1
|
Long Q, Zhang X, Ren F, Wu X, Wang ZM. Identification of novel biomarkers, shared molecular signatures and immune cell infiltration in heart and kidney failure by transcriptomics. Front Immunol 2024; 15:1456083. [PMID: 39351221 PMCID: PMC11439679 DOI: 10.3389/fimmu.2024.1456083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction Heart failure (HF) and kidney failure (KF) are closely related conditions that often coexist, posing a complex clinical challenge. Understanding the shared mechanisms between these two conditions is crucial for developing effective therapies. Methods This study employed transcriptomic analysis to unveil molecular signatures and novel biomarkers for both HF and KF. A total of 2869 shared differentially expressed genes (DEGs) were identified in patients with HF and KF compared to healthy controls. Functional enrichment analysis was performed to explore the common mechanisms underlying these conditions. A protein-protein interaction (PPI) network was constructed, and machine learning algorithms, including Random Forest (RF), Support Vector Machine-Recursive Feature Elimination (SVM-RFE), and Least Absolute Shrinkage and Selection Operator (LASSO), were used to identify key signature genes. These genes were further analyzed using Gene Set Variation Analysis (GSVA) and Gene Set Enrichment Analysis (GSEA), with their diagnostic values validated in both training and validation sets. Molecular docking studies were conducted. Additionally, immune cell infiltration and correlation analyses were performed to assess the relationship between immune responses and the identified biomarkers. Results The functional enrichment analysis indicated that the common mechanisms are associated with cellular homeostasis, cell communication, cellular replication, inflammation, and extracellular matrix (ECM) production, with the PI3K-Akt signaling pathway being notably enriched. The PPI network revealed two key protein clusters related to the cell cycle and inflammation. CDK2 and CCND1 were identified as signature genes for both HF and KF. Their diagnostic value was validated in both training and validation sets. Additionally, docking studies with CDK2 and CCND1 were performed to evaluate potential drug candidates. Immune cell infiltration and correlation analyses highlighted the immune microenvironment, and that CDK2 and CCND1 are associated with immune responses in HF and KF. Discussion This study identifies CDK2 and CCND1 as novel biomarkers linking cell cycle regulation and inflammation in heart and kidney failure. These findings offer new insights into the molecular mechanisms of HF and KF and present potential targets for diagnosis and therapy.
Collapse
Affiliation(s)
- Qingqing Long
- Division of Nephrology and Clinical Immunology, Medical Faculty, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Xinlong Zhang
- Institute for Photogrammetry and Geoinformatics, University of Stuttgart, Stuttgart, Germany
| | - Fangyuan Ren
- Division of Organic Chemistry - Bioorganic Chemistry, Mathematics/Natural Sciences Faculty, Koblenz University, Koblenz, Germany
| | - Xinyu Wu
- Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Ze-Mu Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Ajibowo AO, Okobi OE, Emore E, Soladoye E, Sike CG, Odoma VA, Bakare IO, Kolawole OA, Afolayan A, Okobi E, Chukwu C. Cardiorenal Syndrome: A Literature Review. Cureus 2023; 15:e41252. [PMID: 37529809 PMCID: PMC10389294 DOI: 10.7759/cureus.41252] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2023] [Indexed: 08/03/2023] Open
Abstract
Cardiorenal syndrome (CRS) is a condition characterized by the intricate two-way relationship between the heart and kidneys, which can lead to acute or chronic dysfunction in these organs. The interplay between cardiorenal connectors and both hemodynamic and non-hemodynamic factors is crucial to understanding this syndrome. The clinical importance of these interactions is evident in the changes observed in hemodynamic factors, neurohormonal markers, and inflammatory processes. Identifying and understanding biomarkers associated with CRS is valuable for early detection and enabling intervention before significant organ dysfunction occurs. This comprehensive review focuses on the clinical significance of biomarkers in the diagnosis, prognosis, and management of CRS. Finally, it highlights the necessity for further advancements in managing this condition.
Collapse
Affiliation(s)
| | - Okelue E Okobi
- Family Medicine, Medficient Health Systems, Laurel, USA
- Family Medicine, Lakeside Medical Center, Belle Glade, USA
| | | | | | - Cherechi G Sike
- General Practice, Windsor University School of Medicine, Cayon, KNA
| | - Victor A Odoma
- Cardiology/Oncology, Indiana University (IU) Health, Bloomington, USA
| | - Ibrahim O Bakare
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, USA
| | | | - Adebola Afolayan
- Internal Medicine, Triboro Center for Nursing and Rehabilitation, New York City, USA
| | - Emeka Okobi
- Dentistry, Ahmadu Bello University Teaching Hospital Zaria, Abuja, NGA
| | | |
Collapse
|
3
|
Jefferies JL, Kovesdy CP, Ronco C. Contemporary laboratory assessment of acute cardiorenal syndrome for early diagnosis: A call for action. Am Heart J 2023; 261:75-84. [PMID: 36948370 DOI: 10.1016/j.ahj.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/15/2023] [Accepted: 03/15/2023] [Indexed: 05/26/2023]
Abstract
Acute cardiorenal syndrome (CRS), categorized as CRS type 1 and 3, is defined by the interplay of acute kidney injury or dysfunction and acute cardiac disease. For optimized diagnosis and management of CRS, strategies targeting multi-organ dysfunction must be adopted. Early diagnosis of acute CRS is important to enable timely initiation of appropriate treatment to prevent serious morbidity and mortality; however, traditional biomarkers are suboptimal. Over the past 2 decades, numerous biomarkers have been investigated for a better and more rapid diagnosis of CRS. Yet, the uptake of these contemporary biomarkers has been slow, possibly owing to the use of imperfect gold-standard reference tests. We believe that there is now scope for use of contemporary laboratory test panels to improve the diagnosis of acute CRS. In this review, we briefly discuss a proposed set of biomarkers for the diagnosis of type 1 and type 3 CRS.
Collapse
Affiliation(s)
- John L Jefferies
- Division of Cardiovascular Diseases, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Csaba P Kovesdy
- Division of Nephrology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Claudio Ronco
- Department of Nephrology, Dialysis and Transplantation, San Bortolo Hospital, Vicenza, Italy
| |
Collapse
|
4
|
Barbero NM, Oller J, Sanz AB, Ramos AM, Ortiz A, Ruiz-Ortega M, Rayego-Mateos S. Mitochondrial Dysfunction in the Cardio-Renal Axis. Int J Mol Sci 2023; 24:ijms24098209. [PMID: 37175915 PMCID: PMC10179675 DOI: 10.3390/ijms24098209] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Cardiovascular disease (CVD) frequently complicates chronic kidney disease (CKD). The risk of all-cause mortality increases from 20% to 500% in patients who suffer both conditions; this is referred to as the so-called cardio-renal syndrome (CRS). Preclinical studies have described the key role of mitochondrial dysfunction in cardiovascular and renal diseases, suggesting that maintaining mitochondrial homeostasis is a promising therapeutic strategy for CRS. In this review, we explore the malfunction of mitochondrial homeostasis (mitochondrial biogenesis, dynamics, oxidative stress, and mitophagy) and how it contributes to the development and progression of the main vascular pathologies that could be affected by kidney injury and vice versa, and how this knowledge may guide the development of novel therapeutic strategies in CRS.
Collapse
Affiliation(s)
- Nerea Mendez Barbero
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, 28037 Madrid, Spain
| | - Jorge Oller
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, 28037 Madrid, Spain
| | - Ana B Sanz
- Spain Nephrology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
| | - Adrian M Ramos
- Spain Nephrology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
| | - Alberto Ortiz
- Spain Nephrology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
| | - Marta Ruiz-Ortega
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
| | - Sandra Rayego-Mateos
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
| |
Collapse
|
5
|
Cabrera Cárdenas A, Salanova Villanueva L, Sánchez Horrillo A, Muñoz Ramos P, Ruano P, Quiroga B. Diastolic dysfunction is an independent predictor for cardiovascular events after an acute kidney injury. Nefrologia 2023; 43:224-231. [PMID: 37442710 DOI: 10.1016/j.nefroe.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 11/02/2021] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND AND AIM Acute kidney injury (AKI) conditions several short- and long-term complications. The aim of the present study was to analyse the impact of cardiac function and structure in the cardiovascular prognosis after an in-hospital AKI episode. MATERIAL AND METHODS This is an observational retrospective cohorts study including all in-hospital AKI episodes in 2013 and 2014 in our centre. At baseline, epidemiological values, comorbidities and echocardiography parameters were collected. During a follow-up of 49 ± 28 months, cardiovascular events (CVE) were collected, and associated factors were analysed. RESULTS 1255 patients were included (55% male, age 75 ± 13 years). Of the 676 (54%) that had a previous echocardiogram, 46% had left ventricular hypertrophy, 38% pulmonary hypertension, 38% diastolic dysfunction and 22% systolic dysfunction. During the follow-up, 484 (39%) developed a CVE. Associated factors to VCE were male sex, age, diabetes mellitus, hypertension, dyslipidemia, coronary heart disease, heart failure, atrial fibrillation, neoplasia and chronic kidney disease (also, glomerular filtration rate at baseline and after the AKI episode). Survival curves demonstrated that all the echocardiographic parameters were associated to CVE. An adjusted Cox regression model showed that age (HR 1.017), diabetes (HR 1.576) and diastolic dysfunction (HR 1.358) were independent predictors for CVE. CONCLUSION Diastolic dysfunction is an independent predictor for long-term cardiovascular events after an in-hospital acute kidney injury episode.
Collapse
Affiliation(s)
| | | | | | | | - Pablo Ruano
- Servicio de Nefrología, Hospital Universitario de La Princesa, Madrid, Spain
| | - Borja Quiroga
- Servicio de Nefrología, Hospital Universitario de La Princesa, Madrid, Spain.
| |
Collapse
|
6
|
Liu Y, Guan X, Shao Y, Zhou J, Huang Y. The Molecular Mechanism and Therapeutic Strategy of Cardiorenal Syndrome Type 3. Rev Cardiovasc Med 2023; 24:52. [PMID: 39077418 PMCID: PMC11273121 DOI: 10.31083/j.rcm2402052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 07/31/2024] Open
Abstract
Cardiorenal syndrome type 3 (CRS3) is defined as acute kidney injury (AKI)-induced acute cardiac dysfunction, characterized by high morbidity and mortality. CRS3 often occurs in elderly patients with AKI who need intensive care. Approximately 70% of AKI patients develop into CRS3. CRS3 may also progress towards chronic kidney disease (CKD) and chronic cardiovascular disease (CVD). However, there is currently no effective treatment. Although the major intermediate factors that can mediate cardiac dysfunction remain elusive, recent studies have summarized the AKI biomarkers, identified direct mechanisms, including mitochondrial dysfunction, inflammation, oxidative stress, apoptosis and activation of the sympathetic nervous system (SNS) and renin-angiotensin-aldosterone system (RAAS), inflammasome, as well as indirect mechanisms such as fluid overload, electrolyte imbalances, acidemia and uremic toxins, which are involved in the pathophysiological changes of CRS3. This study reviews the main pathological characteristics, underlying molecular mechanisms, and potential therapeutic strategies of CRS3. Mitochondrial dysfunction and inflammatory factors have been identified as the key initiators and abnormal links between the impaired heart and kidney, which contribute to the formation of a vicious circle, ultimately accelerating the progression of CRS3. Therefore, targeting mitochondrial dysfunction, antioxidants, Klotho, melatonin, gene therapy, stem cells, exosomes, nanodrugs, intestinal microbiota and Traditional Chinese Medicine may serve as promising therapeutic approaches against CRS3.
Collapse
Affiliation(s)
- Yong Liu
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037 Chongqing, China
| | - Xu Guan
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037 Chongqing, China
| | - Yuming Shao
- Medical Division, Xinqiao Hospital, Army Medical University, 400037 Chongqing, China
| | - Jie Zhou
- Department of Oncology, Southwest Cancer Center, Southwest Hospital, Army Medical University, 400038 Chongqing, China
| | - Yinghui Huang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037 Chongqing, China
| |
Collapse
|
7
|
Evaluation of Four eGFR Calculating Formulae in Predicting Postoperative Acute Kidney Injury in Adult Patients Undergoing Open-Heart Surgery with Cardiopulmonary Bypass. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:6929758. [PMID: 35935325 PMCID: PMC9300297 DOI: 10.1155/2022/6929758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/17/2022] [Accepted: 06/27/2022] [Indexed: 11/18/2022]
Abstract
Objective There are four widely-used formulae to calculate the perioperative glomerular filtration rate (GFR) of patients undergoing cardiac surgery. We assessed the predictive values of these formulae in the occurrence of postoperative acute kidney injury (AKI). Methods Patients who underwent open-heart valvular surgery with cardiopulmonary bypass from January 2015 to October 2017 were enrolled in this retrospective study. Demographic data and perioperative serum creatinine levels were collected. The estimated GFR (eGFR) was calculated using four formulae: Cockcroft Gault (CG), body surface area (BSA) corrected CG, simplified modification of diet in renal disease (MDRD), and chronic kidney disease-epidemiology collaboration (CKD-EPI) formula. The incidence of post-operative AKI was calculated and the predictive capability of these formulae was evaluated. Results A total of 290 patients were included. 136 patients (46.90%) developed AKI after surgery. The eGFR in the AKI group was significantly lower than those in the non-AKI group at all investigated time points. In addition, the eGFR in the non-AKI group increased temporarily on the day of operation, then decreased on the following days, and returned to preoperative level about one week after surgery. However, in the AKI group, the eGFR decreased from the day of operation, which still did not recover to the preoperative level by the end of the first week after surgery. The eGFR calculated by the BSA-standardized CG formula had the highest AUC ROC curves of 0.699 and 0.774 before operation and on the day of operation, respectively. While eGFR calculated by CKD-EPI formula had the highest AUC ROC of 0.874 and 0.887 at the first and second postoperative day. Conclusions The eGFR formula is a powerful tool for perioperative renal function assessment. The BSA-corrected CG and CKD-EPI formula have better performance in predicting postoperative AKI after cardiopulmonary bypass than serum creatinine level and other formulae.
Collapse
|
8
|
Gabbin B, Meraviglia V, Mummery CL, Rabelink TJ, van Meer BJ, van den Berg CW, Bellin M. Toward Human Models of Cardiorenal Syndrome in vitro. Front Cardiovasc Med 2022; 9:889553. [PMID: 35694669 PMCID: PMC9177996 DOI: 10.3389/fcvm.2022.889553] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Heart and kidney diseases cause high morbidity and mortality. Heart and kidneys have vital functions in the human body and, interestingly, reciprocally influence each other’s behavior: pathological changes in one organ can damage the other. Cardiorenal syndrome (CRS) is a group of disorders in which there is combined dysfunction of both heart and kidney, but its underlying biological mechanisms are not fully understood. This is because complex, multifactorial, and dynamic mechanisms are likely involved. Effective treatments are currently unavailable, but this may be resolved if more was known about how the disease develops and progresses. To date, CRS has actually only been modeled in mice and rats in vivo. Even though these models can capture cardiorenal interaction, they are difficult to manipulate and control. Moreover, interspecies differences may limit extrapolation to patients. The questions we address here are what would it take to model CRS in vitro and how far are we? There are already multiple independent in vitro (human) models of heart and kidney, but none have so far captured their dynamic organ-organ crosstalk. Advanced in vitro human models can provide an insight in disease mechanisms and offer a platform for therapy development. CRS represents an exemplary disease illustrating the need to develop more complex models to study organ-organ interaction in-a-dish. Human induced pluripotent stem cells in combination with microfluidic chips are one powerful tool with potential to recapitulate the characteristics of CRS in vitro. In this review, we provide an overview of the existing in vivo and in vitro models to study CRS, their limitations and new perspectives on how heart-kidney physiological and pathological interaction could be investigated in vitro for future applications.
Collapse
Affiliation(s)
- Beatrice Gabbin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Viviana Meraviglia
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, Enschede, Netherlands
| | - Ton J. Rabelink
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, Netherlands
- Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Berend J. van Meer
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Cathelijne W. van den Berg
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, Netherlands
- Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
- Department of Biology, University of Padua, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
- *Correspondence: Milena Bellin, ,
| |
Collapse
|
9
|
Shi S, Zhang B, Li Y, Xu X, Lv J, Jia Q, Chai R, Xue W, Li Y, Wang Y, Wu H, Song Q, Hu Y. Mitochondrial Dysfunction: An Emerging Link in the Pathophysiology of Cardiorenal Syndrome. Front Cardiovasc Med 2022; 9:837270. [PMID: 35282359 PMCID: PMC8914047 DOI: 10.3389/fcvm.2022.837270] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/18/2022] [Indexed: 12/24/2022] Open
Abstract
The crosstalk between the heart and kidney is carried out through various bidirectional pathways. Cardiorenal syndrome (CRS) is a pathological condition in which acute or chronic dysfunction in the heart or kidneys induces acute or chronic dysfunction of the other organ. Complex hemodynamic factors and biochemical and hormonal pathways contribute to the development of CRS. In addition to playing a critical role in generating metabolic energy in eukaryotic cells and serving as signaling hubs during several vital processes, mitochondria rapidly sense and respond to a wide range of stress stimuli in the external environment. Impaired adaptive responses ultimately lead to mitochondrial dysfunction, inducing cell death and tissue damage. Subsequently, these changes result in organ failure and trigger a vicious cycle. In vitro and animal studies have identified an important role of mitochondrial dysfunction in heart failure (HF) and chronic kidney disease (CKD). Maintaining mitochondrial homeostasis may be a promising therapeutic strategy to interrupt the vicious cycle between HF and acute kidney injury (AKI)/CKD. In this review, we hypothesize that mitochondrial dysfunction may also play a central role in the development and progression of CRS. We first focus on the role of mitochondrial dysfunction in the pathophysiology of HF and AKI/CKD, then discuss the current research evidence supporting that mitochondrial dysfunction is involved in various types of CRS.
Collapse
Affiliation(s)
- Shuqing Shi
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bingxuan Zhang
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yumeng Li
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xia Xu
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiayu Lv
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiulei Jia
- Beijing University of Chinese Medicine, Beijing, China
| | - Ruoning Chai
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenjing Xue
- Beijing University of Chinese Medicine, Beijing, China
| | - Yuan Li
- Reproductive and Genetic Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yajiao Wang
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huaqin Wu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Huaqin Wu
| | - Qingqiao Song
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Qingqiao Song
| | - Yuanhui Hu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Yuanhui Hu
| |
Collapse
|
10
|
La disfunción diastólica es un predictor independiente de eventos cardiovasculares tras un fracaso renal agudo. Nefrologia 2022. [DOI: 10.1016/j.nefro.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
11
|
Coyle M, Flaherty G, Jennings C. A critical review of chronic kidney disease as a risk factor for coronary artery disease. IJC HEART & VASCULATURE 2021; 35:100822. [PMID: 34179334 PMCID: PMC8213912 DOI: 10.1016/j.ijcha.2021.100822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/14/2021] [Accepted: 06/04/2021] [Indexed: 11/29/2022]
Abstract
Chronic kidney disease (CKD) is a significant risk factor for cardiovascular disease (CVD). In addition to common CVD risk factors, the presence of CKD is independently associated with an elevated cardiovascular (CV) risk. We examined the association between CKD and CVD, focusing on coronary artery disease (CAD) in both primary and secondary CVD. A total of 94 articles were included for this review using search strategies on Pubmed and Google scholar. The main findings of our review included that besides sharing common risk factors, CKD induces several physiological microscopic changes leading to increased CV risk. These microscopic changes manifest macroscopically with evidence of the development of primary CAD in CKD patients, in addition to accelerating CAD in those with pre-established CV pathology, with CKD consequently being a risk factor for both primary and secondary CAD progression. Current CV guideline recommendations do not discriminate between those patients with and without CKD. Future research is needed in this area, examining if there may be a role for tighter modifiable risk factor targets in this high-risk population.
Collapse
Affiliation(s)
- Mark Coyle
- Corresponding author at: National Institute for Prevention and Cardiovascular Health, Galway, Ireland.
| | | | | |
Collapse
|
12
|
Goffredo G, Barone R, Di Terlizzi V, Correale M, Brunetti ND, Iacoviello M. Biomarkers in Cardiorenal Syndrome. J Clin Med 2021; 10:jcm10153433. [PMID: 34362216 PMCID: PMC8348334 DOI: 10.3390/jcm10153433] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 07/29/2021] [Indexed: 01/16/2023] Open
Abstract
Cardiorenal syndrome is a clinical manifestation of the bidirectional interaction between the heart and kidney diseases. Over the last years, in patients with cardiovascular diseases, several biomarkers have been studied in order to better assess renal function as well as to identify patients prone to experiencing chronic or acute worsening of renal function. The aim of this review is to focus on the possible clinical usefulness of the most recent biomarkers in the setting of cardiorenal syndrome.
Collapse
Affiliation(s)
- Giovanni Goffredo
- Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto 1, 71122 Foggia, Italy; (G.G.); (R.B.); (V.D.T.); (M.C.); (N.D.B.)
- Cardiology Unit, University Policlinic Hospital Riuniti, Viale Luigi Pinto 1, 71122 Foggia, Italy
| | - Roberta Barone
- Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto 1, 71122 Foggia, Italy; (G.G.); (R.B.); (V.D.T.); (M.C.); (N.D.B.)
- Cardiology Unit, University Policlinic Hospital Riuniti, Viale Luigi Pinto 1, 71122 Foggia, Italy
| | - Vito Di Terlizzi
- Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto 1, 71122 Foggia, Italy; (G.G.); (R.B.); (V.D.T.); (M.C.); (N.D.B.)
- Cardiology Unit, University Policlinic Hospital Riuniti, Viale Luigi Pinto 1, 71122 Foggia, Italy
| | - Michele Correale
- Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto 1, 71122 Foggia, Italy; (G.G.); (R.B.); (V.D.T.); (M.C.); (N.D.B.)
- Cardiology Unit, University Policlinic Hospital Riuniti, Viale Luigi Pinto 1, 71122 Foggia, Italy
| | - Natale Daniele Brunetti
- Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto 1, 71122 Foggia, Italy; (G.G.); (R.B.); (V.D.T.); (M.C.); (N.D.B.)
- Cardiology Unit, University Policlinic Hospital Riuniti, Viale Luigi Pinto 1, 71122 Foggia, Italy
| | - Massimo Iacoviello
- Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto 1, 71122 Foggia, Italy; (G.G.); (R.B.); (V.D.T.); (M.C.); (N.D.B.)
- Cardiology Unit, University Policlinic Hospital Riuniti, Viale Luigi Pinto 1, 71122 Foggia, Italy
- Correspondence:
| |
Collapse
|
13
|
Rastogi A, Goyal G, Kesavan R, Bal A, Kumar H, Kamath P, Jude EB, Armstrong DG, Bhansali A. Long term outcomes after incident diabetic foot ulcer: Multicenter large cohort prospective study (EDI-FOCUS investigators) epidemiology of diabetic foot complications study: Epidemiology of diabetic foot complications study. Diabetes Res Clin Pract 2020; 162:108113. [PMID: 32165163 DOI: 10.1016/j.diabres.2020.108113] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/20/2020] [Accepted: 03/02/2020] [Indexed: 10/24/2022]
Abstract
AIMS This long-term prospective study evaluated limb amputation and mortality after the first neuropathic diabetic foot ulcer (DFU). METHODS A total of 2880 patients with neuropathic DFU (DFU group) and a similar number of patients of diabetes without DFU (nDFU) matched for age and diabetes duration were prospectively assessed at five referral-centers over 14 years. Pre-defined outcome was death during follow-up. Various diabetic co-morbidities and amputation were assessed as mortality predictors. RESULTS Overall, 501 (17.4%) patients in DFU group died compared to 89 (3.1%) (p < 0.01) in nDFU group during a median follow-up of 7(1-14) years. The 5- and 10-year mortality was 22% and 71% in the DFU group with a median survival of 7.72 (7.37-8.08) years compared to 3% (p < 0.01) and 5% (p < 0.01) and survival of 12.6 (10.5-12.7) years (p < 0.001) in nDFU group. 29.3% patients had limb amputations. The mortality risk was independent of glycemic control [OR 1.03 (0.80-1.32; p = 0.83)]. However, diabetes duration > 10 years [OR 1.31(1.02-1.70, p = 0.035)], nephropathy [OR 1.47 (1.04-2.09, p < 0.030)], minor 1.85 (1.40-2.44; p < 0.001) or major amputation 2.96 (2.01-4.34, p < 0.001)] predicted mortality. CONCLUSIONS Every one-in-three individual with neuropathic DFU has amputation and every sixth individual has an early demise. Prevalent nephropathy and incident amputation following DFU predicts mortality.
Collapse
Affiliation(s)
- Ashu Rastogi
- Deptt. of Endocrinology, PGIMER, Chandigarh, India.
| | - Ghanshyam Goyal
- ILS Hospital, Salt Lake, Kolkata, India; SVS Marwari Hospital, Amhesrst Street, Kolkata, India
| | | | - Arun Bal
- Deptt. of Endocrinology, Amrita Institute of Medical Sciences, Kochi, India
| | - Harish Kumar
- Deptt. of Endocrinology, Amrita Institute of Medical Sciences, Kochi, India
| | - Priyatham Kamath
- Deptt. of Endocrinology, Amrita Institute of Medical Sciences, Kochi, India
| | - Edward B Jude
- Tameside Hospital, NHS Foundation and Trust, Ashton under Lyne, Manchester, UK
| | - David G Armstrong
- Keck School of Medicine of University of Southern California, Los Angeles, USA
| | | |
Collapse
|
14
|
A Clinically Relevant Functional Model of Type-2 Cardio-Renal Syndrome with Paraventricular Changes consequent to Chronic Ischaemic Heart Failure. Sci Rep 2020; 10:1261. [PMID: 31988300 PMCID: PMC6985167 DOI: 10.1038/s41598-020-58071-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/24/2019] [Indexed: 11/25/2022] Open
Abstract
Cardiorenal syndrome, de novo renal pathology arising secondary to cardiac insufficiency, is clinically recognised but poorly characterised. This study establishes and characterises a valid model representative of Type 2 cardiorenal syndrome. Extensive permanent left ventricular infarction, induced by ligation of the left anterior descending coronary artery in Lewis rats, was confirmed by plasma cardiac troponin I, histology and cardiac haemodynamics. Renal function and morphology was assessed 90-days post-ligation when heart failure had developed. The involvement of the paraventricular nucleus was investigated using markers of inflammation, apoptosis, reactive oxygen species and of angiotensin II involvement. An extensive left ventricular infarct was confirmed following coronary artery ligation, resulting in increased left ventricular weight and compromised left ventricular diastolic function and developed pressure. Glomerular filtration was significantly decreased, fractional excretion of sodium and caspase activities were increased and basement membrane thickening, indicating glomerulosclerosis, was evident. Interestingly, angiotensin II receptor I expression and reactive oxygen species levels in the hypothalamic paraventricular nucleus remained significantly increased at 90-days post-coronary artery ligation, suggesting that these hypothalamic changes may represent a novel, valuable pharmacological target. This model provides conclusive morphological, biochemical and functional evidence of renal injury consequent to heart failure, truly representative of Type-2 cardiorenal syndrome.
Collapse
|
15
|
Tesch GH, Ma FY, Nikolic‐Paterson DJ. Targeting apoptosis signal‐regulating kinase 1 in acute and chronic kidney disease. Anat Rec (Hoboken) 2020; 303:2553-2560. [DOI: 10.1002/ar.24373] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/27/2019] [Accepted: 12/07/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Greg H. Tesch
- Department of NephrologyMonash University Victoria Australia
- Department of MedicineMonash Medical Centre Clayton Victoria Australia
| | - Frank Y. Ma
- Department of NephrologyMonash University Victoria Australia
- Department of MedicineMonash Medical Centre Clayton Victoria Australia
| | - David J. Nikolic‐Paterson
- Department of NephrologyMonash University Victoria Australia
- Department of MedicineMonash Medical Centre Clayton Victoria Australia
| |
Collapse
|
16
|
Brito RBDO, Rebello JF, Grabulosa CC, Pinto W, Morales A, Elias RM, Moyses RMA, Dalboni MA. 25-vitamin D reduces inflammation in uremic environment. Sci Rep 2020; 10:128. [PMID: 31924826 PMCID: PMC6954254 DOI: 10.1038/s41598-019-56874-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 09/23/2019] [Indexed: 01/12/2023] Open
Abstract
Chronic kidney disease (CKD) is characterized by loss of renal function and a consequent increase of serum uremic toxins, which contribute to inflammation status. Deficiency of 25-vitamin D, often found in patients with CKD, has been included as an inflammatory factor since it might modulate the immune system. The aim of this study was to investigate the role of 25-vitamin D on inflammatory pathways in healthy and uremic environment. Toll-like receptor 4 (TLR4), oxidative stress (ROS), vitamin D receptor (VDR), 1-α hydroxylase (CYP27), 24 hydroxylase, cathelicidin, and MCP-1 were evaluated in monocytes exposed to a uremic serum pool compared with healthy pool. The human monocytes lineage (U937) was incubated with or without 25-vitamin D (50 ng/ml for 24 hours). TRL4, VDR, CYP27, CYP24, and ROS were evaluated by flow cytometry. We used ELISA to measure IL-6, TNF-α, IL-10, cathelicidin, and MCP-1 in the cell culture supernatant. We observed a higher expression of TRL-4, IL-6, TNF-α, IL-10, cathelicidin and MCP-1 in monocytes incubated with uremic serum when compared with serum from healthy individuals. Supplementation of 25-vitamin D was able to reduce the expression of TRL4, cathelicidin, and MCP-1 in the uremic environment. There was no difference in the expression of VDR, CYP27 and CYP24 intracellular enzymes. This in vitro study showed that the uremic pool activates inflammatory response in monocytes, which was reversed by 25-vitamin D supplementation; this finding suggests that 25-vitamin D has an anti-inflammatory role in the uremic environment.
Collapse
Affiliation(s)
| | | | | | - Walter Pinto
- Universidade Nove de Julho, UNINOVE, Sao Paulo, Brazil
| | | | - Rosilene Motta Elias
- Universidade Nove de Julho, UNINOVE, Sao Paulo, Brazil.,Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Rosa Maria Affonso Moyses
- Universidade Nove de Julho, UNINOVE, Sao Paulo, Brazil.,Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | | |
Collapse
|
17
|
MALAT1: a therapeutic candidate for a broad spectrum of vascular and cardiorenal complications. Hypertens Res 2019; 43:372-379. [PMID: 31853043 DOI: 10.1038/s41440-019-0378-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/15/2019] [Accepted: 11/15/2019] [Indexed: 01/26/2023]
Abstract
Cardiovascular and renal complications cover a wide array of diseases. The most commonly known overlapping complications include cardiac and renal fibrosis, cardiomyopathy, cardiac hypertrophy, hypertension, and cardiorenal failure. The known or reported causes for the abovementioned complications include injury, ischemia, infection, and metabolic stress. To date, various targets have been reported and investigated in detail that are considered to be the cause of these complications. In the past 5 years, the role of noncoding RNAs has emerged in the area of cardiovascular and renal research, especially in relation to metabolic stress. The long noncoding RNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) has shown immense promise among the long noncoding RNA targets for treating cardiorenal complications. In this review, we shed light on the role of MALAT1 as a primary and novel target in treating cardiovascular and renal diseases as a whole.
Collapse
|
18
|
Abstract
Cardiorenal syndrome commonly refers to the collective dysfunction of heart and kidney resulting in a cascade of feedback mechanism causing damage to both the organs and is associated with adverse clinical outcomes. The pathophysiology of cardiorenal syndrome is complex, multifactorial, and dynamic. Improving the understanding of disease mechanisms will aid in developing targeted pharmacologic and nonpharmacologic therapies for the management of this syndrome. This article discusses the various mechanisms involved in the pathophysiology of the cardiorenal syndrome.
Collapse
Affiliation(s)
- Ujjala Kumar
- Division of Nephrology-Hypertension, University of California San Diego, 9500 Gilman Drive# 9111H, La Jolla, CA 92093-9111, USA
| | - Nicholas Wettersten
- Division of Cardiology, University of California San Diego, 9434 Medical Center Drive, La Jolla, CA 92037, USA
| | - Pranav S Garimella
- Division of Nephrology-Hypertension, University of California San Diego, 9500 Gilman Drive# 9111H, La Jolla, CA 92093-9111, USA.
| |
Collapse
|
19
|
Chien TM, Wen H, Huang JW, Hsieh CC, Chen HM, Chiu CC, Chen YF. Significance of preoperative acute kidney injury in patients with acute type A aortic dissection. J Formos Med Assoc 2019; 118:815-820. [DOI: 10.1016/j.jfma.2018.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/28/2018] [Accepted: 09/03/2018] [Indexed: 11/24/2022] Open
|
20
|
Savira F, Magaye R, Hua Y, Liew D, Kaye D, Marwick T, Wang BH. Molecular mechanisms of protein-bound uremic toxin-mediated cardiac, renal and vascular effects: underpinning intracellular targets for cardiorenal syndrome therapy. Toxicol Lett 2019; 308:34-49. [PMID: 30872129 DOI: 10.1016/j.toxlet.2019.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 02/21/2019] [Accepted: 03/05/2019] [Indexed: 02/07/2023]
Abstract
Cardiorenal syndrome (CRS) remains a global health burden with a lack of definitive and effective treatment. Protein-bound uremic toxin (PBUT) overload has been identified as a non-traditional risk factor for cardiac, renal and vascular dysfunction due to significant albumin-binding properties, rendering these solutes non-dialyzable upon the state of irreversible kidney dysfunction. Although limited, experimental studies have investigated possible mechanisms in PBUT-mediated cardiac, renal and vascular effects. The ultimate aim is to identify relevant and efficacious targets that may translate beneficial outcomes in disease models and eventually in the clinic. This review will expand on detailed knowledge on mechanisms involved in detrimental effects of PBUT, specifically affecting the heart, kidney and vasculature, and explore potential effective intracellular targets to abolish their effects in CRS initiation and/or progression.
Collapse
Affiliation(s)
- Feby Savira
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Ruth Magaye
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Yue Hua
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Danny Liew
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - David Kaye
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia
| | - Tom Marwick
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia
| | - Bing Hui Wang
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia; Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia.
| |
Collapse
|
21
|
Liu S. Heart-kidney interactions: mechanistic insights from animal models. Am J Physiol Renal Physiol 2019; 316:F974-F985. [PMID: 30838876 DOI: 10.1152/ajprenal.00624.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pathological changes in the heart or kidney can instigate the release of a cascade of cardiorenal mediators that promote injury in the other organ. Combined dysfunction of heart and kidney is referred to as cardiorenal syndrome (CRS) and has gained considerable attention. CRS has been classified into five distinct entities, each with different major pathophysiological changes. Despite the magnitude of the public health problem of CRS, the underlying mechanisms are incompletely understood, and effective intervention is unavailable. Animal models have allowed us to discover pathogenic molecular changes to clarify the pathophysiological mechanisms responsible for heart-kidney interactions and to enable more accurate risk stratification and effective intervention. Here, this article focuses on the use of currently available animal models to elucidate mechanistic insights in the clinical cardiorenal phenotype arising from primary cardiac injury, primary renal disease with special emphasis of chronic kidney disease-specific risk factors, and simultaneous cardiorenal/renocardiac dysfunction. The development of novel animal models that recapitulate more closely the cardiorenal phenotype in a clinical scenario and discover the molecular basis of this condition will be of great benefit.
Collapse
Affiliation(s)
- Shan Liu
- School of Medicine, South China University of Technology , Guangzhou , China
| |
Collapse
|
22
|
New mechanisms of CCR5-Δ32 carriers' advantage - Impact on progenitor cells and renal function. Int J Biochem Cell Biol 2019; 108:92-97. [PMID: 30648621 DOI: 10.1016/j.biocel.2019.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 11/26/2018] [Accepted: 01/11/2019] [Indexed: 11/21/2022]
Abstract
BACKGROUND CCR5 is a chemokine receptor expressed by various populations including leukocytes, smooth muscle cells and endothelium. Δ32 polymorphism of CCR5 gene has been connected with, inter alia, cardiovascular disease development. The aim of our study was to evaluate impact of CCR5 variant on CD34+ and CD34+VEGFR2+ cells - populations involved in cardiovascular system homeostasis and regeneration. METHODS AND RESULTS We have examined 170 Polish subjects from Pomeranian region. The analysis concerned CCR5 polymorphism and flow cytometry evaluation of whole blood cells. Our results indicate that individuals with at least one CCR5-Δ32 allele are characterized by greater number of CD34+CXCR4+, CD34+VEGFR2+ and CD34+VEGFR2+c-Kit + cells than their wild type counterparts. This group also exhibits more beneficial values of renal function parameters. CONCLUSION Maintaining greater size of CD34+ and CD34+VEGFR2+ populations as well as proper kidney function may constitute mechanisms that connect chemokine receptor polymorphism with cardiovascular system health.
Collapse
|
23
|
Ham O, Jin W, Lei L, Huang HH, Tsuji K, Huang M, Roh J, Rosenzweig A, Lu HAJ. Pathological cardiac remodeling occurs early in CKD mice from unilateral urinary obstruction, and is attenuated by Enalapril. Sci Rep 2018; 8:16087. [PMID: 30382174 PMCID: PMC6208335 DOI: 10.1038/s41598-018-34216-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 10/04/2018] [Indexed: 12/27/2022] Open
Abstract
Cardiovascular disease constitutes the leading cause of mortality in patients with chronic kidney disease (CKD) and end-stage renal disease. Despite increasing recognition of a close interplay between kidney dysfunction and cardiovascular disease, termed cardiorenal syndrome (CRS), the underlying mechanisms of CRS remain poorly understood. Here we report the development of pathological cardiac hypertrophy and fibrosis in early stage non-uremic CKD. Moderate kidney failure was induced three weeks after unilateral urinary obstruction (UUO) in mice. We observed pathological cardiac hypertrophy and increased fibrosis in UUO-induced CKD (UUO/CKD) animals. Further analysis indicated that this cardiac fibrosis was associated with increased expression of transforming growth factor β (TGF-β) along with significant upregulation of Smad 2/3 signaling in the heart. Moreover early treatment of UUO/CKD animals with an angiotensin-converting-enzyme inhibitor (ACE I), Enalapril, significantly attenuated cardiac fibrosis. Enalapril antagonized activation of the TGF-β signaling pathway in the UUO/CKD heart. In summary our study demonstrates the presence of pathological cardiac hypertrophy and fibrosis in mice early in UUO-induced CKD, in association with early activation of the TGF-β/Smad signaling pathway. We also demonstrate the beneficial effect of ACE I in alleviating this early fibrogenic process in the heart in UUO/CKD animals.
Collapse
Affiliation(s)
- Onju Ham
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - William Jin
- College of Arts & Sciences, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Lei Lei
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Hui Hui Huang
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Kenji Tsuji
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Ming Huang
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jason Roh
- Corrigan Minehan Heart Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Anthony Rosenzweig
- Corrigan Minehan Heart Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Hua A Jenny Lu
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
24
|
Claro LM, Moreno-Amaral AN, Gadotti AC, Dolenga CJ, Nakao LS, Azevedo MLV, de Noronha L, Olandoski M, de Moraes TP, Stinghen AEM, Pécoits-Filho R. The Impact of Uremic Toxicity Induced Inflammatory Response on the Cardiovascular Burden in Chronic Kidney Disease. Toxins (Basel) 2018; 10:toxins10100384. [PMID: 30249039 PMCID: PMC6215310 DOI: 10.3390/toxins10100384] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 09/11/2018] [Accepted: 09/18/2018] [Indexed: 01/23/2023] Open
Abstract
Uremic toxin (UT) retention in chronic kidney disease (CKD) affects biological systems. We aimed to identify the associations between UT, inflammatory biomarkers and biomarkers of the uremic cardiovascular response (BUCVR) and their impact on cardiovascular status as well as their roles as predictors of outcome in CKD patients. CKD patients stages 3, 4 and 5 (n = 67) were recruited and UT (indoxyl sulfate/IS, p-cresil sulfate/pCS and indole-3-acetic acid/IAA); inflammatory biomarkers [Interleukin-6 (IL-6), high sensitivity C reactive protein (hsCRP), monocyte chemoattractant protein-1 (MCP-1), soluble vascular adhesion molecule-1 (sVCAM-1), soluble intercellular adhesion molecule-1 (sICAM-1) and soluble Fas (sFas)] and BUCVRs [soluble CD36 (sCD36), soluble receptor for advanced glycation end products (sRAGE), fractalkine] was measured. Patients were followed for 5.2 years and all causes of death was used as the primary outcome. Artery segments collected at the moment of transplantation were used for the immunohistochemistry analysis in a separate cohort. Estimated glomerular filtration rate (eGFR), circulating UT, plasma biomarkers of systemic and vascular inflammation and BUCVR were strongly interrelated. Patients with plaque presented higher signs of UT-induced inflammation and arteries from CKD patients presented higher fractalkine receptor (CX3CR1) tissue expression. Circulating IS (p = 0.03), pCS (p = 0.007), IL-6 (p = 0.026), sFas (p = 0.001), sCD36 (p = 0.01) and fractalkine (p = 0.02) were independent predictors of total mortality risk in CKD patients. Our results reinforce the important role of uremic toxicity in the pathogenesis of cardiovascular disease (CVD) in CKD patients through an inflammatory pathway.
Collapse
Affiliation(s)
- Ligia Maria Claro
- Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, PR 80215-901, Brazil.
| | - Andrea N Moreno-Amaral
- Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, PR 80215-901, Brazil.
| | - Ana Carolina Gadotti
- Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, PR 80215-901, Brazil.
| | - Carla J Dolenga
- Basic Pathology Department, Universidade Federal do Paraná, Curitiba, PR 80050-540, Brazil.
| | - Lia S Nakao
- Basic Pathology Department, Universidade Federal do Paraná, Curitiba, PR 80050-540, Brazil.
| | - Marina L V Azevedo
- Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, PR 80215-901, Brazil.
| | - Lucia de Noronha
- Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, PR 80215-901, Brazil.
| | - Marcia Olandoski
- Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, PR 80215-901, Brazil.
| | - Thyago P de Moraes
- Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, PR 80215-901, Brazil.
| | - Andréa E M Stinghen
- Basic Pathology Department, Universidade Federal do Paraná, Curitiba, PR 80050-540, Brazil.
| | - Roberto Pécoits-Filho
- Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, PR 80215-901, Brazil.
| |
Collapse
|
25
|
Apoptosis signal-regulating kinase 1 inhibition attenuates cardiac hypertrophy and cardiorenal fibrosis induced by uremic toxins: Implications for cardiorenal syndrome. PLoS One 2017; 12:e0187459. [PMID: 29107962 PMCID: PMC5673193 DOI: 10.1371/journal.pone.0187459] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 10/22/2017] [Indexed: 01/16/2023] Open
Abstract
Intracellular accumulation of protein-bound uremic toxins in the setting of cardiorenal syndrome leads to adverse effects on cardiorenal cellular functions, where cardiac hypertrophy and cardiorenal fibrosis are the hallmarks. In this study, we sought to determine if Apoptosis Signal-Regulated Kinase 1 (ASK1), an upstream regulator of cellular stress response, mediates cardiac hypertrophy and cardiorenal fibrosis induced by indoxyl sulfate (IS) and p-cresol sulfate (PCS) in vitro, and whether ASK1 inhibition is beneficial to ameliorate these cellular effects. PCS augmented cardiac myocyte hypertrophy and fibroblast collagen synthesis (as determined by 3H-leucine and 3H-proline incorporation, respectively), similar to our previous finding with IS. IS and PCS also increased collagen synthesis of proximal tubular cells and renal mesangial cells. Pro-hypertrophic (α-skeletal muscle actin and β-MHC) and pro-fibrotic genes (TGF-β1 and ctgf) were induced by both IS and PCS. Western blot analyses revealed the activation of ASK1 and downstream mitogen activated protein kinases (MAPKs) (p38MAPK and ERK1/2) as well as nuclear factor-kappa B (NF-κB) by IS and PCS. ASK1, OAT1/3, ERK1/2 and p38MAPK inhibitors suppressed all these effects. In summary, IS and PCS exhibit pro-hypertrophic and pro-fibrotic properties, at least in part, via the activation of ASK1 and its downstream pathways. ASK1 inhibitor is an effective therapeutic agent to alleviate protein-bound uremic toxin-induced cardiac hypertrophy and cardiorenal fibrosis in vitro, and may be translated further for cardiorenal syndrome therapy.
Collapse
|
26
|
Mafra D, Borges NA, Cardozo LFMDF, Anjos JS, Black AP, Moraes C, Bergman P, Lindholm B, Stenvinkel P. Red meat intake in chronic kidney disease patients: Two sides of the coin. Nutrition 2017; 46:26-32. [PMID: 29290351 DOI: 10.1016/j.nut.2017.08.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/01/2017] [Accepted: 08/28/2017] [Indexed: 12/29/2022]
Abstract
Red meat is an important dietary source of high biological value protein and micronutrients such as vitamins, iron, and zinc that exert many beneficial functions. However, high consumption of animal protein sources, especially red meat, results in an increased intake of saturated fat, cholesterol, iron, and salt, as well as an excessive acid load. Red meat intake may lead to an elevated production of uremic toxins by the gut microbiota, such as trimethylamine n-oxide (TMAO), indoxyl sulfate, and p-cresyl sulfate. These uremic toxins are associated with increased risk for cardiovascular (CV) mortality. Limiting the intake of red meat in patients with chronic kidney disease (CKD) thus may be a good strategy to reduce CV risk, and may slow the progression of kidney disease. In the present review, we discuss the role of red meat in the diet of patients with CKD. Additionally, we report on a pilot study that focused on the effect of a low-protein diet on TMAO plasma levels in nondialysis CKD patients.
Collapse
Affiliation(s)
- Denise Mafra
- Post Graduation Program in Medical Sciences, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil; Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil.
| | - Natalia A Borges
- Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil
| | | | - Juliana S Anjos
- Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil
| | - Ana Paula Black
- Post Graduation Program in Medical Sciences, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil
| | - Cristiane Moraes
- Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil
| | - Peter Bergman
- Department of Laboratory Medicine, Clinical Microbiology, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Bengt Lindholm
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institute, Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
27
|
Yang JY, Huang JW, Chen L, Chen YY, Pai MF, Tung KT, Peng YS, Hung KY. Frequency of Early Predialysis Nephrology Care and Postdialysis Cardiovascular Events. Am J Kidney Dis 2017; 70:164-172. [DOI: 10.1053/j.ajkd.2016.12.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/19/2016] [Indexed: 11/11/2022]
|
28
|
Gao H, Liu S. Role of uremic toxin indoxyl sulfate in the progression of cardiovascular disease. Life Sci 2017; 185:23-29. [PMID: 28754616 DOI: 10.1016/j.lfs.2017.07.027] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/07/2017] [Accepted: 07/24/2017] [Indexed: 12/19/2022]
Abstract
The prevalence of cardiovascular disease (CVD) among patients with chronic kidney disease (CKD) is relatively high. Deterioration of renal function in CKD leads to accumulation of indoxyl sulfate, a tryptophan metabolite produced by gut microbiota. It is acknowledged that indoxyl sulfate is capable to stimulate oxidative stress, which in turn contributes to the progression of vascular disorders and its resultant coronary artery disease. Recent research have demonstrated the adverse effects of indoxyl sulfate on the heart, together with the acceleration of vascular dysfunction, suggesting that indoxyl sulfate might contribute to high prevalence of CVD in CKD. The present mini review has focused on the potential mechanisms by which indoxyl sulfate exerts this pro-oxidant effects on the cardiovascular system. The action of indoxyl sulfate are related to multiple NADPH oxidase-mediated redox signaling pathways, which have been implicated in the pathophysiology of different forms of CVD, including chronic heart failure, arrhythmia, atherosclerotic vascular disease and coronary calcification. Future therapeutic options are discussed, including modulating gut microbial flora and blocking responsible pathophysiologic pathways.
Collapse
Affiliation(s)
- Huichang Gao
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Shan Liu
- School of Medicine, South China University of Technology, Guangzhou 510006, China.
| |
Collapse
|
29
|
Rosenzweig B, Rubinstein ND, Reznik E, Shingarev R, Juluru K, Akin O, Hsieh JJ, Jaimes EA, Russo P, Susztak K, Coleman JA, Hakimi AA. Benign and tumor parenchyma metabolomic profiles affect compensatory renal growth in renal cell carcinoma surgical patients. PLoS One 2017; 12:e0180350. [PMID: 28727768 PMCID: PMC5519040 DOI: 10.1371/journal.pone.0180350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 06/14/2017] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Pre-operative kidney volume is an independent predictor of glomerular filtration rate in renal cell carcinoma patients. Compensatory renal growth (CRG) can ensue prior to nephrectomy in parallel to tumor growth and benign parenchyma loss. We aimed to test whether renal metabolite abundances significantly associate with CRG, suggesting a causative relationship. DESIGN, SETTING, PARTICIPANTS, AND MEASUREMENTS Tissue metabolomics data from 49 patients, with a median age of 60 years, were previously collected and the pre-operative fold-change of their contra to ipsi-lateral benign kidney volume served as a surrogate for their CRG. Contra-lateral kidney volume fold-change within a 3.3 +/- 2.1 years follow-up interval was used as a surrogate for long-term CRG. Using a multivariable statistical model, we identified metabolites whose abundances significantly associate with CRG. RESULTS Our analysis found 13 metabolites in the benign (e.g. L-urobilin, Variable Influence in Projection, VIP, score = 3.02, adjusted p = 0.017) and 163 metabolites in the malignant (e.g. 3-indoxyl-sulfate, VIP score = 1.3, adjusted p = 0.044) tissues that significantly associate with CRG. Benign/tumor fold change in metabolite abundances revealed three additional metabolites with that significantly positively associate with CRG (e.g. p-cresol sulfate, VIP score = 2.945, adjusted p = 0.033). At the pathway level, we show that fatty-acid oxidation is highly enriched with metabolites whose benign tissue abundances strongly positively associate with CRG, both pre-operatively and long term, whereas in the tumor tissue significant enrichment of dipeptides and benzoate (positive association), glycolysis/gluconeogenesis, lysolipid and nucleotide sugar pentose (negative associations) sub-pathways, were observed. CONCLUSION These data suggest that specific biological processes in the benign as well as in the tumor parenchyma strongly influence compensatory renal growth.
Collapse
Affiliation(s)
- Barak Rosenzweig
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Nimrod D. Rubinstein
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Ed Reznik
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Roman Shingarev
- Renal Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Krishna Juluru
- Body Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Oguz Akin
- Body Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - James J. Hsieh
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Edgar A. Jaimes
- Renal Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Paul Russo
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jonathan A. Coleman
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- * E-mail: (AAH); (JAC)
| | - A. Ari Hakimi
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- * E-mail: (AAH); (JAC)
| |
Collapse
|
30
|
Kamiński TW, Pawlak K, Karbowska M, Myśliwiec M, Pawlak D. Indoxyl sulfate - the uremic toxin linking hemostatic system disturbances with the prevalence of cardiovascular disease in patients with chronic kidney disease. BMC Nephrol 2017; 18:35. [PMID: 28122514 PMCID: PMC5267373 DOI: 10.1186/s12882-017-0457-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 01/19/2017] [Indexed: 12/19/2022] Open
Abstract
Background During chronic kidney disease progression, kidney-specific risk factors for cardiovascular disease come into play. The present study investigated the impact of indoxyl sulfate, dietary tryptophan-derived uremic toxin, accumulated in the blood of patients with chronic kidney disease on hemostatic parameters, markers of inflammation, oxidative stress and monocyte to macrophage transition. Methods Fifty-one CKD patients not undergoing hemodialysis were enrolled in the study. Coagulation factors, fibrinolytic parameters, adhesion molecules, endothelial dysfunction markers, oxidative stress as well as inflammation markers were examined using immune-enzymatic method. Indoxyl sulfate levels were assessed using high-performance liquid chromatography. Biochemical parameters were determined by routine laboratory techniques using an automated analyzers. All assessed parameters were compared with controls and subjected to cross-sectional statistical analysis. Results Elevated concentrations of indoxyl sulfate, the vast majority of parameters affecting hemostasis, and markers of renal insufficiency conditions were observed. Part of hemostatic factors, namely tissue factor, von Willebrand factor, thrombomodulin, soluble urokinase-type plasminogen activator receptor, soluble intercellular adhesion molecule-1, soluble vascular cell adhesion protein were correlated with the fraction of indoxyl sulfate. A significant quantity of assessed parameters showed strong correlations with superoxide-dismutase, renal insufficiency rate, C-reactive protein, and neopterin. Levels of indoxyl sulfate were independently associated with markers of impaired endothelial function (thrombomodulin, adhesion molecules), oxidative stress (superoxide-dismutase) and monocytes activation determinant (neopterin), which indicate unconventional links between these systems and the role of indoxyl sulfate. Furthermore, parameters that correlated with the levels of indoxyl sulfate (von Willebrand factor, soluble urokinase-type plasminogen activator receptor, soluble intercellular adhesion molecule-1) were positively associated with the prevalence of cardiovascular disease in a CKD patients. Conclusions The study demonstrated that in conditions of chronic exposure to uremic toxins, indoxyl sulfate seems to be one of the “missing links” between impaired renal function and prevalence of cardiovascular events, especially hemostatic disorders. The main functions of the action appear to be altered monocytes activation, intensified inflammatory process, and augmented oxidative stress by this uremic toxin.
Collapse
Affiliation(s)
- Tomasz W Kamiński
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C Str., 15-222, Białystok, Poland
| | - Krystyna Pawlak
- Department of Monitored Pharmacotherapy, Medical University of Bialystok, Mickiewicza 2C Str., 15-089, Białystok, Poland
| | - Małgorzata Karbowska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C Str., 15-222, Białystok, Poland
| | - Michał Myśliwiec
- Department of Nephrology and Clinical Transplantation, Medical University of Bialystok, Żurawia 14 Str., 15-540, Bialystok, Poland
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C Str., 15-222, Białystok, Poland.
| |
Collapse
|
31
|
Cernea S. Heart Failure and Chronic Kidney Disease in Type 2 Diabetes. JOURNAL OF INTERDISCIPLINARY MEDICINE 2016. [DOI: 10.1515/jim-2016-0066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Abstract
Complex hemodynamic, neurohormonal and biochemical changes occur in heart failure and chronic kidney disease, and hyperglycemia/diabetes further accentuate the multifactorial pathogenetic mechanisms. The acknowledgement of concomitant heart and kidney dysfunction in patients with type 2 diabetes has major clinical implications with regards to prognosis, as they significantly increase the risk of mortality, and to therapeutical strategy of both conditions, as well as of hyperglycemia. A comprehensive interdisciplinary approach is needed in these cases in order to improve the outcomes.
Collapse
Affiliation(s)
- Simona Cernea
- Department M3/Internal Medicine IV, University of Medicine and Pharmacy, Tîrgu Mureş, Romania
- Diabetes, Nutrition and Metabolic Diseases Outpatient Unit, County Emergency Clinical Hospital, Tîrgu Mureş, Romania
| |
Collapse
|
32
|
Chen HH, Cheng PW, Ho WY, Lu PJ, Lai CC, Tseng YM, Fang HC, Sun GC, Hsiao M, Liu CP, Tseng CJ. Renal Denervation Improves the Baroreflex and GABA System in Chronic Kidney Disease-induced Hypertension. Sci Rep 2016; 6:38447. [PMID: 27917928 PMCID: PMC5137107 DOI: 10.1038/srep38447] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/07/2016] [Indexed: 12/18/2022] Open
Abstract
Hypertensive rats with chronic kidney disease (CKD) exhibit enhanced gamma-aminobutyric acid (GABA)B receptor function and regulation within the nucleus tractus solitarii (NTS). For CKD with hypertension, renal denervation (RD) interrupts the afferent renal sympathetic nerves, which are connecting to the NTS. The objective of the present study was to investigate how RD improves CKD-induced hypertension. Rats underwent 5/6 nephrectomy for 8 weeks, which induced CKD and hypertension. RD was induced by applying phenol to surround the renal artery in CKD. RD improved blood pressure (BP) by lowering sympathetic nerve activity and markedly restored the baroreflex response in CKD. The GABAB receptor expression was increased in the NTS of CKD; moreover, the central GABA levels were reduced in the cerebrospinal fluid, and the peripheral GABA levels were increased in the serum. RD restored the glutamic acid decarboxylase activity in the NTS in CKD, similar to the effect observed for central treatment with baclofen, and the systemic administration of gabapentin reduced BP. RD slightly improved renal function and cardiac load in CKD. RD may improve CKD-induced hypertension by modulating the baroreflex response, improving GABA system dysfunction and preventing the development and reducing the severity of cardiorenal syndrome type 4 in CKD rats.
Collapse
Affiliation(s)
- Hsin-Hung Chen
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Yuh-Ing Junior College of Health Care &Management, Kaohsiung, Taiwan
| | - Pei-Wen Cheng
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Yuh-Ing Junior College of Health Care &Management, Kaohsiung, Taiwan
| | - Wen-Yu Ho
- Division of General Internal Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Pei-Jung Lu
- Graduate Institute of Clinical Medicine, National Cheng-Kung University, Tainan, Taiwan
| | - Chi-Cheng Lai
- Cardiovascular Center, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Yang-Ming Tseng
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Hua-Chang Fang
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Gwo-Ching Sun
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Chun-Peng Liu
- Department of Administration, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Section of Cardiology, Department of Medcine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ching-Jiunn Tseng
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
33
|
Pinheiro da Silva AL, Vaz da Silva MJ. Type 4 cardiorenal syndrome. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2016. [DOI: 10.1016/j.repce.2016.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
34
|
Pinheiro da Silva AL, Vaz da Silva MJ. Type 4 cardiorenal syndrome. Rev Port Cardiol 2016; 35:601-616. [PMID: 27712930 DOI: 10.1016/j.repc.2016.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 06/04/2016] [Indexed: 12/25/2022] Open
Abstract
The Acute Dialysis Quality Initiative consensus conference proposed a classification of cardiorenal syndrome (CRS), aiming for a better delineation of each subtype. Although the exact pathophysiology of type 4 CRS is not completely understood, the mechanisms involved are probably multifactorial. There is growing evidence that oxidative stress is a major connector in the development and progression of type 4 CRS. Giving its complexity, poor prognosis and increasing incidence, type 4 CRS is becoming a significant public health problem. Patients with chronic kidney disease are particularly predisposed to cardiac dysfunction, due to the high prevalence of traditional cardiovascular risk factors in this population, but the contribution of risk factors specific to chronic kidney disease should also be taken into account. Much remains to be elucidated about type 4 CRS: despite progress over the last decade, there are still significant questions regarding its pathophysiology and there is as yet no specific therapy. A better understanding of the mechanisms involved may provide potential targets for intervention. The present review will provide a brief description of the definition, epidemiology, diagnosis, prognosis, biomarkers and management strategies of type 4 CRS, and the pathophysiological mechanisms and risk factors presumably involved in its development will be particularly highlighted.
Collapse
|
35
|
Ryom L, Lundgren JD, Ross M, Kirk O, Law M, Morlat P, Fontas E, Smit C, Fux CA, Hatleberg CI, de Wit S, Sabin CA, Mocroft A. Renal Impairment and Cardiovascular Disease in HIV-Positive Individuals: The D:A:D Study. J Infect Dis 2016; 214:1212-20. [PMID: 27485357 DOI: 10.1093/infdis/jiw342] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 07/26/2016] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND While the association between renal impairment and cardiovascular disease (CVD) is well established in the general population, the association remains poorly understood in human immunodeficiency virus (HIV)-positive individuals. METHODS Individuals with ≥2 estimated glomerular filtration rate (eGFR) measurements after 1 February 2004 were followed until CVD, death, last visit plus 6 months, or 1 February 2015. CVD was defined as the occurrence of centrally validated myocardial infarction, stroke, invasive cardiovascular procedures, or sudden cardiac death. RESULTS During a median follow-up duration of 8.0 years (interquartile range, 5.4-8.9 years) 1357 of 35 357 individuals developed CVD (incidence rate, 5.2 cases/1000 person-years [95% confidence interval {CI}, 5.0-5.5]). Confirmed baseline eGFR and CVD were closely related with 1.8% of individuals (95% CI, 1.6%-2.0%) with an eGFR > 90 mL/minute/1.73 m(2) estimated to develop CVD at 5 years, increasing to 21.1% (95% CI, 6.6%-35.6%) among those with an eGFR ≤ 30 mL/minute/1.73 m(2) The strong univariate relationship between low current eGFR and CVD was primarily explained by increasing age in adjusted analyses, although all eGFRs ≤ 80 mL/minute/1.73 m(2) remained associated with 30%-40% increased CVD rates, and particularly high CVD rates among individuals with an eGFR ≤ 30 mL/minute/1.73 m(2) (incidence rate ratio, 3.08 [95% CI, 2.04-4.65]). CONCLUSIONS Among HIV-positive individuals in a large contemporary cohort, a strong relation between confirmed impaired eGFR and CVD was observed. This finding highlights the need for renal preventive measures and intensified monitoring for emerging CVD, particularly in older individuals with continuously low eGFRs.
Collapse
Affiliation(s)
- Lene Ryom
- Department of Infectious Diseases, CHIP, Section 2100, Rigshospitalet, University of Copenhagen, Denmark
| | - Jens D Lundgren
- Department of Infectious Diseases, CHIP, Section 2100, Rigshospitalet, University of Copenhagen, Denmark
| | - Mike Ross
- Division of Nephrology, Mount Sinai School of Medicine, New York
| | - Ole Kirk
- Department of Infectious Diseases, CHIP, Section 2100, Rigshospitalet, University of Copenhagen, Denmark
| | - Matthew Law
- Kirby Institute, University of New South Wales, Sydney, Australia
| | | | - Eric Fontas
- Nephrology Department, Public Health Department, CHU Nice, France
| | - Colette Smit
- Academic Medical Center, Division of Infectious Diseases Department of Global Health, University of Amsterdam HIV Monitoring Foundation, Amsterdam, The Netherlands
| | - Christoph A Fux
- Clinic for Infectious Diseases and Hospital Hygiene, Kantonsspital Aarau, Switzerland
| | - Camilla I Hatleberg
- Department of Infectious Diseases, CHIP, Section 2100, Rigshospitalet, University of Copenhagen, Denmark
| | - Stéphane de Wit
- Department of Infectious Diseases, CHU Saint-Pierre, Brussels, Belgium
| | - Caroline A Sabin
- Research Department of Infection and Population Health, University College London, United Kingdom
| | - Amanda Mocroft
- Research Department of Infection and Population Health, University College London, United Kingdom
| | | |
Collapse
|
36
|
Affiliation(s)
- Louise Robson
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| |
Collapse
|