum versus urine) seems to be a feasible biomarker of CsA replacement to SRL.
Collapse
|
23
|
Domen J, Li Y, Sun L, Simpson P, Gandy K. Rapid tolerance induction by hematopoietic progenitor cells in the absence of donor-matched lymphoid cells. Transpl Immunol 2014; 31:112-8. [PMID: 24794050 DOI: 10.1016/j.trim.2014.04.001] [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: 12/31/2013] [Accepted: 04/03/2014] [Indexed: 01/25/2023]
Abstract
BACKGROUND Donor specific hematopoietic cell transplantation has long been recognized for its potential in tolerance induction for subsequently transplanted organs. We have recently published that co-administration of Myeloid Progenitor (MP) and third party Hematopoietic Stem Cells (HSC) can induce MP-specific tolerance for subsequently transplanted organs [1]. METHODS Mice received an allogeneic HSC and third party MP transplantation simultaneous with placement of a MP-matched skin graft. Variants tested include time of graft placement, MP genotype and source of cells. RESULTS Using B10;B6-Rag2(-/-)Il2rg(-/-) mice, we demonstrate that specific tolerance can be induced by MP given simultaneous with the skin graft in the complete absence of MP-donor-matched lymphoid cells. Ex vivo expanded MP function as well as sorted cells in inducing tolerance. In addition we demonstrate that tolerance can be induced by MP in the context of autologous HSC transplantation. CONCLUSIONS Our results demonstrate that the previously observed expansion of organ donor matched Treg is not essential for tolerance, and that MP tolerance protocols can be envisioned in most clinical settings, including those involving deceased donor organs.
Collapse
Affiliation(s)
- Jos Domen
- Section of Cardiac Surgery, Children's Mercy Hospital and Clinics, Kansas City MO64108, United States; Department of Pediatrics, University of Missouri Kansas City, Kansas City, MO, United States.
| | - Yongwu Li
- Section of Cardiac Surgery, Children's Mercy Hospital and Clinics, Kansas City MO64108, United States.
| | - Lei Sun
- Section of Cardiac Surgery, Children's Mercy Hospital and Clinics, Kansas City MO64108, United States.
| | - Pippa Simpson
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.
| | - Kimberly Gandy
- Section of Cardiac Surgery, Children's Mercy Hospital and Clinics, Kansas City MO64108, United States; Department of Pediatrics, University of Missouri Kansas City, Kansas City, MO, United States; Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.
| |
Collapse
|
24
|
Serum and renal tissue markers of nephropathy in rats under immunosuppressive therapy: cyclosporine versus sirolimus. Transplant Proc 2013; 45:1149-56. [PMID: 23622648 DOI: 10.1016/j.transproceed.2013.02.085] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cyclosporin (CsA) has been progressively replaced by other drugs with putatively fever side effects, including nephrotoxicity and hypertension. Sirolimus (SRL) is one of the main options for management of kidney transplant patients in the post-CsA era. It shows identical efficacy with apparently less cardiorenal side effects than CsA. However, doubts remain concerning the mechanisms of putative renoprotection by SRL as well as the best serum and/or tissue markers for nephropathy, as assessed in this study employing CsA- and SRL-treated rats. Three groups (n = 6) were treated orally during a 6-week protocol: control (vehicle); CsA (5 mg/kg body weight per day Sandimmun Neoral); SRL (1 mg/kg body weight per day Rapamune). Blood pressure and heart rate were assessed with a "tail cuff". Renal dysfunction and morphology were characterized using serum creatinine and blood urea nitrogen (BUN) levels as well as hematoxylin and eosin and periodic acid Schiff staining, respectively. We examined serum concentrations of interleukin (IL)-2, IL-1β, high-sensitivity C-reactive protein, tumor necrosis factor TNF-α, and vascular endothelial growth factor and kidney mRNA expression of interleukin-1β (IL-1β), tumor protein 53 (TP53), mammalian target of rapamycin (mTOR) and proliferating cell nuclear antigen (PCNA), as well as markers of lipid peroxidation in the kidney and serum. Both CsA and SRL induced significant increases in systolic and diastolic blood pressure, but only CsA caused tachycardia. CsA-treated rats also displayed increased serum creatinine and BUN levels, accompanied by mild renal lesions, which were almost absent among SRL-treated rats, which presented hyperlipidemic and hyperglycemic profiles. CsA-induced nephrotoxicity was accompanied by kidney overexpression of inflammatory and proliferative mRNA markers (IL-1β, mTOR and PCNA), which were absent among SRL group. In conclusion, the antiproliferative and antifibrotic character of SRL may explain its less nephrotoxic profile. Renal over expression of mTOR in the CsA-treated group, associated with renal dysfunction and structural damage, reinforces the potential beneft of SRL as a strategy to reduce CsA-evoked nephrotoxicity.
Collapse
|
25
|
Jafari A, Dashti-Khavidaki S, Khalili H, Lessan-Pezeshki M. Potential nephroprotective effects of l-carnitine against drug-induced nephropathy: a review of literature. Expert Opin Drug Saf 2013; 12:523-43. [PMID: 23656498 DOI: 10.1517/14740338.2013.794217] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Drug-induced nephrotoxicity (DIN) has been reported with a great number of medications and contributes to ∼ 20% of hospital admissions. l-carnitine owing to its antioxidant, anti-inflammatory and antiapoptotic properties has been proposed as a candidate for nephroprotection against DIN. Increasing need to use nephrotoxic therapeutic agents necessitated this review. AREAS COVERED The present review covers all published clinical and animal researches on nephroprotective effects of l-carnitine against DIN. l-carnitine significantly ameliorates DIN in animal studies especially against cisplatin-induced renal damage. Inhibition of reactive oxygen species generation, lipid peroxidation, matrix remodeling and apoptosis, anti-inflammatory properties and improvement in carnitine deficiency has been suggested as probable nephroprotective mechanisms of l-carnitine. EXPERT OPINION In spite of the evidences that support the nephroprotective effect of l-carnitine, the main problems in this area are inadequacy of reliable studies in humans and difficulty of translating the experimental results into clinical practice. In most of the described studies, l-carnitine treatment is prophylactically given. Use of l-carnitine as a prophylactic agent in clinical situations with an indication for nephrotoxic therapies is rarely possible except for contrast-induced nephrotoxicity. Development of validated early biomarkers to detect DIN may provide the opportunity to use prophylactic nephroprotective agents at golden time.
Collapse
Affiliation(s)
- Atefeh Jafari
- Tehran University of Medical Science, Resident of Clinical Pharmacy, Faculty of Pharmacy, Tehran, Iran
| | | | | | | |
Collapse
|
26
|
Dillmann J, Popp FC, Fillenberg B, Zeman F, Eggenhofer E, Farkas S, Scherer MN, Koller M, Geissler EK, Deans R, Ladenheim D, Loss M, Schlitt HJ, Dahlke MH. Treatment-emergent adverse events after infusion of adherent stem cells: the MiSOT-I score for solid organ transplantation. Trials 2012; 13:211. [PMID: 23151227 PMCID: PMC3543274 DOI: 10.1186/1745-6215-13-211] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 10/31/2012] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Cellular therapy after organ transplantation is emerging as an intriguing strategy to achieve dose reduction of classical immunosuppressive pharmacotherapy. Here, we introduce a new scoring system to assess treatment-emergent adverse events (TEAEs) of adherent stem cell therapies in the clinical setting of allogeneic liver transplantation (for example, the MiSOT-I trial Eudract CT: 2009-017795-25). METHODS The score consists of three independent modalities (set of parameters) that focus on clinically relevant events early after intravenous or intraportal stem cell infusion: pulmonary toxicity, intraportal-infusional toxicity and systemic toxicity. For each modality, values between 0 (no TEAE) and 3 (severe TEAE) were defined. The score was validated retrospectively on a cohort of n=187 recipients of liver allografts not receiving investigational cell therapy between July 2004 and December 2010. These patients represent a control population for further trials. Score values were calculated for days 1, 4, and 10 after liver transplantation. RESULTS Grade 3 events were most commonly related to the pulmonary system (3.5% of study cohort on day 4). Almost no systemic-related TEAEs were observed during the study period. The relative frequency of grade 3 events never exceeded 5% over all modalities and time points. A subgroup analysis for grade 3 patients provided no descriptors associated with severe TEAEs. CONCLUSION The MiSOT-I score provides an assessment tool to score specific adverse events that may occur after adherent stem cell therapy in the clinical setting of organ transplantation and is thus a helpful tool to conduct a safety study.
Collapse
Affiliation(s)
- Johannes Dillmann
- Department of Surgery, University Hospital Regensburg, Franz Josef Strauss Allee 11, 93053, Regensburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Hershko K, Simhadri VL, Blaisdell A, Hunt RC, Newell J, Tseng SC, Hershko AY, Choi JW, Sauna ZE, Wu A, Bram RJ, Komar AA, Kimchi-Sarfaty C. Cyclosporin A impairs the secretion and activity of ADAMTS13 (a disintegrin and metalloprotease with thrombospondin type 1 repeat). J Biol Chem 2012; 287:44361-71. [PMID: 23144461 DOI: 10.1074/jbc.m112.383968] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The protease ADAMTS13 (a disintegrin and metalloprotease with thrombospondin type 1 repeat) cleaves multimers of von Willebrand factor, thus regulating platelet aggregation. ADAMTS13 deficiency leads to the fatal disorder thrombotic thrombocytopenic purpura (TTP). It has been observed that cyclosporin A (CsA) treatment, particularly in transplant patients, may sometimes be linked to the development of TTP. Until now, the reason for such a link was unclear. Here we provide evidence demonstrating that cyclophilin B (CypB) activity plays an important role in the secretion of active ADAMTS13. We found that CsA, an inhibitor of CypB, reduces the secretion of ADAMTS13 and leads to conformational changes in the protein resulting in diminished ADAMTS13 proteolytic activity. A direct, functional interaction between CypB (which possesses peptidyl-prolyl cis-trans isomerase (PPIase) and chaperone functions) and ADAMTS13 is demonstrated using immunoprecipitation and siRNA knockdown of CypB. Finally, CypB knock-out mice were found to have reduced ADAMTS13 levels. Taken together, our findings indicate that cyclophilin-mediated activity is an important factor affecting secretion and activity of ADAMTS13. The large number of proline residues in ADAMTS13 is consistent with the important role of cis-trans isomerization in the proper folding of this protein. These results altogether provide a novel mechanistic explanation for CsA-induced TTP in transplant patients.
Collapse
Affiliation(s)
- Klilah Hershko
- Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland 20982, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Sagiroglu T, Kanter M, Yagci MA, Sezer A, Erboga M. Protective effect of curcumin on cyclosporin A-induced endothelial dysfunction, antioxidant capacity, and oxidative damage. Toxicol Ind Health 2012; 30:316-27. [PMID: 22903178 DOI: 10.1177/0748233712456065] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cyclosporin A (CsA) is the most widely used immunosuppressive drug for preventing graft rejection and autoimmune disease. However, the therapeutic treatment induces several side effects such as nephrotoxicity, cardiotoxicity, hypertension, and hepatotoxicity. Curcumin has been successfully used as a potent antioxidant against many pathophysiological states. This experimental study was performed to test, during CsA treatment, the alterations of curcumin antioxidant properties against CsA-induced endothelial dysfunction. Rats were divided into four groups: control, curcumin alone, CsA, and CsA + curcumin; each group containing eight animals. The animals in the CsA + curcumin group were treated with CsA (10 days, 25 mg/kg, orally) and curcumin (15 days, 200 mg/kg, orally, starting 5 days before CsA administration). At the end of the treatments, the animals were killed; serum and aorta tissue were treated for biochemical and morphological analyses. The results indicate that CsA-induced aortic endothelial dysfunction was characterized by morphological and ultrastructural alterations in tissue architecture, changes in malondialdehyde and ferric reducing/antioxidant power levels, and increase in endothelial nitric oxide synthase and terminal-deoxynucleotidyl-transferase mediated dUTP nick end labeling (TUNEL) expression. In conclusion, our data suggest that the imbalance between production of free oxygen radicals and antioxidant defence systems, due to CsA administration, is a mechanism responsible for oxidative stress. Moreover, we show that curcumin plays a protective action against CsA-induced endothelial dysfunction and oxidative stress, as supported by biochemical, ultrastructural, immunohistochemical, and TUNEL results.
Collapse
Affiliation(s)
- Tamer Sagiroglu
- 1Department of General Surgery, Faculty of Medicine, Trakya University, Edirne, Turkey
| | | | | | | | | |
Collapse
|
29
|
Denecke C, Reutzel-Selke A, Sawitzki B, Boenisch O, Khalpey Z, Seifert M, Pratschke J, Volk HD, Tullius SG. Low-dose cyclosporine mediates donor hyporesponsiveness in a fully mismatched rat kidney transplant model. Transpl Immunol 2012; 26:176-85. [PMID: 22414756 DOI: 10.1016/j.trim.2012.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 02/23/2012] [Accepted: 02/27/2012] [Indexed: 12/11/2022]
Abstract
Tolerance induction protocols have been successfully tested in animal models, yet their compatibility with immunosuppressive drugs remains to be fully elucidated. Our own previous data have indicated that cyclosporine A (CsA) affects the balance of effector and regulatory mechanisms with low-dose CsA doses promoting hyporesponsiveness. Here, we present a fully mismatched rat kidney model in which low-dose CsA treatment induces donor hyporesponsiveness to secondary renal allografts. Lewis recipients of DA kidney grafts received low, medium or high doses of CsA × 10 days. By 30 days, the primary transplant was removed and a second transplant of donor origin was engrafted. Following low-dose CsA, but not high-dose CsA treatment of the primary recipient, secondary transplants were accepted long-term in the absence of immunosuppression. Regulatory T-cell function was unimpaired and independent of the CyA dosage. Of note, low-dose CsA significantly reduced alloantibody titers in primary recipients. Adoptive transfer of graft infiltrating cells or splenocytes from hyporesponsive recipients supported long-term acceptance of donor kidney allografts. These results demonstrate a dose-dependent and transferable "pro-tolerogenic" effect of low-dose CsA treatment. This model is of clinical relevance to test the interference of CsA with tolerance induction in the absence of additional immunosuppression.
Collapse
Affiliation(s)
- Christian Denecke
- Department of Visceral, Transplantation and Thoracic Surgery, Medizinische Universitaet Innsbruck, Austria; Division of Transplant Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
O’Connell S, Tuite N, Slattery C, Ryan MP, McMorrow T. Cyclosporine A–Induced Oxidative Stress in Human Renal Mesangial Cells: A Role for ERK 1/2 MAPK Signaling. Toxicol Sci 2011; 126:101-13. [DOI: 10.1093/toxsci/kfr330] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
31
|
Popp FC, Fillenberg B, Eggenhofer E, Renner P, Dillmann J, Benseler V, Schnitzbauer AA, Hutchinson J, Deans R, Ladenheim D, Graveen CA, Zeman F, Koller M, Hoogduijn MJ, Geissler EK, Schlitt HJ, Dahlke MH. Safety and feasibility of third-party multipotent adult progenitor cells for immunomodulation therapy after liver transplantation--a phase I study (MISOT-I). J Transl Med 2011; 9:124. [PMID: 21798013 PMCID: PMC3166276 DOI: 10.1186/1479-5876-9-124] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 07/28/2011] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Liver transplantation is the definitive treatment for many end-stage liver diseases. However, the life-long immunosuppression needed to prevent graft rejection causes clinically significant side effects. Cellular immunomodulatory therapies may allow the dose of immunosuppressive drugs to be reduced. In the current protocol, we propose to complement immunosuppressive pharmacotherapy with third-party multipotent adult progenitor cells (MAPCs), a culture-selected population of adult adherent stem cells derived from bone marrow that has been shown to display potent immunomodulatory and regenerative properties. In animal models, MAPCs reduce the need for pharmacological immunosuppression after experimental solid organ transplantation and regenerate damaged organs. METHODS Patients enrolled in this phase I, single-arm, single-center safety and feasibility study (n = 3-24) will receive 2 doses of third-party MAPCs after liver transplantation, on days 1 and 3, in addition to a calcineurin-inhibitor-free "bottom-up" immunosuppressive regimen with basiliximab, mycophenolic acid, and steroids. The study objective is to evaluate the safety and clinical feasibility of MAPC administration in this patient cohort. The primary endpoint of the study is safety, assessed by standardized dose-limiting toxicity events. One secondary endpoint is the time until first biopsy-proven acute rejection, in order to collect first evidence of efficacy. Dose escalation (150, 300, 450, and 600 million MAPCs) will be done according to a 3 + 3 classical escalation design (4 groups of 3-6 patients each). DISCUSSION If MAPCs are safe for patients undergoing liver transplantation in this study, a phase II/III trial will be conducted to assess their clinical efficacy.
Collapse
Affiliation(s)
- Felix C Popp
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
STUDY DESIGN Cell transplantation strategies are gaining increasing interest for spinal cord injury (SCI) with the objective of promoting spinal cord repair. To avoid allogenic graft rejection, an adequate immune suppression is required, and one of the most potent and commonly used immunosuppressives is cyclosporin A (CsA). In SCI, permanent sensory motor loss is combined with modifications of drug absorption, distribution and elimination. OBJECTIVES The objectives of this study were to thoroughly explore histological and functional outcomes of CsA treatment in a rat model of spinal cord compression. SETTING Experiments were carried out at the Institute for Neurosciences of Montpellier (France), the Integrative Biology of Neurodegeneration Laboratory (Spain) and in the Novartis Institutes for BioMedical Research (Switzerland) for CsA blood concentration determination. METHODS We first evaluated histological outcomes of CsA treatment on kidneys and spinal cord after SCI. We then investigated whether SCI modified CsA blood concentration. Finally, using behavioral analysis, we assessed the potential CsA impact on functional recovery. RESULTS When spinal-cord-injured rats were treated with a CsA dose of 10 mg kg(-1) per day, we observed deleterious effects on kidneys, associated with modifications of CsA blood concentration. Adding an antibiotic treatment reduced kidney alteration without modifying CsA blood concentration. Finally, we showed that CsA treatment per se modified neither functional recovery nor lesion extension. CONCLUSION This study pinpoints the absolute requirement of careful CsA monitoring in the clinical setting for patients with SCI to minimize potential unexpected effects and avoid therapeutic failure.
Collapse
|
33
|
Sobel DO, Henzke A, Abbassi V. Cyclosporin and methotrexate therapy induces remission in type 1 diabetes mellitus. Acta Diabetol 2010; 47:243-50. [PMID: 20440520 DOI: 10.1007/s00592-010-0188-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Accepted: 03/28/2010] [Indexed: 01/30/2023]
Abstract
Cyclosporin and methotrexate administration induces remission of type 1 diabetes mellitus. Administration of high-dose cyclosporin (cyclo) has been demonstrated to induce remission of type 1 diabetes mellitus (T1D). Its usefulness was limited by its toxicity. Since methotrexate (mtx) and cyclo synergistically inhibit autoimmune processes, we postulated that low doses of cyclo and mtx could safely induce remission of T1D. In a pilot study, insulin dose requirements and glycemic control were compared in 10 new onset T1D control children with seven children who were administered cyclo at 7.5 mg/kg/day for 6 weeks and then 4 mg/kg/day in addition to mtx 5 mg/kg/wk for 1 year. After 6 weeks, cyclo doses were adjusted to maintain blood cyclo levels 110-220 ng/ml. All children were treated with two daily injections of insulin. Clinical and biochemical toxicity of drug therapy was assessed. There were only very minor adverse effects and no drug induced biochemical test abnormalities. Mean HbA1c levels were similar in the experimental and control groups at baseline and at 3, 6, and 9 months but was lower in the cyclo + mtx group at 12 months. Daily insulin requirements of the groups were similar at baseline but lower in the cyclo + mtx group at 3, 6, 9, and 12 months. Although no control subjects became non-insulin requiring, four of seven cyclo + mtx-treated subjects were entirely off insulin therapy for 2.5, 4.5, 8, and 12 months. Low-dose cyclo and mtx treatment of subjects with new onset T1D can safely induce remission of disease and decrease the amount of required insulin.
Collapse
Affiliation(s)
- Douglas O Sobel
- Department of Pediatrics, Georgetown University Medical Center, Washington, DC 20007, USA.
| | | | | |
Collapse
|
34
|
Recovery of Renal Function in Heart Transplantation Patients After Conversion From a Calcineurin Inhibitor-Based Therapy to Sirolimus. Transplant Proc 2010; 42:542-4. [DOI: 10.1016/j.transproceed.2010.01.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
35
|
van der Windt AE, Jahr H, Farrell E, Verhaar JAN, Weinans H, van Osch GJVM. Calcineurin inhibitors promote chondrogenic marker expression of dedifferentiated human adult chondrocytes via stimulation of endogenous TGFbeta1 production. Tissue Eng Part A 2010; 16:1-10. [PMID: 19604038 DOI: 10.1089/ten.tea.2009.0082] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In vitro chondrocyte expansion is required for several cell-based approaches for the repair of chondral lesions. During expansion, loss of chondrogenic phenotype takes place (dedifferentiation). The objective of this study was to investigate calcineurin (Cn) as a potential target to improve chondrocyte phenotype for cartilage repair purposes. Cn activity in human articular chondrocytes was significantly increased during dedifferentiation and decreased during redifferentiation in vitro. Inhibition of Cn activity by FK506 increased the expression of chondrogenic markers collagen type 2, aggrecan, and SOX9 in culture-expanded cells. Addition of FK506 increased endogenous transforming growth factor 2 (TGF) beta1 expression on both mRNA and protein level. The effect of FK506 on chondrogenic markers was abolished by addition of anti-TGFbeta1 antibody, indicating that the endogenous TGFbeta1 was necessary to increase chondrogenic marker expression. We also showed that chondrocyte redifferentiation by TGFbeta requires calcium influx and does not depend on changes in Cn activity. In conclusion, inhibition of Cn activity by FK506 increases the expression of chondrogenic markers via endogenous TGFbeta1 production in human articular chondrocytes. Cn inhibitors might be an alternative for the application of (recombinant) TGFbeta, to promote chondrocyte phenotype for cell-based cartilage repair procedures.
Collapse
Affiliation(s)
- Anna E van der Windt
- Department of Orthopaedics, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
36
|
Mazzeo AT, Brophy GM, Gilman CB, Alves ÓL, Robles JR, Hayes RL, Povlishock JT, Bullock MR. Safety and tolerability of cyclosporin a in severe traumatic brain injury patients: results from a prospective randomized trial. J Neurotrauma 2009; 26:2195-206. [PMID: 19621985 PMCID: PMC2824218 DOI: 10.1089/neu.2009.1012] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cyclosporin A (CsA) has recently been proposed for use in the early phase after traumatic brain injury (TBI), for its ability to preserve mitochondrial integrity in experimental brain injury models, and thereby provide improved behavioral outcomes as well as significant histological protection. The aim of this prospective, randomized, double-blind, dual-center, placebo-controlled trial was to evaluate the safety, tolerability, and pharmacokinetics of a single intravenous infusion of CsA in patients with severe TBI. Fifty adult severe TBI patients were enrolled over a 22-month period. Within 12 h of the injury patients received 5 mg/kg of CsA infused over 24 h, or placebo. Blood urea nitrogen (BUN), creatinine, hemoglobin, platelets, white blood cell count (WBC), and a hepatic panel were monitored on admission, and at 12, 24, 36, and 48 h, and on days 4 and 7. Potential adverse events (AEs) were also recorded. Neurological outcome was recorded at 3 and 6 months after injury. This study revealed only transient differences in BUN levels at 24 and 48 h and for WBC counts at 24 h between the CsA and placebo patients. These modest differences were not clinically significant in that they did not negatively impact on patient course. Both BUN and creatinine values, markers of renal function, remained within their normal limits over the entire monitoring period. There were no significant differences in other mean laboratory values, or in the incidence of AEs at any other measured time point. Also, no significant difference was demonstrated for neurological outcome. Based on these results, we report a good safety profile of CsA infusion when given at the chosen dose of 5 mg/kg, infused over 24 h, during the early phase after severe head injury in humans, with the aim of neuroprotection.
Collapse
Affiliation(s)
- Anna Teresa Mazzeo
- Department of Neurosciences, Psychiatric and Anesthesiological Sciences, University of Messina, Messina, Italy
| | - Gretchen M. Brophy
- Department of Pharmacy and Neurosurgery, Virginia Commonwealth University, Richmond, Virginia
| | - Charlotte B. Gilman
- Division of Neurosurgery, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia
| | - Óscar Luís Alves
- Serviço de Neurocirurgia, Centro Hospitalar de Vila Nova de Gaia, Faculdade de Medicina da Universitade do Porto, Porto, Portugal
| | - Jaime R. Robles
- Department of Pharmacy, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Ronald L. Hayes
- Department of Neurosurgery, University of Florida, Center of Innovative Research, Banyan Biomarkers, Inc., Alachua, Florida
| | - John T. Povlishock
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia
| | - M. Ross Bullock
- Department of Neurosurgery, University of Miami, Miller School of Medicine, Lois Pope LIFE Center, Miami, Florida
| |
Collapse
|
37
|
|
38
|
McGuire BM, Rosenthal P, Brown CC, Busch AMH, Calcatera SM, Claria RS, Hunt NK, Korenblat KM, Mazariegos GV, Moonka D, Orloff SL, Perry DK, Rosen CB, Scott DL, Sudan DL. Long-term management of the liver transplant patient: recommendations for the primary care doctor. Am J Transplant 2009; 9:1988-2003. [PMID: 19563332 DOI: 10.1111/j.1600-6143.2009.02733.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
No official document has been published for primary care physicians regarding the management of liver transplant patients. With no official source of reference, primary care physicians often question their care of these patients. The following guidelines have been approved by the American Society of Transplantation and represent the position of the association. The data presented are based on formal review and analysis of published literature in the field and the clinical experience of the authors. These guidelines address drug interactions and side effects of immunosuppressive agents, allograft dysfunction, renal dysfunction, metabolic disorders, preventive medicine, malignancies, disability and productivity in the workforce, issues specific to pregnancy and sexual function, and pediatric patient concerns. These guidelines are intended to provide a bridge between transplant centers and primary care physicians in the long-term management of the liver transplant patient.
Collapse
Affiliation(s)
- B M McGuire
- University of Alabama at Birmingham, Birmingham, AL, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
It has recently been recognized that mesenchymal stem cells (MSCs) isolated from adult bone marrow are able to modify the alloimmune response in vitro and in vivo. MSCs can be expanded into large quantities in culture, thereby facilitating potential future applications in solid organ transplantation. To develop novel MSC-based antirejection treatments, the mechanism behind the immunomodulatory ability of MSCs has to be elucidated further. At present, a variety of possible in vitro effects of MSCs on immune system effector cells have been reported, but little is known about their in vivo properties. Here, we discuss recent findings regarding the influence of MSCs on different effector cell populations in vitro and summarize the available data describing their in vivo properties.
Collapse
|
40
|
Cologna AJ, Lima LVDS, Tucci S, Suaid HJ, Reis RB, Tirapelli LF, Rodrigues AA, Martins ACP. Cyclosporine action on kidneys of rats submitted to normothermic ischaemia and reperfusion. Acta Cir Bras 2009; 23 Suppl 1:36-41; discussion 41. [PMID: 18516446 DOI: 10.1590/s0102-86502008000700007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
PURPOSE To verify if rat kidneys lesioned by ischaemia followed by reperfusion are affected by cyclosporine A (CsA). METHODS Male Wistar rats were randomly divided into three groups, control (GS) and experimental (G1 and G2). G1 was subdivided in two: G1A composed of animals submitted to 60 minutes ischaemia and G1C with the same ischaemic procedure associated to 20 mg/kg/day CsA. Group G2 was subdivided and treated in the same way as G1 except that ischaemia was applied only for 40 minutes. Clamping the left renal artery followed by right side nephrectomy induced kidney ischaemia. Serum urea and creatinine were quantified on the day of surgery (D0) and in the following day (D1). Twenty four hours after reperfusion the left kidney was removed and histologically analyzed. RESULTS Group GS had normal values for urea and creatinine both on D0 and D1 and did not show structural alterations. Renal function was not significantly different when G2C was compared to GS (p>0.05). Tissue lesions were smaller in G2C than in the other groups. CONCLUSIONS Renal function was protected by CsA, which also reduced tissue lesions in the kidneys of rats submitted to 40 minutes ischaemia.
Collapse
Affiliation(s)
- Adauto José Cologna
- Department of Surgery and Anatomy, Ribeirão Preto Faculty of Medicine, University of São Paulo, SP, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Cattaneo D, Ruggenenti P, Baldelli S, Motterlini N, Gotti E, Sandrini S, Salvadori M, Segoloni G, Rigotti P, Donati D, Perico N, Remuzzi G. ABCB1 genotypes predict cyclosporine-related adverse events and kidney allograft outcome. J Am Soc Nephrol 2009; 20:1404-1415. [PMID: 19470683 PMCID: PMC2689900 DOI: 10.1681/asn.2008080819] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Accepted: 01/14/2009] [Indexed: 01/11/2023] Open
Abstract
Cyclosporine A (CsA) is a substrate of P-glycoprotein, an efflux transporter encoded by the ABCB1 gene. Compared with carriers of the wild-type gene, carriers of T allelic variants in exons 21 or 26 have reduced P-glycoprotein activity and, secondarily, increased intracellular concentration of CsA; therefore, carriers of T variants might be at increased risk for CsA-related adverse events. We evaluated the associations between ABCB1 genotypes (in exons 12, 21, and 26) and CsA-related outcomes in 147 renal transplant recipients who were receiving CsA-based immunosuppression and were included in the Mycophenolate Steroids Sparing study. During a median of 65.5 mo follow-up, carriers of T allelic variants in exons 21 or 26 had a three-fold risk for delayed graft function (DGF), a trend to slower recovery of renal function and lower GFR at study end, and significantly higher incidences of new-onset diabetes and cytomegalovirus reactivation compared with carriers of the wild-type genotype. T variants in both exons 21 and 26 were independently associated with 3.8- and 3.5-fold higher risk for DGF, respectively (P = 0.022 and P = 0.034). The incidence of acute rejection and the mean CsA dose and blood levels were comparable in genotype groups. In conclusion, renal transplant recipients with T allelic variants in ABCB1 exons 21 or 26 are at increased risk for CsA-related adverse events. Genetic evaluation may help to identify patients at risk and to modulate CsA therapy to optimize graft and patient outcomes.
Collapse
Affiliation(s)
- Dario Cattaneo
- Department of Medicine and Transplantation, Ospedali Riuniti-Mario Negri Institute for Pharmacological Research, Bergamo, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Puigmulé M, López-Hellin J, Suñé G, Tornavaca O, Camaño S, Tejedor A, Meseguer A. Differential proteomic analysis of cyclosporine A-induced toxicity in renal proximal tubule cells. Nephrol Dial Transplant 2009; 24:2672-86. [PMID: 19369687 DOI: 10.1093/ndt/gfp149] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The use of cyclosporine A (CsA) as a potent immunosuppressant has been limited by its severe nephrotoxic effects. The mechanisms involved are haemodynamic but also related to direct toxic effects of CsA on proximal tubule epithelial cells. We focused on defining a proteomic profile in CsA-treated proximal tubule cells to distinguish the direct impact of CsA on these cells from overlapping haemodynamically mediated phenomena that occur in an in vivo system. METHODS By means of high-throughput differential proteomic analyses and mass spectrometry techniques in CsA and vehicle-treated proximal tubule-derived cell lines of human and mouse origin, we determined proteins that change their expression in the presence of CsA. RESULTS CsA-induced toxicity analyses revealed that 10 mM CsA for 24 h was the threshold condition to induce significant changes in cell viability and proteomic profile. We identified 38 differentially expressed proteins on CsA-treated mouse PCT3 and human HK-2 cells, related to protein metabolism, response to damage, cell organization and cytoskeleton, energy metabolism, cell cycle and nucleobase/nucleoside/nucleotidic metabolism. 1D and 2D western blot assays in crude extracts from CsA-treated cells or kidneys with impaired function upon CsA treatment revealed a correlation with proteomic changes or differential isoform expression, in randomly selected proteins. CONCLUSIONS Proteins identified in this work might be useful markers to eventually distinguish CsA toxicity from chronic allograft nephropathy in protocol biopsies of transplanted patients, facilitating the adjustment of CsA doses to non-toxic ranges, as well as to study the impact of potential therapeutic interventions in an animal model.
Collapse
Affiliation(s)
- Marta Puigmulé
- Institut de Recerca Hospital Universitari Vall d'Hebron, Centre d'Investigacions en Bioquimica i Biologia Molecular, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Prediction of high-degree steroid dependency in pediatric idiopathic nephrotic syndrome. Pediatr Nephrol 2008; 23:2221-6. [PMID: 18618150 DOI: 10.1007/s00467-008-0914-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2008] [Revised: 05/05/2008] [Accepted: 05/23/2008] [Indexed: 10/21/2022]
Abstract
Most patients with idiopathic nephrotic syndrome are steroid-responsive, but about 50% relapse and often become steroid-dependent and exposed to long-term steroid complications. The aim of this study was to determine predictive risk factors for steroid and/or cyclosporine A (CyA) dependence. In France, steroid responsiveness is defined as remission after 1 month of oral prednisone (60 mg/m(2) per day) and-in the case of persistent proteinuria on day 30-three methylprednisolone pulses (MPP; 1 g/1.73 m(2) on days 1, 3, and 5). Thirty-five steroid-responsive children, followed between 1999 and 2006, were included in this study. Median age at diagnosis was 4.9 years. All patients initially received prednisone 60 mg/m(2) per day. Twenty-four of the 35 patients were steroid-dependent, with 12 requiring MPP. Of the latter 12 patients, 83.3% were treated with CyA during follow-up; in comparison, only 16.7% of the patients who did not receive MPP required CyA during follow-up (chi-square test, P = 0.001). T risk for steroid dependence was 100% in our cohort if remission was achieved after day 20. Patients who need MPP are at high risk to require CyA to achieve disease control. By identifying these children, we could use adequate immunosuppressive drugs earlier and reduce morbidity related to steroids and multiple relapses.
Collapse
|
45
|
An underlying role for hepatobiliary dysfunction in cyclosporine A nephrotoxicity. Toxicol Appl Pharmacol 2008; 230:126-34. [DOI: 10.1016/j.taap.2008.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Revised: 02/01/2008] [Accepted: 02/06/2008] [Indexed: 11/19/2022]
|
46
|
Josephine A, Amudha G, Veena CK, Preetha SP, Rajeswari A, Varalakshmi P. Beneficial effects of sulfated polysaccharides from Sargassum wightii against mitochondrial alterations induced by Cyclosporine A in rat kidney. Mol Nutr Food Res 2007; 51:1413-22. [PMID: 17918168 DOI: 10.1002/mnfr.200700127] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Sulfated polysaccharides from marine seaweeds are receiving continuous attention owing to their wide therapeutic applications and are known to inhibit free radical generation. It has been well known that mitochondria are the major sources as well as the target of free radicals. The renal tubules have high density of mitochondria and therefore show structural and functional defects in acute renal failure. Hence, the present study is designed to appraise the mitochondrial status during Cyclosporine A (CsA)-induced nephrotoxicity and the effect of sulfated polysaccharides over it. Sulfated polysaccharides (5 mg/kg body weight, subcutaneously) treatment significantly prevented the CsA-induced (25 mg/kg body weight, orally) mitochondrial damage. CsA-induced mitochondrial oxidative stress in rat kidney was evident from increased reactive oxygen species level, decreased antioxidant defense system, coupled with enhanced lipid peroxidation. Further, the activities of tricarboxylic acid cycle and electron transport chain enzymes were decreased in CsA-induced rats, along with a significant increase in the activities of urinary enzymes, thus indicating renal tubular injury. Ultrastructural changes were also in accord with the above aberrations. The above abnormalities were favorably modulated by sulfated polysaccharides supplementation, thus highlighting the significance of sulfated polysaccharides in preventing the renal mitochondrial dysfunction allied with CsA-provoked nephrotoxicity.
Collapse
Affiliation(s)
- Anthony Josephine
- Department of Medical Biochemistry, Dr. ALM. Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | | | | | | | | | | |
Collapse
|
47
|
Gustafsson F, Ross HJ, Delgado MS, Bernabeo G, Delgado DH. Sirolimus-Based Immunosuppression After Cardiac Transplantation: Predictors of Recovery From Calcineurin Inhibitor-Induced Renal Dysfunction. J Heart Lung Transplant 2007; 26:998-1003. [DOI: 10.1016/j.healun.2007.07.034] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2007] [Revised: 07/20/2007] [Accepted: 07/20/2007] [Indexed: 01/09/2023] Open
|
48
|
Schneeberger S, Ninkovic M, Gabl M, Ninkovic M, Hussl H, Rieger M, Loescher W, Zelger B, Brandacher G, Bonatti H, Hautz T, Boesmueller C, Piza-Katzer H, Margreiter R. First forearm transplantation: outcome at 3 years. Am J Transplant 2007; 7:1753-62. [PMID: 17511764 DOI: 10.1111/j.1600-6143.2007.01837.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We here report on the surgical procedure, postoperative course and functional results at 3 years following the first bilateral forearm transplantation. A 41-year-old male underwent bilateral forearm transplantation on February 17, 2003. After ATG induction therapy, tacrolimus, prednisone and MMF were given for maintenance immunosuppression. At 16 months, MMF was switched to everolimus. Hand function, histology, immunohistochemistry, radiomorphology, motor and nerve conduction and somatosensory-evoked potentials were investigated at frequent intervals. A total of six rejection episodes required treatment with either steroids, basiliximab, ATG, alemtuzumab or tacrolimus dose augmentation. At 3 years, the patient is free of clinical signs of rejection despite a persisting minimal perivascular lymphocytic dermal infiltrate. No signs of myointimal proliferation in graft vessels were seen. Motor function continuously improved, resulting in satisfactory hand function. Intrinsic hand muscle function was first observed at 16 months and continues to improve. Although discrimination of hot and cold recovered, overall sensitivity remains poor. The patient is satisfied with the outcome. Bilateral forearm transplantation represents a novel therapeutic option after loss of forearms.
Collapse
Affiliation(s)
- S Schneeberger
- Department of General and Transplant Surgery, Innsbruck Medical University, Innsbruck, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Feitoza CQ, Gonçalves GM, Bertocchi APF, Wang PW, Damião MJ, Cenedeze MA, Teixeira VPA, Dos Reis MA, Pacheco-Silva A, Câmara NOS. A role for HO-1 in renal function impairment in animals subjected to ischemic and reperfusion injury and treated with immunosuppressive drugs. Transplant Proc 2007; 39:424-6. [PMID: 17362747 DOI: 10.1016/j.transproceed.2007.01.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Ischemia/reperfusion injury (IRI) represents the single major antigen-independent factor implicated in pathogenesis of chronic graft dysfunction. Tacrolimus is a calcineurin inhibitor, which has been suggested to be helpful in cyclosporine-related chronic toxicity. Rapamycin has antiproliferative properties that may impair renal regeneration after IRI. Therefore, immunosuppressive drugs might impair renal graft outcome in those organs suffering IRI. MATERIAL AND METHODS C57B1/6 male mice subjected to 45 minutes of renal pedicle ligation were reperfused for 24 hours. Mice were treated with rapamycin, cyclosporine, or tacrolimus. Blood and renal tissue samples were collected at 24 hours after IRI. Urea levels were measured. Heme Oxygenase 1 (HO-1) gene transcript was amplified by a real-time polymerase chain reaction technique. RESULTS Animals treated with cyclosporine and subjected to IRI showed impaired renal function that peaked at 24 hours. Additional pretreatment with rapamycin produced even more impairment of renal function, when compared with controls. However, tacrolimus pretreatment was associated with a better renal outcome. HO-1 expression was upregulated after IRI by 2.6 arbitrary units at 24 hours. Rapamycin showed worse impairment of renal function. CONCLUSION Tacrolimus was not associated with worsening renal function when compared with animals just subjected to IRI. Upregulation of HO-1 may be an attractive approach to limit graft injury.
Collapse
Affiliation(s)
- C Q Feitoza
- Laboratory of Experimental and Clinical Immunology, Nephrology Division, Federal University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Gooch JL, Roberts BR, Cobbs SL, Tumlin JA. Loss of the alpha-isoform of calcineurin is sufficient to induce nephrotoxicity and altered expression of transforming growth factor-beta. Transplantation 2007; 83:439-47. [PMID: 17318077 DOI: 10.1097/01.tp.0000251423.78124.51] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Use of calcineurin inhibitors is frequently limited by fibrosis, closely linked with increased transforming growth factor (TGF)-beta. However, mechanisms of extracellular matrix expansion and TGFbeta regulation following calcineurin inhibition are unknown. Mice lacking specific calcineurin catalytic subunit isoforms may offer important insight into this pathway. METHODS We compared mice lacking the alpha or beta isoform to a model of cyclosporin nephrotoxicity. Histological features common with cyclosporin nephrotoxicity including matrix expansion, arteriole hyalinization, and inflammation were assessed. Next, regulation specifically of fibronectin and TGFbeta was examined in vivo and in vitro. Finally, the role of TGFbeta in upregulation of fibronectin with loss of calcineurin activity was examined. RESULTS Loss of the alpha isoform results in histologic features and matrix expansion similar to cyclosporin, whereas loss of the beta does not. Fibronectin and TGFbeta are increased and renal function is impaired in alpha-null and aged alpha+/-. In primary alpha-/- renal fibroblasts, nuclear translocation of the calcineurin substrate NFATc is normal but regulation is lost in beta-null fibroblasts, confirming that the isoforms have distinct functions. Consistent with in vivo findings, alpha-null cells have increased fibronectin and TGFbeta. However, neutralizing TGFbeta antibody did not reduce fibronectin accumulation. CONCLUSIONS Our data show that calcineurin-alpha is key to regulation of fibrosis and TGFbeta and loss of this isoform reproduces features of cyclosporine nephrotoxicity in vivo and in vitro. In addition, we show that upregulation of TGFbeta and fibronectin likely result from a shared mechanism, but changes in fibronectin expression are independent of TGFbeta in renal fibroblasts.
Collapse
Affiliation(s)
- Jennifer L Gooch
- Department of Medicine, Division of Nephrology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | | | | | | |
Collapse
|