1
|
Yozgat I, Cakır U, Serdar MA, Sahin S, Sezerman OU, Nemutlu E, Baykal AT, Serteser M. Longitudinal non-targeted metabolomic profiling of urine samples for monitoring of kidney transplantation patients. Ren Fail 2024; 46:2300736. [PMID: 38213228 PMCID: PMC10791079 DOI: 10.1080/0886022x.2023.2300736] [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: 08/29/2023] [Accepted: 12/26/2023] [Indexed: 01/13/2024] Open
Abstract
The assessment of kidney function within the first year following transplantation is crucial for predicting long-term graft survival. This study aimed to develop a robust and accurate model using metabolite profiles to predict early long-term outcomes in patient groups at the highest risk of early graft loss. A group of 61 kidney transplant recipients underwent thorough monitoring during a one-year follow-up period, which included a one-week hospital stay and follow-up assessments at three and six months. Based on their 12-month follow-up serum creatinine levels: Group 2 had levels exceeding 1.5 mg/dl, while Group 1 had levels below 1.5 mg/dl. Metabolites were detected by mass spectrometer and first pre-processed. Univariate and multivariate statistical analyses were employed to identify significant differences between the two groups. Nineteen metabolites were found to differ significantly in the 1st week, and seventeen metabolites in the 3rd month (adjusted p-value < 0.05, quality control (QC) < 30, a fold change (FC) > 1.1 or a FC < 0.91, Variable Influence on Projection (VIP) > 1). However, no significant differences were observed in the 6th month. These distinctive metabolites mainly belonged to lipid, fatty acid, and amino acid categories. Ten models were constructed using a backward conditional approach, with the best performance seen in model 5 for Group 2 at the 1st-week mark (AUC 0.900) and model 3 at the 3rd-month mark (AUC 0.924). In conclusion, the models developed in the early stages may offer potential benefits in the management of kidney transplant patients.
Collapse
Affiliation(s)
- Ihsan Yozgat
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Ulkem Cakır
- Department of Nephrology, Acibadem University School of Medicine, Istanbul, Turkey
| | | | - Sevgi Sahin
- Department of Nephrology, Acibadem University School of Medicine, Istanbul, Turkey
| | - Osman Ugur Sezerman
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Acibadem University, Istanbul, Turkey
| | - Emirhan Nemutlu
- Faculty of Pharmacy, Department of Analytical Chemistry, Hacettepe University, Ankara, Türkiye
| | - Ahmet Tarik Baykal
- Department of Medical Biochemistry, Faculty of Medicine, Acibadem University, Istanbul, Turkey
| | - Mustafa Serteser
- Department of Medical Biochemistry, Faculty of Medicine, Acibadem University, Istanbul, Turkey
| |
Collapse
|
2
|
Saberi S, Najafipour H, Rajizadeh MA, Etminan A, Jafari E, Iranpour M. NaHS protects brain, heart, and lungs as remote organs from renal ischemia/reperfusion-induced oxidative stress in male and female rats. BMC Nephrol 2024; 25:373. [PMID: 39438873 PMCID: PMC11515705 DOI: 10.1186/s12882-024-03824-3] [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: 06/17/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Acute Kidney Injury (AKI) is frequently observed in hospitalized patients in intensive care units, often caused by renal ischemia-reperfusion injury (IRI). IRI disrupts the function of various 'remote organs' such as the lungs, pancreas, intestine, liver, heart, and brain through inflammation, oxidative stress, apoptosis, leukocyte infiltration, and increased urea and creatinine levels. Gender differences in renal IRI-induced injury are noted. H2S, an endogenous gaseous modulator, shows potential in vasodilation, bronchodilation, and hypotension and can regulate apoptosis, inflammation, angiogenesis, metabolism, and oxidative stress. This study aims to investigate the protective effects of NaHS on brain, heart, and lung injuries following renal IR and to assess the oxidative system status as a potential mechanism in male and female rats.Forty-eight Wistar rats were randomly divided into eight groups (n = 6): Control/Saline, Sham/Saline, IR/Saline, and IR/NaHS in both sexes. Forty-five minutes of bilateral renal ischemia followed by 24-hour reperfusion was induced in the IR groups. NaHS (100µM/Kg, IP) was administered 10 min before clamp release in treated groups. BUN, SCr, BUN/SCr, albuminuria, histopathology, and oxidative stress biomarkers of the brain, heart, and lung were assessed as remote organs. IR increased serum markers of renal function, albuminuria, malondialdehyde levels, and tissue injury scores while reducing nitrite levels and superoxide dismutase and glutathione peroxidase activities. NaHS treatment reversed the adverse effects of IR in remote organs in both sexes, although it showed limited improvement in renal function. Our findings demonstrate that NaHS has a beneficial effect on remote organ injury following renal IR by mitigating oxidative stress, with noted tissue-specific and gender-specific differences in response. These findings suggest NaHS as a potential therapeutic agent for mitigating multi-organ injury after renal IR, with effects varying by tissue and gender.
Collapse
Affiliation(s)
- Shadan Saberi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Hamid Najafipour
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Amin Rajizadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Endocrinology and Metabolism Research Center, Institute of basic and clinical physiology sciences, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Abbas Etminan
- Physiology Research Center, Departments of Nephrology, Urology and Renal Transplantation, Kerman University of Medical Sciences, Kerman, Iran.
| | - Elham Jafari
- Department of Pathology, Pathology and Stem Cell Research Center, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Iranpour
- Department of Pathology, Pathology and Stem Cell Research Center, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
3
|
Golestaneh L, Basalely A, Linkermann A, El-Achkar TM, Kim RS, Neugarten J. Sex, Acute Kidney Injury, and Age: A Prospective Cohort Study. Am J Kidney Dis 2024:S0272-6386(24)01037-0. [PMID: 39447957 DOI: 10.1053/j.ajkd.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024]
Abstract
RATIONALE & OBJECTIVE Animal models of kidney disease suggest a protective role for female sex hormones but in humans, some authorities assert that female sex is a risk factor for acute kidney injury (AKI). To better understand the risk of AKI, we studied the strength of association between sex and AKI incidence in hormonally distinct age groups across the life span. STUDY DESIGN Prospective cohort study. SETTINGS & PARTICIPANTS All patients hospitalized in the Montefiore Health System between 10/15/2015 and 1/1/2019, excluding those with kidney failure or obstetrics diagnoses. EXPOSURE Male versus female sex. OUTCOMES Acute kidney injury (AKI) occurring during hospitalization based on KDIGO definitions. ANALYTICAL APPROACH Generalized Estimating Equation logistic regression adjusted for comorbidities, socio-demographic factors, and severity of illness. Analyses were stratified into 3 age categories, 6 months to ≤16 years, age >16 years - <55 years, and age ≥55 years. RESULTS A total of 132,667 individuals were hospitalized a total of 235,629 times. The mean age was 55.2 (SD 23.8) years. The counts (%) of hospitalizations for women were 129,912 (55%). Hospitalization counts (%) among Black and Hispanic patients were 71,834 (30.5%) and 24,199 (10.3%), respectively. AKI occurred in 53,926 (22.9%) hospitalizations. In adjusted models, there was a significant interaction between age and sex (p<0.001). Boys and men had higher risk of AKI across all age groups, an association more pronounced in the age group >16 years to <55 years in which the OR for men was 1.7 (95% CI, 1.6-1.8). This age-based pattern remained consistent across prespecified types of hospitalizations. In a sensitivity analysis, women older than 55 years who received prescriptions for estrogen had lower odds of AKI than those without prescriptions. LIMITATIONS Residual confounding. CONCLUSION The greatest relative risk of AKI for males occurred during ages >16 to <55 years. The lower risk among post-menopausal women receiving supplemental estrogen supports a protective role for female sex hormones.
Collapse
Affiliation(s)
- Ladan Golestaneh
- Department of Medicine, Section of Nephrology, Yale University School of Medicine, New Haven, CT; Department of Medicine, Division of Nephrology, Albert Einstein College of Medicine, Bronx, NY.
| | - Abby Basalely
- Department of Pediatrics, Division of Pediatric Nephrology, Northwell Health, New Hyde Park, NY
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; Department of Medicine, Division of Nephrology, Albert Einstein College of Medicine, Bronx, NY
| | - Tarek M El-Achkar
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, and the Roudebush Indianapolis VA, Indianapolis, IN
| | - Ryung S Kim
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Joel Neugarten
- Department of Medicine, Division of Nephrology, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
4
|
Tirtayasa PMW, Situmorang GR, Duarsa GWK, Mahadita GW, Ghinorawa T, Myh E, Nugroho EA, Kandarini Y, Rodjani A, Rasyid N. Risk factors of delayed graft function following living donor kidney transplantation: A meta-analysis. Transpl Immunol 2024; 86:102094. [PMID: 39053613 DOI: 10.1016/j.trim.2024.102094] [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: 11/16/2023] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
INTRODUCTION Delayed graft function (DGF) is a common condition that necessitates dialysis during the first week after transplantation. Although DGF rarely occurs following living-donor kidney transplantation (LDKT), it may eventually lead to acute or chronic graft rejection. This study aimed to assess the risk factors for DGF in patients who underwent LDKT. METHODS A systematic review and meta-analysis of studies published before August 2022 was conducted using the PubMed, Science Direct, Cochrane, and Directory of Open Access Journal (DOAJ) databases. The review included studies that assessed the incidence of DGF following LDKT, and examined its risk factors, while excluding studies involving deceased donors. Potential risk factors were analyzed using pooled mean differences or odds ratios with 95% confidence intervals (CIs). Review Manager 5.3 was used for the meta-analysis. RESULTS Among the 13 included studies, 3685 cases of DGF were identified in a total of 113,261 patients (3.25%). Potential risk factors for DGF following LDKT were examined across several aspects, including donor, recipient, donor/recipient relationship, and immunological and intraoperative factors. The identified risk factors included older donors (P = 0.07), male recipients (P < 0.0001), higher recipient body mass index (BMI) (P < 0.0001), non-white recipients (P < 0.0001), pre-existing diabetes (P < 0.0001), pre-existing hypertension (P = 0.01), history of dialysis (P < 0.0001), re-transplantation (P = 0.004), unrelated donor/recipient (P = 0.02), ABO incompatibility (P < 0.0001), higher panel reactive antibody (PRA) levels (P < 0.0001), utilization of right kidney (P < 0.0001), and longer cold ischemia time (CIT) (P = 0.004). CONCLUSION Several factors related to the donor, recipient, donor/recipient relationship, and immunological and intraoperative aspects were identified as potential risk factors for the development of DGF following LDKT. Addressing and optimizing these factors may improve the long-term outcomes of LDKT.
Collapse
Affiliation(s)
- Pande Made Wisnu Tirtayasa
- Department of Urology, Faculty of Medicine, Universitas Udayana, Universitas Udayana Teaching Hospital, Bali, Indonesia.
| | - Gerhard Reinaldi Situmorang
- Department of Urology, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo National Referral Hospital, Jakarta, Indonesia
| | - Gede Wirya Kusuma Duarsa
- Department of Urology, Faculty of Medicine, Universitas Udayana, Prof. Dr. I.G.N.G. Ngoerah General Hospital, Bali, Indonesia
| | - Gede Wira Mahadita
- Department of Nephrology, Faculty of Medicine, Universitas Udayana, Prof. Dr. I.G.N.G. Ngoerah General Hospital, Bali, Indonesia
| | - Tanaya Ghinorawa
- Department of Urology, Faculty of Medicine, Universitas Gadjah Mada, Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Etriyel Myh
- Department of Urology, Faculty of Medicine, Universitas Andalas, Dr. M.Djamil General Hospital, Padang, Indonesia
| | - Eriawan Agung Nugroho
- Department of Urology, Faculty of Medicine, Universitas Diponegoro, Dr. Kariadi General Hospital, Semarang, Indonesia
| | - Yenny Kandarini
- Department of Nephrology, Faculty of Medicine, Universitas Udayana, Prof. Dr. I.G.N.G. Ngoerah General Hospital, Bali, Indonesia
| | - Arry Rodjani
- Department of Urology, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo National Referral Hospital, Jakarta, Indonesia
| | - Nur Rasyid
- Department of Urology, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo National Referral Hospital, Jakarta, Indonesia
| |
Collapse
|
5
|
Jankowski J, Lee HK, Liu C, Wilflingseder J, Hennighausen L. Sexually dimorphic renal expression of mouse Klotho is directed by a kidney-specific distal enhancer responsive to HNF1b. Commun Biol 2024; 7:1142. [PMID: 39277686 PMCID: PMC11401919 DOI: 10.1038/s42003-024-06855-6] [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: 03/29/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024] Open
Abstract
Transcription enhancers are genomic sequences regulating common and tissue-specific genes and their disruption can contribute to human disease development and progression. Klotho, a sexually dimorphic gene specifically expressed in kidney, is well-linked to kidney dysfunction and its deletion from the mouse genome leads to premature aging and death. However, the sexually dimorphic regulation of Klotho is not understood. Here, we characterize two candidate Klotho enhancers using H3K27ac epigenetic marks and transcription factor binding and investigate their functions, individually and combined, through CRISPR-Cas9 genome engineering. We discovered that only the distal (E1), but not the proximal (E2) candidate region constitutes a functional enhancer, with the double deletion not causing Klotho expression to further decrease. E1 activity is dependent on HNF1b transcription factor binding site within the enhancer. Further, E1 controls the sexual dimorphism of Klotho as evidenced by qPCR and RNA-seq. Despite the sharp reduction of Klotho mRNA, unlike germline Klotho knockouts, mutant mice present normal phenotype, including weight, lifespan, and serum biochemistry. Lastly, only males lacking E1 display more prominent acute, but not chronic kidney injury responses, indicating a remarkable range of potential adaptation to isolated Klotho loss, especially in female E1 knockouts, retaining renoprotection despite over 80% Klotho reduction.
Collapse
Affiliation(s)
- Jakub Jankowski
- Section of Genetics and Physiology, Laboratory of Cellular and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, MD, 20892, USA.
- , 8 Center Drive, Room 107, 20892, Bethesda, MD, USA.
| | - Hye Kyung Lee
- Section of Genetics and Physiology, Laboratory of Cellular and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, MD, 20892, USA
| | - Chengyu Liu
- Transgenic Core, National Heart, Lung, and Blood Institute, US National Institutes of Health, Bethesda, MD, 20892, USA
| | - Julia Wilflingseder
- Department of Physiology and Pathophysiology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Lothar Hennighausen
- Section of Genetics and Physiology, Laboratory of Cellular and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
6
|
Peng H, Liang Z, Huang B, Zhang S, Yang Y. Negative association of serum neurofilament light chain with estimated glomerular filtration rate levels and the impact of gender. Front Neurol 2024; 15:1457984. [PMID: 39323436 PMCID: PMC11422151 DOI: 10.3389/fneur.2024.1457984] [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: 07/01/2024] [Accepted: 08/23/2024] [Indexed: 09/27/2024] Open
Abstract
Background The relationship between kidney function and brain function is complex and poorly understood. This study aims to investigate the association between serum neurofilament light chain (sNfL) and levels of estimated glomerular filtration rate (eGFR), offering new insights into their interactions. Methods Data from the national health and nutrition examination survey (NHANES) in 2013-2014, linked with national death index records, were used. Participants who met specific criteria were analyzed. Baseline characteristics were stratified by tertiles of sNfL levels and compared using weighted Kruskal-Wallis and chi-square tests. Weighted linear regression models, both unadjusted and adjusted, evaluated the relationship between log sNfL and eGFR. Subgroup and interaction analyses validated the findings. Restricted cubic spline, scatter plots, and Spearman correlation confirmed the relationship between log sNfL and eGFR. Results A total of 2,038 eligible participants were included. Higher sNfL levels were significantly associated with lower eGFR (p < 0.01). The highest sNfL tertile had a significantly higher mortality rate (p < 0.01). Fully adjusted multivariable weighted linear regression showed a significant negative correlation between log sNfL and eGFR (per 10-unit increase; β = -0.07, 95% CI: -0.10 to -0.04, p < 0.01). Subgroup analyses consistently supported this negative correlation (p < 0.01). Interaction analysis revealed a significant gender difference (p = 0.032), with males showing a - 0.06 (-0.09, -0.04) decrease and females a - 0.07 (-0.11, -0.04) decrease in log sNfL per 10-unit increase in eGFR. Restricted cubic spline confirmed a linear relationship (p-non-linear = 0.121), and the Spearman correlation coefficient was -0.45. Females had slightly lower log sNfL levels compared to males at equivalent eGFR levels. Conclusion A significant negative correlation was found between log sNfL and eGFR levels. Gender influenced the degree of this negative association. Further research is needed to validate these findings and elucidate their underlying mechanisms.
Collapse
Affiliation(s)
- Hongyan Peng
- Department of Pediatric Intensive Care Unit, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Intensive Care Medicine, Liuzhou Hospital of Affiliated Guangzhou Women and Children's Medical Center, Liuzhou, Guangxi, China
| | - Zhuoxin Liang
- Department of Intensive Care Medicine, Liuzhou Hospital of Affiliated Guangzhou Women and Children's Medical Center, Liuzhou, Guangxi, China
| | - Bolun Huang
- Department of Pediatric Intensive Care Unit, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Senxiong Zhang
- Department of Intensive Care Medicine, Liuzhou Hospital of Affiliated Guangzhou Women and Children's Medical Center, Liuzhou, Guangxi, China
| | - Yiyu Yang
- Department of Pediatric Intensive Care Unit, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Nakano Y, Mandai S, Takahashi D, Ikenouchi K, Mori Y, Ando F, Susa K, Mori T, Iimori S, Naito S, Sohara E, Fushimi K, Uchida S. Sex disparities in the risk of urgent dialysis following acute aortic dissections in Japan. iScience 2024; 27:110577. [PMID: 39211546 PMCID: PMC11357881 DOI: 10.1016/j.isci.2024.110577] [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: 05/08/2024] [Revised: 06/18/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
The global outcome of acute aortic dissection (AD) remains poor, with a high risk of the need for urgent dialysis. This study aimed to clarify the association between sex and the requirement for urgent dialysis within 30 days after admission among patients with AD. This study included 79,998 cases who were hospitalized due to AD in Japan from 2010 to 2020 using an administrative claims database. The association between the risk of urgent dialysis and sex was investigated using the Fine and Gray model. Patients were classified into two groups based on the Stanford classification: type A AD (TAAD) and type B AD (TBAD). The lower subdistribution hazard ratio (SHR) in women was observed in both groups: TAAD (SHR: 0.58, 95% confidence interval [CI]: 0.54-0.62); TBAD (SHR: 0.49, 95% CI: 0.41-0.58). Our study revealed that women had a lower risk of requiring urgent dialysis than men in TAAD and TBAD.
Collapse
Affiliation(s)
- Yuta Nakano
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8519, Japan
| | - Shintaro Mandai
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8519, Japan
| | - Daiei Takahashi
- Department of Nephrology, Musashino Red Cross Hospital, 1-26-1, Kyonann-cho, Musashino-shi, Tokyo 180-8610, Japan
| | - Ken Ikenouchi
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8519, Japan
| | - Yutaro Mori
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8519, Japan
| | - Fumiaki Ando
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8519, Japan
| | - Koichiro Susa
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8519, Japan
| | - Takayasu Mori
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8519, Japan
| | - Soichiro Iimori
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8519, Japan
| | - Shotaro Naito
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8519, Japan
| | - Eisei Sohara
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8519, Japan
| | - Kiyohide Fushimi
- Department of Health Policy and Informatics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8519, Japan
| | - Shinichi Uchida
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8519, Japan
| |
Collapse
|
8
|
Han YK, Lim HJ, Jang G, Jang SY, Park KM. Kidney ischemia/reperfusion injury causes cholangiocytes primary cilia disruption and abnormal bile secretion. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167225. [PMID: 38749218 DOI: 10.1016/j.bbadis.2024.167225] [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: 02/29/2024] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 06/17/2024]
Abstract
BACKGROUND Acute kidney injury (AKI) causes distant liver injury, to date, which causes poor outcomes of patients with AKI. Many studies have been performed to overcome AKI-associated liver injury. However, those studies have mainly focused on hepatocytes, and AKI-induced liver injury still remains a clinical problem. Here, we investigated the implication of cholangiocytes and their primary cilia which are critical in final bile secretion. Cholangiocyte, a lining cell of bile ducts, are the only liver epithelial cell containing primary cilium (a microtubule-based cell surface signal-sensing organelle). METHODS Cystathione γ-lyase (CSE, a transsulfuration enzyme) deficient and wild-type mice were subjected to kidney ischemia followed by reperfusion (KIR). Some mice were administered with N-acetyl-cysteine (NAC). RESULTS KIR damaged hepatocytes and cholagiocytes, disrupted cholangiocytes primary cilia, released the disrupted ciliary fragments into the bile, and caused abnormal bile secretion. Glutathione (GSH) and H2S levels in the livers were significantly reduced by KIR, resulting in increased the ratio oxidized GSH to total GSH, and oxidation of tissue and bile. CSE and cystathione β-synthase (CBS) expression were lowered in the liver after KIR. NAC administration increased total GSH and H2S levels in the liver and attenuated KIR-induced liver injuries. In contrast, Cse deletion caused the reduction of total GSH levels and worsened KIR-induced liver injuries, including primary cilia damage and abnormal bile secretion. CONCLUSIONS These results indicate that KIR causes cholangiocyte damage, cholangiocytes primary cilia disruption, and abnormal bile secretion through reduced antioxidative ability of the liver.
Collapse
Affiliation(s)
- Yong Kwon Han
- Department of Anatomy, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea
| | - Hui Jae Lim
- Department of Anatomy, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea; Department of Biomedical Science and BK21 Plus, The Graduate School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea
| | - GiBong Jang
- Department of Anatomy, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea; Department of Biomedical Science and BK21 Plus, The Graduate School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea
| | - Se Young Jang
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea
| | - Kwon Moo Park
- Department of Anatomy, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea; Department of Biomedical Science and BK21 Plus, The Graduate School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea.
| |
Collapse
|
9
|
Reichardt C, Brandt S, Bernhardt A, Krause A, Lindquist JA, Weinert S, Geffers R, Franz T, Kahlfuss S, Dudeck A, Mathew A, Rana R, Isermann B, Mertens PR. DNA-binding protein-A promotes kidney ischemia/reperfusion injury and participates in mitochondrial function. Kidney Int 2024; 106:241-257. [PMID: 38821446 DOI: 10.1016/j.kint.2024.05.009] [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: 12/08/2023] [Revised: 04/23/2024] [Accepted: 05/08/2024] [Indexed: 06/02/2024]
Abstract
DNA-binding protein-A (DbpA; gene: Ybx3) belongs to the cold shock protein family with known functions in cell cycling, transcription, translation, and tight junction communication. In chronic nephritis, DbpA is upregulated. However, its activities in acute injury models, such as kidney ischemia/reperfusion injury (IRI), are unclear. To study this, mice harboring Ybx3+/+, Ybx3+/- or the Ybx3-/- genotype were characterized over 24 months and following experimental kidney IRI. Mitochondrial function, number and integrity were analyzed by mitochondrial stress tests, MitoTracker staining and electron microscopy. Western Blot, immunohistochemistry and flow cytometry were performed to quantify tubular cell damage and immune cell infiltration. DbpA was found to be dispensable for kidney development and tissue homeostasis under healthy conditions. Furthermore, endogenous DbpA protein localizes within mitochondria in primary tubular epithelial cells. Genetic deletion of Ybx3 elevates the mitochondrial membrane potential, lipid uptake and metabolism, oxygen consumption rates and glycolytic activities of tubular epithelial cells. Ybx3-/- mice demonstrated protection from IRI with less immune cell infiltration, endoplasmic reticulum stress and tubular cell damage. A presumed protective mechanism was identified via upregulated antioxidant activities and reduced ferroptosis, when Ybx3 was deleted. Thus, our studies reveal DbpA acts as a mitochondrial protein with profound adverse effects on cell metabolism and highlights a protective effect against IRI when Ybx3 is genetically deleted. Hence, preemptive DbpA targeting in situations with expected IRI, such as kidney transplantation or cardiac surgery, may preserve post-procedure kidney function.
Collapse
Affiliation(s)
- Charlotte Reichardt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Sabine Brandt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Anja Bernhardt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Anna Krause
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Jonathan A Lindquist
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Sönke Weinert
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Clinic of Cardiology and Angiology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Robert Geffers
- Genome Analytics Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Tobias Franz
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Sascha Kahlfuss
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Medical Microbiology and Hospital Hygiene, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Anne Dudeck
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Akash Mathew
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, Germany
| | - Rajiv Rana
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, Germany
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, Germany
| | - Peter R Mertens
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany.
| |
Collapse
|
10
|
Zhang J, Che T, Wang L, Sun W, Zhao J, Chen J, Liu Y, Pu Q, Zhang Y, Li J, Li Z, Zhu Z, Fu Q, Wang X, Yuan J. Proteomics coupled transcriptomics reveals Slc34a1 and Slc34a3 downregulation as potential features of nephrotoxin-induced acute kidney injury. J Proteomics 2024; 302:105203. [PMID: 38782357 DOI: 10.1016/j.jprot.2024.105203] [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: 08/07/2023] [Revised: 05/11/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Acute kidney injury (AKI) stands as a prevalent and economically burdensome condition worldwide, yet its complex molecular mechanisms remain incompletely understood. To address this gap, our study employs a multifaceted approach, combining mass spectrometry and RNA sequencing technologies, to elucidate the intricate molecular landscape underlying nephrotoxin-induced AKI in mice by cisplatin- and LPS-induced. By examining the protein and RNA expression profiles, we aimed to uncover novel insights into the pathogenesis of AKI and identify potential diagnostic and therapeutic targets. Our results demonstrate significant down-regulation of Slc34a1 and Slc34a3, shedding light on their crucial roles in AKI pathology and highlighting their promise as actionable targets for diagnosis and treatment. This comprehensive analysis not only enhances our understanding of AKI pathophysiology but also offers valuable avenues for the development of targeted interventions to mitigate its clinical impact. SIGNIFICANCE: Nephrotoxicity acute kidney injury (AKI) is a common clinical condition whose pathogenesis is the process by which some drugs, chemicals or other factors cause damage to the kidneys, resulting in impaired kidney function. Although it has been proved that different nephrotoxic substances can affect the kidney through different pathways, whether they have a commonality has not been registered. Here, we combined transcriptomics and proteomics to study the molecular mechanism of LPS and cisplatin-induced nephrotoxic acute kidney injury finding that the down-regulation of Slc34a1 and Slc34a3 may be a critical link in nephrotoxic acute kidney injury, which can be used as a marker for its early diagnosis.
Collapse
Affiliation(s)
- Junying Zhang
- Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China; College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Tiantian Che
- Chongqing Nanan District Center for Diseases Control and Prevention, Chongqing 401336, China
| | - Liting Wang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Wei Sun
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Jing Zhao
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Jiajia Chen
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Yang Liu
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Qi Pu
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Yu Zhang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Jiani Li
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Zhangfu Li
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China; Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, Shenzhen 518036, China
| | - Zhaojing Zhu
- Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China.
| | - Qihuan Fu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China.
| | - Xiaoyang Wang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China..
| | - Jiangbei Yuan
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China; Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, Shenzhen 518036, China.; Center for General Practice Medicine, Department of Infectious Diseases, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Department of Infectious Diseases, Affiliated Banan Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
11
|
Noonan ML, Muto Y, Yoshimura Y, Leckie-Harre A, Wu H, Kalinichenko VV, Humphreys BD, Chang-Panesso M. Injury-induced Foxm1 expression in the mouse kidney drives epithelial proliferation by a cyclin F-dependent mechanism. JCI Insight 2024; 9:e175416. [PMID: 38916959 PMCID: PMC11383596 DOI: 10.1172/jci.insight.175416] [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: 09/05/2023] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
Acute kidney injury (AKI) strongly upregulates the transcription factor Foxm1 in the proximal tubule in vivo, and Foxm1 drives epithelial proliferation in vitro. Here, we report that deletion of Foxm1 either with a nephron-specific Cre driver or by inducible global deletion reduced proximal tubule proliferation after ischemic injury in vivo. Foxm1 deletion led to increased AKI to chronic kidney disease transition, with enhanced fibrosis and ongoing tubule injury 6 weeks after injury. We report ERK mediated FOXM1 induction downstream of the EGFR in primary proximal tubule cells. We defined FOXM1 genomic binding sites by cleavage under targets and release using nuclease (CUT&RUN) and compared the genes located near FOXM1 binding sites with genes downregulated in primary proximal tubule cells after FOXM1 knockdown. The aligned data sets revealed the cell cycle regulator cyclin F (CCNF) as a putative FOXM1 target. We identified 2 cis regulatory elements that bound FOXM1 and regulated CCNF expression, demonstrating that Ccnf is strongly induced after kidney injury and that Foxm1 deletion abrogates Ccnf expression in vivo and in vitro. Knockdown of CCNF also reduced proximal tubule proliferation in vitro. These studies identify an ERK/FOXM1/CCNF signaling pathway that regulates injury-induced proximal tubule cell proliferation.
Collapse
Affiliation(s)
- Megan L Noonan
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Yoshiharu Muto
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Yasuhiro Yoshimura
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Aidan Leckie-Harre
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Haojia Wu
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Vladimir V Kalinichenko
- Phoenix Children's Health Research Institute, Department of Child Health, University of Arizona College of Medicine, Phoenix, Arizona, USA
- Division of Neonatology, Phoenix Children's Hospital, Phoenix, Arizona, USA
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Monica Chang-Panesso
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
12
|
Körner A, Sailer B, Sari-Yavuz S, Haeberle HA, Mirakaj V, Bernard A, Rosenberger P, Koeppen M. Explainable Boosting Machine approach identifies risk factors for acute renal failure. Intensive Care Med Exp 2024; 12:55. [PMID: 38874694 PMCID: PMC11178719 DOI: 10.1186/s40635-024-00639-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/02/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Risk stratification and outcome prediction are crucial for intensive care resource planning. In addressing the large data sets of intensive care unit (ICU) patients, we employed the Explainable Boosting Machine (EBM), a novel machine learning model, to identify determinants of acute kidney injury (AKI) in these patients. AKI significantly impacts outcomes in the critically ill. METHODS An analysis of 3572 ICU patients was conducted. Variables such as average central venous pressure (CVP), mean arterial pressure (MAP), age, gender, and comorbidities were examined. This analysis combined traditional statistical methods with the EBM to gain a detailed understanding of AKI risk factors. RESULTS Our analysis revealed chronic kidney disease, heart failure, arrhythmias, liver disease, and anemia as significant comorbidities influencing AKI risk, with liver disease and anemia being particularly impactful. Surgical factors were also key; lower GI surgery heightened AKI risk, while neurosurgery was associated with a reduced risk. EBM identified four crucial variables affecting AKI prediction: anemia, liver disease, and average CVP increased AKI risk, whereas neurosurgery decreased it. Age was a progressive risk factor, with risk escalating after the age of 50 years. Hemodynamic instability, marked by a MAP below 65 mmHg, was strongly linked to AKI, showcasing a threshold effect at 60 mmHg. Intriguingly, average CVP was a significant predictor, with a critical threshold at 10.7 mmHg. CONCLUSION Using an Explainable Boosting Machine enhance the precision in AKI risk factors in ICU patients, providing a more nuanced understanding of known AKI risks. This approach allows for refined predictive modeling of AKI, effectively overcoming the limitations of traditional statistical models.
Collapse
Affiliation(s)
- Andreas Körner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Benjamin Sailer
- Medical Data Integration Center, University Hospital Tübingen, Tübingen, Germany
| | - Sibel Sari-Yavuz
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Helene A Haeberle
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Valbona Mirakaj
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Alice Bernard
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Peter Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Michael Koeppen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany.
| |
Collapse
|
13
|
Demirjian S, Huml A, Bakaeen F, Poggio E, Geube M, Shaw A, Gillinov AM, Gadegbeku CA. Sex bias in prediction and diagnosis of cardiac surgery associated acute kidney injury. BMC Nephrol 2024; 25:180. [PMID: 38778259 PMCID: PMC11112848 DOI: 10.1186/s12882-024-03614-x] [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: 11/24/2023] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Female sex has been recognized as a risk factor for cardiac surgery associated acute kidney injury (CS-AKI). The current study sought to evaluate whether female sex is a risk factor for CS-AKI, or modifies the association of peri-operative change in serum creatinine with CS-AKI. METHODS Observational study of adult patients undergoing cardiac surgery between 2000 and 2019 in a single U.S. center. The main variable of interest was registered patient sex, identified from electronic medical records. The main outcome was CS-AKI within 2 weeks of surgery. RESULTS Of 58526 patients, 19353 (33%) were female; 12934 (22%) incurred AKI based on ≥ 0.3 mg/dL or ≥ 50% rise in serum creatinine (any AKI), 3320 (5.7%) had moderate to severe AKI, and 1018 (1.7%) required dialysis within 2 weeks of surgery. Female sex was associated with higher risk for AKI in models that were based on preoperative serum creatinine (OR, 1.35; 95% CI, 1.29-1.42), and lower risk with the use of estimated glomerular filtration, (OR, 0.90; 95% CI, 0.86-0.95). The risk for moderate to severe CS-AKI for a given immediate peri-operative change in serum creatinine was higher in female compared to male patients (p < .0001 and p < .0001 for non-linearity), and the association was modified by pre-operative kidney function (p < .0001 for interaction). CONCLUSIONS The association of patient sex with CS-AKI and its direction was dependent on the operational definition of pre-operative kidney function, and differential outcome misclassification due to AKI defined by absolute change in serum creatinine.
Collapse
Affiliation(s)
- Sevag Demirjian
- Department of Kidney Medicine, Cleveland Clinic, 9500 Euclid Avenue, Q7, Cleveland, OH, 44195, USA.
| | - Anne Huml
- Department of Kidney Medicine, Cleveland Clinic, 9500 Euclid Avenue, Q7, Cleveland, OH, 44195, USA
| | - Faisal Bakaeen
- Department of Thoracic and Cardiovascular Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Emilio Poggio
- Department of Kidney Medicine, Cleveland Clinic, 9500 Euclid Avenue, Q7, Cleveland, OH, 44195, USA
| | - Mariya Geube
- Department of Cardiothoracic Anesthesiology, Cleveland Clinic, Cleveland, OH, USA
| | - Andrew Shaw
- Department of Intensive Care and Resuscitation, Cleveland Clinic, Cleveland, OH, USA
| | - A Marc Gillinov
- Department of Thoracic and Cardiovascular Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Crystal A Gadegbeku
- Department of Kidney Medicine, Cleveland Clinic, 9500 Euclid Avenue, Q7, Cleveland, OH, 44195, USA
| |
Collapse
|
14
|
Zhang Y, Liu M, Zhu Z, Chen H. Proton pump inhibitors use is associated with a higher prevalence of kidney stones: NHANES 2007-2018. BMC Public Health 2024; 24:1215. [PMID: 38698372 PMCID: PMC11067170 DOI: 10.1186/s12889-024-18710-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 04/24/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Proton pump inhibitors (PPIs) are widely used throughout the world as an effective gastrointestinal drug. Nevertheless, according to the existing literature, PPIs can reduce the excretion of magnesium, calcium and other components in urine, which may promote the formation of kidney stones. We used the National Health and Nutrition Examination Survey (NHANES) database to further investigate the association between the use of PPIs and the prevalence of kidney stones. METHODS We performed a cross-sectional analysis using data from 2007 to 2018 NHANES. PPIs use information of 29,910 participants was obtained by using prescription medications in the preceding month, and kidney stones were presented by a standard questionnaire. Multiple regression analysis and stratified analysis were used to estimate the association between PPIs use and kidney stones after an adjustment for potential confounders. RESULTS The multiple logistic regression indicated that the PPIs exposure group (P1) had a significantly higher risk of nephrolithiasis than the PPIs non-exposure group (P0) in Model 3 (OR 1.24, 95% CI 1.10-1.39, P < 0.001). The stratified analyses indicated there were significant statistical differences between PPIs use and kidney stones among females (OR 1.36, 95% CI 1.15-1.62, P < 0.001), non-Hispanic whites (OR 1.27, 95% CI 1.09-1.48, P = 0.002), individuals with an education level than 11th grade (OR 1.41, 95% CI 1.13-1.76, P = 0.002) and individuals with an annual family income of $0 to $19,999 (OR 1.32, 95% CI 1.06-1.65, P = 0.014) and $20,000 to $44,999 (OR 1.25, 95% CI 1.02-1.54, P = 0.033) in Model 3. CONCLUSIONS Our study revealed that PPIs use is associated with a higher prevalence of kidney stones for the US population, primarily among women, non-Hispanic whites, individuals with low education levels and individuals with low household income levels. Further studies are required to confirm our findings.
Collapse
Affiliation(s)
- Youjie Zhang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Minghui Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zewu Zhu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Hequn Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
15
|
Fossum M, Kaefer M, Bagli D, Harper L, Herbst KW, Nelson CP, Braga LH, Nieuwhof-Leppink A, Kalfa N, Garriboli M, Cascio S. The importance of animal specificity in animal experimentation, part I: Anatomy in relation to pediatric urology. J Pediatr Urol 2024; 20:312-314. [PMID: 38199905 DOI: 10.1016/j.jpurol.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 12/11/2023] [Indexed: 01/12/2024]
Abstract
Why and when is animal experimentation relevant? The answer to this question depends on the research question. In this short educational article we aim to raise awareness of the importance of formulating a very specific research question before choosing an animal species. An awareness of anatomical and physiological differences vis-a-vis similarities between species, will increase the potential for obtaining data that is relevant for translation to human conditions.
Collapse
Affiliation(s)
- Magdalena Fossum
- Department of Pediatric Surgery, Center of Cancer and Organ Diseases, Rigshospitalet, Copenhagen University, Copenhagen, Denmark; Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.
| | - Martin Kaefer
- Riley Hospital for Children, Indiana University, Indianapolis, IN, United States
| | - Darius Bagli
- Division of Urology, Departments of Surgery and Physiology, University of Toronto, Developmental and Stem Cell Biology, The Hospital for Sick Children and Research Institute, Toronto, Ontario, Canada
| | - Luke Harper
- Service de Chirurgie Pédiatrique, Hôpital Pellegrin-Enfants, CHU de Bordeaux, France
| | - Katherine W Herbst
- Division of Urology, Research Institute, Connecticut Children's Medical Center, Hartford, CT, United States
| | - Caleb P Nelson
- Department of Urology, Boston Children's Hospital and Harvard Medical School, MA, United States
| | - Luis H Braga
- Department of Surgery, Division of Urology, McMaster University, McMaster Children's Hospital, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| | - Anka Nieuwhof-Leppink
- Department Urotherapy, Wilhelmina Children's Hospital, Part of University Medical Center Utrecht, Netherlands
| | - Nicolas Kalfa
- Department of Pediatric Surgery and Urology, Lapeyronie Hospital, CHU Montpellier, France; UMR1302 Institute Desbrest of Epidemiology and Public Health, INSERM, University Montpellier, Montpellier, France
| | - Massimo Garriboli
- Department of Paediatric Urology, Evelina London Children's Hospital, Westminster Bridge Road, London, SE1 7EH, United Kingdom
| | - Salvatore Cascio
- Department of Pediatric Surgery, School of Medicine, University College Dublin and Children's Health Ireland at Temple Street, Dublin, Ireland
| |
Collapse
|
16
|
Kan H, Zhao M, Wang W, Sun B. Understanding Propofol's Protective Mechanism in Tubular Epithelial Cells: Mitigating Pyroptosis via the miR-143-3p/ATPase Na + /K + Transporting Subunit Alpha 2 Pathway in Renal Ischemia-Reperfusion. Mol Biotechnol 2024:10.1007/s12033-024-01116-7. [PMID: 38498283 DOI: 10.1007/s12033-024-01116-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 02/02/2024] [Indexed: 03/20/2024]
Abstract
Propofol (Pro), a prevalent intravenous anesthetic, has recently been recognized for its potential in mitigating ischemia-reperfusion (I/R) injuries. Despite a plethora of evidence suggesting the beneficial effects of low-dose Pro in renal I/R injury (RI/R), its role in modulating pyroptosis in renal tubular epithelial cells consequent to RI/R has not been thoroughly elucidated. In our investigation, we explored the therapeutic potential of Pro against pyroptosis in renal tubular epithelial cells under the duress of RI/R, employing both in vivo and in vitro models, while deciphering the intricate molecular pathways involved. Our results demonstrate an elevation in the expression of miR-143-3p, contrasted by a diminution in ATPase Na + /K + Transporting Subunit Alpha 2 (ATP1A2) under RI/R conditions. Pro effectively mitigates apoptosis in renal tubular epithelial cells induced by RI/R, principally characterized by the inhibition of pro-inflammatory cytokines interleukin (IL-)-1β and IL-18, enhancement of cellular viability, reduction in the ratio of pyroptotic cells, and suppression of nucleotide-binding domain and leucine-rich repeat-related family, pyrin domain containing 3 inflammasome activation along with the expression of cleaved caspase-1, and gasdermin D. Both knockdown and overexpression studies of miR-143-3p revealed its pivotal role in modulating RI/R-induced tubular cell pyroptosis. Notably, Pro's capacity to inhibit pyroptosis in renal tubular epithelial cells was found to be reversible following ATP1A2 knockdown. Furthermore, our study unveils miR-143-3p as a targeted regulator of ATP1A2 expression. From a mechanistic standpoint, Pro's therapeutic efficacy is attributed to its regulatory influence on miR-143-3p and ATP1A2 expression levels. In conclusion, our findings pioneer the understanding that Pro can significantly ameliorate pyroptosis in renal tubular epithelial cells in the context of RI/R, predominantly through the modulation of the miR-143-3p/ATP1A2 axis. This novel insight furnishes robust empirical support for the development of targeted therapeutics and clinical strategies in addressing RI/R.
Collapse
Affiliation(s)
- Hongjun Kan
- Department of Anesthesiology, Shandong Second Provincial General Hospital, No.4 Duanxing West Road, Huaiyin District, Jinan City, 250022, Shandong Province, China
| | - Miaomiao Zhao
- Department of Paediatrics, Pediatrics, Zaozhuang Traditional Chinese Medicine Hospital, Zaozhuang City, 277100, Shandong Province, China
| | - Wei Wang
- Anesthesia and Perioperative Medicine, Zaozhuang Municipal Hospital, Zaozhuang City, 277000, Shandong Province, China
| | - Baozhong Sun
- Department of Anesthesiology, Shandong Second Provincial General Hospital, No.4 Duanxing West Road, Huaiyin District, Jinan City, 250022, Shandong Province, China.
| |
Collapse
|
17
|
Demirjian S, Huml A, Bakaeen F, Poggio E, Geube M, Shaw A, Gillinov AM, Gadegbeku CA. Sex Bias in Prediction and Diagnosis of Cardiac Surgery Associated Acute Kidney Injury. RESEARCH SQUARE 2024:rs.3.rs-3660617. [PMID: 38558997 PMCID: PMC10980107 DOI: 10.21203/rs.3.rs-3660617/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Background Female sex has been recognized as a risk factor for cardiac surgery associated acute kidney injury (CS-AKI). The current study sought to evaluate whether female sex is a risk factor for CS-AKI, or modifies the association of peri-operative change in serum creatinine with CS-AKI. Methods Observational study of adult patients undergoing cardiac surgery between 2000 and 2019 in a single U.S. center. The main variable of interest was registered patient sex, identified from electronic medical records. The main outcome was CS-AKI within 2 weeks of surgery. Results Of 58526 patients, 19353 (33%) were female; 12934 (22%) incurred AKI based on ≥ 0.3 mg/dL or ≥ 50% rise in serum creatinine (any AKI), 3320 (5.7%) had moderate to severe AKI, and 1018 (1.7%) required dialysis within 2 weeks of surgery. Female sex was associated with higher risk for AKI in models that were based on preoperative serum creatinine (OR, 1.35; 95% CI, 1.29-1.42), and lower risk with the use of estimated glomerular filtration, (OR, 0.90; 95% CI, 0.86-0.95). The risk for moderate to severe CS-AKI for a given immediate peri-operative change in serum creatinine was higher in female compared to male patients (p < .0001 and p < .0001 for non-linearity), and the association was modified by pre-operative kidney function (p < .0001 for interaction). Conclusions The association of patient sex with CS-AKI and its direction was dependent on the operational definition of pre-operative kidney function, and differential outcome misclassification due to AKI defined by absolute change in serum creatinine.
Collapse
|
18
|
Rodrigues FG, Van Der Plas WY, Sotomayor CG, Van Der Vaart A, Kremer D, Pol RA, Kruijff S, Heilberg IP, Bakker SJL, De Borst MH. Pre-Transplant Hyperparathyroidism and Graft or Patient Outcomes After Kidney Transplantation. Transpl Int 2024; 37:11916. [PMID: 38384325 PMCID: PMC10880800 DOI: 10.3389/ti.2024.11916] [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: 08/12/2023] [Accepted: 01/23/2024] [Indexed: 02/23/2024]
Abstract
The impact of pre-transplant parathyroid hormone (PTH) levels on early or long-term kidney function after kidney transplantation is subject of debate. We assessed whether severe hyperparathyroidism is associated with delayed graft function (DGF), death-censored graft failure (DCGF), or all-cause mortality. In this single-center cohort study, we studied the relationship between PTH and other parameters related to bone and mineral metabolism, including serum alkaline phosphatase (ALP) at time of transplantation with the subsequent risk of DGF, DCGF and all-cause mortality using multivariable logistic and Cox regression analyses. In 1,576 kidney transplant recipients (51.6 ± 14.0 years, 57.3% male), severe hyperparathyroidism characterized by pre-transplant PTH ≥771 pg/mL (>9 times the upper limit) was present in 121 patients. During 5.2 [0.2-30.0] years follow-up, 278 (15.7%) patients developed DGF, 150 (9.9%) DCGF and 432 (28.6%) died. A higher pre-transplant PTH was not associated with DGF (HR 1.06 [0.90-1.25]), DCGF (HR 0.98 [0.87-1.13]), or all-cause mortality (HR 1.02 [0.93-1.11]). Results were consistent in sensitivity analyses. The same applied to other parameters related to bone and mineral metabolism, including ALP. Severe pre-transplant hyperparathyroidism was not associated with an increased risk of DGF, DCGF or all-cause mortality, not supporting the need of correction before kidney transplantation to improve graft or patient survival.
Collapse
Affiliation(s)
- Fernanda Guedes Rodrigues
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Nutrition Post Graduation Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Willemijn Y. Van Der Plas
- Department of Surgery, Division of Surgical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Camilo German Sotomayor
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Amarens Van Der Vaart
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Daan Kremer
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Robert A. Pol
- Department of Surgery, Division of Surgical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Schelto Kruijff
- Department of Surgery, Division of Surgical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Surgery, Martini Hospital Groningen, Groningen, Netherlands
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ita Pfeferman Heilberg
- Nutrition Post Graduation Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- Nephrology Division, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Stephan J. L. Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Martin H. De Borst
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
19
|
Beamish JA, Telang AC, McElliott MC, Al-Suraimi A, Chowdhury M, Ference-Salo JT, Otto EA, Menon R, Soofi A, Weinberg JM, Patel SR, Dressler GR. Pax protein depletion in proximal tubules triggers conserved mechanisms of resistance to acute ischemic kidney injury preventing transition to chronic kidney disease. Kidney Int 2024; 105:312-327. [PMID: 37977366 PMCID: PMC10958455 DOI: 10.1016/j.kint.2023.10.022] [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: 06/08/2023] [Revised: 09/18/2023] [Accepted: 10/25/2023] [Indexed: 11/19/2023]
Abstract
Acute kidney injury (AKI) is a common condition that lacks effective treatments. In part, this shortcoming is due to an incomplete understanding of the genetic mechanisms that control pathogenesis and recovery. Identifying the molecular and genetic regulators unique to nephron segments that dictate vulnerability to injury and regenerative potential could lead to new therapeutic targets to treat ischemic kidney injury. Pax2 and Pax8 are homologous transcription factors with overlapping functions that are critical for kidney development and are re-activated in AKI. Here, we examined the role of Pax2 and Pax8 in recovery from ischemic AKI and found them upregulated after severe AKI and correlated with chronic injury. Surprisingly, proximal-tubule-selective deletion of Pax2 and Pax8 resulted in a less severe chronic injury phenotype. This effect was mediated by protection against the acute insult, similar to pre-conditioning. Prior to injury, Pax2 and Pax8 mutant mice develop a unique subpopulation of proximal tubule cells in the S3 segment that displayed features usually seen only in acute or chronic injury. The expression signature of these cells was strongly enriched with genes associated with other mechanisms of protection against ischemic AKI including caloric restriction, hypoxic pre-conditioning, and female sex. Thus, our results identified a novel role for Pax2 and Pax8 in mature proximal tubules that regulates critical genes and pathways involved in both the injury response and protection from ischemic AKI.
Collapse
Affiliation(s)
- Jeffrey A Beamish
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| | - Asha C Telang
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Madison C McElliott
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Anas Al-Suraimi
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Mahboob Chowdhury
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jenna T Ference-Salo
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Edgar A Otto
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Rajasree Menon
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Abdul Soofi
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Joel M Weinberg
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Sanjeevkumar R Patel
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Gregory R Dressler
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
20
|
Porter A, Vorndran HE, Marciszyn A, Mutchler SM, Subramanya AR, Kleyman TR, Hendershot LM, Brodsky JL, Buck TM. Excess dietary sodium partially restores salt and water homeostasis caused by loss of the endoplasmic reticulum molecular chaperone, GRP170, in the mouse nephron. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.13.575426. [PMID: 38260467 PMCID: PMC10802592 DOI: 10.1101/2024.01.13.575426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The maintenance of fluid and electrolyte homeostasis by the kidney requires proper folding and trafficking of ion channels and transporters in kidney epithelia. Each of these processes requires a specific subset of a diverse class of proteins termed molecular chaperones. One such chaperone is GRP170, which is an Hsp70-like, endoplasmic reticulum (ER)-localized chaperone that plays roles in protein quality control and protein folding in the ER. We previously determined that loss of GRP170 in the mouse nephron leads to hypovolemia, electrolyte imbalance, and rapid weight loss. In addition, GRP170-deficient mice develop an AKI-like phenotype, typified by tubular injury, elevation of clinical kidney injury markers, and induction of the unfolded protein response (UPR). By using an inducible GRP170 knockout cellular model, we confirmed that GRP170 depletion induces the UPR, triggers an apoptotic response, and disrupts protein homeostasis. Based on these data, we hypothesized that UPR induction underlies hyponatremia and volume depletion in rodents, but that these and other phenotypes might be rectified by supplementation with high salt. To test this hypothesis, control and GRP170 tubule-specific knockout mice were provided with a diet containing 8% sodium chloride. We discovered that sodium supplementation improved electrolyte imbalance and reduced clinical kidney injury markers, but was unable to restore weight or tubule integrity. These results are consistent with UPR induction contributing to the kidney injury phenotype in the nephron-specific GR170 knockout model, and that the role of GRP170 in kidney epithelia is essential to both maintain electrolyte balance and cellular protein homeostasis.
Collapse
Affiliation(s)
- Aidan Porter
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA
- Division of Pediatric Nephrology, University of Pittsburgh, Pittsburgh, PA
| | - Hannah E. Vorndran
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Allison Marciszyn
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Stephanie M. Mutchler
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Arohan R. Subramanya
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA
| | - Thomas R. Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA
| | - Linda M. Hendershot
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN 30105
| | - Jeffrey L. Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Teresa M. Buck
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
21
|
Curtis LM. Sex and Gender Differences in AKI. KIDNEY360 2024; 5:160-167. [PMID: 37990360 PMCID: PMC10833607 DOI: 10.34067/kid.0000000000000321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 11/14/2023] [Indexed: 11/23/2023]
Abstract
Sex differences in AKI continue to be identified. Generally, women are protected from AKI when compared to men. Much of the protection exhibited in women is diminished after menopause. These sex and age effects have also been noted in animal models of AKI. Gonadal hormones, as modifiers of incidence, severity, and progression of AKI, have been offered as likely contributors to this sex and age effect. In animal models of AKI, estrogen and testosterone seem to modulate susceptibility. Questions remain however regarding cellular and molecular changes that are initiated by modulation of these hormones because both estrogen and testosterone have effects across cell types that play a role in AKI. Although findings have largely been informed by studies in males, molecular pathways that are involved in the initiation and progression of AKI may be modulated by gonadal hormones. Compounding the hormone-receptor effects are developmental effects of sex chromosomal complement and epigenetic influences that may confer sex-based baseline differences in gene and protein expression, and gene dosage effects of X inactivation and escape on molecular pathways. Elucidation of sex-based protection may afford a more complete view of AKI and potential therapeutic interventions. Furthermore, the effect on susceptibility to AKI in transgender patients, who receive life-altering and essential gender-affirming hormone therapy, requires greater attention. In this review, several potential contributors to the sex differences observed in humans and animal models are discussed.
Collapse
Affiliation(s)
- Lisa M Curtis
- Division of Nephrology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
22
|
Hou Y, Lin S, Xia J, Zhang Y, Yin Y, Huang M, Xu Y, Yang W, Zhu Y. Alleviation of ischemia-reperfusion induced renal injury by chemically modified SOD2 mRNA delivered via lipid nanoparticles. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102067. [PMID: 38028193 PMCID: PMC10652142 DOI: 10.1016/j.omtn.2023.102067] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023]
Abstract
Ischemia-reperfusion injury (IRI) is a major cause of acute kidney injury, which is a serious clinical condition with no effective pharmacological treatment. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) significantly alleviate kidney IRI; however, the underlying mechanisms and key molecules conferring renoprotection remain elusive. In this study, we characterized the protein composition of MSC-EVs using a proteomics approach and found that mitochondrial protein superoxide dismutase 2 (SOD2) was enriched in MSC-EVs. Using lipid nanoparticles (LNP), we successfully delivered chemically modified SOD2 mRNA into kidney cells and mice with kidney IRI. We demonstrated that SOD2 mRNA-LNP treatment decreased cellular reactive oxygen species (ROS) in cultured cells and ameliorated renal damage in IRI mice, as indicated by reduced levels of serum creatinine and restored tissue integrity compared with the control mRNA-LNP-injected group. Thus, the modulation of mitochondrial ROS levels through SOD2 upregulation by SOD2 mRNA-LNP delivery could be a novel therapeutic method for ischemia-reperfusion-induced acute kidney injury.
Collapse
Affiliation(s)
- 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 200025, P.R. China
| | - Sihao Lin
- Department of Urology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201803, P.R. China
| | - Jia Xia
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Yu Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Yanan Yin
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. 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 200025, P.R. 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 200025, P.R. China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
- RNAcure Biopharma, Shanghai, P.R. China
- Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai, P.R. 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 200025, P.R. China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, P.R. China
| | - Yingjian Zhu
- Department of Urology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201803, P.R. China
| |
Collapse
|
23
|
Wiener SV. Effects of the environment on the evolution of the vertebrate urinary tract. Nat Rev Urol 2023; 20:719-738. [PMID: 37443264 DOI: 10.1038/s41585-023-00794-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2023] [Indexed: 07/15/2023]
Abstract
Evolution of the vertebrate urinary system occurs in response to numerous selective pressures, which have been incompletely characterized. Developing research into urinary evolution led to the occurrence of clinical applications and insights in paediatric urology, reproductive medicine, urolithiasis and other domains. Each nephron segment and urinary organ has functions that can be contextualized within an evolutionary framework. For example, the structure and function of the glomerulus and proximal tubule are highly conserved, enabling blood cells and proteins to be retained, and facilitating the elimination of oceanic Ca+ and Mg+. Urea emerged as an osmotic mediator during evolution, as cells of large organisms required increased precision in the internal regulation of salinity and solutes. As the first vertebrates moved from water to land, acid-base regulation was shifted from gills to skin and kidneys in amphibians. In reptiles and birds, solute regulation no longer occurred through the skin but through nasal salt glands and post-renally, within the cloaca and the rectum. In placental mammals, nasal salt glands are absent and the rectum and urinary tracts became separate, which limited post-renal urine concentration and led to the necessity of a kidney capable of high urine concentration. Considering the evolutionary and environmental selective pressures that have contributed to renal evolution can help to gain an increased understanding of renal physiology.
Collapse
Affiliation(s)
- Scott V Wiener
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
24
|
Dahiya A, Pannu N, Soranno DE. Sex as a biological variable in acute kidney injury. Curr Opin Crit Care 2023; 29:529-533. [PMID: 37861211 DOI: 10.1097/mcc.0000000000001091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide an overview of the preclinical and clinical studies investigating sex as a biological variable, as well as the impact of gender, on the development of and progression of acute kidney injury (AKI). RECENT FINDINGS Despite a matched degree of ischemia-reperfusion AKI based on measured glomerular filtration rates, male and female mice demonstrated important sex biases in cardiorenal outcomes (1). Although the 2012 Kidney Disease Improving Global Outcomes (KDIGO) Clinical Practice Guideline for AKI reported that female sex is associated with increased rates of hospital acquired AKI, subsequent meta-analyses do not show increased risk of AKI in women. Recent large scale, multicenter epidemiologic studies suggest males have higher rates of hospital acquired AKI. However, women have been consistently shown to have worse renal outcomes after AKI. There may be also be gender-based differences in presentation to care and management. SUMMARY Sex is an important biological variable in animal models of acute kidney injury. The impact of sex on AKI likely varies based on the etiology of AKI. Preclinical studies demonstrate the nuances of sex chromosomes, sex hormones and epigenetic factors on AKI, however these have not been well studied in humans. Gender may also impact processes of care, treatment and clinical outcomes related to AKI. The scientific rigor and reproducibility of translational studies benefit from the consideration of sex and gender.
Collapse
Affiliation(s)
- Anita Dahiya
- Division of Nephrology, University of Alberta, Department of Medicine, Alberta, Canada
| | - Neesh Pannu
- Division of Nephrology, University of Alberta, Department of Medicine, Alberta, Canada
| | - Danielle E Soranno
- Department of Pediatrics, Pediatric Nephrology, Indiana University School of Medicine, Indianapolis
- Department of Bioengineering, Purdue University, Weldon School of Engineering, West Lafayette, Indiana, USA
| |
Collapse
|
25
|
Packialakshmi B, Burmeister DM, Anderson JA, Morgan J, Cannon G, Kiang JG, Feng Y, Lee S, Stewart IJ, Zhou X. A clinically-relevant mouse model that displays hemorrhage exacerbates tourniquet-induced acute kidney injury. Front Physiol 2023; 14:1240352. [PMID: 38028812 PMCID: PMC10663317 DOI: 10.3389/fphys.2023.1240352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Hemorrhage is a leading cause of death in trauma. Tourniquets are effective at controlling extremity hemorrhage and have saved lives. However, tourniquets can cause ischemia reperfusion injury of limbs, leading to systemic inflammation and other adverse effects, which results in secondary damage to the kidney, lung, and liver. A clinically relevant animal model is critical to understanding the pathophysiology of this process and developing therapeutic interventions. Despite the importance of animal models, tourniquet-induced lower limb ischemia/reperfusion (TILLIR) models to date lack a hemorrhage component. We sought to develop a new TILLIR model that included hemorrhage and analyze the subsequent impact on kidney, lung and liver injuries. Four groups of mice were examined: group 1) control, group 2) hemorrhage, group 3) tourniquet application, and group 4) hemorrhage and tourniquet application. The hemorrhagic injury consisted of the removal of 15% of blood volume through the submandibular vein. The tourniquet injury consisted of orthodontic rubber bands applied to the inguinal area bilaterally for 80 min. Mice were then placed in metabolic cages individually for 22 h to collect urine. Hemorrhage alone did not significantly affect transcutaneous glomerular filtration rate (tGFR), blood urea nitrogen (BUN) or urinary kidney injury molecule-1 (KIM-1) levels. Without hemorrhage, TILLIR decreased tGFR by 46%, increased BUN by 162%, and increased KIM-1 by 27% (p < 0.05 for all). With hemorrhage, TILLIR decreased the tGFR by 72%, increased BUN by 395%, and increased urinary KIM-1 by 37% (p < 0.05 for all). These differences were statistically significant (p < 0.05). While hemorrhage had no significant effect on TILLIR-induced renal tubular degeneration and necrosis, it significantly increased TILLIR-induced lung total injury scores and congestion, and fatty liver. In conclusion, hemorrhage exacerbates TILLIR-induced acute kidney injury and structural damage in the lung and liver.
Collapse
Affiliation(s)
- Balamurugan Packialakshmi
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - David M. Burmeister
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Joseph A. Anderson
- Department of Laboratory Animal Resources, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Judah Morgan
- Internal Medicine Residency Program at Madigan Army Medical Center, Joint Base Lewis-McChord, Tacoma, WA, United States
| | - Georgetta Cannon
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Juliann G. Kiang
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Yuanyi Feng
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Sang Lee
- Department of Laboratory Animal Resources, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Ian J. Stewart
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Xiaoming Zhou
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
26
|
Nørgård MØ, Svenningsen P. Acute Kidney Injury by Ischemia/Reperfusion and Extracellular Vesicles. Int J Mol Sci 2023; 24:15312. [PMID: 37894994 PMCID: PMC10607034 DOI: 10.3390/ijms242015312] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Acute kidney injury (AKI) is often caused by ischemia-reperfusion injury (IRI). IRI significantly affects kidney metabolism, which elicits pro-inflammatory responses and kidney injury. The ischemia/reperfusion of the kidney is associated with transient high mitochondrial-derived reactive oxygen species (ROS) production rates. Excessive mitochondrial-derived ROS damages cellular components and, together with other pathogenic mechanisms, elicits a range of acute injury mechanisms that impair kidney function. Mitochondrial-derived ROS production also stimulates epithelial cell secretion of extracellular vesicles (EVs) containing RNAs, lipids, and proteins, suggesting that EVs are involved in AKI pathogenesis. This literature review focuses on how EV secretion is stimulated during ischemia/reperfusion and how cell-specific EVs and their molecular cargo may modify the IRI process. Moreover, critical pitfalls in the analysis of kidney epithelial-derived EVs are described. In particular, we will focus on how the release of kidney epithelial EVs is affected during tissue analyses and how this may confound data on cell-to-cell signaling. By increasing awareness of methodological pitfalls in renal EV research, the risk of false negatives can be mitigated. This will improve future EV data interpretation regarding EVs contribution to AKI pathogenesis and their potential as biomarkers or treatments for AKI.
Collapse
Affiliation(s)
| | - Per Svenningsen
- Department of Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark;
| |
Collapse
|
27
|
Beamish JA, Telang AC, McElliott MC, Al-Suraimi A, Chowdhury M, Ference-Salo JT, Otto EA, Menon R, Soofi A, Weinberg JM, Patel SR, Dressler GR. Pax Protein Depletion in Proximal Tubules Triggers Conserved Mechanisms of Resistance to Acute Ischemic Kidney Injury and Prevents Transition to Chronic Kidney Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.03.559511. [PMID: 37873377 PMCID: PMC10592940 DOI: 10.1101/2023.10.03.559511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Acute kidney injury (AKI) is a common condition that lacks effective treatments. In part this shortcoming is due to an incomplete understanding of the genetic mechanisms that control pathogenesis and recovery. Pax2 and Pax8 are homologous transcription factors with overlapping functions that are critical for kidney development and are re-activated in AKI. In this report, we examined the role of Pax2 and Pax8 in recovery from ischemic AKI. We found that Pax2 and Pax8 are upregulated after severe AKI and correlate with chronic injury. Surprisingly, we then discovered that proximal-tubule-selective deletion of Pax2 and Pax8 resulted in a less severe chronic injury phenotype. This effect was mediated by protection against the acute insult, similar to preconditioning. Prior to injury, Pax2 and Pax8 mutant mice develop a unique subpopulation of S3 proximal tubule cells that display features usually seen only in acute or chronic injury. The expression signature of these cells was strongly enriched with genes associated with other mechanisms of protection against ischemic AKI including caloric restriction, hypoxic preconditioning, and female sex. Taken together, our results identify a novel role for Pax2 and Pax8 in mature proximal tubules that regulates critical genes and pathways involved in both injury response and protection from ischemic AKI. TRANSLATIONAL STATEMENT Identifying the molecular and genetic regulators unique to the nephron that dictate vulnerability to injury and regenerative potential could lead to new therapeutic targets to treat ischemic kidney injury. Pax2 and Pax8 are two homologous nephron-specific transcription factors that are critical for kidney development and physiology. Here we report that proximal-tubule-selective depletion of Pax2 and Pax8 protects against both acute and chronic injury and induces an expression profile in the S3 proximal tubule with common features shared among diverse conditions that protect against ischemia. These findings highlight a new role for Pax proteins as potential therapeutic targets to treat AKI.
Collapse
|
28
|
Vazquez G, Sfakianos M, Coppa G, Jacob A, Wang P. NOVEL PS-OME MIRNA130B-3P REDUCES INFLAMMATION AND INJURY AND IMPROVES SURVIVAL AFTER RENAL ISCHEMIA-REPERFUSION INJURY. Shock 2023; 60:613-620. [PMID: 37594792 PMCID: PMC10592167 DOI: 10.1097/shk.0000000000002211] [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] [Indexed: 08/19/2023]
Abstract
ABSTRACT Introduction : Acute kidney injury (AKI) is a prevalent medical disorder characterized by a sudden decline in kidney function, often because of ischemia/reperfusion (I/R) events. It is associated with significant chronic complications, and currently available therapies are limited to supportive measures. Extracellular cold-inducible RNA-binding protein (eCIRP) has been identified as a mediator that potentiates inflammation after I/R injury. However, it has been discovered that miRNA 130b-3p acts as an endogenous inhibitor of eCIRP. To address the inherent instability of miRNA in vivo , a chemically modified miRNA mimic called PS-OME miR130 was developed. We hypothesize that administration of PS-OME miR130 after renal I/R can lead to reduced inflammation and injury in a murine model of AKI. Methods : C57BL/6 male mice underwent renal I/R by clamping of bilateral renal hilum for 30 min or sham operation. Immediately after closure, mice were intravenously administered vehicle (phosphate-buffered saline) or PS-OME miR130 at a dose of 12.5 nmol/mouse. Blood and kidneys were collected after 24 h for further analysis. Separately, mice underwent renal I/R and administered vehicle or treatment and, survival was monitored for 10 days. Results : After renal I/R, mice receiving vehicle showed a significant increase in serum markers of kidney injury and inflammation including blood urea nitrogen, NGAL, KIM-1, and IL-6. After treatment with PS-OME miR130, these markers were significantly decreased. Kidney tissue mRNA expression for injury and inflammation markers including NGAL, KIM-1, KC, and MIP-2 were increased after renal I/R; however, these markers showed a significant reduction with PS-OME miR130 treatment. Histologically, treatment with PS-OME miR130 showed a significant decrease in neutrophil infiltration and injury severity score, and decreased apoptosis. In the 10-day survival study, mice in the treatment group showed a significant reduction in mortality as compared with vehicle group. Conclusion : In a murine renal I/R model, the administration of PS-OME miR130, a direct eCIRP antagonistic miRNA mimic, resulted in the reduction of kidney inflammation and injury, and improved survival. PS-OME miR130 holds promise to be developed as novel therapeutic for AKI as an adjunct to the standard of care.
Collapse
Affiliation(s)
- Gustavo Vazquez
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Maria Sfakianos
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Gene Coppa
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Asha Jacob
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| |
Collapse
|
29
|
Späth MR, Hoyer-Allo KJR, Seufert L, Höhne M, Lucas C, Bock T, Isermann L, Brodesser S, Lackmann JW, Kiefer K, Koehler FC, Bohl K, Ignarski M, Schiller P, Johnsen M, Kubacki T, Grundmann F, Benzing T, Trifunovic A, Krüger M, Schermer B, Burst V, Müller RU. Organ Protection by Caloric Restriction Depends on Activation of the De Novo NAD+ Synthesis Pathway. J Am Soc Nephrol 2023; 34:772-792. [PMID: 36758124 PMCID: PMC10125653 DOI: 10.1681/asn.0000000000000087] [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: 03/20/2022] [Accepted: 01/10/2023] [Indexed: 02/11/2023] Open
Abstract
SIGNIFICANCE STATEMENT AKI is a major clinical complication leading to high mortality, but intensive research over the past decades has not led to targeted preventive or therapeutic measures. In rodent models, caloric restriction (CR) and transient hypoxia significantly prevent AKI and a recent comparative transcriptome analysis of murine kidneys identified kynureninase (KYNU) as a shared downstream target. The present work shows that KYNU strongly contributes to CR-mediated protection as a key player in the de novo nicotinamide adenine dinucleotide biosynthesis pathway. Importantly, the link between CR and NAD+ biosynthesis could be recapitulated in a human cohort. BACKGROUND Clinical practice lacks strategies to treat AKI. Interestingly, preconditioning by hypoxia and caloric restriction (CR) is highly protective in rodent AKI models. However, the underlying molecular mechanisms of this process are unknown. METHODS Kynureninase (KYNU) knockout mice were generated by Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and comparative transcriptome, proteome and metabolite analyses of murine kidneys pre- and post-ischemia-reperfusion injury in the context of CR or ad libitum diet were performed. In addition, acetyl-lysin enrichment and mass spectrometry were used to assess protein acetylation. RESULTS We identified KYNU as a downstream target of CR and show that KYNU strongly contributes to the protective effect of CR. The KYNU-dependent de novo nicotinamide adenine dinucleotide (NAD+) biosynthesis pathway is necessary for CR-associated maintenance of NAD+ levels. This finding is associated with reduced protein acetylation in CR-treated animals, specifically affecting enzymes in energy metabolism. Importantly, the effect of CR on de novo NAD+ biosynthesis pathway metabolites can be recapitulated in humans. CONCLUSIONS CR induces the de novo NAD+ synthesis pathway in the context of IRI and is essential for its full nephroprotective potential. Differential protein acetylation may be the molecular mechanism underlying the relationship of NAD+, CR, and nephroprotection.
Collapse
Affiliation(s)
- Martin R. Späth
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - K. Johanna R. Hoyer-Allo
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lisa Seufert
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Martin Höhne
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Christina Lucas
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Theresa Bock
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Genetics, University of Cologne, Cologne, Germany
| | - Lea Isermann
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Medical Faculty, Institute for Mitochondrial Diseases and Aging, University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Susanne Brodesser
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jan-Wilm Lackmann
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Katharina Kiefer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Felix C. Koehler
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Katrin Bohl
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Michael Ignarski
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Petra Schiller
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Marc Johnsen
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Torsten Kubacki
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Franziska Grundmann
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Aleksandra Trifunovic
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Medical Faculty, Institute for Mitochondrial Diseases and Aging, University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Marcus Krüger
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Genetics, University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Volker Burst
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Emergency Department, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Roman-Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
30
|
Miao J, Huang J, Liang Y, Zhang Y, Li J, Meng P, Shen W, Li X, Wu Q, Wang X, Niu H, Tang Y, Zhou S, Zhou L. Sirtuin 6 is a key contributor to gender differences in acute kidney injury. Cell Death Discov 2023; 9:134. [PMID: 37185276 PMCID: PMC10130034 DOI: 10.1038/s41420-023-01432-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/31/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Acute kidney injury (AKI) is rapidly increasing nowadays and at a high risk to progress into chronic kidney disease (CKD). Of note, men are more susceptive to AKI, suggesting gender differences in AKI patients. However, the underlying mechanisms remain largely unclear. To test it, we adopted two experimental models of AKI, including ischemia/reperfusion injury and rhabdomyolysis, which were constructed in age-matched male and female mice. We found severe damages of tubular apoptosis, mitochondrial dysfunction, and loss of renal function showing in male mice, while female mice only had very mild injury. We further tested the expression of Sirtuins, and found that female mice could preserve more Sirtuin members' expression in case of kidney damage. Among Sirtuin family, Sirtuin 6 was maximally preserved in injured kidney in female mice, suggesting its important role involved in the gender differences of AKI pathogenesis. We then found that knockdown of androgen receptor (AR) attenuated tubular damage, mitochondrial dysfunction and retarded the loss of renal function. Overexpression of Sirtuin 6 also showed similar results. Furthermore, in cultured tubular cells, dihydrotestosterone (DHT) decreased Sirtuin 6 expression and exacerbated cell apoptosis. Ectopic expression of Sirtuin 6 sufficiently inhibited DHT-induced cell apoptosis. Mechanically, we found AR inhibited Sirtuin 6, leading to the repression of binding of Sirtuin 6 with PGC-1α. This resulted in acetylation of PGC-1α and inhibition of its activity, further triggered the loss of mitochondrial homeostasis. Our results provided new insights to the underlying mechanisms of gender differences in AKI, suggesting Sirtuin 6 maybe a new therapeutic target for preventing AKI in male patients.
Collapse
Affiliation(s)
- Jinhua Miao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiewu Huang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Liang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yunfang Zhang
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Jiemei Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ping Meng
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Weiwei Shen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaolong Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qinyu Wu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxu Wang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongxin Niu
- Department of General Practice, Special Medical Service Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Ying Tang
- Department of Nephrology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| | - Shan Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
31
|
Rowe CJ, Walsh SA, Dragon AH, Rhodes AM, Pak OL, Ronzier E, Levi B, Potter BK, Spreadborough PJ, Davis TA. Tourniquet-induced ischemia creates increased risk of organ dysfunction and mortality following delayed limb amputation. Injury 2023:S0020-1383(23)00179-1. [PMID: 36906480 DOI: 10.1016/j.injury.2023.02.047] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/06/2023] [Accepted: 02/23/2023] [Indexed: 03/13/2023]
Abstract
Tourniquets are critical for the control of traumatic extremity hemorrhage. In this study, we sought to determine, in a rodent blast-related extremity amputation model, the impact of prolonged tourniquet application and delayed limb amputation on survival, systemic inflammation, and remote end organ injury. Adult male Sprague Dawley rats were subjected to blast overpressure (120±7 kPa) and orthopedic extremity injury consisting femur fracture, one-minute soft tissue crush injury (20 psi), ± 180 min of tourniquet-induced hindlimb ischemia followed by delayed (60 min of reperfusion) hindlimb amputation (dHLA). All animals in the non-tourniquet group survived whereas 7/21 (33%) of the animals in the tourniquet group died within the first 72 h with no deaths observed between 72 and 168 h post-injury. Tourniquet induced ischemia-reperfusion injury (tIRI) likewise resulted in a more robust systemic inflammation (cytokines and chemokines) and concomitant remote pulmonary, renal, and hepatic dysfunction (BUN, CR, ALT. AST, IRI/inflammation-mediated genes). These results indicate prolonged tourniquet application and dHLA increases risk of complications from tIRI, leading to greater risk of local and systemic complications including organ dysfunction or death. We thus need enhanced strategies to mitigate the systemic effects of tIRI, particularly in the military prolonged field care (PFC) setting. Furthermore, future work is needed to extend the window within which tourniquet deflation to assess limb viability remains feasible, as well as new, limb-specific or systemic point of care tests to better assess the risks of tourniquet deflation with limb preservation in order to optimize patient care and save both limb and life.
Collapse
Affiliation(s)
- Cassie J Rowe
- Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, United States; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, United States
| | - Sarah A Walsh
- Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, United States
| | - Andrea H Dragon
- Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, United States; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, United States
| | - Alisha M Rhodes
- Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, United States; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, United States
| | - Olivia L Pak
- Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, United States; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, United States
| | - Elsa Ronzier
- Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, United States; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, United States
| | - Benjamin Levi
- Center for Organogenesis Research and Trauma, University of Texas Southwestern, Dallas, TX, United States
| | - Benjamin K Potter
- Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, United States
| | - Philip J Spreadborough
- Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, United States; Academic Department of Military Surgery and Trauma, Royal Centre for Defence Medicine, Birmingham, United Kingdom
| | - Thomas A Davis
- Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, United States.
| |
Collapse
|
32
|
The Presence of Testis Determines Aristolochic Acid-Induced Nephrotoxicity in Mice. Toxins (Basel) 2023; 15:toxins15020118. [PMID: 36828432 PMCID: PMC9962534 DOI: 10.3390/toxins15020118] [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: 12/12/2022] [Revised: 01/12/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
Aristolochic acid (AA) is notorious for inducing nephrotoxicity, but the influence of sex on AA-induced kidney injury was not clear. This study sought to investigate sex differences in kidney dysfunction and tubular injury induced by AA. Male and female mice were bilaterally orchiectomized and ovariectomized, respectively. Fourteen days after gonadectomy, the mice were intraperitoneally injected with AA (10 mg/kg body weight/day) daily for 2 days and sacrificed 7 days after the first injection. Body weight, kidney function, and tubular structure were assessed. When compared between male and female non-gonadectomized mice, AA-induced body weight loss was greater in male mice than in female mice. Functional and structural damages in male kidneys were markedly induced by AA injection, but kidneys in AA-injected female mice showed no or mild damages. Ovariectomy had no effect on AA-induced nephrotoxic acute kidney injury in female mice. However, orchiectomy significantly reduced body weight loss, kidney dysfunction, and tubular injury in AA-induced nephrotoxicity in male mice. This study has demonstrated that testis causes AA-induced nephrotoxic acute kidney injury.
Collapse
|
33
|
Privratsky JR, Fuller M, Raghunathan K, Ohnuma T, Bartz RR, Schroeder R, Price TM, Martinez MR, Sigurdsson MI, Mathis MR, Naik B, Krishnamoorthy V. Postoperative Acute Kidney Injury by Age and Sex: A Retrospective Cohort Association Study. Anesthesiology 2023; 138:184-194. [PMID: 36512724 PMCID: PMC10439699 DOI: 10.1097/aln.0000000000004436] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Acute kidney injury (AKI) after noncardiac surgery is common and has substantial health impact. Preclinical and clinical studies examining the influence of sex on AKI have yielded conflicting results, although they typically do not account for age-related changes. The objective of the study was to determine the association of age and sex groups on postoperative AKI. The authors hypothesized that younger females would display lower risk of postoperative AKI than males of similar age, and the protection would be lost in older females. METHODS This was a multicenter retrospective cohort study across 46 institutions between 2013 and 2019. Participants included adult inpatients without pre-existing end-stage kidney disease undergoing index major noncardiac, nonkidney/urologic surgeries. The authors' primary exposure was age and sex groups defined as females 50 yr or younger, females older than 50 yr, males 50 yr or younger, and males older than 50 yr. The authors' primary outcome was development of AKI by Kidney Disease-Improving Global Outcomes serum creatinine criteria. Exploratory analyses included associations of ascending age groups and hormone replacement therapy home medications with postoperative AKI. RESULTS Among 390,382 patients, 25,809 (6.6%) developed postoperative AKI (females 50 yr or younger: 2,190 of 58,585 [3.7%]; females older than 50 yr: 9,320 of 14,4047 [6.5%]; males 50 yr or younger: 3,289 of 55,503 [5.9%]; males older than 50 yr: 11,010 of 132,447 [8.3%]). When adjusted for AKI risk factors, compared to females younger than 50 yr (odds ratio, 1), the odds of AKI were higher in females older than 50 yr (odds ratio, 1.51; 95% CI, 1.43 to 1.59), males younger than 50 yr (odds ratio, 1.90; 95% CI, 1.79 to 2.01), and males older than 50 yr (odds ratio, 2.06; 95% CI, 1.96 to 2.17). CONCLUSIONS Younger females display a lower odds of postoperative AKI that gradually increases with age. These results suggest that age-related changes in women should be further studied as modifiers of postoperative AKI risk after noncardiac surgery. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Jamie R. Privratsky
- Critical Care and Perioperative Population Health Research (CAPER) Unit, Department of Anesthesiology, Duke University Medical Center, Durham, NC
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Matthew Fuller
- Critical Care and Perioperative Population Health Research (CAPER) Unit, Department of Anesthesiology, Duke University Medical Center, Durham, NC
| | - Karthik Raghunathan
- Critical Care and Perioperative Population Health Research (CAPER) Unit, Department of Anesthesiology, Duke University Medical Center, Durham, NC
- Durham VA Medical Center, Durham, NC, USA
| | - Tetsu Ohnuma
- Critical Care and Perioperative Population Health Research (CAPER) Unit, Department of Anesthesiology, Duke University Medical Center, Durham, NC
| | - Raquel R. Bartz
- Critical Care and Perioperative Population Health Research (CAPER) Unit, Department of Anesthesiology, Duke University Medical Center, Durham, NC
| | - Rebecca Schroeder
- Critical Care and Perioperative Population Health Research (CAPER) Unit, Department of Anesthesiology, Duke University Medical Center, Durham, NC
- Durham VA Medical Center, Durham, NC, USA
| | - Thomas M. Price
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | - Michael R. Martinez
- Critical Care and Perioperative Population Health Research (CAPER) Unit, Department of Anesthesiology, Duke University Medical Center, Durham, NC
| | - Martin I. Sigurdsson
- Division of Anesthesia and Intensive Care Medicine, Landspitali -The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik Iceland
| | - Michael R. Mathis
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Bhiken Naik
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA
| | - Vijay Krishnamoorthy
- Critical Care and Perioperative Population Health Research (CAPER) Unit, Department of Anesthesiology, Duke University Medical Center, Durham, NC
| |
Collapse
|
34
|
Uduagbamen P, Sanusi M, Udom O. Original research gender differences in recovering from cardiac and vascular surgery associated acute kidney injury: A six-year retrospective comparative study in Nigeria. Ann Afr Med 2023; 22:160-166. [PMID: 37026196 DOI: 10.4103/aam.aam_81_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
Introduction Gender differences exist in the demographic, clinical characteristic and outcome of patients with cardiac and vascular surgery (CVS) associated acute kidney injury (AKI). Materials and Methods This retrospective study had a total of 88 participants for which socio-demographic, clinical and laboratory (serum electrolyte, full blood count, urine analysis and urine volume, creatinine, and glomerular filtration rate) data of participants were taken preoperative and postoperative days 1, 7, and 30. Results A total of 88 (66 males and 22 females) participants were studied. Diseases of the heart valves were more common in females than males. The mean age of the participants was 65.9 ± 6.9 years, with males 65.1 ± 7.6 years and females 68.3 ± 8.4 years, P = 0.02. Before surgery, a significantly greater proportion of females had kidney dysfunction compared to males, P = 0.003. Valvular surgery and coronary bypass were the most common surgeries. The proportion of emergency surgeries and admissions <7 days were significantly higher in females than males, P = 0.04 and P = 0.02, respectively. Full recovering from AKI was significantly higher in males as partial recovery and death were significantly lower in them, P = 0.02. Of the 35 (39.8%) who had dialysis, 85.7% recovered fully, 5.7% became dialysis, and dependent while 8.6% died. The predictors of nonrecovery from CVS-AKI were female gender, elderly, preoperative kidney dysfunction and AKI stage 3. Conclusion Males with AKI were younger than the females. Valvular surgeries were most common. Background kidney dysfunction and advance age were risk factors for AKI. Postoperative, AKI was commoner in males who were more likely to recover full kidney function. Optimizing patient preparation could reduce the incidence of CVS-AKI.
Collapse
|
35
|
McEvoy CM, Murphy JM, Zhang L, Clotet-Freixas S, Mathews JA, An J, Karimzadeh M, Pouyabahar D, Su S, Zaslaver O, Röst H, Arambewela R, Liu LY, Zhang S, Lawson KA, Finelli A, Wang B, MacParland SA, Bader GD, Konvalinka A, Crome SQ. Single-cell profiling of healthy human kidney reveals features of sex-based transcriptional programs and tissue-specific immunity. Nat Commun 2022; 13:7634. [PMID: 36496458 PMCID: PMC9741629 DOI: 10.1038/s41467-022-35297-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 11/27/2022] [Indexed: 12/13/2022] Open
Abstract
Knowledge of the transcriptional programs underpinning the functions of human kidney cell populations at homeostasis is limited. We present a single-cell perspective of healthy human kidney from 19 living donors, with equal contribution from males and females, profiling the transcriptome of 27677 cells to map human kidney at high resolution. Sex-based differences in gene expression within proximal tubular cells were observed, specifically, increased anti-oxidant metallothionein genes in females and aerobic metabolism-related genes in males. Functional differences in metabolism were confirmed in proximal tubular cells, with male cells exhibiting higher oxidative phosphorylation and higher levels of energy precursor metabolites. We identified kidney-specific lymphocyte populations with unique transcriptional profiles indicative of kidney-adapted functions. Significant heterogeneity in myeloid cells was observed, with a MRC1+LYVE1+FOLR2+C1QC+ population representing a predominant population in healthy kidney. This study provides a detailed cellular map of healthy human kidney, and explores the complexity of parenchymal and kidney-resident immune cells.
Collapse
Affiliation(s)
- Caitriona M McEvoy
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, ON, Canada
| | - Julia M Murphy
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Lin Zhang
- Department of Statistical Sciences, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Sergi Clotet-Freixas
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Jessica A Mathews
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - James An
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Mehran Karimzadeh
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Vector Institute, Toronto, ON, Canada
| | - Delaram Pouyabahar
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Shenghui Su
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Olga Zaslaver
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Hannes Röst
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Rangi Arambewela
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Lewis Y Liu
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Sally Zhang
- Division of Urology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Keith A Lawson
- Division of Urology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Antonio Finelli
- Division of Urology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Bo Wang
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Vector Institute, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Sonya A MacParland
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Gary D Bader
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Ana Konvalinka
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada.
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, ON, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| | - Sarah Q Crome
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada.
- Department of Immunology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
36
|
Jing H, Liao M, Tang S, Lin S, Ye L, Zhong J, Wang H, Zhou J. Predicting the risk of acute kidney injury after cardiopulmonary bypass: development and assessment of a new predictive nomogram. BMC Anesthesiol 2022; 22:379. [PMID: 36476178 PMCID: PMC9727998 DOI: 10.1186/s12871-022-01925-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a common and severe complication of cardiac surgery with cardiopulmonary bypass (CPB). This study aimed to establish a model to predict the probability of postoperative AKI in patients undergoing cardiac surgery with CPB. METHODS We conducted a retrospective, multicenter study to analyze 1082 patients undergoing cardiac surgery under CPB. The least absolute shrinkage and selection operator regression model was used to optimize feature selection for the AKI model. Multivariable logistic regression analysis was applied to build a prediction model incorporating the feature selected in the previously mentioned model. Finally, we used multiple methods to evaluate the accuracy and clinical applicability of the model. RESULTS Age, gender, hypertension, CPB duration, intraoperative 5% bicarbonate solution and red blood cell transfusion, urine volume were identified as important factors. Then, these risk factors were created into nomogram to predict the incidence of AKI after cardiac surgery under CPB. CONCLUSION We developed a nomogram to predict the incidence of AKI after cardiac surgery. This model can be used as a reference tool for evaluating early medical intervention to prevent postoperative AKI.
Collapse
Affiliation(s)
- Huan Jing
- grid.413107.0The Third Affiliated Hospital of Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangdong Province Guangzhou City, China
| | - Meijuan Liao
- grid.452881.20000 0004 0604 5998The First People’s Hospital of Foshan, 81 Lingnan Avenue, Chancheng District, Guangdong Province Foshan City, China
| | - Simin Tang
- grid.413107.0The Third Affiliated Hospital of Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangdong Province Guangzhou City, China
| | - Sen Lin
- grid.452881.20000 0004 0604 5998The First People’s Hospital of Foshan, 81 Lingnan Avenue, Chancheng District, Guangdong Province Foshan City, China
| | - Li Ye
- grid.452881.20000 0004 0604 5998The First People’s Hospital of Foshan, 81 Lingnan Avenue, Chancheng District, Guangdong Province Foshan City, China
| | - Jiying Zhong
- grid.452881.20000 0004 0604 5998The First People’s Hospital of Foshan, 81 Lingnan Avenue, Chancheng District, Guangdong Province Foshan City, China
| | - Hanbin Wang
- grid.452881.20000 0004 0604 5998The First People’s Hospital of Foshan, 81 Lingnan Avenue, Chancheng District, Guangdong Province Foshan City, China
| | - Jun Zhou
- grid.413107.0The Third Affiliated Hospital of Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangdong Province Guangzhou City, China
| |
Collapse
|
37
|
Ciarambino T, Crispino P, Giordano M. Gender and Renal Insufficiency: Opportunities for Their Therapeutic Management? Cells 2022; 11:cells11233820. [PMID: 36497080 PMCID: PMC9740491 DOI: 10.3390/cells11233820] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
Acute kidney injury (AKI) is a major clinical problem associated with increased morbidity and mortality. Despite intensive research, the clinical outcome remains poor, and apart from supportive therapy, no other specific therapy exists. Furthermore, acute kidney injury increases the risk of developing chronic kidney disease (CKD) and end-stage renal disease. Acute tubular injury accounts for the most common intrinsic cause of AKI. The main site of injury is the proximal tubule due to its high workload and energy demand. Upon injury, an intratubular subpopulation of proximal epithelial cells proliferates and restores the tubular integrity. Nevertheless, despite its strong regenerative capacity, the kidney does not always achieve its former integrity and function and incomplete recovery leads to persistent and progressive CKD. Clinical and experimental data demonstrate sexual differences in renal anatomy, physiology, and susceptibility to renal diseases including but not limited to ischemia-reperfusion injury. Some data suggest the protective role of female sex hormones, whereas others highlight the detrimental effect of male hormones in renal ischemia-reperfusion injury. Although the important role of sex hormones is evident, the exact underlying mechanisms remain to be elucidated. This review focuses on collecting the current knowledge about sexual dimorphism in renal injury and opportunities for therapeutic manipulation, with a focus on resident renal progenitor stem cells as potential novel therapeutic strategies.
Collapse
Affiliation(s)
- Tiziana Ciarambino
- Internal Medicine Department, Hospital of Marcianise, ASL Caserta, 81031 Caserta, Italy
- Correspondence: (T.C.); (M.G.)
| | - Pietro Crispino
- Emergency Department, Hospital of Latina, ASL Latina, 04100 Latina, Italy
| | - Mauro Giordano
- Department of Advanced Medical and Surgical Science, University of Campania, Luigi Vanvitelli, 80138 Naples, Italy
- Correspondence: (T.C.); (M.G.)
| |
Collapse
|
38
|
Saleh H, Salama M, Hussein RM. Polyethylene glycol capped gold nanoparticles ameliorate renal ischemia-reperfusion injury in diabetic mice through AMPK-Nrf2 signaling pathway. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:77884-77907. [PMID: 35688972 PMCID: PMC9581836 DOI: 10.1007/s11356-022-21235-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/29/2022] [Indexed: 05/09/2023]
Abstract
The aim of this study is to investigate the protective effect of polyethylene glycol capped gold nanoparticles (PEG-AuNPs) on renal ischemia-reperfusion injury (I/R)-induced acute kidney injury (AKI) in diabetic mice via the activation of adenosine 5' monophosphate-activated protein kinase-nuclear factor erythroid-2-related factor-2 (AMPK-Nrf2) pathway. Diabetes was induced in male mice (12/group) by streptozotocin (50 mg/kg) for 5 consecutive days. After 4 weeks, the mice have intravenously received doses of PEG-AuNPs (40, 150, and 400 µg/kg body weight) for 3 consecutive days, and then animals were subjected to 30 min ischemia and 48 h reperfusion. Following the treatment with three different doses of PEG-AuNPs, the levels of blood urea nitrogen (BUN) and creatinine were reduced. Obvious reduction in renal tubular atrophy, glomerular damage, mitochondrial damage, and necrotic area were ultra-structurally detected, and renal interstitial inflammation and apoptosis were diminished. Moreover, PEG-AuNPs increased the recovering of damaged renal cells, suppressed significantly levels of malondialdehyde (MDA), downregulated significantly the level of inflammatory cytokines (TNF-α and IL-1β), and upregulated the AMPK-Nrf2 pathway. PEG-AuNPs exhibited a promising alternative therapeutic target for diabetic renal I/R-induced AKI through upregulation of AMPK/PI3K/AKT path which additionally stimulated Nrf2-regulated antioxidant enzymes in a dose-dependent manner.
Collapse
Affiliation(s)
- Hanan Saleh
- Department of Zoology, Faculty of Science, Cairo University, P.O. Box 12613, Giza, Egypt
| | - Mohamed Salama
- Textile Research and and Technology Institute, National Research Centre, El Buhouth street Dokki, P.O. Box 12622, Giza, Egypt
| | - Rehab Mohamed Hussein
- Department of Zoology, Faculty of Science, Cairo University, P.O. Box 12613, Giza, Egypt
| |
Collapse
|
39
|
Testosterone deficiency in male organ transplant recipients. Int J Impot Res 2022; 34:679-684. [PMID: 35013565 DOI: 10.1038/s41443-021-00513-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 11/13/2021] [Accepted: 11/26/2021] [Indexed: 11/09/2022]
Abstract
Testosterone deficiency is known to affect men with increasing incidence throughout their lifespan. The clinical manifestations of testosterone deficiency, in turn, negatively impact men's quality of life and perception of overall health. The interaction of chronic systemic disease and androgen deficiency represent an area for potential intervention. Here, we explore the topic of testosterone deficiency amongst men with end-stage organ failure requiring transplantation in order to elucidate the underlying pathophysiology of androgen deficiency of chronic disease and discuss whether intervention, including testosterone replacement and organ transplantation, improve patients' outcomes and quality of life.
Collapse
|
40
|
Semerdzhieva NE, Tsakova AD, Gospodinova M, Dimitrov SI, Denchev S. Total Testosterone as a Specific Marker of Acute Kidney Injury in Male Patients With Myocardial Infarction. Cureus 2022; 14:e28682. [PMID: 36199650 PMCID: PMC9526781 DOI: 10.7759/cureus.28682] [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] [Accepted: 08/31/2022] [Indexed: 11/18/2022] Open
Abstract
Aim The aim of the present study was to assess the significance of total testosterone (T) as a marker of acute kidney injury (AKI) in patients with acute myocardial infarction (MI). Patients and methods The study was a retrospective, single-center cohort study that included 55 consecutive male patients diagnosed with acute MI who were admitted to the Cardiology Clinic of Alexandrovska University Hospital (Sofia, Bulgaria) between July 2011 and December 2013. The plasma total T levels, measured at admission, the peak levels of myocardial necrosis markers, high-sensitive C-reactive protein (hsCRP), and the left ventricular ejection fraction (LVEF) were analyzed in relation to the incidence of AKI. Results The occurrence of AKI was positively predicted by reduced EF (OR=0.825; CI=0.724-0.942; P=0.004), advanced age (OR=1.077; CI=1.038-1.151; P=0.029), and low levels of total T (OR=0.837; CI=0.707-0.990; P=0.037). Reduced systolic function (OR=0.861; 95% CI=0.758-0.978; P=0.022 for EF) and marginally age (OR=1.094; 95% CI=1.000-1.197; P=0.051) contributed to the incidence of AKI in a multivariate model. Total T was not an independent factor (OR=0.841; 95% CI=0.669-1.058; P=0.139) for AKI. The total T levels were significantly inversely correlated with the peak of hsCRP (r= -0.153; P=0.009) and showed a tendency to inverse relation with the SYNTAX score (r= -0.235; P=0.083). Conclusion The total T levels are significantly inversely related to the peak of hsCRP and as a tendency to the SYNTAX score in male patients with acute MI. A low level of plasma total T is not an independent marker of AKI in acute MI. Advanced age and low EF are independent factors for AKI discrimination in a small cohort of patients with acute MI.
Collapse
Affiliation(s)
| | - Adelina D Tsakova
- Central Clinical Laboratory, Medical University of Sofia, Aleksandrovska University Hospital, Sofia, BGR
| | | | | | - Stefan Denchev
- Cardiology Department, Medical Center 'Mediva', Sofia, BGR
| |
Collapse
|
41
|
Liu M, Cui Z, Chen J, Gao M, Zhu Z, Chen H. Dietary selenium intake and the risk of kidney stones in adults, an analysis of 2007-2018 National Health and Nutrition Examination Survey, a cross-sectional study. Front Nutr 2022; 9:877917. [PMID: 36034902 PMCID: PMC9400542 DOI: 10.3389/fnut.2022.877917] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose To evaluate the association between dietary selenium intake and the risk of kidney stones in adults. Materials and methods We performed a cross-sectional analysis using data from 2007 to 2018 National Health and Nutrition Examination Survey (NHANES). Dietary intake information of 30,184 participants was obtained using first 24-h dietary recall interview, and kidney stones were presented by a standard questionnaire. The quartile analysis, stratified analysis and non-linearity analysis were used to estimate the association between dietary selenium intake and kidney stones after an adjustment for potential confounders. Results The multiple logistic regression indicated that the fourth quantile (Q4) of dietary selenium intake had a lower risk of kidney stones than the first quantile (Q1) in Model 3 (OR 0.82, P < 0.05). The stratified analyses indicated there were statistical differences between dietary selenium intake and kidney stones among younger (age < 50) (OR 0.65, P < 0.01), male (OR 0.73, P < 0.01) and overweight/obese (BMI ≥ 25.0) (OR 0.80, P < 0.05) individuals in Model 3. The non-linear relationship was founded between dietary selenium intake and kidney stones in all participants, younger, male and overweight/obese individuals after adjusting for confounding factors. Conclusion Our study revealed an inverse relation between the level of dietary selenium intake and the risk of kidney stones for the United States population, especially for younger (age < 50), male and overweight/obese (BMI ≥ 25.0) individuals. The study provides preliminary guidance on dietary selenium intake for the prevention of kidney stones in different populations. Further studies are required to confirm our findings and clarified the biological mechanisms.
Collapse
Affiliation(s)
- Minghui Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhongxiao Cui
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jinbo Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Meng Gao
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zewu Zhu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hequn Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
42
|
Cornelius RJ, Nelson JW, Su XT, Yang CL, Ellison DH. COP9 signalosome deletion promotes renal injury and distal convoluted tubule remodeling. Am J Physiol Renal Physiol 2022; 323:F4-F19. [PMID: 35532068 PMCID: PMC9236871 DOI: 10.1152/ajprenal.00436.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cullin-RING ligases are a family of E3 ubiquitin ligases that control cellular processes through regulated degradation. Cullin 3 targets with-no-lysine kinase 4 (WNK4), a kinase that activates the Na+-Cl- cotransporter (NCC), the main pathway for Na+ reabsorption in the distal convoluted tubule (DCT). Mutations in the cullin 3 gene lead to familial hyperkalemic hypertension by increasing WNK4 abundance. The constitutive photomorphogenesis 9 (COP9) signalosome (CSN) regulates the activity of cullin-RING ligases by removing the ubiquitin-like protein neural precursor cell expressed developmentally downregulated protein 8. Genetic deletion of the catalytically active CSN subunit, Jab1, along the nephron in mice (KS-Jab1-/-) led to increased WNK4 abundance; however, NCC abundance was substantially reduced. We hypothesized that the reduction in NCC resulted from a cortical injury that led to hypoplasia of the segment, which counteracted WNK4 activation of NCC. To test this, we studied KS-Jab1-/- mice at weekly intervals over a period of 3 wk. The results showed that NCC abundance was unchanged until 3 wk after Jab1 deletion, at which time other DCT-specific proteins were also reduced. The kidney injury markers kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin demonstrated kidney injury immediately after Jab1 deletion; however, the damage was initially limited to the medulla. The injury progressed and expanded into the cortex 3 wk after Jab1 deletion coinciding with loss of the DCT. The data indicate that nephron-specific disruption of the cullin-RING ligase system results in a complex progression of tubule injury that leads to hypoplasia of the DCT.NEW & NOTEWORTHY Cullin 3 (CUL3) targets with-no-lysine-kinase 4 (WNK4), which activates Na+-Cl- cotransporter (NCC) in the distal convoluted tubule (DCT) of the kidney. Renal-specific genetic deletion of the constitutive photomorphogenesis 9 signalosome, an upstream regulator of CUL3, resulted in a reduction of NCC due to DCT hypoplasia, which coincided with cortical kidney injury. The data indicate that nephron-specific disruption of the cullin-RING ligase system results in a complex progression of tubule injury leading to hypoplasia of the DCT.
Collapse
Affiliation(s)
- Ryan J. Cornelius
- 1Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Jonathan W. Nelson
- 1Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Xiao-Tong Su
- 1Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Chao-Ling Yang
- 1Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - David H. Ellison
- 1Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon,2Veterans Affairs Portland Health Care System, Portland, Oregon
| |
Collapse
|
43
|
Schiffl H, Lang SM. Dimorphic Response of Sex and Hospital-acquired Acute Kidney Injury. SAUDI JOURNAL OF KIDNEY DISEASES AND TRANSPLANTATION 2022; 33:574-581. [PMID: 37929551 DOI: 10.4103/1319-2442.388192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023] Open
Abstract
The risk of hospital-acquired acute kidney injury (HA-AKI) depends on a person's intrinsic susceptibility, the presence of risk factors, and on the type and extent of exposure to kidney insults. Older cohort studies have focused on male-only or mostly male populations, assuming a lower incidence of HA-AKI in women. Insufficient statistical power suggested that female sex was a shared susceptibility factor for HA-AKI. It was included as a risk factor in risk prediction models of HA-AKI. With the inclusion of women in clinical research studies, this presumption was challenged. Recent meta-analyses of sex-stratified studies showed that the risk for HA-AKI was significantly higher in men. These results suggested a protective role of female sex hormones. However, these studies were complicated by the inclusion of women across an age spectrum that includes the menopausal shift. Preliminary clinical and basic research data suggest that postmenopausal women lose their protection from HA-AKI. The number, size, and quality of reported clinical studies are low. There is an unmet need to characterize the susceptibility factor sex, to assess its clinical relevance and to evaluate renoprotection by sex hormone administration.
Collapse
Affiliation(s)
- Helmut Schiffl
- Department of Internal Medicine IV, University Hospital LMU Munich, Munich, Germany
| | - Susanne M Lang
- Department of Pneumology, Clinic of Internal Medicine V, University Hospital Jena, Jena, Germany
| |
Collapse
|
44
|
Chen HW, Hsu CN, Lee YT, Fu CM, Wang SW, Huang CC, Li LC. Comparative Adverse Kidney Outcomes in Women Receiving Raloxifene and Denosumab in a Real-World Setting. Biomedicines 2022; 10:biomedicines10071494. [PMID: 35884799 PMCID: PMC9313089 DOI: 10.3390/biomedicines10071494] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
Both osteoporosis and kidney diseases are common and intercorrelate to increase morbidity and mortality in elderly women. This study aimed to compare adverse kidney outcome between women initiated with denosumab and a matched group of raloxifene initiators using propensity score matching methods in a large healthcare delivery system in Taiwan. The risks of adverse kidney outcomes were estimated using Cox proportional hazard regression and the change in kidney function over time was analyzed using the linear mixed model. A total of 9444 (4722 in each group) women were identified who matched the inclusion criteria between January 2003 and December, 2018. Denosumab use was significantly associated with higher risk of eGFR decline ≥ 30% from baseline than raloxifene use (aHR: 1.26; 95% CI: 1.16−1.36, p < 0.0001). The mean change in eGFR over time was 1.24 mL/min/1.73 m2 per year in the denosumab group and 0.45 mL/min/1.73 m2 per year in the raloxifene group (p = 0.0004). However, the risks of acute kidney injury (10.53%) and chronic dialysis (0.66%) in this study cohort were not significantly different for the two anti-osteoporosis treatments. Close monitoring of the residual kidney function and treatment effect is needed in those with denosumab.
Collapse
Affiliation(s)
- Hsin-Wei Chen
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-W.C.); (Y.-T.L.); (C.-M.F.); (C.-C.H.)
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (C.-N.H.); (S.-W.W.)
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yueh-Ting Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-W.C.); (Y.-T.L.); (C.-M.F.); (C.-C.H.)
| | - Chung-Ming Fu
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-W.C.); (Y.-T.L.); (C.-M.F.); (C.-C.H.)
| | - Shih-Wei Wang
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (C.-N.H.); (S.-W.W.)
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chiang-Chi Huang
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-W.C.); (Y.-T.L.); (C.-M.F.); (C.-C.H.)
| | - Lung-Chih Li
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-W.C.); (Y.-T.L.); (C.-M.F.); (C.-C.H.)
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Correspondence: ; Tel.: +886-7-7317123 (ext. 8306)
| |
Collapse
|
45
|
Caloric restriction reduces the pro-inflammatory eicosanoid 20- hydroxyeicosatetraenoic acid to protect from acute kidney injury. Kidney Int 2022; 102:560-576. [PMID: 35654224 DOI: 10.1016/j.kint.2022.04.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/13/2022] [Accepted: 04/22/2022] [Indexed: 11/22/2022]
Abstract
Acute kidney injury is a frequent complication in the clinical setting and associated with significant morbidity and mortality. Preconditioning with short-term caloric restriction is highly protective against kidney injury in rodent ischemia reperfusion injury models. However, the underlying mechanisms are unknown hampering clinical translation. Here, we examined the molecular basis of caloric restriction-mediated protection to elucidate the principles of kidney stress resistance. Analysis of an RNAseq dataset after caloric restriction identified Cyp4a12a, a cytochrome exclusively expressed in male mice, to be strongly downregulated after caloric restriction. Kidney ischemia reperfusion injury robustly induced acute kidney injury in male mice and this damage could be markedly attenuated by pretreatment with caloric restriction. In females, damage was significantly less pronounced and preconditioning with caloric restriction had only little effect. Tissue concentrations of the metabolic product of Cyp4a12a, 20-hydroxyeicosatetraenoic acid (20-HETE), were found to be significantly reduced by caloric restriction. Conversely, intraperitoneal supplementation of 20-HETE in preconditioned males partly abrogated the protective potential of caloric restriction. Interestingly, this effect was accompanied by a partial reversal of caloric restriction-induced changes in protein but not RNA expression pointing towards inflammation, endoplasmic reticulum stress and lipid metabolism. Thus, our findings provide an insight into the mechanisms underlying kidney protection by caloric restriction. Hence, understanding the mediators of preconditioning is an important pre-requisite for moving towards translation to the clinical setting.
Collapse
|
46
|
Aal-Aaboda M, Abu Raghif AR, Almudhafer RH, Hadi NR. Lipopolysaccharide from Rhodobacter spheroids modulate toll-like receptors expression and tissue damage in an animal model of bilateral renal ischemic reperfusion injury. J Med Life 2022; 15:685-697. [PMID: 35815074 PMCID: PMC9262262 DOI: 10.25122/jml-2021-0255] [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: 09/04/2021] [Accepted: 12/10/2021] [Indexed: 11/19/2022] Open
Abstract
Ischemic reperfusion injury (IRI) of the kidneys is a direct sequela of surgical procedures associated with the interruption of blood supply. The pathophysiology of IRI is complicated, and several inflammatories, apoptosis, and oxidative stress pathways are implicated. Among the major receptors directly involved in renal IRI are the toll-like receptors (TLRs), specifically TLR2 and TLR4. In this study, we investigated the effects of Lipopolysaccharide from Rhodobacter Sphaeroides (TLR2 and TLR4 antagonist, LPS-RS) and the ultrapure form (pure TLR4 antagonist, ULPS-RS) on the histopathological changes and TLRs expression in an animal model of bilateral renal IRI. Forty-eight adult male rats were allocated into six groups (N=8) as follows: sham group (negative control without IRI), control group (rats underwent bilateral renal ischemia for 30 minutes and 2 hours of reperfusion), vehicle group (IRI+ vehicle), LPS-RS group (IRI+ 0.5 mg/kg of LPS-RS), ULPS-RS group (IRI+ 0.1 mg/kg of ULPS-RS), ULPS-RSH group (IRI+ 0.2 mg/kg of ULPS-RS). Significant improvement in the histopathological damages induced by renal IRI was found in the ULPS-RS treated groups at both doses compared with the control group. The protective effect of ULPS-RS was associated with significantly reduced TLR4 expression without affecting TLR2. Regarding LPS-RS, the tested dose adversely affected the renal tissues as manifested by the histopathological findings, although it similarly affected TLRs expression as ULPS-RS. Our results demonstrated that ULPS-RS was renoprotective while LPS-RS had no protective effect against the tissue damages induced by renal IRI.
Collapse
Affiliation(s)
- Munaf Aal-Aaboda
- Department of Pharmacology, Faculty of Pharmacy, University of Misan, Amarah, Iraq
| | | | - Rihab Hameed Almudhafer
- Middle Euphrates Unit for Cancer Research, Faculty of Medicine, University of Kufa, Kufa, Iraq
| | - Najah Riesh Hadi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq,Corresponding Author: Najah Riesh Hadi, Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq. E-mail:
| |
Collapse
|
47
|
Nemours S, Castro L, Ribatallada-Soriano D, Semidey ME, Aranda M, Ferrer M, Sanchez A, Morote J, Cantero-Recasens G, Meseguer A. Temporal and sex-dependent gene expression patterns in a renal ischemia-reperfusion injury and recovery pig model. Sci Rep 2022; 12:6926. [PMID: 35484379 PMCID: PMC9051203 DOI: 10.1038/s41598-022-10352-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/30/2022] [Indexed: 12/30/2022] Open
Abstract
Men are more prone to acute kidney injury (AKI) and chronic kidney disease (CKD), progressing to end-stage renal disease (ESRD) than women. Severity and capacity to regenerate after AKI are important determinants of CKD progression, and of patient morbidity and mortality in the hospital setting. To determine sex differences during injury and recovery we have generated a female and male renal ischemia/reperfusion injury (IRI) pig model, which represents a major cause of AKI. Although no differences were found in blood urea nitrogen (BUN) and serum creatinine (SCr) levels between both sexes, females exhibited higher mononuclear infiltrates at basal and recovery, while males showed more tubular damage at injury. Global transcriptomic analyses of kidney biopsies from our IRI pig model revealed a sexual dimorphism in the temporal regulation of genes and pathways relevant for kidney injury and repair, which was also detected in human samples. Enrichment analysis of gene sets revealed five temporal and four sexual patterns governing renal IRI and recovery. Overall, this study constitutes an extensive characterization of the time and sex differences occurring during renal IRI and recovery at gene expression level and offers a template of translational value for further study of sexual dimorphism in kidney diseases.
Collapse
Affiliation(s)
- Stéphane Nemours
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Luis Castro
- Biomedical Research in Urology Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Didac Ribatallada-Soriano
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Maria E Semidey
- Department of Pathology, Hospital Vall d'Hebron, Barcelona, Spain
| | - Miguel Aranda
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Marina Ferrer
- Rodent Platform, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Sanchez
- Unitat d'Estadística I Bioinformàtica, (UEB), Vall d'Hebron Research Institute, Barcelona, Spain
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Barcelona, Spain
| | - Joan Morote
- Biomedical Research in Urology Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Gerard Cantero-Recasens
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Anna Meseguer
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
- Departament de Bioquímica I Biologia Molecular, Unitat de Bioquímica de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain.
- Red de Investigación Renal (REDINREN), Instituto Carlos III-FEDER, Madrid, Spain.
| |
Collapse
|
48
|
Porter AW, Nguyen DN, Clayton DR, Ruiz WG, Mutchler SM, Ray EC, Marciszyn AL, Nkashama LJ, Subramanya AR, Gingras S, Kleyman TR, Apodaca G, Hendershot LM, Brodsky JL, Buck TM. The molecular chaperone GRP170 protects against ER stress and acute kidney injury in mice. JCI Insight 2022; 7:e151869. [PMID: 35104250 PMCID: PMC8983141 DOI: 10.1172/jci.insight.151869] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 01/28/2022] [Indexed: 01/26/2023] Open
Abstract
Molecular chaperones are responsible for maintaining cellular homeostasis, and one such chaperone, GRP170, is an endoplasmic reticulum (ER) resident that oversees both protein biogenesis and quality control. We previously discovered that GRP170 regulates the degradation and assembly of the epithelial sodium channel (ENaC), which reabsorbs sodium in the distal nephron and thereby regulates salt-water homeostasis and blood pressure. To define the role of GRP170 - and, more generally, molecular chaperones in kidney physiology - we developed an inducible, nephron-specific GRP170-KO mouse. Here, we show that GRP170 deficiency causes a dramatic phenotype: profound hypovolemia, hyperaldosteronemia, and dysregulation of ion homeostasis, all of which are associated with the loss of ENaC. Additionally, the GRP170-KO mouse exhibits hallmarks of acute kidney injury (AKI). We further demonstrate that the unfolded protein response (UPR) is activated in the GRP170-deficient mouse. Notably, the UPR is also activated in AKI when originating from various other etiologies, including ischemia, sepsis, glomerulonephritis, nephrotic syndrome, and transplant rejection. Our work establishes the central role of GRP170 in kidney homeostasis and directly links molecular chaperone function to kidney injury.
Collapse
Affiliation(s)
- Aidan W. Porter
- Department of Biological Sciences
- Department of Pediatrics, Nephrology Division
| | | | | | - Wily G. Ruiz
- Department of Medicine, Renal-Electrolyte Division
| | | | - Evan C. Ray
- Department of Medicine, Renal-Electrolyte Division
| | | | | | | | | | - Thomas R. Kleyman
- Department of Medicine, Renal-Electrolyte Division
- Department of Cell Biology, and
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Linda M. Hendershot
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | | | | |
Collapse
|
49
|
Abstract
Female sex confers renoprotection in chronic progressive kidney disease. It is less well recognized that sexual dimorphism also is evident in the development of ischemic and nephrotoxic acute kidney injury (AKI). Animal studies consistently have shown that female sex protects against the development of renal injury in experimental models of ischemic AKI. However, the consensus opinion is that in human beings, female sex is an independent risk factor for AKI. Based on a systematic review of experimental and clinical literature, we present data to support the conclusion that, contrary to consensus opinion, it is male sex, not female sex, that is associated with the development of AKI.
Collapse
Affiliation(s)
- Joel Neugarten
- Renal Division, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY.
| | - Ladan Golestaneh
- Renal Division, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
50
|
Packialakshmi B, Stewart IJ, Burmeister DM, Feng Y, McDaniel DP, Chung KK, Zhou X. Tourniquet-induced lower limb ischemia/reperfusion reduces mitochondrial function by decreasing mitochondrial biogenesis in acute kidney injury in mice. Physiol Rep 2022; 10:e15181. [PMID: 35146957 PMCID: PMC8831939 DOI: 10.14814/phy2.15181] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 06/14/2023] Open
Abstract
The mechanisms by which lower limb ischemia/reperfusion induces acute kidney injury (AKI) remain largely uncharacterized. We hypothesized that tourniquet-induced lower limb ischemia/reperfusion (TILLIR) would inhibit mitochondrial function in the renal cortex. We used a murine model to show that TILLIR of the high thigh regions inflicted time-dependent AKI as determined by renal function and histology. This effect was associated with decreased activities of mitochondrial complexes I, II, V and citrate synthase in the kidney cortex. Moreover, TILLIR reduced mRNA levels of a master regulator of mitochondrial biogenesis PGC-1α, and its downstream genes NDUFS1 and ATP5o in the renal cortex. TILLIR also increased serum corticosterone concentrations. TILLIR did not significantly affect protein levels of the critical regulators of mitophagy PINK1 and PARK2, mitochondrial transport proteins Tom20 and Tom70, or heat-shock protein 27. TILLIR had no significant effect on mitochondrial oxidative stress as determined by mitochondrial ability to generate reactive oxygen species, protein carbonylation, or protein levels of MnSOD and peroxiredoxin1. However, TILLIR inhibited classic autophagic flux by increasing p62 protein abundance and preventing the conversion of LC3-I to LC3-II. TILLIR increased phosphorylation of cytosolic and mitochondrial ERK1/2 and mitochondrial AKT1, as well as mitochondrial SGK1 activity. In conclusion, lower limb ischemia/reperfusion induces distal AKI by inhibiting mitochondrial function through reducing mitochondrial biogenesis. This AKI occurs without significantly affecting PINK1-PARK2-mediated mitophagy or mitochondrial oxidative stress in the kidney cortex.
Collapse
Affiliation(s)
- Balamurugan Packialakshmi
- Department of MedicineUniformed Services University of the Health SciencesBethesdaMarylandUSA
- The Henry Jackson M. Foundation for the Advancement of Military MedicineBethesdaMarylandUSA
| | - Ian J. Stewart
- Department of MedicineUniformed Services University of the Health SciencesBethesdaMarylandUSA
| | - David M. Burmeister
- Department of MedicineUniformed Services University of the Health SciencesBethesdaMarylandUSA
| | - Yuanyi Feng
- Department of BiochemistryUniformed Services University of the Health SciencesBethesdaMarylandUSA
| | - Dennis P. McDaniel
- Biomedical Instrumentation CenterUniformed Services University of the Health SciencesBethesdaMarylandUSA
| | - Kevin K. Chung
- Department of MedicineUniformed Services University of the Health SciencesBethesdaMarylandUSA
| | - Xiaoming Zhou
- Department of MedicineUniformed Services University of the Health SciencesBethesdaMarylandUSA
| |
Collapse
|