1
|
O'Sullivan ED, Mylonas KJ, Xin C, Baird DP, Carvalho C, Docherty MH, Campbell R, Matchett KP, Waddell SH, Walker AD, Gallagher KM, Jia S, Leung S, Laird A, Wilflingseder J, Willi M, Reck M, Finnie S, Pisco A, Gordon-Keylock S, Medvinsky A, Boulter L, Henderson NC, Kirschner K, Chandra T, Conway BR, Hughes J, Denby L, Bonventre JV, Ferenbach DA. Indian Hedgehog release from TNF-activated renal epithelia drives local and remote organ fibrosis. Sci Transl Med 2023; 15:eabn0736. [PMID: 37256934 DOI: 10.1126/scitranslmed.abn0736] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 05/10/2023] [Indexed: 06/02/2023]
Abstract
Progressive fibrosis is a feature of aging and chronic tissue injury in multiple organs, including the kidney and heart. Glioma-associated oncogene 1 expressing (Gli1+) cells are a major source of activated fibroblasts in multiple organs, but the links between injury, inflammation, and Gli1+ cell expansion and tissue fibrosis remain incompletely understood. We demonstrated that leukocyte-derived tumor necrosis factor (TNF) promoted Gli1+ cell proliferation and cardiorenal fibrosis through induction and release of Indian Hedgehog (IHH) from renal epithelial cells. Using single-cell-resolution transcriptomic analysis, we identified an "inflammatory" proximal tubular epithelial (iPT) population contributing to TNF- and nuclear factor κB (NF-κB)-induced IHH production in vivo. TNF-induced Ubiquitin D (Ubd) expression was observed in human proximal tubular cells in vitro and during murine and human renal disease and aging. Studies using pharmacological and conditional genetic ablation of TNF-induced IHH signaling revealed that IHH activated canonical Hedgehog signaling in Gli1+ cells, which led to their activation, proliferation, and fibrosis within the injured and aging kidney and heart. These changes were inhibited in mice by Ihh deletion in Pax8-expressing cells or by pharmacological blockade of TNF, NF-κB, or Gli1 signaling. Increased amounts of circulating IHH were associated with loss of renal function and higher rates of cardiovascular disease in patients with chronic kidney disease. Thus, IHH connects leukocyte activation to Gli1+ cell expansion and represents a potential target for therapies to inhibit inflammation-induced fibrosis.
Collapse
Affiliation(s)
- Eoin D O'Sullivan
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, Queensland 4029, Australia
| | - Katie J Mylonas
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Cuiyan Xin
- Renal Division and Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - David P Baird
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Cyril Carvalho
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Marie-Helena Docherty
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Ross Campbell
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Kylie P Matchett
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Scott H Waddell
- Cancer Research UK Scotland Centre and MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Alexander D Walker
- Cancer Research UK Scotland Centre and MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Kevin M Gallagher
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
- Department of Urology, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Siyang Jia
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Steve Leung
- Department of Urology, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Alexander Laird
- Department of Urology, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Julia Wilflingseder
- Renal Division and Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Department of Physiology and Pathophysiology, University of Veterinary Medicine, Veterinärplatz 1, 1210 Vienna, Austria
| | - Michaela Willi
- Laboratory of Genetics and Physiology, NIDDK, NIH, Bethesda, MD 20892, USA
| | - Maximilian Reck
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Sarah Finnie
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Angela Pisco
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | | | - Alexander Medvinsky
- Centre for Regenerative Medicine. University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Luke Boulter
- Cancer Research UK Scotland Centre and MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Neil C Henderson
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
- Cancer Research UK Scotland Centre and MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Kristina Kirschner
- School of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Tamir Chandra
- Cancer Research UK Scotland Centre and MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Bryan R Conway
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Jeremy Hughes
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Laura Denby
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Joseph V Bonventre
- Renal Division and Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - David A Ferenbach
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
- Renal Division and Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
2
|
Abdelmageed MM, Kefaloyianni E, Arthanarisami A, Komaru Y, Atkinson JJ, Herrlich A. TNF or EGFR inhibition equally block AKI-to-CKD transition: opportunities for etanercept treatment. Nephrol Dial Transplant 2023; 38:1139-1150. [PMID: 36269313 PMCID: PMC10157768 DOI: 10.1093/ndt/gfac290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Inflammation is a key driver of the transition of acute kidney injury to progressive fibrosis and chronic kidney disease (AKI-to-CKD transition). Blocking a-disintegrin-and-metalloprotease-17 (ADAM17)-dependent ectodomain shedding, in particular of epidermal growth factor receptor (EGFR) ligands and of the type 1 inflammatory cytokine tumor necrosis factor (TNF), reduces pro-inflammatory and pro-fibrotic responses after ischemic AKI or unilateral ureteral obstruction (UUO), a classical fibrosis model. Metalloprotease or EGFR inhibition show significant undesirable side effects in humans. In retrospective studies anti-TNF biologics reduce the incidence and progression of CKD in humans. Whether TNF has a role in AKI-to-CKD transition and how TNF inhibition compares to EGFR inhibition is largely unknown. METHODS Mice were subjected to bilateral renal ischemia-reperfusion injury or unilateral ureteral obstruction. Kidneys were analyzed by histology, immunohistochemistry, qPCR, western blot, mass cytometry, scRNA sequencing, and cytokine profiling. RESULTS Here we show that TNF or EGFR inhibition reduce AKI-to-CKD transition and fibrosis equally by about 25%, while combination has no additional effect. EGFR inhibition reduced kidney TNF expression by about 50% largely by reducing accumulation of TNF expressing immune cells in the kidney early after AKI, while TNF inhibition did not affect EGFR activation or immune cell accumulation. Using scRNAseq data we show that TNF is predominantly expressed by immune cells in AKI but not in proximal tubule cells (PTC), and PTC-TNF knockout did not affect AKI-to-CKD transition in UUO. Thus, the anti-inflammatory and anti-fibrotic effects of the anti-TNF biologic etanercept in AKI-to-CKD transition rely on blocking TNF that is released from immune cells recruited or accumulating in response to PTC-EGFR signals. CONCLUSION Short-term anti-TNF biologics during or after AKI could be helpful in the prevention of AKI-to-CKD transition.
Collapse
Affiliation(s)
- Mai M Abdelmageed
- Washington University School of Medicine in Saint Louis, Department of Medicine, St. Louis, MO, USA
- Division of Nephrology
| | - Eirini Kefaloyianni
- Washington University School of Medicine in Saint Louis, Department of Medicine, St. Louis, MO, USA
- Division of Nephrology
| | - Akshayakeerthi Arthanarisami
- Washington University School of Medicine in Saint Louis, Department of Medicine, St. Louis, MO, USA
- Division of Nephrology
| | - Yohei Komaru
- Washington University School of Medicine in Saint Louis, Department of Medicine, St. Louis, MO, USA
- Division of Nephrology
| | - Jeffrey J Atkinson
- Washington University School of Medicine in Saint Louis, Department of Medicine, St. Louis, MO, USA
- Division of Pulmonary and Critical Care Medicine
| | - Andreas Herrlich
- Washington University School of Medicine in Saint Louis, Department of Medicine, St. Louis, MO, USA
- Division of Nephrology
| |
Collapse
|
3
|
Rayego-Mateos S, Rodrigues-Diez RR, Fernandez-Fernandez B, Mora-Fernández C, Marchant V, Donate-Correa J, Navarro-González JF, Ortiz A, Ruiz-Ortega M. Targeting inflammation to treat diabetic kidney disease: the road to 2030. Kidney Int 2023; 103:282-296. [PMID: 36470394 DOI: 10.1016/j.kint.2022.10.030] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/05/2022] [Accepted: 10/31/2022] [Indexed: 12/07/2022]
Abstract
Diabetic kidney disease (DKD) is one of the fastest growing causes of chronic kidney disease and associated morbidity and mortality. Preclinical research has demonstrated the involvement of inflammation in its pathogenesis and in the progression of kidney damage, supporting clinical trials designed to explore anti-inflammatory strategies. However, the recent success of sodium-glucose cotransporter-2 inhibitors and the nonsteroidal mineralocorticoid receptor antagonist finerenone has changed both guidelines and standard of care, rendering obsolete older studies directly targeting inflammatory mediators and the clinical development was discontinued for most anti-inflammatory drugs undergoing clinical trials for DKD in 2016. Given the contribution of inflammation to the pathogenesis of DKD, we review the impact on kidney inflammation of the current standard of care, therapies undergoing clinical trials, or repositioned drugs for DKD. Moreover, we review recent advances in the molecular regulation of inflammation in DKD and discuss potential novel therapeutic strategies with clinical relevance. Finally, we provide a road map for future research aimed at integrating the growing knowledge on inflammation and DKD into clinical practice to foster improvement of patient outcomes.
Collapse
Affiliation(s)
- Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma, Madrid, Spain; Ricord2040, Instituto de Salud Carlos II, Spain
| | - Raul R Rodrigues-Diez
- Ricord2040, Instituto de Salud Carlos II, Spain; Translational Immunology, Instituto de Investigación Sanitaria del Principado de Asturias ISPA, Oviedo, Asturias, Spain
| | - Beatriz Fernandez-Fernandez
- Ricord2040, Instituto de Salud Carlos II, Spain; Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma, Madrid, Spain
| | - Carmen Mora-Fernández
- Ricord2040, Instituto de Salud Carlos II, Spain; Research Unit, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Vanessa Marchant
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma, Madrid, Spain; Ricord2040, Instituto de Salud Carlos II, Spain
| | - Javier Donate-Correa
- Ricord2040, Instituto de Salud Carlos II, Spain; Research Unit, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Juan F Navarro-González
- Ricord2040, Instituto de Salud Carlos II, Spain; Research Unit, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain; Nephrology Service, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Alberto Ortiz
- Ricord2040, Instituto de Salud Carlos II, Spain; Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma, Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma, Madrid, Spain; Ricord2040, Instituto de Salud Carlos II, Spain.
| |
Collapse
|
4
|
Kawahito Y, Morinobu A, Kaneko Y, Kohno M, Hirata S, Kishimoto M, Seto Y, Sugihara T, Tanaka E, Ito H, Kojima T, Matsushita I, Nishida K, Mori M, Murashima A, Yamanaka H, Nakayama T, Kojima M, Harigai M. Drug treatment algorithm and recommendations from the 2020 update of the Japan College of Rheumatology clinical practice guidelines for the management of rheumatoid arthritis-secondary publication. Mod Rheumatol 2023; 33:21-35. [PMID: 35297492 DOI: 10.1093/mr/roac017] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/05/2022] [Accepted: 02/09/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE The aim of this study was to update the Japan College of Rheumatology (JCR) clinical practice guidelines (CPG) for the management of rheumatoid arthritis (RA; JCR CPG for RA) according to recent changes in the medical environment in Japan. This article is a digest version of the guidance. METHODS We used the Grading of Recommendations, Assessment, Development, and Evaluation method to update the 2014 JCR CPG for RA. A consensus was formed by CPG panel members. RESULTS We identified 36 important clinical questions regarding drug treatment and developed corresponding recommendations for RA. The recommendations included the following RA medications: non-steroidal anti-inflammatory drugs, corticosteroids, conventional synthetic disease-modifying antirheumatic drugs, biological disease-modifying antirheumatic drugs, anti-receptor activator for nuclear factor-κB ligand antibodies, and Janus kinase inhibitors, as well as the tapering and discontinuation of these medications. Recommendations regarding the efficacy and safety of treatments in the elderly and patients with comorbidities were also developed. Finally, we used these recommendations to create an original algorithm for drug treatment for RA based on the Treat-to-Target approach. CONCLUSION The 2020 JCR CPG for RA provides a useful tool for rheumatologists, health care professionals, and patients with RA, enabling shared decision-making in a variety of clinical situations.
Collapse
Affiliation(s)
- Yutaka Kawahito
- Inflammation and Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akio Morinobu
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuko Kaneko
- Department of Internal Medicine, Division of Rheumatology, Keio University School of Medicine, Tokyo, Japan
| | - Masataka Kohno
- Inflammation and Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shintaro Hirata
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Mitsumasa Kishimoto
- Department of Nephrology and Rheumatology, Kyorin University School of Medicine, Tokyo, Japan
| | - Yohei Seto
- Department of Rheumatology, Yachiyo Medical Center, Tokyo Women's Medical University, Yachiyo, Japan
| | - Takahiko Sugihara
- Division of Rheumatology and Allergology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Eiichi Tanaka
- Department of Internal Medicine, Division of Rheumatology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Hiromu Ito
- Department of Orthopedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Orthopaedic Surgery, Kurashiki Central Hospital, Kurashiki, Japan
| | - Toshihisa Kojima
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Isao Matsushita
- Department of Rehabilitation Medicine, Kanazawa Medical University, Ishikawa, Japan
| | - Keiichiro Nishida
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Density and Pharmaceutical Sciences, Okayama, Japan
| | - Masaaki Mori
- Division of Rheumatology and Allergology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Atsuko Murashima
- Center for Maternal-Fetal, Neonatal and Reproductive Medicine/Japan Drug Information Institute in Pregnancy, National Center for Child Health and Development, Tokyo, Japan
| | | | - Takeo Nakayama
- Department of Health Informatics, Kyoto University School of Public Health, Kyoto, Japan
| | - Masayo Kojima
- Department of Frailty Research, Center for Gerontology and Social Science, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Masayoshi Harigai
- Department of Internal Medicine, Division of Rheumatology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| |
Collapse
|
5
|
Ramos-Vera C, Barrientos AS, Vallejos-Saldarriaga J, Calizaya-Milla YE, Saintila J. Network Structure of Comorbidity Patterns in U.S. Adults with Depression: A National Study Based on Data from the Behavioral Risk Factor Surveillance System. DEPRESSION RESEARCH AND TREATMENT 2023; 2023:9969532. [PMID: 37096248 PMCID: PMC10122603 DOI: 10.1155/2023/9969532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/14/2023] [Accepted: 03/21/2023] [Indexed: 04/26/2023]
Abstract
Background People with depression are at increased risk for comorbidities; however, the clustering of comorbidity patterns in these patients is still unclear. Objective The aim of the study was to identify latent comorbidity patterns and explore the comorbidity network structure that included 12 chronic conditions in adults diagnosed with depressive disorder. Methods A cross-sectional study was conducted based on secondary data from the 2017 behavioral risk factor surveillance system (BRFSS) covering all 50 American states. A sample of 89,209 U.S. participants, 29,079 men and 60,063 women aged 18 years or older, was considered using exploratory graphical analysis (EGA), a statistical graphical model that includes algorithms for grouping and factoring variables in a multivariate system of network relationships. Results The EGA findings show that the network presents 3 latent comorbidity patterns, i.e., that comorbidities are grouped into 3 factors. The first group was composed of 7 comorbidities (obesity, cancer, high blood pressure, high blood cholesterol, arthritis, kidney disease, and diabetes). The second pattern of latent comorbidity included the diagnosis of asthma and respiratory diseases. The last factor grouped 3 conditions (heart attack, coronary heart disease, and stroke). Hypertension reported higher measures of network centrality. Conclusion Associations between chronic conditions were reported; furthermore, they were grouped into 3 latent dimensions of comorbidity and reported network factor loadings. The implementation of care and treatment guidelines and protocols for patients with depressive symptomatology and multimorbidity is suggested.
Collapse
Affiliation(s)
- Cristian Ramos-Vera
- Research Area, Faculty of Health Sciences, Universidad César Vallejo, Lima, Peru
| | | | | | - Yaquelin E. Calizaya-Milla
- Research Group for Nutrition and Lifestyle, School of Human Nutrition, Universidad Peruana Unión, Lima, Peru
| | - Jacksaint Saintila
- Research Group for Nutrition and Lifestyle, School of Human Nutrition, Universidad Peruana Unión, Lima, Peru
| |
Collapse
|
6
|
Swart IAP, Visman IM, Heslinga M, van der Horst-Bruinsma IE, van Denderen JC, Nurmohamed MT. The effect of anti-TNF on renal function in patients with ankylosing spondylitis: a prospective cohort study. Clin Rheumatol 2022; 41:3747-3752. [PMID: 35962246 PMCID: PMC9652256 DOI: 10.1007/s10067-022-06330-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/28/2022] [Accepted: 08/05/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Biologicals, such as anti-tumor necrosis factor (anti-TNF), reduce cardiovascular disease (CVD) in patients with inflammatory rheumatic diseases. Impaired renal function is a known predictor of CVD and elevated in ankylosing spondylitis (AS). OBJECTIVE To assess the effect of anti-TNF on renal function in patients with AS and whether anti-TNF use is safe in AS patients with pre-existing risk factors for renal decline. METHOD Biological-naïve consecutive AS patients treated with etanercept or adalimumab were prospectively followed from 2005 to 2014. Renal function was determined by calculation of the estimated glomerular filtration rate (eGFR), estimated with the abbreviated modification of diet in renal disease (MDRD) formula. The effect of anti-TNF on eGFR was analyzed using mixed model analysis. RESULTS 211 AS patients were followed for a median of 156 (36-286) weeks. Overall mixed model analyses showed a significant decrease of eGFR over time (β = - 0.040, p = 0.000), although this association did not remain significant after adjustment for responding to anti-TNF, alcohol use, disease duration, body mass index (BMI), C-reactive protein (CRP), and disease activity (β = - 0.018, p = 0.094). However, patients with pre-existing risk factors for renal decline did have a significant change in eGFR over time (β = - 0.029, p = 0.006). CONCLUSIONS We found a significant change in eGFR over time, although this small decrease was not clinically relevant. This study further demonstrates that anti-TNF does not affect renal function in AS patients with and without existing risk factors for renal decline, which means that use of anti-TNF is safe concerning renal function in patients with AS. Key Points • Previous studies showed that biologicals, such as anti-tumor necrosis factor (anti-TNF), reduce cardiovascular disease (CVD) in patients with inflammatory rheumatic diseases, such as ankylosing spondylitis (AS). • Impaired renal function is a known predictor of CVD, and also a known concern for many AS patients. • Use of anti-TNF is safe with regard to renal function in patients with AS. • The effect of anti-TNF on CVD in AS patients does not seem to be mediated by changes in renal function.
Collapse
Affiliation(s)
- I A P Swart
- Amsterdam Rheumatology and Immunology Center, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - I M Visman
- Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Jan van Breemenstraat 2, Reade, Amsterdam, 1056 AB, The Netherlands.
| | - M Heslinga
- Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Jan van Breemenstraat 2, Reade, Amsterdam, 1056 AB, The Netherlands
| | - I E van der Horst-Bruinsma
- Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Jan van Breemenstraat 2, Reade, Amsterdam, 1056 AB, The Netherlands
| | - J C van Denderen
- Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Jan van Breemenstraat 2, Reade, Amsterdam, 1056 AB, The Netherlands
| | - M T Nurmohamed
- Amsterdam Rheumatology and Immunology Center, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Jan van Breemenstraat 2, Reade, Amsterdam, 1056 AB, The Netherlands
| |
Collapse
|
7
|
Smirnova LA, Simonova OV, Sukhikh EN, Shilyaeva AO. Case of successful use of sarilumab in secondary renal amyloidosis in a patient with rheumatoid arthritis. RHEUMATOLOGY SCIENCE AND PRACTICE 2022. [DOI: 10.47360/1995-4484-2022-599-602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Clinical observation of the successful use of the interleukin 6 (IL6) inhibitor sarilumab in secondary renal amyloidosis in a patient with active seropositive rheumatoid arthritis, is presented. This complication was confirmed by biopsy of rectum. The presented clinical example demonstrates a fairly rapid, within five years from the onset of the disease, the development of secondary renal amyolidosis with the formation of a persistent nephrotic syndrome that is resistant to therapy with cyclophosphamide and rituximab. Prescription of the IL6 inhibitor tocilizumab contributed to a decrease in the clinical and laboratory activity of the underlying disease, a decrease in the severity of daily proteinuria, but did not allow to achieve the full effect. The use of another IL6 inhibitor – sarilumab, led to a complete regression of nephrotic syndrome with normalization of general urine analysis, biochemical blood tests – total protein, albumin, total cholesterol, against the background of stable clinical and laboratory remission of rheumatoid arthritis.
Collapse
|
8
|
New mechanisms for the kidney-protective effect of alkali in chronic kidney disease. Clin Sci (Lond) 2022; 136:1433-1437. [DOI: 10.1042/cs20220395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022]
Abstract
Abstract
Worldwide, more than one in ten adults are estimated to have chronic kidney disease (CKD). As CKD progresses, both the cost of treatment and associated risk of morbidity and mortality increase exponentially. As such, there is a great need for therapies that effectively slow CKD progression. Evidence from several small clinical trials indicates that alkali therapy may slow the rate of CKD progression. The biological mechanisms underlying this protective effect, however, remain unknown. In their recently published manuscript, Pastor Arroyo et al. (Clin Sci (Lond) (2022) 136(8): https://doi.org/10.1042/CS20220095) demonstrate that the alkali sodium bicarbonate protects against loss of renal function in a crystal nephropathy model in mice. Using unbiased approaches in both mice and human tissue, the authors go on to identify two novel mechanisms that may underly this protection. The first pathway is through promoting pathways of cell metabolism, which they speculate helps the remaining functional nephrons adapt to the greater metabolic needs required to maintain kidney filtration. The second pathway is by restoration of α-Klotho levels, which may limit the expression of adhesion molecules in the injured kidney. This, the authors speculate, may prevent inflammation from driving the functional decline of the kidney. Identifying these novel pathways represents an important step forward harnessing the potential benefits of alkali therapy in CKD.
Collapse
|
9
|
Capolongo G, Capasso G, Viggiano D. A Shared Nephroprotective Mechanism for Renin-Angiotensin-System Inhibitors, Sodium-Glucose Co-Transporter 2 Inhibitors, and Vasopressin Receptor Antagonists: Immunology Meets Hemodynamics. Int J Mol Sci 2022; 23:3915. [PMID: 35409276 PMCID: PMC8999762 DOI: 10.3390/ijms23073915] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 02/01/2023] Open
Abstract
A major paradigm in nephrology states that the loss of filtration function over a long time is driven by a persistent hyperfiltration state of surviving nephrons. This hyperfiltration may derive from circulating immunological factors. However, some clue about the hemodynamic effects of these factors derives from the effects of so-called nephroprotective drugs. Thirty years after the introduction of Renin-Angiotensin-system inhibitors (RASi) into clinical practice, two new families of nephroprotective drugs have been identified: the sodium-glucose cotransporter 2 inhibitors (SGLT2i) and the vasopressin receptor antagonists (VRA). Even though the molecular targets of the three-drug classes are very different, they share the reduction in the glomerular filtration rate (GFR) at the beginning of the therapy, which is usually considered an adverse effect. Therefore, we hypothesize that acute GFR decline is a prerequisite to obtaining nephroprotection with all these drugs. In this study, we reanalyze evidence that RASi, SGLT2i, and VRA reduce the eGFR at the onset of therapy. Afterward, we evaluate whether the extent of eGFR reduction correlates with their long-term efficacy. The results suggest that the extent of initial eGFR decline predicts the nephroprotective efficacy in the long run. Therefore, we propose that RASi, SGLT2i, and VRA delay kidney disease progression by controlling maladaptive glomerular hyperfiltration resulting from circulating immunological factors. Further studies are needed to verify their combined effects.
Collapse
Affiliation(s)
- Giovanna Capolongo
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (G.C.); (G.C.)
- BioGeM, Institute of Molecular Biology and Genetics, 83031 Ariano Irpino, Italy
| | - Giovambattista Capasso
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (G.C.); (G.C.)
- BioGeM, Institute of Molecular Biology and Genetics, 83031 Ariano Irpino, Italy
| | - Davide Viggiano
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (G.C.); (G.C.)
- BioGeM, Institute of Molecular Biology and Genetics, 83031 Ariano Irpino, Italy
| |
Collapse
|
10
|
Lousa I, Reis F, Santos-Silva A, Belo L. The Signaling Pathway of TNF Receptors: Linking Animal Models of Renal Disease to Human CKD. Int J Mol Sci 2022; 23:3284. [PMID: 35328704 PMCID: PMC8950598 DOI: 10.3390/ijms23063284] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 11/25/2022] Open
Abstract
Chronic kidney disease (CKD) has been recognized as a global public health problem. Despite the current advances in medicine, CKD-associated morbidity and mortality remain unacceptably high. Several studies have highlighted the contribution of inflammation and inflammatory mediators to the development and/or progression of CKD, such as tumor necrosis factor (TNF)-related biomarkers. The inflammation pathway driven by TNF-α, through TNF receptors 1 (TNFR1) and 2 (TNFR2), involves important mediators in the pathogenesis of CKD. Circulating levels of TNFRs were associated with changes in other biomarkers of kidney function and injury, and were described as predictors of disease progression, cardiovascular morbidity, and mortality in several cohorts of patients. Experimental studies describe the possible downstream signaling pathways induced upon TNFR activation and the resulting biological responses. This review will focus on the available data on TNFR1 and TNFR2, and illustrates their contributions to the pathophysiology of kidney diseases, their cellular and molecular roles, as well as their potential as CKD biomarkers. The emerging evidence shows that TNF receptors could act as biomarkers of renal damage and as mediators of the disease. Furthermore, it has been suggested that these biomarkers could significantly improve the discrimination of clinical CKD prognostic models.
Collapse
Affiliation(s)
- Irina Lousa
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (I.L.); (A.S.-S.)
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Flávio Reis
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-075 Coimbra, Portugal
| | - Alice Santos-Silva
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (I.L.); (A.S.-S.)
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Luís Belo
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (I.L.); (A.S.-S.)
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
11
|
Hasankhani A, Bahrami A, Sheybani N, Aria B, Hemati B, Fatehi F, Ghaem Maghami Farahani H, Javanmard G, Rezaee M, Kastelic JP, Barkema HW. Differential Co-Expression Network Analysis Reveals Key Hub-High Traffic Genes as Potential Therapeutic Targets for COVID-19 Pandemic. Front Immunol 2021; 12:789317. [PMID: 34975885 PMCID: PMC8714803 DOI: 10.3389/fimmu.2021.789317] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/26/2021] [Indexed: 01/08/2023] Open
Abstract
Background The recent emergence of COVID-19, rapid worldwide spread, and incomplete knowledge of molecular mechanisms underlying SARS-CoV-2 infection have limited development of therapeutic strategies. Our objective was to systematically investigate molecular regulatory mechanisms of COVID-19, using a combination of high throughput RNA-sequencing-based transcriptomics and systems biology approaches. Methods RNA-Seq data from peripheral blood mononuclear cells (PBMCs) of healthy persons, mild and severe 17 COVID-19 patients were analyzed to generate a gene expression matrix. Weighted gene co-expression network analysis (WGCNA) was used to identify co-expression modules in healthy samples as a reference set. For differential co-expression network analysis, module preservation and module-trait relationships approaches were used to identify key modules. Then, protein-protein interaction (PPI) networks, based on co-expressed hub genes, were constructed to identify hub genes/TFs with the highest information transfer (hub-high traffic genes) within candidate modules. Results Based on differential co-expression network analysis, connectivity patterns and network density, 72% (15 of 21) of modules identified in healthy samples were altered by SARS-CoV-2 infection. Therefore, SARS-CoV-2 caused systemic perturbations in host biological gene networks. In functional enrichment analysis, among 15 non-preserved modules and two significant highly-correlated modules (identified by MTRs), 9 modules were directly related to the host immune response and COVID-19 immunopathogenesis. Intriguingly, systemic investigation of SARS-CoV-2 infection identified signaling pathways and key genes/proteins associated with COVID-19's main hallmarks, e.g., cytokine storm, respiratory distress syndrome (ARDS), acute lung injury (ALI), lymphopenia, coagulation disorders, thrombosis, and pregnancy complications, as well as comorbidities associated with COVID-19, e.g., asthma, diabetic complications, cardiovascular diseases (CVDs), liver disorders and acute kidney injury (AKI). Topological analysis with betweenness centrality (BC) identified 290 hub-high traffic genes, central in both co-expression and PPI networks. We also identified several transcriptional regulatory factors, including NFKB1, HIF1A, AHR, and TP53, with important immunoregulatory roles in SARS-CoV-2 infection. Moreover, several hub-high traffic genes, including IL6, IL1B, IL10, TNF, SOCS1, SOCS3, ICAM1, PTEN, RHOA, GDI2, SUMO1, CASP1, IRAK3, HSPA5, ADRB2, PRF1, GZMB, OASL, CCL5, HSP90AA1, HSPD1, IFNG, MAPK1, RAB5A, and TNFRSF1A had the highest rates of information transfer in 9 candidate modules and central roles in COVID-19 immunopathogenesis. Conclusion This study provides comprehensive information on molecular mechanisms of SARS-CoV-2-host interactions and identifies several hub-high traffic genes as promising therapeutic targets for the COVID-19 pandemic.
Collapse
Affiliation(s)
- Aliakbar Hasankhani
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Abolfazl Bahrami
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
- Biomedical Center for Systems Biology Science Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Negin Sheybani
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran
| | - Behzad Aria
- Department of Physical Education and Sports Science, School of Psychology and Educational Sciences, Yazd University, Yazd, Iran
| | - Behzad Hemati
- Biotechnology Research Center, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Farhang Fatehi
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | | | - Ghazaleh Javanmard
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Mahsa Rezaee
- Department of Medical Mycology, School of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - John P. Kastelic
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Herman W. Barkema
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
12
|
Wu W, Liang D. Expression and related mechanisms of miR-330-3p and S100B in an animal model of cartilage injury. J Int Med Res 2021; 49:3000605211039471. [PMID: 34590918 PMCID: PMC8489778 DOI: 10.1177/03000605211039471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Objective To investigate the roles of and relationship between microRNA (miR)-330-3p and S100 calcium-binding protein B (S100B) in an animal model of cartilage injury. Methods This study included 30 New Zealand male rabbits randomly divided into three groups: an intervention group, a model group and a sham surgery control group. Modelling was performed in the intervention and model groups, but in the sham surgery group, only the skin was cut. After modelling, the intervention and model groups were injected with the miR-330-3p overexpression vector GV268-miR-330-3p or the control GV268-N-ODN vector, respectively, twice a week for 7 weeks. Results Levels of interleukin-1β and tumour necrosis factor-α in the synovial fluid were significantly higher in the model group than in the intervention and control groups. The level of miR-330-3p in the cartilage tissue was significantly higher in the control group than in the model group but it was significantly lower compared with the intervention group. Levels of S100B, fibroblast growth factor receptor 1 and fibroblast growth factor-2 in the cartilage tissue of rabbits in the model group were significantly higher compared with the control and intervention groups. Conclusion These findings demonstrate that the upregulation of miR-330-3p can inhibit the expression of S100B.
Collapse
Affiliation(s)
- Wenming Wu
- Department of Orthopaedics, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Dongming Liang
- Lirimax (Tianjin) Medical Technical Co., Ltd, Tianjin, China
| |
Collapse
|
13
|
Adhikari A, Mondal S, Chatterjee T, Das M, Biswas P, Ghosh R, Darbar S, Alessa H, Althakafy JT, Sayqal A, Ahmed SA, Das AK, Bhattacharyya M, Pal SK. Redox nanomedicine ameliorates chronic kidney disease (CKD) by mitochondrial reconditioning in mice. Commun Biol 2021; 4:1013. [PMID: 34446827 PMCID: PMC8390471 DOI: 10.1038/s42003-021-02546-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 08/02/2021] [Indexed: 12/29/2022] Open
Abstract
Targeting reactive oxygen species (ROS) while maintaining cellular redox signaling is crucial in the development of redox medicine as the origin of several prevailing diseases including chronic kidney disease (CKD) is linked to ROS imbalance and associated mitochondrial dysfunction. Here, we have shown that a potential nanomedicine comprising of Mn3O4 nanoparticles duly functionalized with biocompatible ligand citrate (C-Mn3O4 NPs) can maintain cellular redox balance in an animal model of oxidative injury. We developed a cisplatin-induced CKD model in C57BL/6j mice with severe mitochondrial dysfunction and oxidative distress leading to the pathogenesis. Four weeks of treatment with C-Mn3O4 NPs restored renal function, preserved normal kidney architecture, ameliorated overexpression of pro-inflammatory cytokines, and arrested glomerulosclerosis and interstitial fibrosis. A detailed study involving human embryonic kidney (HEK 293) cells and isolated mitochondria from experimental animals revealed that the molecular mechanism behind the pharmacological action of the nanomedicine involves protection of structural and functional integrity of mitochondria from oxidative damage, subsequent reduction in intracellular ROS, and maintenance of cellular redox homeostasis. To the best of our knowledge, such studies that efficiently treated a multifaceted disease like CKD using a biocompatible redox nanomedicine are sparse in the literature. Successful clinical translation of this nanomedicine may open a new avenue in redox-mediated therapeutics of several other diseases (e.g., diabetic nephropathy, neurodegeneration, and cardiovascular disease) where oxidative distress plays a central role in pathogenesis.
Collapse
Affiliation(s)
- Aniruddha Adhikari
- Department of Chemical, Biological and Macromolecular Sciences, S. N. Bose National Centre for Basic Sciences, Kolkata, India
| | - Susmita Mondal
- Department of Chemical, Biological and Macromolecular Sciences, S. N. Bose National Centre for Basic Sciences, Kolkata, India
| | | | - Monojit Das
- Department of Zoology, Uluberia College, University of Calcutta, Uluberia, Howrah, India
- Department of Zoology, Vidyasagar University, Rangamati, Midnapore, India
| | - Pritam Biswas
- Department of Microbiology, St. Xavier's College, Kolkata, India
| | - Ria Ghosh
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Soumendra Darbar
- Research & Development Division, Dey's Medical Stores (Mfg.) Ltd, Kolkata, India
| | - Hussain Alessa
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Jalal T Althakafy
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ali Sayqal
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Saleh A Ahmed
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
- Chemistry Department, Faculty of Science, Assiut University, Assiut, Egypt
| | - Anjan Kumar Das
- Department of Pathology, Calcutta National Medical College and Hospital, Kolkata, India
| | | | - Samir Kumar Pal
- Department of Chemical, Biological and Macromolecular Sciences, S. N. Bose National Centre for Basic Sciences, Kolkata, India.
- Department of Zoology, Uluberia College, University of Calcutta, Uluberia, Howrah, India.
| |
Collapse
|
14
|
Cantero-Navarro E, Rayego-Mateos S, Orejudo M, Tejedor-Santamaria L, Tejera-Muñoz A, Sanz AB, Marquez-Exposito L, Marchant V, Santos-Sanchez L, Egido J, Ortiz A, Bellon T, Rodrigues-Diez RR, Ruiz-Ortega M. Role of Macrophages and Related Cytokines in Kidney Disease. Front Med (Lausanne) 2021; 8:688060. [PMID: 34307414 PMCID: PMC8295566 DOI: 10.3389/fmed.2021.688060] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/11/2021] [Indexed: 12/14/2022] Open
Abstract
Inflammation is a key characteristic of kidney disease, but this immune response is two-faced. In the acute phase of kidney injury, there is an activation of the immune cells to fight against the insult, contributing to kidney repair and regeneration. However, in chronic kidney diseases (CKD), immune cells that infiltrate the kidney play a deleterious role, actively participating in disease progression, and contributing to nephron loss and fibrosis. Importantly, CKD is a chronic inflammatory disease. In early CKD stages, patients present sub-clinical inflammation, activation of immune circulating cells and therefore, anti-inflammatory strategies have been proposed as a common therapeutic target for renal diseases. Recent studies have highlighted the plasticity of immune cells and the complexity of their functions. Among immune cells, monocytes/macrophages play an important role in all steps of kidney injury. However, the phenotype characterization between human and mice immune cells showed different markers; therefore the extrapolation of experimental studies in mice could not reflect human renal diseases. Here we will review the current information about the characteristics of different macrophage phenotypes, mainly focused on macrophage-related cytokines, with special attention to the chemokine CCL18, and its murine functional homolog CCL8, and the macrophage marker CD163, and their role in kidney pathology.
Collapse
Affiliation(s)
- Elena Cantero-Navarro
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Rayego-Mateos
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Macarena Orejudo
- Renal, Vascular and Diabetes Research Laboratory, Fundación IIS -Fundación Jiménez Díaz, Universidad Autónoma, Madrid, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Lucía Tejedor-Santamaria
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Tejera-Muñoz
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Belén Sanz
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
- Laboratory of Nephrology and Hypertension, Fundación IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Laura Marquez-Exposito
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Vanessa Marchant
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Santos-Sanchez
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, Fundación IIS -Fundación Jiménez Díaz, Universidad Autónoma, Madrid, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Alberto Ortiz
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
- Laboratory of Nephrology and Hypertension, Fundación IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Teresa Bellon
- La Paz Hospital Health Research Institute, Madrid, Spain
| | - Raúl R Rodrigues-Diez
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
15
|
Shah A, Lindquist JA, Rosendahl L, Schmitz I, Mertens PR. Novel Insights into YB-1 Signaling and Cell Death Decisions. Cancers (Basel) 2021; 13:3306. [PMID: 34282755 PMCID: PMC8269159 DOI: 10.3390/cancers13133306] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022] Open
Abstract
YB-1 belongs to the evolutionarily conserved cold-shock domain protein family of RNA binding proteins. YB-1 is a well-known transcriptional and translational regulator, involved in cell cycle progression, DNA damage repair, RNA splicing, and stress responses. Cell stress occurs in many forms, e.g., radiation, hyperthermia, lipopolysaccharide (LPS) produced by bacteria, and interferons released in response to viral infection. Binding of the latter factors to their receptors induces kinase activation, which results in the phosphorylation of YB-1. These pathways also activate the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), a well-known transcription factor. NF-κB is upregulated following cellular stress and orchestrates inflammatory responses, cell proliferation, and differentiation. Inflammation and cancer are known to share common mechanisms, such as the recruitment of infiltrating macrophages and development of an inflammatory microenvironment. Several recent papers elaborate the role of YB-1 in activating NF-κB and signaling cell survival. Depleting YB-1 may tip the balance from survival to enhanced apoptosis. Therefore, strategies that target YB-1 might be a viable therapeutic option to treat inflammatory diseases and improve tumor therapy.
Collapse
Affiliation(s)
- Aneri Shah
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University, 39120 Magdeburg, Germany; (A.S.); (L.R.); (P.R.M.)
| | - Jonathan A. Lindquist
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University, 39120 Magdeburg, Germany; (A.S.); (L.R.); (P.R.M.)
| | - Lars Rosendahl
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University, 39120 Magdeburg, Germany; (A.S.); (L.R.); (P.R.M.)
| | - Ingo Schmitz
- Department of Molecular Immunology, ZKF2, Ruhr-University Bochum, 44801 Bochum, Germany;
| | - Peter R. Mertens
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University, 39120 Magdeburg, Germany; (A.S.); (L.R.); (P.R.M.)
| |
Collapse
|
16
|
Ixekizumab May Improve Renal Function in Psoriasis. Healthcare (Basel) 2021; 9:healthcare9050543. [PMID: 34066917 PMCID: PMC8148436 DOI: 10.3390/healthcare9050543] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/22/2021] [Accepted: 04/27/2021] [Indexed: 01/09/2023] Open
Abstract
Background: Psoriasis is a chronic dermatological condition characterized by lesions on extensor surfaces, hands, feet, and genital areas. Chronic renal failure is often associated with metabolic syndrome and inflammatory conditions, such as psoriasis. Case report: In this paper, we report a patient with stage-three chronic renal failure that improved his renal condition after treatment with ixekizumab, an anti-IL17A drug used in the treatment of various cutaneous and rheumatological conditions. Conclusions: IL17A blockage may help to treat various autoimmune and inflammatory conditions, such as psoriasis, that may lead to renal impairment. Further investigation is necessary in order to prove the effectiveness of this drug in renal conditions.
Collapse
|
17
|
Robinson PC, Liew DFL, Liew JW, Monaco C, Richards D, Shivakumar S, Tanner HL, Feldmann M. The Potential for Repurposing Anti-TNF as a Therapy for the Treatment of COVID-19. MED 2020; 1:90-102. [PMID: 33294881 PMCID: PMC7713589 DOI: 10.1016/j.medj.2020.11.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19) currently has few effective treatments. Given the uncertainty surrounding the effectiveness and uptake of a vaccine, it is important that the search for treatments continue. An exaggerated inflammatory state is likely responsible for much of the morbidity and mortality in COVID-19. Elevated levels of tumor necrosis factor (TNF), a key pro-inflammatory cytokine, have been shown to be associated with increased COVID-19 mortality. In patients with rheumatoid arthritis, TNF blockade reduces not only biologically active TNF but other pro-inflammatory cytokines important in COVID-19 hyperinflammation. Observational data from patients already on anti-TNF therapy show a reduced rate of COVID-19 poor outcomes and death compared with other immune-suppressing therapies. Anti-TNF has a long history of safe use, including in special at-risk populations, and is widely available. The case to adequately assess anti-TNF as a treatment for COVID-19 is compelling.
Collapse
Affiliation(s)
- Philip C Robinson
- University of Queensland Faculty of Medicine, Herston, Queensland, Australia
- Department of Medicine, Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Herston, Queensland, Australia
| | - David F L Liew
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
- Department of Clinical Pharmacology and Therapeutics, Austin Health, Heidelberg, Victoria, Australia
| | - Jean W Liew
- Section of Rheumatology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford OX3 7LD, UK
| | - Duncan Richards
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford OX3 7LD, UK
- Oxford Clinical Trials Research Unit, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford OX3 7LD, UK
| | - Senthuran Shivakumar
- Department of Clinical Pharmacology and Therapeutics, Austin Health, Heidelberg, Victoria, Australia
| | - Helen L Tanner
- University of Queensland Faculty of Medicine, Herston, Queensland, Australia
- Department of Medicine, Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Herston, Queensland, Australia
| | - Marc Feldmann
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford OX3 7LD, UK
| |
Collapse
|
18
|
Diena D, Priora M, Barreca A, Parisi S, Colla L, Biancone L, Fusaro E. Double Glomerulonephritis in a Patient with Ankylosing Spondylitis Treated with Biologic Agent: Extrarticolar Involvement or Anti-Tumor Necrosis Factor Alpha Injury? A Case-Based Review. CLINICAL MEDICINE INSIGHTS-CASE REPORTS 2020; 13:1179547620974672. [PMID: 33281463 PMCID: PMC7683916 DOI: 10.1177/1179547620974672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/24/2020] [Indexed: 11/15/2022]
Abstract
With the widespreading use of biologic drugs, reports of renal injury are increasing, most of which belong to the spectrum of secondary autoimmune syndromes. We present the case of a young man affected by Ankylosing Spondylitis, treated with tumor necrosis factor alpha inhibitors (Anti-TNF) that develop a peculiar renal damage: a coexistence of 2 glomerulonephritis due to different noxae, an IgA nephropaty with a Membranous nephropathy. The first one probably related to the rheumatologic disease, the second one related to Anti-TNF. Despite the underlying mechanisms, the renal involvement both related to Ankylosing Spondylitis and secondary to biologic treatment are currently rare and not predictable. Regular control of renal function and urinalysis during treatment with anti-TNF is mandatory. A concomitant treatment with Disease Modifying Anti Rheumatic Drugs or eventually a low dose of steroids may prevent the formation of anti-drug antibodies and could limit the renal damage related to this phenomenon.
Collapse
Affiliation(s)
- Davide Diena
- Division of Nephrology Dialysis and Transplantation, Department of Medical Sciences, University of Turin, Italy University Hospital "Città della Salute e della Scienza," Turin, Italy
| | - Marta Priora
- Division of Rheumatology, University Hospital "Città della Salute e della Scienza," Turin, Italy
| | - Antonella Barreca
- Division of Pathology, Department of Medical Sciences, University Hospital "Città della Salute e della Scienza," Turin, Italy
| | - Simone Parisi
- Division of Rheumatology, University Hospital "Città della Salute e della Scienza," Turin, Italy
| | - Loredana Colla
- Division of Nephrology Dialysis and Transplantation, Department of Medical Sciences, University of Turin, Italy University Hospital "Città della Salute e della Scienza," Turin, Italy
| | - Luigi Biancone
- Division of Nephrology Dialysis and Transplantation, Department of Medical Sciences, University of Turin, Italy University Hospital "Città della Salute e della Scienza," Turin, Italy
| | - Enrico Fusaro
- Division of Rheumatology, University Hospital "Città della Salute e della Scienza," Turin, Italy
| |
Collapse
|
19
|
Mannon EC, O'Connor PM. Alkali supplementation as a therapeutic in chronic kidney disease: what mediates protection? Am J Physiol Renal Physiol 2020; 319:F1090-F1104. [PMID: 33166183 DOI: 10.1152/ajprenal.00343.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Sodium bicarbonate (NaHCO3) has been recognized as a possible therapy to target chronic kidney disease (CKD) progression. Several small clinical trials have demonstrated that supplementation with NaHCO3 or other alkalizing agents slows renal functional decline in patients with CKD. While the benefits of NaHCO3 treatment have been thought to result from restoring pH homeostasis, a number of studies have now indicated that NaHCO3 or other alkalis may provide benefit regardless of the presence of metabolic acidosis. These data have raised questions as to how NaHCO3 protects the kidneys. To date, the physiological mechanism(s) that mediates the reported protective effect of NaHCO3 in CKD remain unclear. In this review, we first examine the evidence from clinical trials in support of a beneficial effect of NaHCO3 and other alkali in slowing kidney disease progression and their relationship to acid-base status. Then, we discuss the physiological pathways that have been proposed to underlie these renoprotective effects and highlight strengths and weaknesses in the data supporting each pathway. Finally, we discuss how answering key questions regarding the physiological mechanism(s) mediating the beneficial actions of NaHCO3 therapy in CKD is likely to be important in the design of future clinical trials. We conclude that basic research in animal models is likely to be critical in identifying the physiological mechanisms underlying the benefits of NaHCO3 treatment in CKD. Gaining an understanding of these pathways may lead to the improved implementation of NaHCO3 as a therapy in CKD and perhaps other disease states.
Collapse
Affiliation(s)
- Elinor C Mannon
- Department of Physiology, Augusta University, Augusta, Georgia
| | - Paul M O'Connor
- Department of Physiology, Augusta University, Augusta, Georgia
| |
Collapse
|
20
|
Hessman CL, Hildebrandt J, Shah A, Brandt S, Bock A, Frye BC, Raffetseder U, Geffers R, Brunner-Weinzierl MC, Isermann B, Mertens PR, Lindquist JA. YB-1 Interferes with TNFα-TNFR Binding and Modulates Progranulin-Mediated Inhibition of TNFα Signaling. Int J Mol Sci 2020; 21:ijms21197076. [PMID: 32992926 PMCID: PMC7583764 DOI: 10.3390/ijms21197076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/23/2022] Open
Abstract
Inflammation and an influx of macrophages are common elements in many diseases. Among pro-inflammatory cytokines, tumor necrosis factor α (TNFα) plays a central role by amplifying the cytokine network. Progranulin (PGRN) is a growth factor that binds to TNF receptors and interferes with TNFα-mediated signaling. Extracellular PGRN is processed into granulins by proteases released from immune cells. PGRN exerts anti-inflammatory effects, whereas granulins are pro-inflammatory. The factors coordinating these ambivalent functions remain unclear. In our study, we identify Y-box binding protein-1 (YB-1) as a candidate for this immune-modulating activity. Using a yeast-2-hybrid assay with YB-1 protein as bait, clones encoding for progranulin were selected using stringent criteria for strong interaction. We demonstrate that at physiological concentrations, YB-1 interferes with the binding of TNFα to its receptors in a dose-dependent manner using a flow cytometry-based binding assay. We show that YB-1 in combination with progranulin interferes with TNFα-mediated signaling, supporting the functionality with an NF-κB luciferase reporter assay. Together, we show that YB-1 displays immunomodulating functions by affecting the binding of TNFα to its receptors and influencing TNFα-mediated signaling via its interaction with progranulin.
Collapse
Affiliation(s)
- Christopher L. Hessman
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University, 39120 Magdeburg, Germany; (C.L.H.); (J.H.); (A.S.); (S.B.); (A.B.)
| | - Josephine Hildebrandt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University, 39120 Magdeburg, Germany; (C.L.H.); (J.H.); (A.S.); (S.B.); (A.B.)
| | - Aneri Shah
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University, 39120 Magdeburg, Germany; (C.L.H.); (J.H.); (A.S.); (S.B.); (A.B.)
| | - Sabine Brandt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University, 39120 Magdeburg, Germany; (C.L.H.); (J.H.); (A.S.); (S.B.); (A.B.)
| | - Antonia Bock
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University, 39120 Magdeburg, Germany; (C.L.H.); (J.H.); (A.S.); (S.B.); (A.B.)
| | - Björn C. Frye
- Department of Nephrology and Clinical Immunology, RWTH Aachen University, 52074 Aachen, Germany; (B.C.F.); (U.R.)
| | - Ute Raffetseder
- Department of Nephrology and Clinical Immunology, RWTH Aachen University, 52074 Aachen, Germany; (B.C.F.); (U.R.)
| | - Robert Geffers
- Genome Analytics Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany;
| | | | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany;
| | - Peter R. Mertens
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University, 39120 Magdeburg, Germany; (C.L.H.); (J.H.); (A.S.); (S.B.); (A.B.)
- Correspondence: (P.R.M.); (J.A.L.); Tel.: +49-391-6713236 (P.R.M.); +49-391-6724703 (J.A.L.)
| | - Jonathan A. Lindquist
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University, 39120 Magdeburg, Germany; (C.L.H.); (J.H.); (A.S.); (S.B.); (A.B.)
- Correspondence: (P.R.M.); (J.A.L.); Tel.: +49-391-6713236 (P.R.M.); +49-391-6724703 (J.A.L.)
| |
Collapse
|
21
|
Chebotareva NV, Gulyaev SV, Androsova TV, Popova EN, Gurova DV, Novikov PI, Milovanova LY, Moiseev SV. [Clinicopatological variants and risk factors for chronic kidney disease in rheumatoid arthritis]. TERAPEVT ARKH 2020; 92:55-60. [PMID: 32598776 DOI: 10.26442/00403660.2020.05.000604] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Indexed: 11/22/2022]
Abstract
Recent studies have shown a high risk of chronic kidney disease and associated cardiovascular complications in patients with rheumatoid arthritis (RA), which determines the prognosis. However, the prevalence of chronic kidney disease (CKD) in RA has not been established in the Russians. AIM Study was to examine the prevalence, risk factors and histological variants of CKD in RA. MATERIALS AND METHODS 180 patients with rheumatoid arthritis were observed in the Tareev clinic of nephrology, for the period from 2014 to 2019 years. Age, gender, duration of RA, drug therapy, ESR, CRP, DAS28, renal function, proteinuria, histological variants were analyzed. Of the common population risk factors for CKD arterial hypertension, weight index, serum lipids and glucose levels were also assessed. RESULTS The prevalence of CKD in RA was 19.7%. Age, presence and stage of arterial hypertension, an increase in body mass index, as well as high rates of disease activity ESR, CRP, DAS28 score and duration of RA were risk factors of CKD in RA. Age, duration of the disease, stage of AH and hypercholesterolemia were risk factors in multifactorial regression analysis. Amyloidosis was the most common histologic pattern (50.0%), followed by chronic glomerulonephritis (30.4%) and tubulo-interstitial nephritis (19.6%). Among chronic glomerulonephritis mesangial glomerulonephritis was the most frequent. Renal amyloidosis was associated with a duration of RA, presence of systemic symptoms and CRP level. An isolated decrease in GFR of less than 60 ml/min was detected in 31 (36.0%) out of 86 patients. Сonclusion. The risk factors for CKD in patients with RA are activity and duration of the disease In addition to common population factors. Amyloidosis was the most common histologic pattern associated with duration of RA and inflammatory proteins levels.
Collapse
Affiliation(s)
- N V Chebotareva
- Sechenov First Moscow State Medical University (Sechenov University)
| | - S V Gulyaev
- Sechenov First Moscow State Medical University (Sechenov University)
| | - T V Androsova
- Sechenov First Moscow State Medical University (Sechenov University)
| | - E N Popova
- Sechenov First Moscow State Medical University (Sechenov University)
| | - D V Gurova
- Sechenov First Moscow State Medical University (Sechenov University)
| | - P I Novikov
- Sechenov First Moscow State Medical University (Sechenov University)
| | - L Y Milovanova
- Sechenov First Moscow State Medical University (Sechenov University)
| | - S V Moiseev
- Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
22
|
Maghfour J, Elliott E, Gill F, Stumpf B, Murina A. Effect of biologic drugs on renal function in psoriasis patients with chronic kidney disease. J Am Acad Dermatol 2019; 82:1249-1251. [PMID: 31881298 DOI: 10.1016/j.jaad.2019.12.043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 12/02/2019] [Accepted: 12/13/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Jalal Maghfour
- Tulane University School of Medicine, Department of Dermatology, New Orleans, Louisiana
| | - Erika Elliott
- Tulane University School of Medicine, Department of Dermatology, New Orleans, Louisiana
| | - Frances Gill
- Tulane University School of Medicine, Department of Dermatology, New Orleans, Louisiana
| | - Brittany Stumpf
- Tulane University School of Medicine, Department of Dermatology, New Orleans, Louisiana
| | - Andrea Murina
- Tulane University School of Medicine, Department of Dermatology, New Orleans, Louisiana.
| |
Collapse
|
23
|
Chebotareva NV, Guliaev SV, Androsova TV, Milivanova LU. Chronic kidney disease in rheumatoid arthritis patients: prevalence, risks factors, histopathological variants. TERAPEVT ARKH 2019; 91:129-133. [DOI: 10.26442/00403660.2019.05.000255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Indexed: 01/04/2023]
Abstract
The present review is focused on risk factors of chronic kidney disease in rheumatoid arthritis (RA). According to recent data, the chronic kidney disease (CKD) in RA patients is more often than at patients without RA. It is closely associated with risk of cardiovascular disease and high mortality. Besides of general population risk factors of CKD, the activity of the disease is independent predictors of reduction in glomerular filtration rate less than 60 ml/min/1.73 m2. In the review, histopathological variants and mechanisms of CKD on basis of international experience are also considered. Suppression of inflammation by basic therapy of RA and biological therapy have changed outcomes RA, prevalence, and structure of kidney involvement in recent years.
Collapse
|
24
|
Toxines urémiques de moyen poids moléculaire : un véritable regain d’intérêt. Nephrol Ther 2019; 15:82-90. [DOI: 10.1016/j.nephro.2018.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/02/2018] [Indexed: 01/20/2023]
|
25
|
Tam LS, Wei JCC, Aggarwal A, Baek HJ, Cheung PP, Chiowchanwisawakit P, Dans L, Gu J, Hagino N, Kishimoto M, Reyes HM, Soroosh S, Stebbings S, Whittle S, Yeap SS, Lau CS. 2018 APLAR axial spondyloarthritis treatment recommendations. Int J Rheum Dis 2019; 22:340-356. [PMID: 30816645 DOI: 10.1111/1756-185x.13510] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Despite the availability of axial spondyloarthritis (SpA) recommendations proposed by various rheumatology societies, we considered that a region-specific guideline was of substantial added value to clinicians of the Asia-Pacific region, given the wide variations in predisposition to infections and other patient factors, local practice patterns, and access to treatment across countries. MATERIALS AND METHODS Systematic reviews were undertaken of English-language articles published between 2000 and 2016, identified from MEDLINE using PubMed, EMBASE and Cochrane databases. The strength of available evidence was graded using the Grading of Recommendations, Assessment, Development and Evaluations (GRADE) approach. Recommendations were developed through consensus using the Delphi technique. RESULTS Fourteen axial SpA treatment recommendations were developed based on evidence summaries and consensus. The first 2 recommendations cover non-pharmacological approaches to management. Recommendations 3 to 5 describe the following: the use of non-steroidal anti-inflammatory drugs as first-line symptomatic treatment; the avoidance of long-term corticosteroid use; and the utility of conventional synthetic disease-modifying anti-rheumatic drugs (csDMARDs) for peripheral or extra-articular manifestations. Recommendation 6 refers to the indications for biological DMARDs (bDMARDs). Recommendation 7 deals specifically with screening for infections endemic to Asia, prior to use of bDMARDs. Recommendations 7 to 13 cover the role of bDMARDs in the treatment of active axial SpA and include related issues such as continuing therapy and use in special populations. Recommendation 14 deals with the utility of surgical intervention in axial SpA. CONCLUSION These recommendations provide up-to-date guidance for treatment of axial SpA to help meet the needs of patients and clinicians in the Asia-Pacific region.
Collapse
Affiliation(s)
- Lai Shan Tam
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - James Cheng-Chung Wei
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Amita Aggarwal
- Department of Clinical Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Han Joo Baek
- Division of Rheumatology, Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Peter P Cheung
- Division of Rheumatology, National University Hospital and Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | | | - Leonila Dans
- Department of Pediatrics and Clinical Epidemiology, Philippine General Hospital, University of the Philippines, Manila, Philippines
| | - Jieruo Gu
- Department of Rheumatology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Noboru Hagino
- Division of Hematology and Rheumatology, Teikyo University Chiba Medical Center, Chiba, Japan
| | - Mitsumasa Kishimoto
- Immuno-Rheumatology Center, St Luke`s International Hospital, St Luke`s International University, Tokyo, Japan
| | - Heizel Manapat Reyes
- Division of Rheumatology, Department of Medicine, Philippine General Hospital, University of the Philippines, Manila, Philippines
| | - Soosan Soroosh
- AJA University of Medical Sciences, Rheumatology Research Center, Tehran, Iran
| | - Simon Stebbings
- Department of Medicine Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Samuel Whittle
- The Queen Elizabeth Hospital, University of Adelaide, Adelaide, South Australia, Australia
| | - Swan Sim Yeap
- Department of Medicine, Subang Jaya Medical Centre, Subang Jaya, Malaysia
| | - Chak Sing Lau
- Division of Rheumatology and Clinical Immunology, Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
26
|
Smykiewicz P, Segiet A, Keag M, Żera T. Proinflammatory cytokines and ageing of the cardiovascular-renal system. Mech Ageing Dev 2018; 175:35-45. [DOI: 10.1016/j.mad.2018.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 07/01/2018] [Accepted: 07/19/2018] [Indexed: 12/11/2022]
|
27
|
Kim SK, Choe JY. Gender Is a Risk Factor for Annual Decline in Estimated Glomerular Filtration Rate in Patients Treated with Biological DMARDs in Rheumatoid Arthritis and Ankylosing Spondylitis: a Retrospective Observational Study. J Korean Med Sci 2018; 33:e188. [PMID: 30034303 PMCID: PMC6052330 DOI: 10.3346/jkms.2018.33.e188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 05/08/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND This study identified the risk factors of changes in renal function in patients with rheumatoid arthritis (RA) and ankylosing spondylitis (AS) treated with biological disease-modifying anti-rheumatic drugs (bDMARDs). METHODS We retrospectively enrolled patients with RA (n = 293) and AS (n = 125) treated with bDMARDs. The estimated glomerular filter rate (eGFR) using the Modification of Diet in Renal Disease equation was applied for assessment of annual changes in renal function between initiation and last visit after bDMARD therapy. The annual change in eGFR was used as an indicator for change in renal function. Statistical significance was assessed by Mann-Whitney test, Spearman's correlation coefficient, and multivariate linear regression analysis. RESULTS The positive annual change in eGFR in women was significantly noted, compared to that in men (P = 0.004). The annual change in eGFR was different between men and women (P = 0.038) in RA, but not in AS patients (P = 0.126). In multivariate linear regression analysis, women patients and increased serum creatinine at baseline were closely associated with positive annual change in eGFR in both RA and AS patients. In RA patients, younger age and lower ESR level were considered risk factors of positive annual change in eGFR (P = 0.013 and P = 0.022, respectively). However, disease duration and duration of bDMARD use were not associated with annual change in eGFR. CONCLUSION This study found that gender, especially men, might be responsible for annual decline in eGFR in RA and AS patients treated with bDMARDs.
Collapse
Affiliation(s)
- Seong-Kyu Kim
- Division of Rheumatology, Department of Internal Medicine, Arthritis and Autoimmunity Research Center, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Jung-Yoon Choe
- Division of Rheumatology, Department of Internal Medicine, Arthritis and Autoimmunity Research Center, Daegu Catholic University School of Medicine, Daegu, Korea
| |
Collapse
|
28
|
Vanholder R, Van Laecke S, Glorieux G, Verbeke F, Castillo-Rodriguez E, Ortiz A. Deleting Death and Dialysis: Conservative Care of Cardio-Vascular Risk and Kidney Function Loss in Chronic Kidney Disease (CKD). Toxins (Basel) 2018; 10:E237. [PMID: 29895722 PMCID: PMC6024824 DOI: 10.3390/toxins10060237] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 05/11/2018] [Indexed: 02/07/2023] Open
Abstract
The uremic syndrome, which is the clinical expression of chronic kidney disease (CKD), is a complex amalgam of accelerated aging and organ dysfunctions, whereby cardio-vascular disease plays a capital role. In this narrative review, we offer a summary of the current conservative (medical) treatment options for cardio-vascular and overall morbidity and mortality risk in CKD. Since the progression of CKD is also associated with a higher cardio-vascular risk, we summarize the interventions that may prevent the progression of CKD as well. We pay attention to established therapies, as well as to novel promising options. Approaches that have been considered are not limited to pharmacological approaches but take into account lifestyle measures and diet as well. We took as many randomized controlled hard endpoint outcome trials as possible into account, although observational studies and post hoc analyses were included where appropriate. We also considered health economic aspects. Based on this information, we constructed comprehensive tables summarizing the available therapeutic options and the number and kind of studies (controlled or not, contradictory outcomes or not) with regard to each approach. Our review underscores the scarcity of well-designed large controlled trials in CKD. Nevertheless, based on the controlled and observational data, a therapeutic algorithm can be developed for this complex and multifactorial condition. It is likely that interventions should be aimed at targeting several modifiable factors simultaneously.
Collapse
Affiliation(s)
- Raymond Vanholder
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, 9000 Ghent, Belgium.
| | - Steven Van Laecke
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, 9000 Ghent, Belgium.
| | - Griet Glorieux
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, 9000 Ghent, Belgium.
| | - Francis Verbeke
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, 9000 Ghent, Belgium.
| | | | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, 28040 Madrid, Spain.
| |
Collapse
|
29
|
The Effects of MicroRNAs on Key Signalling Pathways and Epigenetic Modification in Fibroblast-Like Synoviocytes of Rheumatoid Arthritis. Mediators Inflamm 2018; 2018:9013124. [PMID: 29861659 PMCID: PMC5971246 DOI: 10.1155/2018/9013124] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 02/03/2018] [Accepted: 04/08/2018] [Indexed: 01/08/2023] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression at the posttranscriptional level via direct binding to the 3′-untranslated region (UTR) of target mRNAs. Emerging evidence shows that miRNAs play crucial roles in controlling and modulating immune system-related diseases. This review focuses on the role played by miRNAs in fibroblast-like synoviocytes (FLS), which is a key cellular component within synovia, during the establishment and maintenance of rheumatoid arthritis (RA), a systemic inflammatory autoimmune disease. It also provides an overview and classification of known functional miRNAs in RA FLS and summarizes the potential uses of these small molecules in RA diagnosis and treatment.
Collapse
|
30
|
Ray SC, Baban B, Tucker MA, Seaton AJ, Chang KC, Mannon EC, Sun J, Patel B, Wilson K, Musall JB, Ocasio H, Irsik D, Filosa JA, Sullivan JC, Marshall B, Harris RA, O'Connor PM. Oral NaHCO 3 Activates a Splenic Anti-Inflammatory Pathway: Evidence That Cholinergic Signals Are Transmitted via Mesothelial Cells. THE JOURNAL OF IMMUNOLOGY 2018; 200:3568-3586. [PMID: 29661827 DOI: 10.4049/jimmunol.1701605] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 03/10/2018] [Indexed: 12/15/2022]
Abstract
We tested the hypothesis that oral NaHCO3 intake stimulates splenic anti-inflammatory pathways. Following oral NaHCO3 loading, macrophage polarization was shifted from predominantly M1 (inflammatory) to M2 (regulatory) phenotypes, and FOXP3+CD4+ T-lymphocytes increased in the spleen, blood, and kidneys of rats. Similar anti-inflammatory changes in macrophage polarization were observed in the blood of human subjects following NaHCO3 ingestion. Surprisingly, we found that gentle manipulation to visualize the spleen at midline during surgical laparotomy (sham splenectomy) was sufficient to abolish the response in rats and resulted in hypertrophy/hyperplasia of the capsular mesothelial cells. Thin collagenous connections lined by mesothelial cells were found to connect to the capsular mesothelium. Mesothelial cells in these connections stained positive for the pan-neuronal marker PGP9.5 and acetylcholine esterase and contained many ultrastructural elements, which visually resembled neuronal structures. Both disruption of the fragile mesothelial connections or transection of the vagal nerves resulted in the loss of capsular mesothelial acetylcholine esterase staining and reduced splenic mass. Our data indicate that oral NaHCO3 activates a splenic anti-inflammatory pathway and provides evidence that the signals that mediate this response are transmitted to the spleen via a novel neuronal-like function of mesothelial cells.
Collapse
Affiliation(s)
- Sarah C Ray
- Department of Physiology, Augusta University, Augusta, GA 30912
| | - Babak Baban
- Department of Oral Biology, Augusta University, Augusta, GA 30912
| | - Matthew A Tucker
- Georgia Prevention Institute, Augusta University, Augusta, GA 30912; and
| | - Alec J Seaton
- Department of Physiology, Augusta University, Augusta, GA 30912
| | - Kyu Chul Chang
- Department of Physiology, Augusta University, Augusta, GA 30912
| | - Elinor C Mannon
- Department of Physiology, Augusta University, Augusta, GA 30912
| | - Jingping Sun
- Department of Physiology, Augusta University, Augusta, GA 30912
| | - Bansari Patel
- Department of Physiology, Augusta University, Augusta, GA 30912
| | - Katie Wilson
- Department of Physiology, Augusta University, Augusta, GA 30912
| | | | - Hiram Ocasio
- Department of Physiology, Augusta University, Augusta, GA 30912
| | - Debra Irsik
- Department of Physiology, Augusta University, Augusta, GA 30912
| | | | | | - Brendan Marshall
- Department of Cell Biology and Anatomy, Augusta University, Augusta, GA 30912
| | - Ryan A Harris
- Georgia Prevention Institute, Augusta University, Augusta, GA 30912; and
| | - Paul M O'Connor
- Department of Physiology, Augusta University, Augusta, GA 30912;
| |
Collapse
|
31
|
Sumida K, Molnar MZ, Potukuchi PK, Hassan F, Thomas F, Yamagata K, Kalantar-Zadeh K, Kovesdy CP. Treatment of rheumatoid arthritis with biologic agents lowers the risk of incident chronic kidney disease. Kidney Int 2018; 93:1207-1216. [PMID: 29409725 DOI: 10.1016/j.kint.2017.11.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 11/05/2017] [Accepted: 11/30/2017] [Indexed: 02/06/2023]
Abstract
Rheumatoid arthritis is associated with reduced kidney function, possibly due to chronic inflammation or the use of nephrotoxic therapies. However, little is known about the effects of using the newer novel non-nephrotoxic biologic agents on the risk of incident chronic kidney disease (CKD). To study this we used a cohort of 20,757 United States veterans diagnosed with rheumatoid arthritis with an estimated glomerular filtration rate (eGFR) of 60 mL/min/1.73m2 or more, recruited between October 2004 and September 2006, and followed through 2013. The associations of biologic use with incident CKD (eGFR under 60 with a decrease of at least 25% from baseline, and eGFR under 45 mL/min/1.73m2) and change in eGFR (<-3, -3 to <0 [reference], and ≥0 mL/min/1.73m2/year) were examined in propensity-matched patients based on their likelihood to initiate biologic treatment, using Cox models and multinomial logistic regression models, respectively. Among 20,757 patients, 4,617 started biologic therapy. In the propensity-matched cohort, patients treated (versus not treated) with biologic agents had a lower risk of incident CKD (hazard ratios 0.95, 95% confidence interval [0.82-1.10] and 0.71 [0.53-0.94] for decrease in eGFR under 60 and under 45 mL/min/1.73m2, respectively) and progressive eGFR decline (multinomial odds ratios [95% CI] for eGFR slopes <-3 and ≥0 [versus -3 to <0] mL/min/1.73m2/year, 0.67 [0.58-0.79] and 0.76 [0.69-0.83], respectively). A significant deceleration of eGFR decline was also observed after biologic administration in patients treated with biologics (-1.0 versus -0.4 [mL/min/1.73m2/year] before and after biologic use). Thus, biologic agent administration was independently associated with lower risk of incident CKD and progressive eGFR decline.
Collapse
Affiliation(s)
- Keiichi Sumida
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA; Nephrology Center, Toranomon Hospital Kajigaya, Kanagawa, Japan; Department of Nephrology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Miklos Z Molnar
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Transplantation and Surgery, Semmelweis University, Budapest, Hungary; Division of Transplant Surgery, Methodist University Hospital Transplant Institute, Memphis, Tennessee, USA; Division of Transplant Surgery, Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Praveen K Potukuchi
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Fatima Hassan
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Fridtjof Thomas
- Division of Biostatistics, Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Kunihiro Yamagata
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kamyar Kalantar-Zadeh
- Harold Simmons Center for Chronic Disease Research and Epidemiology, Division of Nephrology and Hypertension, University of California-Irvine, Orange, California, USA
| | - Csaba P Kovesdy
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA; Nephrology Section, Memphis VA Medical Center, Memphis, Tennessee, USA.
| |
Collapse
|
32
|
Babino G, Esposito M, Bianchi L, Giunta A. Long-term treatment with etanercept monotherapy in a hemodialyzed patient with moderate-to-severe plaque-type psoriasis and psoriatic arthritis. GIORN ITAL DERMAT V 2017; 153:882-884. [PMID: 29144096 DOI: 10.23736/s0392-0488.17.05586-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Graziella Babino
- Department of Dermatology, Tor Vergata University, Rome, Italy -
| | - Maria Esposito
- Department of Dermatology, Tor Vergata University, Rome, Italy
| | - Luca Bianchi
- Department of Dermatology, Tor Vergata University, Rome, Italy
| | | |
Collapse
|
33
|
Distinct Patterns of Dietary Intake in Different Functional Classes of Patients With Rheumatoid Arthritis. TOP CLIN NUTR 2017. [DOI: 10.1097/tin.0000000000000099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
34
|
Inflammatory Cytokines as Uremic Toxins: "Ni Son Todos Los Que Estan, Ni Estan Todos Los Que Son". Toxins (Basel) 2017; 9:toxins9040114. [PMID: 28333114 PMCID: PMC5408188 DOI: 10.3390/toxins9040114] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/13/2017] [Accepted: 03/16/2017] [Indexed: 01/03/2023] Open
Abstract
Chronic kidney disease is among the fastest growing causes of death worldwide. An increased risk of all-cause and cardiovascular death is thought to depend on the accumulation of uremic toxins when glomerular filtration rate falls. In addition, the circulating levels of several markers of inflammation predict mortality in patients with chronic kidney disease. Indeed, a number of cytokines are listed in databases of uremic toxins and uremic retention solutes. They include inflammatory cytokines (IL-1β, IL-18, IL-6, TNFα), chemokines (IL-8), and adipokines (adiponectin, leptin and resistin), as well as anti-inflammatory cytokines (IL-10). We now critically review the cytokines that may be considered uremic toxins. We discuss the rationale to consider them uremic toxins (mechanisms underlying the increased serum levels and evidence supporting their contribution to CKD manifestations), identify gaps in knowledge, discuss potential therapeutic implications to be tested in clinical trials in order to make this knowledge useful for the practicing physician, and identify additional cytokines, cytokine receptors and chemokines that may fulfill the criteria to be considered uremic toxins, such as sIL-6R, sTNFR1, sTNFR2, IL-2, CXCL12, CX3CL1 and others. In addition, we suggest that IL-10, leptin, adiponectin and resistin should not be considered uremic toxins toxins based on insufficient or contradictory evidence of an association with adverse outcomes in humans or preclinical data not consistent with a causal association.
Collapse
|
35
|
Chiu HY, Huang HL, Li CH, Chen HA, Yeh CL, Chiu SH, Lin WC, Cheng YP, Tsai TF, Ho SY. Increased Risk of Chronic Kidney Disease in Rheumatoid Arthritis Associated with Cardiovascular Complications - A National Population-Based Cohort Study. PLoS One 2015; 10:e0136508. [PMID: 26406879 PMCID: PMC4583248 DOI: 10.1371/journal.pone.0136508] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/04/2015] [Indexed: 11/22/2022] Open
Abstract
Background and Objectives There have been few large population-based studies of the association between rheumatoid arthritis (RA) and chronic kidney disease (CKD) and glomerulonephritis. This nationwide cohort study investigated the risks of developing CKD and glomerulonephritis in patients with RA, and the associated risks for cardiovascular complications. Methods From the Taiwan National Health Insurance Research Database, we identified a study cohort of 12,579 patients with RA and randomly selected 37,737 subjects without RA as a control cohort. Each subject was individually followed for up for 5 years, and the risk of CKD was analyzed using Cox proportional hazards regression models. Results During the follow-up period, after adjusting for traditional cardiovascular risk factors RA was independently associated with a significantly increased risk of CKD (adjusted hazard ratio [aHR] 1.31; 95% confidence interval [CI] 1.23–1.40) and glomerulonephritis (aHR 1.55; 95% CI 1.37–1.76). Increased risk of CKD was also associated with the use of non-steroidal anti-inflammatory drugs, cyclosporine, glucocorticoids, mycophenolate mofetil, and cyclophosphamide. Patients with comorbidities had even greater increased risk of CKD. Moreover, RA patients with concurrent CKD had significantly higher likelihood of developing ischemic heart disease and stroke. Conclusions RA patients had higher risk of developing CKD and glomerulonephritis, independent of traditional cardiovascular risk factors. Their increased risk of CKD may be attributed to glomerulonephritis, chronic inflammation, comorbidities, and renal toxicity of antirheumatic drugs. Careful monitoring of renal function in RA patients and tight control of their comorbid diseases and cardiovascular risk factors are warranted.
Collapse
Affiliation(s)
- Hsien-Yi Chiu
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
- Department of Dermatology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Hui-Ling Huang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Chien-Hsun Li
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Hung-An Chen
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
| | - Chia-Lun Yeh
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
| | - Shih-Hsiang Chiu
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
| | - Wei-Chun Lin
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
| | - Yu-Pin Cheng
- Department of Dermatology, Cathay General Hospital, Taipei, Taiwan
| | - Tsen-Fang Tsai
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- * E-mail: (T-FT); (S-YH)
| | - Shinn-Ying Ho
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- * E-mail: (T-FT); (S-YH)
| |
Collapse
|