1
|
Du J, Zhao X, Ding X, Ren Q, Wang H, Han Q, Song C, Wang X, Zhang D, Zhu H. Gut microbiota and their metabolites in hemodialysis patients. Chin Med J (Engl) 2025; 138:502-504. [PMID: 39809700 PMCID: PMC11845176 DOI: 10.1097/cm9.0000000000003423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Indexed: 01/16/2025] Open
Affiliation(s)
- Junxia Du
- Department of Nephrology, First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People’s Liberation Army, Beijing 100853, China
| | - Xiaolin Zhao
- Department of Nephrology, First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Xiaonan Ding
- Department of Nephrology, First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People’s Liberation Army, Beijing 100853, China
| | - Qinqin Ren
- Department of Nephrology, First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Haoran Wang
- Department of Nephrology, First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Qiuxia Han
- Department of Nephrology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Chenwen Song
- Department of Nephrology, First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People’s Liberation Army, Beijing 100853, China
| | - Xiaochen Wang
- Department of Nephrology, First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People’s Liberation Army, Beijing 100853, China
| | - Dong Zhang
- Department of Nephrology, First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Hanyu Zhu
- Department of Nephrology, First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| |
Collapse
|
2
|
Flynn CK, Adams JB, Krajmalnik-Brown R, Khoruts A, Sadowsky MJ, Nirmalkar K, Takyi E, Whiteley P. Review of Elevated Para-Cresol in Autism and Possible Impact on Symptoms. Int J Mol Sci 2025; 26:1513. [PMID: 40003979 PMCID: PMC11855632 DOI: 10.3390/ijms26041513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Para-cresol (p-cresol), and its primary human metabolite p-cresol sulfate (pCS), are among the most studied gut-derived metabolites relevant to autism spectrum disorder (ASD). P-cresol is produced by bacterial modification of phenylalanine or tyrosine and is one of many potentially deleterious metabolites produced by the gut microbiota. Seventeen studies have observed p-cresol and/or p-cresol sulfate as being higher in the urine of children with autism spectrum disorder (ASD) vs. controls. P-cresol has harmful effects on the body, including within the gut, brain, kidneys, liver, immune system, and mitochondria. Some of these effects may contribute to autism and comorbid symptoms. In the gut, p-cresol acts as an antibiotic, altering the gut microbiome to favor the bacteria that produce it. In the mitochondria, p-cresol disrupts ATP production and increases oxidative stress, which is also common in autism. In the brain, p-cresol impairs neuronal development. P-cresol inactivates dopamine beta-hydroxylase, which converts dopamine to noradrenaline. P-cresol sulfate impairs kidney function and is linked to chronic kidney disease (CKD), which is more common in ASD adults. P-cresol also interferes with immune function. Three animal studies have demonstrated that p-cresol causes autism-related symptoms in mice, and that mice can be recovered by the administration of fecal microbiota transplant from healthy mice. Similarly, it was found that microbiota transplant therapy treatment in children with ASD significantly reduced p-cresol sulfate levels to normal and led to significant improvements in gastrointestinal (GI) and ASD symptoms. In summary, p-cresol and pCS likely contribute to ASD core symptoms in a substantial subset of children with ASD.
Collapse
Affiliation(s)
- Christina K. Flynn
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ 85287, USA; (C.K.F.)
- School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85287, USA
| | - James B. Adams
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ 85287, USA; (C.K.F.)
- School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85287, USA
| | - Rosa Krajmalnik-Brown
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ 85287, USA; (C.K.F.)
- School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, AZ 85281, USA
| | - Alexander Khoruts
- Department of Medicine, Division of Gastroenterology, Center for Immunology and BioTechnology Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael J. Sadowsky
- Department of Medicine and BioTechnology Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Khemlal Nirmalkar
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ 85287, USA; (C.K.F.)
| | - Evelyn Takyi
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ 85287, USA; (C.K.F.)
| | | |
Collapse
|
3
|
Devraj K, Kulkarni O, Liebner S. Regulation of the blood-brain barrier function by peripheral cues in health and disease. Metab Brain Dis 2024; 40:61. [PMID: 39671124 PMCID: PMC11645320 DOI: 10.1007/s11011-024-01468-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/12/2024] [Indexed: 12/14/2024]
Abstract
The blood-brain barrier (BBB) is formed by microvascular endothelial cells which are ensembled with pericytes, astrocytes, microglia and neurons in the neurovascular unit (NVU) that is crucial for neuronal function. Given that the NVU and the BBB are highly dynamic and regulated structures, their integrity is continuously challenged by intrinsic and extrinsic factors. Herein, factors from peripheral organs such as gonadal and adrenal hormones may influence vascular function also in CNS endothelial cells in a sex- and age-dependent manner. The communication between the periphery and the CNS likely takes place in specific areas of the brain among which the circumventricular organs have a central position due to their neurosensory or neurosecretory function, owing to physiologically leaky blood vessels. In acute and chronic pathological conditions like liver, kidney, pulmonary disease, toxins and metabolites are generated that reach the brain via the circulation and may directly or indirectly affect BBB functionality via the activation of the immunes system. For example, chronic kidney disease (CKD) currently affects more than 840 million people worldwide and is likely to increase along with western world comorbidities of the cardio-vascular system in continuously ageing societies. Toxins leading to the uremic syndrome, may further lead to neurological complications such as cognitive impairment and uremic encephalopathy. Here we summarize the effects of hormones, toxins and inflammatory reactions on the brain vasculature, highlighting the urgent demand for mechanistically exploring the communication between the periphery and the CNS, focusing on the BBB as a last line of defense for brain protection.
Collapse
Affiliation(s)
- Kavi Devraj
- Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Hyderabad, 500078, Telangana, India.
| | - Onkar Kulkarni
- Metabolic Disorders and Neuroscience Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad, 500078, Telangana, India
| | - Stefan Liebner
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Partner Site Frankfurt, Frankfurt am Main, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt/Mainz, Frankfurt, Germany.
| |
Collapse
|
4
|
Du J, Zhao X, Ding X, Han Q, Duan Y, Ren Q, Wang H, Song C, Wang X, Zhang D, Zhu H. The Role of the Gut Microbiota in Complications among Hemodialysis Patients. Microorganisms 2024; 12:1878. [PMID: 39338552 PMCID: PMC11434415 DOI: 10.3390/microorganisms12091878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
The composition of the gut microbiota varies among end-stage renal disease (ESRD) patients on the basis of their mode of renal replacement therapy (RRT), with notably more pronounced dysbiosis occurring in those undergoing hemodialysis (HD). Interventions such as dialysis catheters, unstable hemodynamics, strict dietary restrictions, and pharmacotherapy significantly alter the intestinal microenvironment, thus disrupting the gut microbiota composition in HD patients. The gut microbiota may influence HD-related complications, including cardiovascular disease (CVD), infections, anemia, and malnutrition, through mechanisms such as bacterial translocation, immune regulation, and the production of gut microbial metabolites, thereby affecting both the quality of life and the prognosis of patients. This review focuses on alterations in the gut microbiota and its metabolites in HD patients. Additionally, understanding the impact of the gut microbiota on the complications of HD could provide insights into the development of novel treatment strategies to prevent or alleviate complications in HD patients.
Collapse
Affiliation(s)
- Junxia Du
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Xiaolin Zhao
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Xiaonan Ding
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Qiuxia Han
- Department of Nephrology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yingjie Duan
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Qinqin Ren
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Haoran Wang
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Chenwen Song
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Xiaochen Wang
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Dong Zhang
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Hanyu Zhu
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| |
Collapse
|
5
|
Xu C, Tsihlis G, Chau K, Trinh K, Rogers NM, Julovi SM. Novel Perspectives in Chronic Kidney Disease-Specific Cardiovascular Disease. Int J Mol Sci 2024; 25:2658. [PMID: 38473905 PMCID: PMC10931927 DOI: 10.3390/ijms25052658] [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: 01/19/2024] [Revised: 02/18/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Chronic kidney disease (CKD) affects > 10% of the global adult population and significantly increases the risk of cardiovascular disease (CVD), which remains the leading cause of death in this population. The development and progression of CVD-compared to the general population-is premature and accelerated, manifesting as coronary artery disease, heart failure, arrhythmias, and sudden cardiac death. CKD and CV disease combine to cause multimorbid cardiorenal syndrome (CRS) due to contributions from shared risk factors, including systolic hypertension, diabetes mellitus, obesity, and dyslipidemia. Additional neurohormonal activation, innate immunity, and inflammation contribute to progressive cardiac and renal deterioration, reflecting the strong bidirectional interaction between these organ systems. A shared molecular pathophysiology-including inflammation, oxidative stress, senescence, and hemodynamic fluctuations characterise all types of CRS. This review highlights the evolving paradigm and recent advances in our understanding of the molecular biology of CRS, outlining the potential for disease-specific therapies and biomarker disease detection.
Collapse
Affiliation(s)
- Cuicui Xu
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; (C.X.); (K.T.)
| | - George Tsihlis
- Renal and Transplantation Medicine, Westmead Hospital, Westmead, NSW 2145, Australia;
| | - Katrina Chau
- Department of Renal Services, Blacktown Hospital, Blacktown, NSW 2148, Australia;
- Blacktown Clinical School, School of Medicine, Western Sydney University, Sydney, NSW 2148, Australia
| | - Katie Trinh
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; (C.X.); (K.T.)
- Department of Renal Services, Blacktown Hospital, Blacktown, NSW 2148, Australia;
| | - Natasha M. Rogers
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; (C.X.); (K.T.)
- Renal and Transplantation Medicine, Westmead Hospital, Westmead, NSW 2145, Australia;
- Faculty of Medicine and Health, The University of Sydney, Science Rd., Camperdown, NSW 2050, Australia
| | - Sohel M. Julovi
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; (C.X.); (K.T.)
- Faculty of Medicine and Health, The University of Sydney, Science Rd., Camperdown, NSW 2050, Australia
| |
Collapse
|
6
|
Nemet I, Funabashi M, Li XS, Dwidar M, Sangwan N, Skye SM, Romano KA, Cajka T, Needham BD, Mazmanian SK, Hajjar AM, Rey FE, Fiehn O, Tang WHW, Fischbach MA, Hazen SL. Microbe-derived uremic solutes enhance thrombosis potential in the host. mBio 2023; 14:e0133123. [PMID: 37947418 PMCID: PMC10746243 DOI: 10.1128/mbio.01331-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/25/2023] [Indexed: 11/12/2023] Open
Abstract
IMPORTANCE Alterations in gut microbial composition and function have been linked to numerous diseases. Identifying microbial pathways responsible for producing molecules that adversely impact the host is an important first step in the development of therapeutic interventions. Here, we first use large-scale clinical observations to link blood levels of defined microbial products to cardiovascular disease risks. Notably, the previously identified uremic toxins p-cresol sulfate and indoxyl sulfate were shown to predict 5-year mortality risks. After identifying the microbes and microbial enzymes involved in the generation of these uremic toxins, we used bioengineering technologies coupled with colonization of germ-free mice to show that the gut microbial genes that generate p-cresol and indole are sufficient to confer p-cresol sulfate and indoxyl sulfate formation, and a pro-thrombotic phenotype in vivo. The findings and tools developed serve as a critical step in both the study and targeting of these gut microbial pathways in vivo.
Collapse
Affiliation(s)
- Ina Nemet
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland, Ohio, USA
- Center for Microbiome & Human Health, Cleveland Clinic, Cleveland, Ohio, USA
| | - Masanori Funabashi
- Department of Bioengineering, Stanford University, Stanford, California, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, California, USA
- ChEM-H Institute, Stanford University, Stanford, California, USA
| | - Xinmin S. Li
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland, Ohio, USA
- Center for Microbiome & Human Health, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mohammed Dwidar
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland, Ohio, USA
- Center for Microbiome & Human Health, Cleveland Clinic, Cleveland, Ohio, USA
| | - Naseer Sangwan
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland, Ohio, USA
- Center for Microbiome & Human Health, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sarah M. Skye
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland, Ohio, USA
- Center for Microbiome & Human Health, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kymberleigh A. Romano
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland, Ohio, USA
- Center for Microbiome & Human Health, Cleveland Clinic, Cleveland, Ohio, USA
| | - Tomas Cajka
- West Coast Metabolomics Center, University of California, Davis, California, USA
| | - Brittany D. Needham
- Departments of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA
| | - Sarkis K. Mazmanian
- Departments of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA
| | - Adeline M. Hajjar
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland, Ohio, USA
- Center for Microbiome & Human Health, Cleveland Clinic, Cleveland, Ohio, USA
| | - Federico E. Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, California, USA
| | - W. H. Wilson Tang
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland, Ohio, USA
- Center for Microbiome & Human Health, Cleveland Clinic, Cleveland, Ohio, USA
- Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Michael A. Fischbach
- Department of Bioengineering, Stanford University, Stanford, California, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, California, USA
- ChEM-H Institute, Stanford University, Stanford, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | - Stanley L. Hazen
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland, Ohio, USA
- Center for Microbiome & Human Health, Cleveland Clinic, Cleveland, Ohio, USA
- Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
7
|
Caldarelli M, Franza L, Rio P, Gasbarrini A, Gambassi G, Cianci R. Gut-Kidney-Heart: A Novel Trilogy. Biomedicines 2023; 11:3063. [PMID: 38002063 PMCID: PMC10669427 DOI: 10.3390/biomedicines11113063] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
The microbiota represents a key factor in determining health and disease. Its role in inflammation and immunological disorders is well known, but it is also involved in several complex conditions, ranging from neurological to psychiatric, from gastrointestinal to cardiovascular diseases. It has recently been hypothesized that the gut microbiota may act as an intermediary in the close interaction between kidneys and the cardiovascular system, leading to the conceptualization of the "gut-kidney-heart" axis. In this narrative review, we will discuss the impact of the gut microbiota on each system while also reviewing the available data regarding the axis itself. We will also describe the role of gut metabolites in this complex interplay, as well as potential therapeutical perspectives.
Collapse
Affiliation(s)
- Mario Caldarelli
- Department of Translational Medicine and Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (M.C.); (P.R.); (A.G.); (G.G.)
| | - Laura Franza
- Emergency Medicine Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy;
| | - Pierluigi Rio
- Department of Translational Medicine and Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (M.C.); (P.R.); (A.G.); (G.G.)
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (M.C.); (P.R.); (A.G.); (G.G.)
| | - Giovanni Gambassi
- Department of Translational Medicine and Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (M.C.); (P.R.); (A.G.); (G.G.)
| | - Rossella Cianci
- Department of Translational Medicine and Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (M.C.); (P.R.); (A.G.); (G.G.)
| |
Collapse
|
8
|
Falconi CA, Fogaça-Ruiz F, da Silva JV, Neres-Santos RS, Sanz CL, Nakao LS, Stinghen AEM, Junho CVC, Carneiro-Ramos MS. Renocardiac Effects of p-Cresyl Sulfate Administration in Acute Kidney Injury Induced by Unilateral Ischemia and Reperfusion Injury In Vivo. Toxins (Basel) 2023; 15:649. [PMID: 37999512 PMCID: PMC10674368 DOI: 10.3390/toxins15110649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
The precise mechanisms underlying the cardiovascular complications due to acute kidney injury (AKI) and the retention of uremic toxins like p-cresyl sulfate (PCS) remain incompletely understood. The objective of this study was to evaluate the renocardiac effects of PCS administration in animals subjected to AKI induced by ischemia and reperfusion (IR) injury. C57BL6 mice were subjected to distinct protocols: (i) administration with PCS (20, 40, or 60 mg/L/day) for 15 days and (ii) AKI due to unilateral IR injury associated with PCS administration for 15 days. The 20 mg/L dose of PCS led to a decrease in renal mass, an increase in the gene expression of Cystatin C and kidney injury molecule 1 (KIM-1), and a decrease in the α-actin in the heart. During AKI, PCS increased the renal injury biomarkers compared to control; however, it did not exacerbate these markers. Furthermore, PCS did not enhance the cardiac hypertrophy observed after 15 days of IR. An increase, but not potentialized, in the cardiac levels of interleukin (IL)-1β and IL-6 in the IR group treated with PCS, as well as in the injured kidney, was also noticed. In short, PCS administration did not intensify kidney injury, inflammation, and cardiac outcomes after AKI.
Collapse
Affiliation(s)
- Carlos Alexandre Falconi
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-170, SP, Brazil; (C.A.F.); (F.F.-R.); (J.V.d.S.); (R.S.N.-S.)
| | - Fernanda Fogaça-Ruiz
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-170, SP, Brazil; (C.A.F.); (F.F.-R.); (J.V.d.S.); (R.S.N.-S.)
| | - Jéssica Verônica da Silva
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-170, SP, Brazil; (C.A.F.); (F.F.-R.); (J.V.d.S.); (R.S.N.-S.)
| | - Raquel Silva Neres-Santos
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-170, SP, Brazil; (C.A.F.); (F.F.-R.); (J.V.d.S.); (R.S.N.-S.)
| | - Carmen Lucía Sanz
- Department of Basic Pathology, Universidade Federal do Paraná, Curitiba 81530-000, PR, Brazil; (C.L.S.); (L.S.N.)
| | - Lia Sumie Nakao
- Department of Basic Pathology, Universidade Federal do Paraná, Curitiba 81530-000, PR, Brazil; (C.L.S.); (L.S.N.)
| | - Andréa Emília Marques Stinghen
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, PR, Brazil;
| | - Carolina Victoria Cruz Junho
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-170, SP, Brazil; (C.A.F.); (F.F.-R.); (J.V.d.S.); (R.S.N.-S.)
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Marcela Sorelli Carneiro-Ramos
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-170, SP, Brazil; (C.A.F.); (F.F.-R.); (J.V.d.S.); (R.S.N.-S.)
| |
Collapse
|
9
|
André C, Bodeau S, Kamel S, Bennis Y, Caillard P. The AKI-to-CKD Transition: The Role of Uremic Toxins. Int J Mol Sci 2023; 24:16152. [PMID: 38003343 PMCID: PMC10671582 DOI: 10.3390/ijms242216152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
After acute kidney injury (AKI), renal function continues to deteriorate in some patients. In a pro-inflammatory and profibrotic environment, the proximal tubules are subject to maladaptive repair. In the AKI-to-CKD transition, impaired recovery from AKI reduces tubular and glomerular filtration and leads to chronic kidney disease (CKD). Reduced kidney secretion capacity is characterized by the plasma accumulation of biologically active molecules, referred to as uremic toxins (UTs). These toxins have a role in the development of neurological, cardiovascular, bone, and renal complications of CKD. However, UTs might also cause CKD as well as be the consequence. Recent studies have shown that these molecules accumulate early in AKI and contribute to the establishment of this pro-inflammatory and profibrotic environment in the kidney. The objective of the present work was to review the mechanisms of UT toxicity that potentially contribute to the AKI-to-CKD transition in each renal compartment.
Collapse
Affiliation(s)
- Camille André
- Department of Clinical Pharmacology, Amiens Medical Center, 80000 Amiens, France; (S.B.); (Y.B.)
- GRAP Laboratory, INSERM UMR 1247, University of Picardy Jules Verne, 80000 Amiens, France
| | - Sandra Bodeau
- Department of Clinical Pharmacology, Amiens Medical Center, 80000 Amiens, France; (S.B.); (Y.B.)
- MP3CV Laboratory, UR UPJV 7517, University of Picardy Jules Verne, 80000 Amiens, France; (S.K.); (P.C.)
| | - Saïd Kamel
- MP3CV Laboratory, UR UPJV 7517, University of Picardy Jules Verne, 80000 Amiens, France; (S.K.); (P.C.)
- Department of Clinical Biochemistry, Amiens Medical Center, 80000 Amiens, France
| | - Youssef Bennis
- Department of Clinical Pharmacology, Amiens Medical Center, 80000 Amiens, France; (S.B.); (Y.B.)
- MP3CV Laboratory, UR UPJV 7517, University of Picardy Jules Verne, 80000 Amiens, France; (S.K.); (P.C.)
| | - Pauline Caillard
- MP3CV Laboratory, UR UPJV 7517, University of Picardy Jules Verne, 80000 Amiens, France; (S.K.); (P.C.)
- Department of Nephrology, Dialysis and Transplantation, Amiens Medical Center, 80000 Amiens, France
| |
Collapse
|
10
|
Nemet I, Li XS, Haghikia A, Li L, Wilcox J, Romano KA, Buffa JA, Witkowski M, Demuth I, König M, Steinhagen-Thiessen E, Bäckhed F, Fischbach MA, Tang WHW, Landmesser U, Hazen SL. Atlas of gut microbe-derived products from aromatic amino acids and risk of cardiovascular morbidity and mortality. Eur Heart J 2023; 44:3085-3096. [PMID: 37342006 PMCID: PMC10481777 DOI: 10.1093/eurheartj/ehad333] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 05/01/2023] [Accepted: 05/16/2023] [Indexed: 06/22/2023] Open
Abstract
AIMS Precision microbiome modulation as a novel treatment strategy is a rapidly evolving and sought goal. The aim of this study is to determine relationships among systemic gut microbial metabolite levels and incident cardiovascular disease risks to identify gut microbial pathways as possible targets for personalized therapeutic interventions. METHODS AND RESULTS Stable isotope dilution mass spectrometry methods to quantitatively measure aromatic amino acids and their metabolites were used to examine sequential subjects undergoing elective diagnostic cardiac evaluation in two independent cohorts with longitudinal outcome data [US (n = 4000) and EU (n = 833) cohorts]. It was also used in plasma from humans and mice before vs. after a cocktail of poorly absorbed antibiotics to suppress gut microbiota. Multiple aromatic amino acid-derived metabolites that originate, at least in part, from gut bacteria are associated with incident (3-year) major adverse cardiovascular event (MACE) risks (myocardial infarction, stroke, or death) and all-cause mortality independent of traditional risk factors. Key gut microbiota-derived metabolites associated with incident MACE and poorer survival risks include: (i) phenylacetyl glutamine and phenylacetyl glycine (from phenylalanine); (ii) p-cresol (from tyrosine) yielding p-cresol sulfate and p-cresol glucuronide; (iii) 4-OH-phenyllactic acid (from tyrosine) yielding 4-OH-benzoic acid and 4-OH-hippuric acid; (iv) indole (from tryptophan) yielding indole glucuronide and indoxyl sulfate; (v) indole-3-pyruvic acid (from tryptophan) yielding indole-3-lactic acid and indole-3-acetyl-glutamine, and (vi) 5-OH-indole-3-acetic acid (from tryptophan). CONCLUSION Key gut microbiota-generated metabolites derived from aromatic amino acids independently associated with incident adverse cardiovascular outcomes are identified, and thus will help focus future studies on gut-microbial metabolic outputs relevant to host cardiovascular health.
Collapse
Affiliation(s)
- Ina Nemet
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Xinmin S Li
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Arash Haghikia
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin 12203, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin 10785, Germany
- Biomedical Innovation Academy, Berlin Institute of Health (BIH), Berlin 10178, Germany
| | - Lin Li
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Jennifer Wilcox
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Kymberleigh A Romano
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Jennifer A Buffa
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Marco Witkowski
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Ilja Demuth
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
- Center for Regenerative Therapies, Berlin Institute of Health (BIH), Berlin 13353, Germany
| | - Maximilian König
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
| | | | - Fredrik Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg SE-413 45, Sweden
| | - Michael A Fischbach
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
- ChEM-H Institute, Stanford University, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - W H Wilson Tang
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ulf Landmesser
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin 12203, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin 10785, Germany
- Biomedical Innovation Academy, Berlin Institute of Health (BIH), Berlin 10178, Germany
| | - Stanley L Hazen
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
11
|
Abstract
Homeostasis is a prerequisite for health. When homeostasis becomes disrupted, dysfunction occurs. This is especially the case for the gut microbiota, which under normal conditions lives in symbiosis with the host. As there are as many microbial cells in and on our body as human cells, it is unlikely they would not contribute to health or disease. The gut bacterial metabolism generates numerous beneficial metabolites but also uremic toxins and their precursors, which are transported into the circulation. Barrier function in the intestine, the heart, and the kidneys regulates metabolite transport and concentration and plays a role in inter-organ and inter-organism communication via small molecules. This communication is analyzed from the perspective of the remote sensing and signaling theory, which emphasizes the role of a large network of multispecific, oligospecific, and monospecific transporters and enzymes in regulating small-molecule homeostasis. The theory provides a systems biology framework for understanding organ cross talk and microbe-host communication involving metabolites, signaling molecules, nutrients, antioxidants, and uremic toxins. This remote small-molecule communication is critical for maintenance of homeostasis along the gut-heart-kidney axis and for responding to homeostatic perturbations. Chronic kidney disease is characterized by gut dysbiosis and accumulation of toxic metabolites. This slowly impacts the body, affecting the cardiovascular system and contributing to the progression of kidney dysfunction, which in its turn influences the gut microbiota. Preserving gut homeostasis and barrier functions or restoring gut dysbiosis and dysfunction could be a minimally invasive way to improve patient outcomes and quality of life in many diseases, including cardiovascular and kidney disease.
Collapse
Affiliation(s)
- Griet Glorieux
- Nephrology Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Gent, Belgium (G.G., R.V., F.V.)
| | - Sanjay K Nigam
- Department of Pediatrics (S.K.N.), University of California San Diego, La Jolla, CA
- Division of Nephrology, Department of Medicine (S.K.N.), University of California San Diego, La Jolla, CA
| | - Raymond Vanholder
- Nephrology Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Gent, Belgium (G.G., R.V., F.V.)
| | - Francis Verbeke
- Nephrology Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Gent, Belgium (G.G., R.V., F.V.)
| |
Collapse
|
12
|
Liao YC, Chang CC, Chen CY, Liu CC, Liao CC, Shih YRV, Lin CS. Preoperative renal insufficiency predicts postoperative adverse outcomes in a mixed surgical population: a retrospective matched cohort study using the NSQIP database. Int J Surg 2023; 109:752-759. [PMID: 36974714 PMCID: PMC10389524 DOI: 10.1097/js9.0000000000000278] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 01/27/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND The incidence of chronic kidney disease is increasing, but most cases are not diagnosed until the accidental finding of abnormal laboratory data or the presentation of severe symptoms. Patients with chronic kidney disease are reported to have an increased risk of postoperative mortality and morbidities, but previous studies mainly targeted populations undergoing cardiovascular surgery. The authors aimed to evaluate the risk of postoperative mortality and complications in a surgical population with preoperative renal insufficiency (RI). MATERIALS AND METHODS This retrospective cohort study used data from the National Surgical Quality Improvement Program database between 2013 and 2018 to evaluate the risk of postoperative morbidity and mortality in the surgical population. Patients with estimated glomerular filtration rate less than 60 ml/min/1.73 m 2 were defined as the RI group. Propensity score matching methods and multivariate logistic regression were used to calculate the risk of postoperative morbidity and mortality. RESULTS After propensity score matching, 502 281 patients were included in the RI and non-RI groups. The RI group had a higher risk of 30-day in-hospital mortality (odds ratio: 1.54, 95% CI: 1.49-1.58) than the non-RI group. The RI group was associated with a higher risk of postoperative complications, including myocardial infarction, stroke, pneumonia, septic shock, and postoperative bleeding. The RI group was also associated with an increased risk of prolonged ventilator use for over 48 h, readmission, and reoperation. CONCLUSION Patients with preoperative RI have an increased risk of postoperative 30-day mortality and complications. RI group patients with current dialysis, estimated glomerular filtration rate less than or equal to 30 ml/min/1.73 m 2 or concomitant anemia had an elevated risk of postoperative mortality.
Collapse
Affiliation(s)
| | - Chuen-Chau Chang
- Department of Anesthesiology
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Yu Chen
- Department of Anesthesiology
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Chung Liu
- Department of Anesthesiology
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Chang Liao
- Department of Anesthesiology
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ru Vernon Shih
- Department of Orthopedic Surgery, Duke University, Durham, North Carolina, USA
| | - Chao-Shun Lin
- Department of Anesthesiology
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
13
|
Caggiano G, Stasi A, Franzin R, Fiorentino M, Cimmarusti MT, Deleonardis A, Palieri R, Pontrelli P, Gesualdo L. Fecal Microbiota Transplantation in Reducing Uremic Toxins Accumulation in Kidney Disease: Current Understanding and Future Perspectives. Toxins (Basel) 2023; 15:toxins15020115. [PMID: 36828429 PMCID: PMC9965504 DOI: 10.3390/toxins15020115] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/21/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
During the past decades, the gut microbiome emerged as a key player in kidney disease. Dysbiosis-related uremic toxins together with pro-inflammatory mediators are the main factors in a deteriorating kidney function. The toxicity of uremic compounds has been well-documented in a plethora of pathophysiological mechanisms in kidney disease, such as cardiovascular injury (CVI), metabolic dysfunction, and inflammation. Accumulating data on the detrimental effect of uremic solutes in kidney disease supported the development of many strategies to restore eubiosis. Fecal microbiota transplantation (FMT) spread as an encouraging treatment for different dysbiosis-associated disorders. In this scenario, flourishing studies indicate that fecal transplantation could represent a novel treatment to reduce the uremic toxins accumulation. Here, we present the state-of-the-art concerning the application of FMT on kidney disease to restore eubiosis and reverse the retention of uremic toxins.
Collapse
|
14
|
Stachulski AV, Knausenberger TBA, Shah SN, Hoyles L, McArthur S. A host-gut microbial amino acid co-metabolite, p-cresol glucuronide, promotes blood-brain barrier integrity in vivo. Tissue Barriers 2023; 11:2073175. [PMID: 35596559 PMCID: PMC9870004 DOI: 10.1080/21688370.2022.2073175] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The sequential activity of gut microbial and host processes can exert a powerful modulatory influence on dietary components, as exemplified by the metabolism of the amino acids tyrosine and phenylalanine to p-cresol by gut microbes, and then to p-cresol glucuronide (pCG) by host enzymes. Although such glucuronide conjugates are classically thought to be biologically inert, there is accumulating evidence that this may not always be the case. We investigated the activity of pCG, studying its interactions with the cerebral vasculature and the brain in vitro and in vivo. Male C57Bl/6 J mice were used to assess blood-brain barrier (BBB) permeability and whole-brain transcriptomic changes in response to pCG treatment. Effects were then further explored using the human cerebromicrovascular endothelial cell line hCMEC/D3, assessing paracellular permeability, transendothelial electrical resistance and barrier protein expression. Mice exposed to pCG showed reduced BBB permeability and significant changes in whole-brain transcriptome expression. Surprisingly, treatment of hCMEC/D3 cells with pCG had no notable effects until co-administered with bacterial lipopolysaccharide, at which point it was able to prevent the permeabilizing effects of endotoxin. Further analysis suggested that pCG acts as an antagonist at the principal lipopolysaccharide receptor TLR4. The amino acid phase II metabolic product pCG is biologically active at the BBB, antagonizing the effects of constitutively circulating lipopolysaccharide. These data add to the growing literature showing glucuronide conjugates to be more than merely metabolic waste products and highlight the complexity of gut microbe to host communication pathways underlying the gut-brain axis.
Collapse
Affiliation(s)
- Andrew V. Stachulski
- Department of Chemistry, Robert Robinson Laboratories, University of Liverpool, Liverpool, UK,contact Lesley Hoyles Department of Bioscience, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK
| | - Tobias B-A Knausenberger
- Institute of Dentistry, Faculty of Medicine & Dentistry, Queen Mary, University of London, Blizard Institute, London, UK
| | - Sita N. Shah
- Institute of Dentistry, Faculty of Medicine & Dentistry, Queen Mary, University of London, Blizard Institute, London, UK
| | - Lesley Hoyles
- Department of Bioscience, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK,CONTACT Simon McArthur Institute of Dentistry, Faculty of Medicine & Dentistry, Queen Mary, University of London, Blizard Institute, 4, Newark Street, LondonE1 2AT, UK
| | - Simon McArthur
- Institute of Dentistry, Faculty of Medicine & Dentistry, Queen Mary, University of London, Blizard Institute, London, UK,Andrew V. Stachulski Department of Chemistry, Robert Robinson Laboratories, University of Liverpool, LiverpoolL69 7ZD, UK
| |
Collapse
|
15
|
|
16
|
Xia J, Hou Y, Cai A, Xu Y, Yang W, Huang M, Mou S. An integrated co-expression network analysis reveals novel genetic biomarkers for immune cell infiltration in chronic kidney disease. Front Immunol 2023; 14:1129524. [PMID: 36875100 PMCID: PMC9981626 DOI: 10.3389/fimmu.2023.1129524] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/06/2023] [Indexed: 02/19/2023] Open
Abstract
Background Chronic kidney disease (CKD) is characterized by persistent damage to kidney function or structure. Progression to end-stage leads to adverse effects on multiple systems. However, owing to its complex etiology and long-term cause, the molecular basis of CKD is not completely known. Methods To dissect the potential important molecules during the progression, based on CKD databases from Gene Expression Omnibus, we used weighted gene co-expression network analysis (WGCNA) to identify the key genes in kidney tissues and peripheral blood mononuclear cells (PBMC). Correlation analysis of these genes with clinical relevance was evaluated based on Nephroseq. Combined with a validation cohort and receiver operating characteristic curve (ROC), we found the candidate biomarkers. The immune cell infiltration of these biomarkers was evaluated. The expression of these biomarkers was further detected in folic acid-induced nephropathy (FAN) murine model and immunohistochemical staining. Results In total, eight genes (CDCP1, CORO1C, DACH1, GSTA4, MAFB, TCF21, TGFBR3, and TGIF1) in kidney tissue and six genes (DDX17, KLF11, MAN1C1, POLR2K, ST14, and TRIM66) in PBMC were screened from co-expression network. Correlation analysis of these genes with serum creatinine levels and estimated glomerular filtration rate from Nephroseq showed a well clinical relevance. Validation cohort and ROC identified TCF21, DACH1 in kidney tissue and DDX17 in PBMC as biomarkers for the progression of CKD. Immune cell infiltration analysis revealed that DACH1 and TCF21 were correlated with eosinophil, activated CD8 T cell, activated CD4 T cell, while the DDX17 was correlated with neutrophil, type-2 T helper cell, type-1 T helper cell, mast cell, etc. FAN murine model and immunohistochemical staining confirmed that these three molecules can be used as genetic biomarkers to distinguish CKD patients from healthy people. Moreover, the increase of TCF21 in kidney tubules might play important role in the CKD progression. Discussion We identified three promising genetic biomarkers which could play important roles in the progression of CKD.
Collapse
Affiliation(s)
- Jia Xia
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yutong Hou
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Anxiang Cai
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingjie Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Yang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Masha Huang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shan Mou
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Colombo G, Astori E, Landoni L, Garavaglia ML, Altomare A, Lionetti MC, Gagliano N, Giustarini D, Rossi R, Milzani A, Dalle‐Donne I. Effects of the uremic toxin indoxyl sulphate on human microvascular endothelial cells. J Appl Toxicol 2022; 42:1948-1961. [PMID: 35854198 PMCID: PMC9796800 DOI: 10.1002/jat.4366] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 01/07/2023]
Abstract
Indoxyl sulphate (IS) is a uremic toxin accumulating in the plasma of chronic kidney disease (CKD) patients. IS accumulation induces side effects in the kidneys, bones and cardiovascular system. Most studies assessed IS effects on cell lines by testing higher concentrations than those measured in CKD patients. Differently, we exposed a human microvascular endothelial cell line (HMEC-1) to the IS concentrations measured in the plasma of healthy subjects (physiological) or CKD patients (pathological). Pathological concentrations reduced cell proliferation rate but did not increase long-term oxidative stress level. Indeed, total protein thiols decreased only after 24 h of exposure in parallel with an increased Nrf-2 protein expression. IS induced actin cytoskeleton rearrangement with formation of stress fibres. Proteomic analysis supported this hypothesis as many deregulated proteins are related to actin filaments organization or involved in the endothelial to mesenchymal transition. Interestingly, two proteins directly linked to cardiovascular diseases (CVD) in in vitro and in vivo studies underwent deregulation: COP9 signalosome complex subunit 9 and thrombomodulin. Future experiments will be needed to investigate the role of these proteins and the signalling pathways in which they are involved to clarify the possible link between CKD and CVD.
Collapse
Affiliation(s)
- Graziano Colombo
- Department of Biosciences (Department of Excellence 2018–2022)Università degli Studi di MilanoMilanItaly
| | - Emanuela Astori
- Department of Biosciences (Department of Excellence 2018–2022)Università degli Studi di MilanoMilanItaly
| | - Lucia Landoni
- Department of Biosciences (Department of Excellence 2018–2022)Università degli Studi di MilanoMilanItaly
| | - Maria L. Garavaglia
- Department of Biosciences (Department of Excellence 2018–2022)Università degli Studi di MilanoMilanItaly
| | - Alessandra Altomare
- Department of Pharmaceutical SciencesUniversità degli Studi di MilanoMilanItaly
| | - Maria C. Lionetti
- Department of Biosciences (Department of Excellence 2018–2022)Università degli Studi di MilanoMilanItaly
| | - Nicoletta Gagliano
- Department of Biomedical Sciences for HealthUniversità degli Studi di MilanoMilanItaly
| | - Daniela Giustarini
- Department of Life Sciences, Laboratory of Pharmacology and ToxicologyUniversity of SienaSienaItaly
| | - Ranieri Rossi
- Department of Life Sciences, Laboratory of Pharmacology and ToxicologyUniversity of SienaSienaItaly
| | - Aldo Milzani
- Department of Biosciences (Department of Excellence 2018–2022)Università degli Studi di MilanoMilanItaly
| | - Isabella Dalle‐Donne
- Department of Biosciences (Department of Excellence 2018–2022)Università degli Studi di MilanoMilanItaly
| |
Collapse
|
18
|
Berberine ameliorates chronic kidney disease through inhibiting the production of gut-derived uremic toxins in the gut microbiota. Acta Pharm Sin B 2022; 13:1537-1553. [PMID: 37139409 PMCID: PMC10149897 DOI: 10.1016/j.apsb.2022.12.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/08/2022] [Accepted: 11/18/2022] [Indexed: 12/24/2022] Open
Abstract
At present, clinical interventions for chronic kidney disease are very limited, and most patients rely on dialysis to sustain their lives for a long time. However, studies on the gut-kidney axis have shown that the gut microbiota is a potentially effective target for correcting or controlling chronic kidney disease. This study showed that berberine, a natural drug with low oral availability, significantly ameliorated chronic kidney disease by altering the composition of the gut microbiota and inhibiting the production of gut-derived uremic toxins, including p-cresol. Furthermore, berberine reduced the content of p-cresol sulfate in plasma mainly by lowering the abundance of g_Clostridium_sensu_stricto_1 and inhibiting the tyrosine-p-cresol pathway of the intestinal flora. Meanwhile, berberine increased the butyric acid producing bacteria and the butyric acid content in feces, while decreased the renal toxic trimethylamine N-oxide. These findings suggest that berberine may be a therapeutic drug with significant potential to ameliorate chronic kidney disease through the gut-kidney axis.
Collapse
|
19
|
Zhu L, Tong G, Yang F, Zhao Y, Chen G. The role of neuroimmune and inflammation in pediatric uremia-induced neuropathy. Front Immunol 2022; 13:1013562. [PMID: 36189322 PMCID: PMC9520989 DOI: 10.3389/fimmu.2022.1013562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Uremic neuropathy in children encompasses a wide range of central nervous system (CNS), peripheral nervous system (PNS), autonomic nervous system (ANS), and psychological abnormalities, which is associated with progressive renal dysfunction. Clinically, the diagnosis of uremic neuropathy in children is often made retrospectively when symptoms improve after dialysis or transplantation, due to there is no defining signs or laboratory and imaging findings. These neurological disorders consequently result in increased morbidity and mortality among children population, making uremia an urgent public health problem worldwide. In this review, we discuss the epidemiology, potential mechanisms, possible treatments, and the shortcomings of current research of uremic neuropathy in children. Mechanistically, the uremic neuropathy may be caused by retention of uremic solutes, increased oxidative stress, neurotransmitter imbalance, and disturbance of the blood-brain barrier (BBB). Neuroimmune, including the change of inflammatory factors and immune cells, may also play a crucial role in the progression of uremic neuropathy. Different from the invasive treatment of dialysis and kidney transplantation, intervention in neuroimmune and targeted anti-inflammatory therapy may provide a new insight for the treatment of uremia.
Collapse
Affiliation(s)
- Linfeng Zhu
- Department of Urology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Guoqin Tong
- Department of Neurology, The First People’s Hospital of XiaoShan District, Hangzhou, China
| | - Fan Yang
- Department of Urology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yijun Zhao
- Department of Urology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Guangjie Chen
- Department of Urology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- *Correspondence: Guangjie Chen,
| |
Collapse
|
20
|
Effects of Chronic Kidney Disease on Nanomechanics of the Endothelial Glycocalyx Are Mediated by the Mineralocorticoid Receptor. Int J Mol Sci 2022; 23:ijms231810659. [PMID: 36142571 PMCID: PMC9503126 DOI: 10.3390/ijms231810659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/31/2022] [Accepted: 09/03/2022] [Indexed: 02/03/2023] Open
Abstract
Endothelial mechanics control vascular reactivity and are regulated by the mineralocorticoid receptor (MR) and its downstream target, the epithelial Na+ channel (ENaC). Endothelial dysfunction is a hallmark of chronic kidney disease (CKD), but its mechanisms are poorly understood. We hypothesized that CKD disrupts endothelial mechanics in an MR/ENaC-dependent process. METHODS Primary human endothelial cells were cultured with uremic serum derived from children with stage 3-5 (predialysis) CKD or adult hemodialysis (HD) patients or healthy controls. The height and stiffness of the endothelial glycocalyx (eGC) and cortex were monitored by atomic force microscopy (AFM) using an ultrasensitive mechanical nanosensor. RESULTS In a stage-dependent manner, sera from children with CKD induced a significant increase in eGC and cortex stiffness and an incremental reduction of the eGC height. AFM measurements were significantly associated with individual pulse wave velocity and serum concentrations of gut-derived uremic toxins. Serum from HD patients increased MR expression and mechanical stiffness of the endothelial cortex, an effect reversed by MR and ENaC antagonists, decreased eNOS expression and NO bioavailability, and augmented monocyte adhesion. CONCLUSION These data indicate progressive structural damage of the endothelial surface with diminishing kidney function and identify the MR as a mediator of CKD-induced endothelial dysfunction.
Collapse
|
21
|
The Role of Gut-Derived, Protein-Bound Uremic Toxins in the Cardiovascular Complications of Acute Kidney Injury. Toxins (Basel) 2022; 14:toxins14050336. [PMID: 35622583 PMCID: PMC9143532 DOI: 10.3390/toxins14050336] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/04/2022] [Accepted: 05/07/2022] [Indexed: 02/04/2023] Open
Abstract
Acute kidney injury (AKI) is a frequent disease encountered in the hospital, with a higher incidence in intensive care units. Despite progress in renal replacement therapy, AKI is still associated with early and late complications, especially cardiovascular events and mortality. The role of gut-derived protein-bound uremic toxins (PBUTs) in vascular and cardiac dysfunction has been extensively studied during chronic kidney disease (CKD), in particular, that of indoxyl sulfate (IS), para-cresyl sulfate (PCS), and indole-3-acetic acid (IAA), resulting in both experimental and clinical evidence. PBUTs, which accumulate when the excretory function of the kidneys is impaired, have a deleterious effect on and cause damage to cardiovascular tissues. However, the link between PBUTs and the cardiovascular complications of AKI and the pathophysiological mechanisms potentially involved are unclear. This review aims to summarize available data concerning the participation of PBUTs in the early and late cardiovascular complications of AKI.
Collapse
|
22
|
Indoxyl sulfate- and P-cresol-induced monocyte adhesion and migration is mediated by integrin-linked kinase-dependent podosome formation. Exp Mol Med 2022; 54:226-238. [PMID: 35246616 PMCID: PMC8980039 DOI: 10.1038/s12276-022-00738-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 10/22/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular disease is an important cause of death in patients with chronic kidney disease (CKD). Protein-bound uremic toxins, such as p-cresyl and indoxyl sulfate (IS), are poorly removed during hemodialysis, leading to vascular endothelial dysfunction and leukocyte extravasation. These processes can be related to dynamic adhesion structures called podosomes. Several studies have indicated the role of integrin-linked kinase (ILK) in the accumulation of integrin-associated proteins in podosomes. Here, we investigated the involvement of ILK and podosome formation in the adhesion and extravasation of monocytes under p-cresol (pc) and IS exposure. Incubation of THP-1 human monocyte cells with these toxins upregulated ILK kinase activity. Together, both toxins increased cell adhesion, podosome formation, extracellular matrix degradation, and migration of THP-1 cells, whereas ILK depletion with specific small interfering RNAs suppressed these processes. Interestingly, F-actin colocalized with cortactin in podosome cores, while ILK was colocalized in podosome rings under toxin stimulation. Podosome Wiskott-Aldrich syndrome protein (WASP)-interacting protein (WIP) and AKT protein depletion demonstrated that monocyte adhesion depends on podosome formation and that the ILK/AKT signaling pathway is involved in these processes. Ex vivo experiments showed that both toxins induced adhesion and podosome formation in leukocytes from wild-type mice, whereas these effects were not observed in leukocytes of conditional ILK-knockdown animals. In summary, under pc and IS stimulation, monocytes increase podosome formation and transmigratory capacity through an ILK/AKT signaling pathway-dependent mechanism, which could lead to vascular injury. Therefore, ILK could be a potential therapeutic target for the treatment of vascular damage associated with CKD. An enzyme involved in organizing structural proteins into protrusions of the cell membrane helps facilitate the movement of white blood cells that occurs in chronic kidney disease and can lead to cardiovascular damage. Laura Calleros from the University of Alcalá, Madrid, Spain, and colleagues showed how toxic metabolites that build up in the bloodstream as kidneys fail induce the activity of an enzyme called integrin-linked kinase (ILK). This enzyme in turn spurs white blood cells to form protrusions called podosomes that make the cells more mobile, enabling them to move out of blood vessels and into surrounding tissues where they can cause inflammation -mediated injury. ILK inactivation reversed these effects. Therapies that target the enzyme could therefore help limit the cardiovascular complications of chronic kidney disease.
Collapse
|
23
|
p-Cresyl Sulfate Predicts Ischemic Stroke among Patients on Hemodialysis: A Prospective Cohort Study. DISEASE MARKERS 2022; 2022:1358419. [PMID: 35140818 PMCID: PMC8820900 DOI: 10.1155/2022/1358419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/21/2021] [Accepted: 12/30/2021] [Indexed: 11/17/2022]
Abstract
Background and Purpose. Hemodialysis patients face a higher risk of ischemic stroke. p-Cresyl sulfate is a typical protein-bound uremic toxin that contributes to chronic kidney disease and cardiovascular disease progression, as well as mortality in hemodialysis patients. The present study was aimed at elucidating the association between p-cresyl sulfate and the risk of ischemic stroke in hemodialysis patients. Method. Patients on hemodialysis over 6 months were enrolled in this prospective cohort study and were divided into 2 groups based on plasma p-cresyl sulfate level. The primary end point was the first episode of ischemic stroke during follow-up. The association between p-cresyl sulfate and ischemic stroke incidence was analyzed by Kaplan-Meier method and Cox proportional hazard model. Results. 220 patients were enrolled in this study. 44 patients experienced episodes of first ischemic stroke during follow-up for 87.8 (47.6-119.5) months. Kaplan-Meier analysis demonstrated that the incidence of ischemic stroke in the high p-cresyl sulfate group was significantly higher than that in the low p-cresyl sulfate group (Log-Rank
). Cox regression analysis as well proved that p-cresyl sulfate level was significantly associated with the first incidence of ischemic stroke (HR (hazard ratio) 2.332, 95% CI (95% confidence interval) 1.236-4.399,
). After being adjusted for other confounding risk factors, the results persisted significant (model 11: HR 2.061, 95% CI 1.030-4.125,
). Conclusion. Plasma p-cresyl sulfate predicts the first incidence of ischemic stroke in hemodialysis patients.
Collapse
|
24
|
Rosner MH, Reis T, Husain-Syed F, Vanholder R, Hutchison C, Stenvinkel P, Blankestijn PJ, Cozzolino M, Juillard L, Kashani K, Kaushik M, Kawanishi H, Massy Z, Sirich TL, Zuo L, Ronco C. Classification of Uremic Toxins and Their Role in Kidney Failure. Clin J Am Soc Nephrol 2021; 16:1918-1928. [PMID: 34233920 PMCID: PMC8729494 DOI: 10.2215/cjn.02660221] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Advances in our understanding of uremic retention solutes, and improvements in hemodialysis membranes and other techniques designed to remove uremic retention solutes, offer opportunities to readdress the definition and classification of uremic toxins. A consensus conference was held to develop recommendations for an updated definition and classification scheme on the basis of a holistic approach that incorporates physicochemical characteristics and dialytic removal patterns of uremic retention solutes and their linkage to clinical symptoms and outcomes. The major focus is on the removal of uremic retention solutes by hemodialysis. The identification of representative biomarkers for different classes of uremic retention solutes and their correlation to clinical symptoms and outcomes may facilitate personalized and targeted dialysis prescriptions to improve quality of life, morbidity, and mortality. Recommendations for areas of future research were also formulated, aimed at improving understanding of uremic solutes and improving outcomes in patients with CKD.
Collapse
Affiliation(s)
- Mitchell H. Rosner
- Division of Nephrology, University of Virginia Health System, Charlottesville, Virginia
| | - Thiago Reis
- Department of Nephrology, University of Brazil, Brasília, Brazil
- National Academy of Medicine, Rio de Janeiro, Brazil
| | - Faeq Husain-Syed
- Department of Internal Medicine II, Justus-Liebig-University Giessen, Giessen, Germany
| | - Raymond Vanholder
- Department of Internal Medicine and Pediatrics, University Hospital, Ghent, Belgium
| | - Colin Hutchison
- Faculty of Medicine, University of Queensland, Herston, Australia
- Department of Medicine, Hawke's Bay District Health Board, Hastings, New Zealand
| | - Peter Stenvinkel
- Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Peter J. Blankestijn
- Department of Nephrology and Hypertension, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Mario Cozzolino
- Renal Division, Università degli Studi di Milano, Milan, Italy
| | - Laurent Juillard
- University of Lyon, Villeurbanne, France
- Hôpital E. Herriot, Lyon, France
| | - Kianoush Kashani
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Manish Kaushik
- Department of Renal Medicine, Singapore General Hospital, Singapore, Singapore
| | - Hideki Kawanishi
- Department of Artificial Organs, Tsuchiya General Hospital, Hiroshima, Japan
| | - Ziad Massy
- INSERM U1018, Villejuif, France
- Service de Néphrologie et Dialyse, Hôpital Universitaire Ambroise Paré, Boulogne-Billancourt, France
| | - Tammy Lisa Sirich
- Nephrology Section, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
- Division of Nephrology, Stanford University School of Medicine, Stanford, California
| | - Li Zuo
- Department of Nephrology, Peking University People's Hospital, Beijing, China
| | - Claudio Ronco
- Department of Medicine, University of Padova, Padova, Italy
- Department of Nephrology, Dialysis and Transplantation, International Renal Research Institute of Vicenza, Vicenza, Italy
| |
Collapse
|
25
|
Asai A, Hatayama N, Kamiya K, Yamauchi M, Kinashi H, Yamaguchi M, Katsuno T, Nobata H, Watanabe K, Wakatsuki A, Aten J, Maruyama S, Ishimoto T, Hirai S, Naito M, Ito Y. Roles of glomerular endothelial hyaluronan in the development of proteinuria. Physiol Rep 2021; 9:e15019. [PMID: 34472715 PMCID: PMC8411502 DOI: 10.14814/phy2.15019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/01/2021] [Accepted: 08/04/2021] [Indexed: 12/29/2022] Open
Abstract
Vascular endothelial cells are covered with glycocalyx comprising heparan sulfate, hyaluronan, chondroitin sulfate, and associated proteins. Glomerular endothelial glycocalyx is involved in protecting against induction of proteinuria and structural damage, but the specific components in glycocalyx that represent therapeutic targets remain unclear. Anti-vascular endothelial growth factor (VEGF) therapy is associated with an increased risk of glomerular endothelial injury. This study investigated whether hyaluronan could provide a therapeutic target to protect against proteinuria. We conducted ex vivo and in vivo experiments to explore the effects of degrading glomerular hyaluronan by administering hyaluronidase and of supplementation with hyaluronan. We investigated hyaluronan expression using biotin-labeled hyaluronan-binding protein (HABP) in human kidney specimens or serum hyaluronan in endothelial injuries under inhibition of VEGF signaling. We directly demonstrated hyaluronan in glomerular endothelial layers using HABP staining. Ex vivo and in vivo experiments showed the development of proteinuria after digestion of hyaluronan in glomerular capillaries. Supplementation with hyaluronan after hyaluronidase treatment suppressed proteinuria. Mice in the in vivo study developed albuminuria after intraperitoneal injection of hyaluronidase with decreased glomerular hyaluronan and increased serum hyaluronan. In human kidneys with endothelial cell dysfunction and proteinuria due to inhibition of VEGF, glomerular expression of hyaluronan was reduced even in normal-appearing glomeruli. Serum hyaluronan levels were elevated in patients with pre-eclampsia with VEGF signaling inhibition. Our data suggest that hyaluronan itself plays crucial roles in preventing proteinuria and preserving the integrity of endothelial cells. Hyaluronan could provide a therapeutic target for preventing glomerular endothelial glycocalyx damage, including VEGF signaling inhibition.
Collapse
Affiliation(s)
- Akimasa Asai
- Department of Nephrology and RheumatologyAichi Medical UniversityNagakuteAichiJapan
| | - Naoyuki Hatayama
- Department of AnatomyAichi Medical University School of MedicineNagakuteAichiJapan
| | - Keisuke Kamiya
- Department of Nephrology and RheumatologyAichi Medical UniversityNagakuteAichiJapan
| | - Mai Yamauchi
- Department of Nephrology and RheumatologyAichi Medical UniversityNagakuteAichiJapan
| | - Hiroshi Kinashi
- Department of Nephrology and RheumatologyAichi Medical UniversityNagakuteAichiJapan
| | - Makoto Yamaguchi
- Department of Nephrology and RheumatologyAichi Medical UniversityNagakuteAichiJapan
| | - Takayuki Katsuno
- Department of Nephrology and RheumatologyAichi Medical UniversityNagakuteAichiJapan
| | - Hironobu Nobata
- Department of Nephrology and RheumatologyAichi Medical UniversityNagakuteAichiJapan
| | - Kazushi Watanabe
- Department of Obstetrics and GynecologyAichi Medical UniversityNagakuteAichiJapan
| | - Akihiko Wakatsuki
- Department of Obstetrics and GynecologyAichi Medical UniversityNagakuteAichiJapan
| | - Jan Aten
- Department of PathologyAmsterdam University Medical Center (Location AMC) University of AmsterdamAmsterdamThe Netherlands
| | - Shoichi Maruyama
- Department of NephrologyNagoya University Graduate School of MedicineNagoyaAichiJapan
| | - Takuji Ishimoto
- Department of NephrologyNagoya University Graduate School of MedicineNagoyaAichiJapan
| | - Shuichi Hirai
- Department of AnatomyAichi Medical University School of MedicineNagakuteAichiJapan
| | - Munekazu Naito
- Department of AnatomyAichi Medical University School of MedicineNagakuteAichiJapan
| | - Yasuhiko Ito
- Department of Nephrology and RheumatologyAichi Medical UniversityNagakuteAichiJapan
| |
Collapse
|
26
|
Abstract
When chronic kidney disease develops, the capacity of the kidneys to clear metabolic waste products from the body is gradually lost. This process results in the retention of a large array of compounds affecting biochemical and biological functions (uremic toxins), of which several can cause cardiovascular damage. This article reviews the main cardiotoxic mechanisms related to uremic toxin retention (endothelial dysfunction, vascular smooth muscle cell alterations, inflammation, mineral bone disorder, insulin resistance, and thrombogenicity) and the main responsible retention compounds. Therapeutic options are reviewed, such as influencing solute generation by intestinal microbiota.
Collapse
Affiliation(s)
- Sophie Valkenburg
- Nephrology Section, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Griet Glorieux
- Nephrology Section, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Raymond Vanholder
- Nephrology Section, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Corneel Heymanslaan 10, Gent 9000, Belgium.
| |
Collapse
|
27
|
Falconi CA, Junho CVDC, Fogaça-Ruiz F, Vernier ICS, da Cunha RS, Stinghen AEM, Carneiro-Ramos MS. Uremic Toxins: An Alarming Danger Concerning the Cardiovascular System. Front Physiol 2021; 12:686249. [PMID: 34054588 PMCID: PMC8160254 DOI: 10.3389/fphys.2021.686249] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 04/19/2021] [Indexed: 12/14/2022] Open
Abstract
The kidneys and heart share functions with the common goal of maintaining homeostasis. When kidney injury occurs, many compounds, the so-called "uremic retention solutes" or "uremic toxins," accumulate in the circulation targeting other tissues. The accumulation of uremic toxins such as p-cresyl sulfate, indoxyl sulfate and inorganic phosphate leads to a loss of a substantial number of body functions. Although the concept of uremic toxins is dated to the 1960s, the molecular mechanisms capable of leading to renal and cardiovascular injuries are not yet known. Besides, the greatest toxic effects appear to be induced by compounds that are difficult to remove by dialysis. Considering the close relationship between renal and cardiovascular functions, an understanding of the mechanisms involved in the production, clearance and overall impact of uremic toxins is extremely relevant for the understanding of pathologies of the cardiovascular system. Thus, the present study has as main focus to present an extensive review on the impact of uremic toxins in the cardiovascular system, bringing the state of the art on the subject as well as clinical implications related to patient's therapy affected by chronic kidney disease, which represents high mortality of patients with cardiac comorbidities.
Collapse
Affiliation(s)
- Carlos Alexandre Falconi
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André, Brazil
| | - Carolina Victoria da Cruz Junho
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André, Brazil
| | - Fernanda Fogaça-Ruiz
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André, Brazil
| | - Imara Caridad Stable Vernier
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André, Brazil
| | - Regiane Stafim da Cunha
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba, Brazil
| | | | - Marcela Sorelli Carneiro-Ramos
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André, Brazil
| |
Collapse
|
28
|
Uremic Toxins, Oxidative Stress, Atherosclerosis in Chronic Kidney Disease, and Kidney Transplantation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6651367. [PMID: 33628373 PMCID: PMC7895596 DOI: 10.1155/2021/6651367] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/25/2021] [Accepted: 01/29/2021] [Indexed: 12/21/2022]
Abstract
Patients with chronic kidney disease (CKD) are at a high risk for cardiovascular disease (CVD), and approximately half of all deaths among patients with CKD are a direct result of CVD. The premature cardiovascular disease extends from mild to moderate CKD stages, and the severity of CVD and the risk of death increase with a decline in kidney function. Successful kidney transplantation significantly decreases the risk of death relative to long-term dialysis treatment; nevertheless, the prevalence of CVD remains high and is responsible for approximately 20-35% of mortality in renal transplant recipients. The prevalence of traditional and nontraditional risk factors for CVD is higher in patients with CKD and transplant recipients compared with the general population; however, it can only partly explain the highly increased cardiovascular burden in CKD patients. Nontraditional risk factors, unique to CKD patients, include proteinuria, disturbed calcium, and phosphate metabolism, anemia, fluid overload, and accumulation of uremic toxins. This accumulation of uremic toxins is associated with systemic alterations including inflammation and oxidative stress which are considered crucial in CKD progression and CKD-related CVD. Kidney transplantation can mitigate the impact of some of these nontraditional factors, but they typically persist to some degree following transplantation. Taking into consideration the scarcity of data on uremic waste products, oxidative stress, and their relation to atherosclerosis in renal transplantation, in the review, we discussed the impact of uremic toxins on vascular dysfunction in CKD patients and kidney transplant recipients. Special attention was paid to the role of native and transplanted kidney function.
Collapse
|
29
|
Glorieux G, Vanholder R, Van Biesen W, Pletinck A, Schepers E, Neirynck N, Speeckaert M, De Bacquer D, Verbeke F. Free p-cresyl sulfate shows the highest association with cardiovascular outcome in chronic kidney disease. Nephrol Dial Transplant 2021; 36:998-1005. [DOI: 10.1093/ndt/gfab004] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Indexed: 12/13/2022] Open
Abstract
Abstract
Background
Several protein-bound uraemic toxins (PBUTs) have been associated with cardiovascular (CV) and all-cause mortality in chronic kidney disease (CKD) but the degree to which this is the case per individual PBUT and the pathophysiological mechanism have only partially been unraveled.
Methods
We compared the prognostic value of both total and free concentrations of five PBUTs [p-cresyl sulfate (pCS), p-cresyl glucuronide, indoxyl sulfate, indole acetic acid and hippuric acid] in a cohort of 523 patients with non-dialysis CKD Stages G1–G5. Patients were followed prospectively for the occurrence of a fatal or non-fatal CV event as the primary endpoint and a number of other major complications as secondary endpoints. In addition, association with and the prognostic value of nine markers of endothelial activation/damage was compared.
Results
After a median follow-up of 5.5 years, 149 patients developed the primary endpoint. In multivariate Cox regression models adjusted for age, sex, systolic blood pressure, diabetes mellitus and estimated glomerular filtration rate, and corrected for multiple testing, only free pCS was associated with the primary endpoint {hazard ratio [HR]1.39 [95% confidence interval (CI) 1.14–1.71]; P = 0.0014}. Free pCS also correlated with a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (r = −0.114, P < 0.05), angiopoietin-2 (ANGPT2) (r = 0.194, P < 0.001), matrix metallopeptidase 7 (MMP-7; (r = 0.238, P < 0.001) and syndecan 1 (r = 0.235, P < 0.001). Of these markers of endothelial activation/damage, ANGPT2 [HR 1.46 (95% CI 1.25–1.70); P < 0.0001] and MMP-7 [HR 1.31 (95% CI 1.08–1.59); P = 0.0056] were also predictive of the primary outcome.
Conclusions
Among PBUTs, free pCS shows the highest association with CV outcome in non-dialysed patients with CKD. Two markers of endothelial activation/damage that were significantly correlated with free pCS, ANGPT2 and MMP-7 were also associated with CV outcome. The hypothesis that free pCS exerts its CV toxic effects by an adverse effect on endothelial function deserves further exploration.
Collapse
Affiliation(s)
- Griet Glorieux
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Raymond Vanholder
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Wim Van Biesen
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Anneleen Pletinck
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Eva Schepers
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Nathalie Neirynck
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Marijn Speeckaert
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Dirk De Bacquer
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Francis Verbeke
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
30
|
Liew H, Roberts MA, Pope A, McMahon LP. Endothelial glycocalyx damage in kidney disease correlates with uraemic toxins and endothelial dysfunction. BMC Nephrol 2021; 22:21. [PMID: 33423673 PMCID: PMC7798348 DOI: 10.1186/s12882-020-02219-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/21/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Damage to the endothelial glycocalyx is an early indicator of vascular damage and a potential marker of endothelial dysfunction. This study aimed to assess the relationship between markers of glycocalyx damage, endothelial dysfunction, and uraemic toxins in patients with chronic kidney disease. METHODS Healthy controls, CKD patients, dialysis patients, and kidney transplant recipients had biochemical markers of glycocalyx damage (syndecan-1 and hyaluronan), endothelial dysfunction (von Willebrand factor; vWF and vascular cell adhesion molecule; VCAM-1), and uraemic toxins (indoxyl sulphate and p-cresyl sulphate) measured. In addition, Sidestream Darkfield imaging was performed using the novel GlycoCheck™ device to measure glycocalyx width by the perfused boundary region (PBR) in the sublingual microcirculation. RESULTS Serum markers of glycocalyx damage were highest in the dialysis group (n = 33), followed by CKD patients (n = 32) and kidney transplant recipients (n = 30) compared to controls (n = 30): hyaluronan: 137 (16-1414), 79 (11-257), 57 (14-218) and 23 (8-116) ng/mL, respectively, p < 0.0001; syndecan-1: 81 (40-529), 46 (21-134), 39 (23-72), and 30 (12-138) ng/mL, respectively, p < 0.0001. Markers of endothelial dysfunction followed a similar pattern. No difference in the width of the PBR was detected between these groups (2.01 ± 0.35, 2.07 ± 0.27, 2.06 ± 0.28, and 2.05 ± 0.3 µm, respectively, p = 0.89). Glycocalyx damage correlated with markers of endothelial dysfunction (log-hyaluronan and log-VCAM-1: r = 0.64, p < 0.001) and levels of uraemic toxins (log-hyaluronan and log-indoxyl sulphate: r = 0.48, p < 0.001). CONCLUSIONS Levels of biochemical markers of glycocalyx and endothelial cell damage are highest in patients receiving dialysis. Glycocalyx and endothelial damage markers correlated with each other, and with uraemic toxins. Although we could not demonstrate a change in PBR, the biochemical markers suggest that glycocalyx damage is most marked in patients with higher levels of uraemic toxins.
Collapse
Affiliation(s)
- Hui Liew
- Department of Renal Medicine, Eastern Health, 3128, Box Hill, Victoria, Australia. .,Eastern Health Clinical School, Monash University, 3128, Box Hill, Victoria, Australia.
| | - Matthew A Roberts
- Department of Renal Medicine, Eastern Health, 3128, Box Hill, Victoria, Australia.,Eastern Health Clinical School, Monash University, 3128, Box Hill, Victoria, Australia
| | - Alun Pope
- Eastern Health Clinical School, Monash University, 3128, Box Hill, Victoria, Australia
| | - Lawrence P McMahon
- Department of Renal Medicine, Eastern Health, 3128, Box Hill, Victoria, Australia.,Eastern Health Clinical School, Monash University, 3128, Box Hill, Victoria, Australia
| |
Collapse
|
31
|
Targeting Uremic Toxins to Prevent Peripheral Vascular Complications in Chronic Kidney Disease. Toxins (Basel) 2020; 12:toxins12120808. [PMID: 33419312 PMCID: PMC7765928 DOI: 10.3390/toxins12120808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/24/2022] Open
Abstract
Chronic kidney disease (CKD) exhibits progressive kidney dysfunction and leads to disturbed homeostasis, including accumulation of uremic toxins, activated renin-angiotensin system, and increased oxidative stress and proinflammatory cytokines. Patients with CKD are prone to developing the peripheral vascular disease (PVD), leading to poorer outcomes than those without CKD. Cumulative evidence has showed that the synergy of uremic milieu and PVD could exaggerate vascular complications such as limb ischemia, amputation, stenosis, or thrombosis of a dialysis vascular access, and increase mortality risk. The role of uremic toxins in the pathogenesis of vascular dysfunction in CKD has been investigated. Moreover, growing evidence has shown the promising role of uremic toxins as a therapeutic target for PVD in CKD. This review focused on uremic toxins in the pathophysiology, in vitro and animal models, and current novel clinical approaches in reducing the uremic toxin to prevent peripheral vascular complications in CKD patients.
Collapse
|
32
|
Trojanowicz B, Ulrich C, Girndt M. Uremic Apelin and Leucocytic Angiotensin-Converting Enzyme 2 in CKD Patients. Toxins (Basel) 2020; 12:toxins12120742. [PMID: 33255902 PMCID: PMC7760850 DOI: 10.3390/toxins12120742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 11/16/2022] Open
Abstract
Apelin peptides (APLN) serve as second substrates for angiotensin-converting enzyme 2 (ACE2) and, in contrast to angiotensin II (AngII), exert blood-pressure lowering and vasodilatation effects through binding to G-coupled APLN receptor (APLNR). ACE2-mediated cleavage of the APLN may reduce its vasodilatory effects, but decreased ACE2 may potentiate the hypotensive properties of APLN. The role of APLN in uremia is unclear. We investigated the correlations between serum-APLN, leucocytic APLNR, and ACE2 in 32 healthy controls (NP), 66 HD, and 24 CKD3-5 patients, and the impact of APLN peptides on monocytic behavior and ACE2 expression under uremic conditions in vitro. We observed that serum APLN and leucocytic APLNR or SLCO2B1 were significantly elevated in uremic patients and correlated with decreased ACE2 on uremic leucocytes. APLN-treated THP-1 monocytes revealed significantly increased APLNR and ACE2, and reduced TNFa, IL-6, and MCSF. Uremic toxins induced a dramatic increase of miR-421 followed by significant reduction of ACE2 transcripts, partially counteracted with APLN-13 and -36. APLN-36 triggered the most potent transmigration and reduction of endothelial adhesion. These results suggest that although APLN peptides may partly protect against the decay of monocytic ACE2 transcripts, uremic milieu is the most dominant modulator of local ACE2, and likely to contribute to the progression of atherosclerosis.
Collapse
|
33
|
Lin TY, Chou HH, Huang HL, Hung SC. Indoxyl Sulfate and Incident Peripheral Artery Disease in Hemodialysis Patients. Toxins (Basel) 2020; 12:toxins12110696. [PMID: 33147880 PMCID: PMC7693838 DOI: 10.3390/toxins12110696] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 10/28/2020] [Accepted: 10/31/2020] [Indexed: 11/17/2022] Open
Abstract
Peripheral artery disease (PAD) is highly prevalent among patients with chronic kidney disease (CKD) and portends a very poor prognosis. Indoxyl sulfate has been shown to induce atherothrombosis and impaired neovascularization in uremic mice. However, there is no clinical evidence regarding the role of indoxyl sulfate in PAD associated with CKD. We examined associations between indoxyl sulfate and incident symptomatic lower extremity PAD events as well as major adverse cardiovascular events (MACE) and all-cause mortality using Cox proportional hazards models in a prospective cohort of 200 hemodialysis patients free of PAD at baseline. Patients were considered as having PAD if they developed PAD symptoms confirmed by an ankle-brachial index with waveforms, duplex ultrasound or angiography, and/or major adverse limb events including revascularization and amputation. During a median follow-up of 6.5 years, 37 patients (18.5%) experienced incident symptomatic PAD. MACE occurred in 52 patients, and a total of 85 patients died. After adjusting for traditional risk factors for PAD, including age, current smoking, diabetes, and cardiovascular disease, indoxyl sulfate was significantly associated with the risk of PAD (hazard ratio (HR), 1.19 for every 10-μg/mL increase in indoxyl sulfate; 95% confidence interval (CI), 1.05–1.35). However, indoxyl sulfate was not associated with risk of MACE (HR, 1.00; 95% CI, 0.90–1.12) or death from any cause (HR, 0.98; 95% CI, 0.90–1.07). Indoxyl sulfate was associated with incident symptomatic PAD but not with MACE or all-cause mortality, suggesting that indoxyl sulfate toxicity may be unique to PAD among hemodialysis patients.
Collapse
Affiliation(s)
- Ting-Yun Lin
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and School of Medicine, Tzu Chi University, Hualien 970, Taiwan;
| | - Hsin-Hua Chou
- Division of Cardiology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and School of Medicine, Tzu Chi University, Hualien 970, Taiwan;
| | - Hsuan-Li Huang
- Division of Cardiology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and School of Medicine, Tzu Chi University, Hualien 970, Taiwan;
- Correspondence: (H.-L.H.); (S.-C.H.); Tel.: +886-2-6628-9779 (H.-L.H. & S.-C.H.)
| | - Szu-Chun Hung
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and School of Medicine, Tzu Chi University, Hualien 970, Taiwan;
- Correspondence: (H.-L.H.); (S.-C.H.); Tel.: +886-2-6628-9779 (H.-L.H. & S.-C.H.)
| |
Collapse
|
34
|
Abstract
Cardiovascular disease and infections are major causes for the high incidence of morbidity and mortality of patients with chronic kidney disease. Both complications are directly or indirectly associated with disturbed functions or altered apoptotic rates of polymorphonuclear leukocytes, monocytes, lymphocytes, and dendritic cells. Normal responses of immune cells can be reduced, leading to infectious diseases or pre-activated/primed, giving rise to inflammation and subsequently to cardiovascular disease. This review summarizes the impact of kidney dysfunction on the immune system. Renal failure results in disturbed renal metabolic activities with reduced renin, erythropoietin, and vitamin D production, which adversely affects the immune system. Decreased kidney function also leads to reduced glomerular filtration and the retention of uremic toxins. A large number of uremic toxins with detrimental effects on immune cells have been identified. Besides small water-soluble and protein-bound compounds originating from the intestinal microbiome, several molecules in the middle molecular range, e.g., immunoglobulin light chains, retinol-binding protein, the neuropeptides Met-enkephalin and neuropeptide Y, endothelin-1, and the adipokines leptin and resistin, adversely affect immune cells. Posttranslational modifications such as carbamoylation, advanced glycation products, and oxidative modifications contribute to uremic toxicity. Furthermore, high-density lipoprotein from uremic patients has an altered protein profile and thereby loses its anti-inflammatory properties.
Collapse
Affiliation(s)
- Gerald Cohen
- Department of Nephrology and Dialysis, Medical University of Vienna, Vienna A-1090, Austria
| |
Collapse
|
35
|
Espi M, Koppe L, Fouque D, Thaunat O. Chronic Kidney Disease-Associated Immune Dysfunctions: Impact of Protein-Bound Uremic Retention Solutes on Immune Cells. Toxins (Basel) 2020; 12:toxins12050300. [PMID: 32384617 PMCID: PMC7291164 DOI: 10.3390/toxins12050300] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 12/19/2022] Open
Abstract
Regardless of the primary disease responsible for kidney failure, patients suffering from chronic kidney disease (CKD) have in common multiple impairments of both the innate and adaptive immune systems, the pathophysiology of which has long remained enigmatic. CKD-associated immune dysfunction includes chronic low-grade activation of monocytes and neutrophils, which induces endothelial damage and increases cardiovascular risk. Although innate immune effectors are activated during CKD, their anti-bacterial capacity is impaired, leading to increased susceptibility to extracellular bacterial infections. Finally, CKD patients are also characterized by profound alterations of cellular and humoral adaptive immune responses, which account for an increased risk for malignancies and viral infections. This review summarizes the recent emerging data that link the pathophysiology of CKD-associated immune dysfunctions with the accumulation of microbiota-derived metabolites, including indoxyl sulfate and p-cresyl sulfate, the two best characterized protein-bound uremic retention solutes.
Collapse
Affiliation(s)
- Maxime Espi
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, Hospices Civils de Lyon, 69000 Lyon, France;
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, 69000 Lyon, France
| | - Laetitia Koppe
- Département de Néphrologie-Dialyse-Nutrition, Centre Hôpital Lyon Sud, Hospices Civils de Lyon, 69310 Pierre Bénite, France; (L.K.); (D.F.)
- CarMeN, INSERM U1060, INRA 1397, 69310 Pierre-Bénite, France
- Lyon-Sud Medical Faculty, Université de Lyon, 69000 Lyon, France
| | - Denis Fouque
- Département de Néphrologie-Dialyse-Nutrition, Centre Hôpital Lyon Sud, Hospices Civils de Lyon, 69310 Pierre Bénite, France; (L.K.); (D.F.)
- CarMeN, INSERM U1060, INRA 1397, 69310 Pierre-Bénite, France
- Lyon-Sud Medical Faculty, Université de Lyon, 69000 Lyon, France
| | - Olivier Thaunat
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, Hospices Civils de Lyon, 69000 Lyon, France;
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, 69000 Lyon, France
- Lyon-Est Medical Faculty, Université de Lyon, 69000 Lyon, France
- Correspondence:
| |
Collapse
|
36
|
Ward RA, Beck W, Bernardo AA, Alves FC, Stenvinkel P, Lindholm B. Hypoalbuminemia: a price worth paying for improved dialytic removal of middle-molecular-weight uremic toxins? Nephrol Dial Transplant 2020; 34:901-907. [PMID: 30102329 DOI: 10.1093/ndt/gfy236] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Indexed: 01/17/2023] Open
Abstract
Hemodiafiltration (HDF) increases the removal of middle-molecular-weight uremic toxins and may improve outcomes in patients with end-stage kidney disease (ESKD), but it requires complex equipment and comes with risks associated with infusion of large volumes of substitution solution. New high-flux hemodialysis membranes with improved diffusive permeability profiles do not have these limitations and offer an attractive alternative to HDF. However, both strategies are associated with increased albumin loss into the dialysate, raising concerns about the potential for decreased serum albumin concentrations that have been associated with poor outcomes in ESKD. Many factors can contribute to hypoalbuminemia in ESKD, including protein energy wasting, inflammation, volume expansion, renal loss and loss into the dialysate; of these factors, loss into the dialysate is not necessarily the most important. Furthermore, recent studies suggest that mild hypoalbuminemia per se is not an independent predictor of increased mortality in dialysis patients, but in combination with inflammation it is a poor prognostic sign. Thus, whether hypoalbuminemia predisposes to increased morbidity and mortality may depend on the presence or absence of inflammation. In this review we summarize recent findings on the role of dialysate losses in hypoalbuminemia and the importance of concomitant inflammation on outcomes in patients with ESKD. Based on these findings, we discuss whether hypoalbuminemia may be a price worth paying for increased dialytic removal of middle-molecular-weight uremic toxins.
Collapse
Affiliation(s)
| | - Werner Beck
- R&D, Baxter International Inc., Hechingen, Germany
| | | | - Filipa C Alves
- Department of Nephrology, Hospital Espírito Santo, Évora, Portugal.,Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Lindholm
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
37
|
Glorieux G, Gryp T, Perna A. Gut-Derived Metabolites and Their Role in Immune Dysfunction in Chronic Kidney Disease. Toxins (Basel) 2020; 12:toxins12040245. [PMID: 32290429 PMCID: PMC7232434 DOI: 10.3390/toxins12040245] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/08/2020] [Accepted: 04/10/2020] [Indexed: 02/07/2023] Open
Abstract
Several of the uremic toxins, which are difficult to remove by dialysis, originate from the gut bacterial metabolism. This opens opportunities for novel targets trying to decrease circulating levels of these toxins and their pathophysiological effects. The current review focuses on immunomodulatory effects of these toxins both at their side of origin and in the circulation. In the gut end products of the bacterial metabolism such as p-cresol, trimethylamine and H2S affect the intestinal barrier structure and function while in the circulation the related uremic toxins stimulate cells of the immune system. Both conditions contribute to the pro-inflammatory status of patients with chronic kidney disease (CKD). Generation and/or absorption of these toxin precursors could be targeted to decrease plasma levels of their respective uremic toxins and to reduce micro-inflammation in CKD.
Collapse
Affiliation(s)
- Griet Glorieux
- Nephrology Division, Ghent University Hospital and Ghent University, 9000 Ghent, Belgium;
- Correspondence: ; Tel.: +32-9-3324511
| | - Tessa Gryp
- Nephrology Division, Ghent University Hospital and Ghent University, 9000 Ghent, Belgium;
| | - Alessandra Perna
- First Division of Nephrology, Department of Translational Medical Sciences, School of Medicine, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy;
| |
Collapse
|
38
|
Indoxyl Sulfate, a Uremic Endotheliotoxin. Toxins (Basel) 2020; 12:toxins12040229. [PMID: 32260489 PMCID: PMC7232210 DOI: 10.3390/toxins12040229] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 12/15/2022] Open
Abstract
Chronic kidney disease (CKD) is associated with a high prevalence of cardiovascular diseases. During CKD, the uremic toxin indoxyl sulfate (IS)—derived from tryptophan metabolism—accumulates. IS is involved in the pathophysiology of cardiovascular complications. IS can be described as an endotheliotoxin: IS induces endothelial dysfunction implicated in cardiovascular morbidity and mortality during CKD. In this review, we describe clinical and experimental evidence for IS endothelial toxicity and focus on the various molecular pathways implicated. In patients with CKD, plasma concentrations of IS correlate with cardiovascular events and mortality, with vascular calcification and atherosclerotic markers. Moreover, IS induces a prothrombotic state and impaired neovascularization. IS reduction by AST-120 reverse these abnormalities. In vitro, IS induces endothelial aryl hydrocarbon receptor (AhR) activation and proinflammatory transcription factors as NF-κB or AP-1. IS has a prooxidant effect with reduction of nitric oxide (NO) bioavailability. Finally, IS alters endothelial cell and endothelial progenitor cell migration, regeneration and control vascular smooth muscle cells proliferation. Reducing IS endothelial toxicity appears to be necessary to improve cardiovascular health in CKD patients.
Collapse
|
39
|
Vanommeslaeghe F, Josipovic I, Boone M, Dhondt A, Van Biesen W, Eloot S. A randomized cross-over study with objective quantification of the performance of an asymmetric triacetate and a polysulfone dialysis membrane using different anticoagulation strategies. Clin Kidney J 2019; 14:398-407. [PMID: 33564444 PMCID: PMC7857793 DOI: 10.1093/ckj/sfz163] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/02/2019] [Indexed: 02/07/2023] Open
Abstract
Background Different strategies can be used to counteract coagulation of extracorporeal systems. Systemic anticoagulation is most widely used in routine clinical practice, but can be contraindicated in specific settings. The Solacea™ dialyser, containing the asymmetric triacetate membrane, claims improved biocompatibility, which should result in decreased tendency for coagulation. We quantified the performance of the Solacea™ versus the FX800CORDIAX dialyser regarding resistance to fibre blocking as assessed by micro-computed tomography (CT). Methods This cross-over study with four arms randomized consecutively 10 maintenance haemodialysis patients to a 4-h post-dilution haemodiafiltration session at midweek, using either Solacea™ 19 H or FX800CORDIAX, with either regular or half dose of anticoagulation (EC2017/1459-NCT03820401). Dialyser fibre blocking was visualized in the dialyser outlet potting using a 3D CT scanning technique on micrometre resolution. Extraction ratios of middle molecules [myoglobin, lambda and kappa free light chains (FLCs)] were determined. Results The relative number of open fibres post-dialysis was lower in FX800CORDIAX versus Solacea™ dialyser, and this was irrespective of the anticoagulation dose used or the threshold for counting open fibres. Extraction ratios of FLCs were not different at regular anticoagulation between Solacea™ and FX800CORDIAX (21% ± 4% for kappa and 32% ± 8% for lambda with Solacea™ versus 23% ± 7% and 38% ± 6% for FX800CORDIAX), but were superior with the Solacea™ (34% ± 12% versus 22% ± 8% with FX800CORDIAX; P = 0.02) for myoglobin in case of halving anticoagulation dose. No clinically relevant albumin loss was detected. Conclusions The Solacea™ dialyser seems to be promising for use in conditions where systemic anticoagulation is contraindicated, as even under conditions of low systemic anticoagulation, virtually no signs of fibre blocking could be observed using the sensitive micro-CT scanning technique. This finding is in line with its presumed good performance in terms of biocompatibility.
Collapse
Affiliation(s)
| | - Iván Josipovic
- Centre for X-ray Tomography, Physics and Astronomy, Ghent University, Ghent, Belgium
| | - Matthieu Boone
- Centre for X-ray Tomography, Physics and Astronomy, Ghent University, Ghent, Belgium
| | - Annemie Dhondt
- Nephrology Department, Ghent University Hospital, Ghent, Belgium
| | - Wim Van Biesen
- Nephrology Department, Ghent University Hospital, Ghent, Belgium
| | - Sunny Eloot
- Nephrology Department, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
40
|
Indoxyl sulfate associates with cardiovascular phenotype in children with chronic kidney disease. Pediatr Nephrol 2019; 34:2571-2582. [PMID: 31428929 DOI: 10.1007/s00467-019-04331-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 05/09/2019] [Accepted: 08/06/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Cardiovascular disease is the leading cause of death in children with chronic kidney disease (CKD). Serum levels of gut-derived uremic toxins increase with deterioration of kidney function and are associated with cardiac comorbidities in adult CKD patients. METHODS Indoxyl sulfate (IS) and p-cresyl sulfate (pCS) were measured by high-performance liquid chromatography in serum of children participating in the Cardiovascular Comorbidity in Children with CKD (4C) Study. Results were correlated with measurements of the carotid intima-media thickness (cIMT), central pulse wave velocity (PWV), and left ventricular mass index (LVMI) in children aged 6-17 years with initial eGFR of 10-60 ml/min per 1.73 m2. RESULTS The median serum levels of total IS and of pCS, measured in 609 patients, were 5.3 μmol/l (8.7) and 17.0 μmol/l (21.6), respectively. In a multivariable regression model, IS and pCS showed significant positive associations with urea and negative associations with eGFR and uric acid. Furthermore, positive associations of pCS with age, serum albumin, and non-Mediterranean residency and a negative association with glomerular disease were observed. By multivariable regression analysis, only IS was significantly associated with a higher cIMT SDS at baseline and progression of PWV SDS within 12 months, independent of other risk factors. CONCLUSIONS Serum levels of gut-derived uremic toxins IS and pCS correlated inversely with eGFR in children. Only IS was significantly associated with surrogate markers of cardiovascular disease in this large pediatric CKD cohort.
Collapse
|
41
|
The Signaling of Cellular Senescence in Diabetic Nephropathy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7495629. [PMID: 31687085 PMCID: PMC6794967 DOI: 10.1155/2019/7495629] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 07/03/2019] [Accepted: 07/23/2019] [Indexed: 12/13/2022]
Abstract
Diabetic nephropathy is the leading cause of chronic kidney disease (CKD) in western countries. Notably, it has a rapidly rising prevalence in China. The patients, commonly complicated with cardiovascular diseases and neurologic disorders, are at high risk to progress into end-stage renal disease (ESRD) and death. However, the pathogenic mechanisms of diabetic nephropathy have not been determined. Cellular senescence, which recently has gained broad attention, is thought to be an important player in the onset and development of diabetic nephropathy. In this issue, we generally review the mechanisms of cellular senescence in diabetic nephropathy, which involve telomere attrition, DNA damage, epigenetic alterations, mitochondrial dysfunction, loss of Klotho, Wnt/β-catenin signaling activation, persistent inflammation, and accumulation of uremic toxins. Moreover, we highlight the potential therapeutic targets of cellular senescence in diabetic nephropathy and provide important clues for clinical strategies.
Collapse
|
42
|
New Insights into the Roles of Monocytes/Macrophages in Cardiovascular Calcification Associated with Chronic Kidney Disease. Toxins (Basel) 2019; 11:toxins11090529. [PMID: 31547340 PMCID: PMC6784181 DOI: 10.3390/toxins11090529] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) is an important cause of death in patients with chronic kidney disease (CKD), and cardiovascular calcification (CVC) is one of the strongest predictors of CVD in this population. Cardiovascular calcification results from complex cellular interactions involving the endothelium, vascular/valvular cells (i.e., vascular smooth muscle cells, valvular interstitial cells and resident fibroblasts), and monocyte-derived macrophages. Indeed, the production of pro-inflammatory cytokines and oxidative stress by monocyte-derived macrophages is responsible for the osteogenic transformation and mineralization of vascular/valvular cells. However, monocytes/macrophages show the ability to modify their phenotype, and consequently their functions, when facing environmental modifications. This plasticity complicates efforts to understand the pathogenesis of CVC-particularly in a CKD setting, where both uraemic toxins and CKD treatment may affect monocyte/macrophage functions and thereby influence CVC. Here, we review (i) the mechanisms by which each monocyte/macrophage subset either promotes or prevents CVC, and (ii) how both uraemic toxins and CKD therapies might affect these monocyte/macrophage functions.
Collapse
|
43
|
Valdivielso JM, Rodríguez-Puyol D, Pascual J, Barrios C, Bermúdez-López M, Sánchez-Niño MD, Pérez-Fernández M, Ortiz A. Atherosclerosis in Chronic Kidney Disease: More, Less, or Just Different? Arterioscler Thromb Vasc Biol 2019; 39:1938-1966. [PMID: 31412740 DOI: 10.1161/atvbaha.119.312705] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Patients with chronic kidney disease (CKD) are at an increased risk of premature mortality, mainly from cardiovascular causes. The association between CKD on hemodialysis and accelerated atherosclerosis was described >40 years ago. However, more recently, it has been suggested that the increase in atherosclerosis risk is actually observed in early CKD stages, remaining stable thereafter. In this regard, interventions targeting the pathogenesis of atherosclerosis, such as statins, successful in the general population, have failed to benefit patients with very advanced CKD. This raises the issue of the relative contribution of atherosclerosis versus other forms of cardiovascular injury such as arteriosclerosis or myocardial injury to the increased cardiovascular risk in CKD. In this review, the pathophysiogical contributors to atherosclerosis in CKD that are shared with the general population, or specific to CKD, are discussed. The NEFRONA study (Observatorio Nacional de Atherosclerosis en NEFrologia) prospectively assessed the prevalence and progression of subclinical atherosclerosis (plaque in vascular ultrasound), confirming an increased prevalence of atherosclerosis in patients with moderate CKD. However, the adjusted odds ratio for subclinical atherosclerosis increased with CKD stage, suggesting a contribution of CKD itself to subclinical atherosclerosis. Progression of atherosclerosis was closely related to CKD progression as well as to the baseline presence of atheroma plaque, and to higher phosphate, uric acid, and ferritin and lower 25(OH) vitamin D levels. These insights may help design future clinical trials of stratified personalized medicine targeting atherosclerosis in patients with CKD. Future primary prevention trials should enroll patients with evidence of subclinical atherosclerosis and should provide a comprehensive control of all known risk factors in addition to testing any additional intervention or placebo.
Collapse
Affiliation(s)
- José M Valdivielso
- From the Vascular & Renal Translational Research Group and UDETMA, IRBLleida. Spanish Research Network for Renal Diseases (RedInRen. ISCIII), Lleida, Spain (J.M.V., M.B.-L.)
| | - Diego Rodríguez-Puyol
- Nephrology Unit, Fundación para la investigación del Hospital Universitario Príncipe de Asturias, RedInRen, Alcalá de Henares, Madrid, Spain (D.R.-P.)
| | - Julio Pascual
- Department of Nephrology, Institute Mar for Medical Research, Hospital del Mar, RedInRen, Barcelona, Spain (J.P., C.B.)
| | - Clara Barrios
- Department of Nephrology, Institute Mar for Medical Research, Hospital del Mar, RedInRen, Barcelona, Spain (J.P., C.B.)
| | - Marcelino Bermúdez-López
- From the Vascular & Renal Translational Research Group and UDETMA, IRBLleida. Spanish Research Network for Renal Diseases (RedInRen. ISCIII), Lleida, Spain (J.M.V., M.B.-L.)
| | - Maria Dolores Sánchez-Niño
- IIS-Fundacion Jimenez Diaz, School of Medicine, University Autonoma of Madrid, FRIAT and RedInRen, Madrid, Spain (M.D.S.-N., A.O.)
| | | | - Alberto Ortiz
- IIS-Fundacion Jimenez Diaz, School of Medicine, University Autonoma of Madrid, FRIAT and RedInRen, Madrid, Spain (M.D.S.-N., A.O.)
| |
Collapse
|
44
|
Hobby GP, Karaduta O, Dusio GF, Singh M, Zybailov BL, Arthur JM. Chronic kidney disease and the gut microbiome. Am J Physiol Renal Physiol 2019; 316:F1211-F1217. [PMID: 30864840 DOI: 10.1152/ajprenal.00298.2018] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The gut microbiome is composed of a diverse population of bacteria that have beneficial and adverse effects on human health. The microbiome has recently gained attention and is increasingly noted to play a significant role in health and a number of disease states. Increasing urea concentration during chronic kidney disease (CKD) leads to alterations in the intestinal flora that can increase production of gut-derived toxins and alter the intestinal epithelial barrier. These changes can lead to an acceleration of the process of kidney injury. A number of strategies have been proposed to interrupt this pathway of injury in CKD. The purpose of this review is to summarize the role of the gut microbiome in CKD, tools used to study this microbial population, and attempts to alter its composition for therapeutic purposes.
Collapse
Affiliation(s)
- Gerren P Hobby
- Division of Nephrology, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | - Oleg Karaduta
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | - Giuseppina F Dusio
- Division of Nephrology, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | - Manisha Singh
- Division of Nephrology, University of Arkansas for Medical Sciences , Little Rock, Arkansas.,Central Arkansas Veterans Health System, Little Rock, Arkansas
| | - Boris L Zybailov
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | - John M Arthur
- Division of Nephrology, University of Arkansas for Medical Sciences , Little Rock, Arkansas.,Central Arkansas Veterans Health System, Little Rock, Arkansas
| |
Collapse
|
45
|
Snelson M, Kellow NJ, Coughlan MT. Modulation of the Gut Microbiota by Resistant Starch as a Treatment of Chronic Kidney Diseases: Evidence of Efficacy and Mechanistic Insights. Adv Nutr 2019; 10:303-320. [PMID: 30668615 PMCID: PMC6416045 DOI: 10.1093/advances/nmy068] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/17/2018] [Accepted: 08/12/2018] [Indexed: 12/14/2022] Open
Abstract
Chronic kidney disease (CKD) has been associated with changes in gut microbial ecology, or "dysbiosis," which may contribute to disease progression. Recent studies have focused on dietary approaches to favorably alter the composition of the gut microbial communities as a treatment method in CKD. Resistant starch (RS), a prebiotic that promotes proliferation of gut bacteria such as Bifidobacteria and Lactobacilli, increases the production of metabolites including short-chain fatty acids, which confer a number of health-promoting benefits. However, there is a lack of mechanistic insight into how these metabolites can positively influence renal health. Emerging evidence shows that microbiota-derived metabolites can regulate the incretin axis and mitigate inflammation via expansion of regulatory T cells. Studies from animal models and patients with CKD show that RS supplementation attenuates the concentrations of uremic retention solutes, including indoxyl sulfate and p-cresyl sulfate. Here, we present the current state of knowledge linking the microbiome to CKD, we explore the efficacy of RS in animal models of CKD and in humans with the condition, and we discuss how RS supplementation could be a promising dietary approach for slowing CKD progression.
Collapse
Affiliation(s)
- Matthew Snelson
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Nicole J Kellow
- Be Active Sleep & Eat (BASE) Facility, Department of Nutrition, Dietetics, and Food, Monash University, Notting Hill, Victoria, Australia
| | - Melinda T Coughlan
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Baker Heart Research Institute, Melbourne, Victoria, Australia
| |
Collapse
|
46
|
Trojanowicz B, Ulrich C, Fiedler R, Martus P, Storr M, Boehler T, Werner K, Hulko M, Zickler D, Willy K, Schindler R, Girndt M. Modulation of leucocytic angiotensin-converting enzymes expression in patients maintained on high-permeable haemodialysis. Nephrol Dial Transplant 2018; 33:34-43. [PMID: 28992224 DOI: 10.1093/ndt/gfx206] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 04/29/2017] [Indexed: 01/22/2023] Open
Abstract
Background High mortality of haemodialysis patients is associated with systemic chronic inflammation and overactivation of the renin-angiotensin system (RAS). Insufficient elimination of pro-inflammatory immune mediators, especially in the molecular weight range of 15-45 kDa, may be one of the reasons for this. Employment of haemodialysis membranes with increased permeability was shown to ameliorate the inflammatory response and might modulate the effects of local RAS. In this study, we tested the impact of high cut-off (HCO), medium cut-off (MCO) and high-flux (HF) dialysis on leucocytic transcripts of angiotensin-converting enzymes (ACE and ACE2). Additionally, the impact of HCO, MCO and HF sera and dialysates on local ACEs and inflammation markers was tested in THP-1 monocytes. Methods Patients' leucocytes were obtained from our recent clinical studies comparing HCO and MCO dialysers with HF. The cells were subjected to quantitaive polymerase chain reaction (qPCR) analyses with TaqMan probes specific for ACE, ACE2 and angiotensin II (AngII) and Ang1-7 receptors. Sera and dialysates from the clinical trials as well as samples from in vitro dialysis were tested on THP-1 monocytic cells. The cells were subjected to qPCR analyses with TaqMan probes specific for ACE, ACE2, interleukin-6 and tumour necrosis factor α and immunocytochemistry with ACE and ACE2 antibodies. Results Leucocytes obtained from patients treated with HCO or MCO demonstrated decreased transcript expression of ACE, while ACE2 was significantly upregulated as compared with HF. Receptors for AngII and Ang1-7 remained unchanged. THP-1 monocytes preconditioned with HCO and MCO patients' or in vitro dialysis sera reflected the same expressional regulation of ACE and ACE2 as those observed in HCO and MCO leucocytes. As a complementary finding, treatment with HCO and MCO in vitro dialysates induced a pro-inflammatory response of the cells as demonstrated by elevated messenger RNA expression of tumour necrosis factor α and interleukin-6, as well as upregulation of ACE and decreased levels of ACE2. Conclusions Taken together, these data demonstrate that employment of membranes with high permeability eliminates a spectrum of mediators from circulation that affect the RAS components in leucocytes, especially ACE/ACE2.
Collapse
Affiliation(s)
- Bogusz Trojanowicz
- Department of Internal Medicine II, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Christof Ulrich
- Department of Internal Medicine II, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Roman Fiedler
- Department of Internal Medicine II, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Peter Martus
- Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, Tübingen, Germany
| | - Markus Storr
- Department of Research and Development, Gambro Dialysatoren, Hechingen, Germany
| | - Torsten Boehler
- Department of Research and Development, Gambro Dialysatoren, Hechingen, Germany
| | - Kristin Werner
- Department of Research and Development, Gambro Dialysatoren, Hechingen, Germany
| | - Michael Hulko
- Department of Research and Development, Gambro Dialysatoren, Hechingen, Germany
| | - Daniel Zickler
- Department of Nephrology and Internal Intensive Care Medicine, Charité-Universitaetsmedizin Berlin, Campus Virchow Clinic, Berlin, Germany
| | - Kevin Willy
- Department of Nephrology and Internal Intensive Care Medicine, Charité-Universitaetsmedizin Berlin, Campus Virchow Clinic, Berlin, Germany
| | - Ralf Schindler
- Department of Nephrology and Internal Intensive Care Medicine, Charité-Universitaetsmedizin Berlin, Campus Virchow Clinic, Berlin, Germany
| | - Matthias Girndt
- Department of Internal Medicine II, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
47
|
Ishigami J, Matsushita K. Clinical epidemiology of infectious disease among patients with chronic kidney disease. Clin Exp Nephrol 2018; 23:437-447. [PMID: 30178234 PMCID: PMC6435626 DOI: 10.1007/s10157-018-1641-8] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/24/2018] [Indexed: 12/20/2022]
Abstract
Infectious disease is recognized as an important complication among patients with end-stage renal disease, contributing to excess morbidity and health care costs. However, recent epidemiological studies have revealed that even mild to moderate stages of chronic kidney disease (CKD) substantially increase risk of infection. Regarding underlying mechanisms, evidence suggests various aspects of altered immune response in patients with CKD including impaired function of T cells, B cells and neutrophil. Multiple conditions surrounding CKD, such as older age, diabetes, and cardiovascular disease are important contributors in the increased susceptibility to infection in this population. In addition, several mechanisms impairing immune function have been hypothesized including accumulated uremic toxins, increased oxidative stress, endothelial dysfunction, low-grade inflammation, and mineral and bone disorders. In terms of prevention strategies, influenza and pneumococcal vaccines are most feasible and important. Nevertheless, the extent of vaccine utilization in CKD has not been well documented. In addition, antibody response to vaccination may be reduced in CKD patients, and thus a vaccine delivery strategy (e.g., dose and frequency) may need to be optimized among patients with CKD. Through this review, we demonstrate that infection is a major but underrecognized complication of CKD. As CKD is recognized as a serious public health issue, dedicated research is needed to better characterize the burden of infectious disease associated with CKD, understand the pathophysiology of infection in patients with CKD, and develop effective strategies to prevent infection and its sequela in this high risk population.
Collapse
Affiliation(s)
- Junichi Ishigami
- Department of Epidemiology, Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, 2024 E. Monument St., Suite 2-600, Baltimore, MD, 21287, USA.
| | - Kunihiro Matsushita
- Department of Epidemiology, Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, 2024 E. Monument St., Suite 2-600, Baltimore, MD, 21287, USA
| |
Collapse
|
48
|
Lekawanvijit S. Cardiotoxicity of Uremic Toxins: A Driver of Cardiorenal Syndrome. Toxins (Basel) 2018; 10:toxins10090352. [PMID: 30200452 PMCID: PMC6162485 DOI: 10.3390/toxins10090352] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 08/19/2018] [Accepted: 08/30/2018] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) is highly prevalent in the setting of chronic kidney disease (CKD). Such coexistence of CVD and CKD—the so-called “cardiorenal or renocardiac syndrome”—contributes to exponentially increased risk of cardiovascular (CV) mortality. Uremic cardiomyopathy is a characteristic cardiac pathology commonly found in CKD. CKD patients are also predisposed to heart rhythm disorders especially atrial fibrillation. Traditional CV risk factors as well as known CKD-associated CV risk factors such as anemia are insufficient to explain CV complications in the CKD population. Accumulation of uremic retention solutes is a hallmark of impaired renal excretory function. Many of them have been considered inert solutes until their biological toxicity is unraveled and they become accepted as “uremic toxins”. Direct cardiotoxicity of uremic toxins has been increasingly demonstrated in recent years. This review offers a mechanistic insight into the pathological cardiac remodeling and dysfunction contributed by uremic toxins with a main focus on fibroblastic growth factor-23, an emerging toxin playing a central role in the chronic kidney disease–mineral bone disorder, and the two most investigated non-dialyzable protein-bound uremic toxins, indoxyl sulfate and p-cresyl sulfate. Potential therapeutic strategies that could address these toxins and their relevant mediated pathways since pre-dialysis stages are also discussed.
Collapse
Affiliation(s)
- Suree Lekawanvijit
- Department of Pathology, Faculty of Medicine, Chiang Mai University, 110 Intawaroros Rd, Sribhoom, Chiang Mai 50200, Thailand.
| |
Collapse
|
49
|
The Impact of Uremic Toxins on Cerebrovascular and Cognitive Disorders. Toxins (Basel) 2018; 10:toxins10070303. [PMID: 30037144 PMCID: PMC6071092 DOI: 10.3390/toxins10070303] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 07/17/2018] [Accepted: 07/19/2018] [Indexed: 12/21/2022] Open
Abstract
Individuals at all stages of chronic kidney disease (CKD) have a higher risk of developing cognitive disorders and dementia. Stroke is also highly prevalent in this population and is associated with a higher risk of neurological deterioration, in-hospital mortality, and poor functional outcomes. Evidence from in vitro studies and in vivo animal experiments suggests that accumulation of uremic toxins may contribute to the pathogenesis of stroke and amplify vascular damage, leading to cognitive disorders and dementia. This review summarizes current evidence on the mechanisms by which uremic toxins may favour the occurrence of cerebrovascular diseases and neurological complications in CKD.
Collapse
|
50
|
Velasquez MT, Centron P, Barrows I, Dwivedi R, Raj DS. Gut Microbiota and Cardiovascular Uremic Toxicities. Toxins (Basel) 2018; 10:E287. [PMID: 29997362 PMCID: PMC6071268 DOI: 10.3390/toxins10070287] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/05/2018] [Accepted: 07/06/2018] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) remains a major cause of high morbidity and mortality in patients with chronic kidney disease (CKD). Numerous CVD risk factors in CKD patients have been described, but these do not fully explain the high pervasiveness of CVD or increased mortality rates in CKD patients. In CKD the loss of urinary excretory function results in the retention of various substances referred to as "uremic retention solutes". Many of these molecules have been found to exert toxicity on virtually all organ systems of the human body, leading to the clinical syndrome of uremia. In recent years, an increasing body of evidence has been accumulated that suggests that uremic toxins may contribute to an increased cardiovascular disease (CVD) burden associated with CKD. This review examined the evidence from several clinical and experimental studies showing an association between uremic toxins and CVD. Special emphasis is addressed on emerging data linking gut microbiota with the production of uremic toxins and the development of CKD and CVD. The biological toxicity of some uremic toxins on the myocardium and the vasculature and their possible contribution to cardiovascular injury in uremia are also discussed. Finally, various therapeutic interventions that have been applied to effectively reduce uremic toxins in patients with CKD, including dietary modifications, use of prebiotics and/or probiotics, an oral intestinal sorbent that adsorbs uremic toxins and precursors, and innovative dialysis therapies targeting the protein-bound uremic toxins are also highlighted. Future studies are needed to determine whether these novel therapies to reduce or remove uremic toxins will reduce CVD and related cardiovascular events in the long-term in patients with chronic renal failure.
Collapse
Affiliation(s)
- Manuel T Velasquez
- Division of Renal Diseases and Hypertension, The George Washington University, Washington, DC 20037, USA.
| | - Patricia Centron
- Division of Renal Diseases and Hypertension, The George Washington University, Washington, DC 20037, USA.
| | - Ian Barrows
- Department of Medicine, Georgetown University, Washington, DC 20007, USA.
| | - Rama Dwivedi
- Division of Renal Diseases and Hypertension, The George Washington University, Washington, DC 20037, USA.
- United States Food and Drug Administration, Silver Spring, MD 20993, USA.
| | - Dominic S Raj
- Division of Renal Diseases and Hypertension, The George Washington University, Washington, DC 20037, USA.
| |
Collapse
|