1
|
Liu L, Cai H, Yang H, Wang S, Li Y, Huang Y, Gao M, Zhang X, Zhang X, Wang H, Qiu G. Targeted metabolomics identified novel metabolites, predominantly phosphatidylcholines and docosahexaenoic acid-containing lipids, predictive of incident chronic kidney disease in middle-to-elderly-aged Chinese adults. Metabolism 2025; 163:156085. [PMID: 39608488 DOI: 10.1016/j.metabol.2024.156085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/29/2024] [Accepted: 11/22/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Evidence is limited regarding the association of circulating metabolites with decline of kidney function, letting alone their value in prediction of development of chronic kidney disease (CKD). METHODS This study included 3802 participants aged 64.1 ± 7.4 years from the Dongfeng-Tongji cohort, among whom 3327 were CKD-free at baseline (estimated glomerular filtration rate [eGFR] > 60 ml/min per 1.73 m2). We measured baseline levels of 211 metabolites with liquid chromatography coupled with mass spectrometry, including 25 amino acids, 12 acyl-carnitines, 161 lipids, and 13 other metabolites. RESULTS The mean (SD) absolute annual change in eGFR was -0.14 ± 4.11 ml/min per 1.73 m2 per year, and a total of 472 participants who were free of CKD at baseline developed incident CKD during follow-up of 4.6 ± 0.2 years (14.2 %). We identified a total of 22 metabolites associated with annual eGFR change and survived Bonferroni correction for multiple testing, including seven metabolites associated with eGFR increase (six being docosahexaenoic acid [DHA]-containing lipids) and 15 associated with eGFR decline (nine being phosphatidylcholines [PCs]). Among them, eight metabolites obtained non-zero coefficients in least absolute shrinkage and selection operator (LASSO) regression on incident CKD, indicating predictive potential, including one amino acid (arginine), one acyl-carnitine (C2), one lysophosphatidylcholine (LPC 22:6), two PCs (32:1 and 34:3), one triacylglycerol (TAG 56:8 [22:6]) and two other metabolites (inosine, niacinamide), and the composite score of these eight metabolites showed an odds ratio (OR) of 8.79 (95 % confidence interval [CI]: 7.49, 10.32; P < 0.001) per SD increase in association with incident CKD. The addition of the metabolite score increased the c-statistic of the reference model of traditional risk factors (including baseline eGFR) by 0.065 (95 % CI: 0.046 to 0.084; P = 3.39 × 10-11) to 0.765 (0.742 to 0.788) in 1000 repetitions of 10-fold cross-validation, while the application of two advanced machine learning algorithms, random forest (RF), and extreme gradient boosting (XGBoost) models produced similar c-statistics, to 0.753 (0.729 to 0.777) and 0.778 (0.733 to 0.824) with increases of 0.074 (0.055 to 0.093; P = 4.11 × 10-14) and 0.073 (0.032 to 0.114; P = 4.00 × 10-4), respectively. CONCLUSIONS In this study, we identified 22 metabolites associated with longitudinal eGFR change, nine of which were PCs and six were DHA-containing lipids. We screened out a panel of eight metabolites which improved prediction for the development of CKD by 9 % beyond traditional risk factors including baseline eGFR. Our findings highlighted involvement of lipid metabolism in kidney function impairment, and provided novel predictors for CKD risk.
Collapse
Affiliation(s)
- Ling Liu
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hao Cai
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Handong Yang
- Department of Cardiovascular Disease, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan 442008, China
| | - Sihan Wang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yingmei Li
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yacan Huang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mingjing Gao
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaogang Zhang
- SClEX Application Support Center, Shanghai 200050, China
| | - Xiaomin Zhang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hao Wang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Gaokun Qiu
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
2
|
Wang RX, Zhou HB, Gao JX, Bai WF, Wang J, Bai YC, Jiang SY, Chang H, Shi SL. Metagenomics and metabolomics to investigate the effect of Amygdalus mongolica oil on intestinal microbiota and serum metabolites in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156335. [PMID: 39709798 DOI: 10.1016/j.phymed.2024.156335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Renal fibrosis (RF) is an inevitable consequence of multiple manifestations of progressive chronic kidney diseases (CKDs). Mechanism of Amygdalus mongolica (Maxim.) in the treatment of RF needs further investigation. PURPOSE The study further investigated the potential mechanism of A. mongolica in the treatment of RF. METHODS A rat model of RF was induced by unilateral ureteral obstruction (UUO), followed by treatment with varying dosages of A. mongolica oil for 4 weeks. Body weight was measured weekly. We detected serum levels of interleukin (IL)-6, IL-1β, type Ⅲ procollagen (Col-Ⅲ), type IV collagen (Col-Ⅳ), laminin (LN), hyaluronidase (HA), and tissue levels of albumin (ALB), blood urea nitrogen (BUN), creatinine (Cre), superoxide dismutase (SOD), malondialdehyde (MDA), and hydroxyproline (HYP). Shotgun metagenomics analyzed the composition of the intestinal microbiota. High-performance liquid chromatography coupled with a quadrupole-exactive mass spectrometer (HPLC-Q-Exactive-MS) monitored changes in metabolite levels in serum and gut. Multiple reaction monitoring-mass spectrometry (MRM-MS) determined the levels of amino acids in serum. RESULTS A. mongolica oil significantly alleviated indicators related to RF (p < 0.05). A. mongolica oil reduced the ratio of Firmicutes to Bacteroidetes and restored the balance of intestinal microbiota in rats with RF. A. mongolica oil modulated levels of metabolites in gut content and serum. It regulated 11 metabolic pathways including arachidonic acid metabolism. Targeted metabolomics of amino acids showed that 17 amino acids were significantly changed by A. mongolica oil, including L-glycine, L-serine and L-glutamine. CONCLUSION A. mongolica oil regulates intestinal microbiota and metabolites, restoring amino acid metabolism to treat RF.
Collapse
Affiliation(s)
- Run-Xi Wang
- Department of Pharmacy, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014040, China
| | - Hong-Bing Zhou
- Department of Pharmacy, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014040, China; Institute of Bioactive Substance and Function of Chinese Materia Medica and Mongolian Medicine, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014040, China
| | - Jia-Xing Gao
- Department of Pharmacy, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014040, China
| | - Wan-Fu Bai
- Department of Pharmacy, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014040, China
| | - Jia Wang
- Department of Pharmacy, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014040, China
| | - Ying-Chun Bai
- Department of Pharmacy, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014040, China
| | - Shu-Yuan Jiang
- Department of Pharmacy, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014040, China
| | - Hong Chang
- Department of Pharmacy, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014040, China; Institute of Bioactive Substance and Function of Chinese Materia Medica and Mongolian Medicine, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014040, China.
| | - Song-Li Shi
- Department of Pharmacy, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014040, China; Institute of Bioactive Substance and Function of Chinese Materia Medica and Mongolian Medicine, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014040, China.
| |
Collapse
|
3
|
Iwanek G, Ponikowska B, Salah H, Fudim M, Guzik M, Zymliński R, Aleksandrowicz K, Ponikowska B, Biegus J. A Tri-Component (Glomerular, Tubular, and Metabolic) Assessment of Renal Function in Acute Heart Failure. J Clin Med 2024; 13:7796. [PMID: 39768719 PMCID: PMC11727872 DOI: 10.3390/jcm13247796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/06/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Background: Despite the prevalence of impaired renal function in acute heart failure (AHF) patients, the intricate relationship between glomerular, tubular, and metabolic renal function remains unexplored. We aimed to investigate the co-occurrence of glomerular, tubular, and metabolic renal dysfunction in AHF and their impact on prognosis. Methods: eGFR, spot urine sodium, and HCO3- were measured in 243 patients hospitalized for AHF. The population was stratified by the 4-point renal dysfunction score and linked with outcomes. Results: Glomerular dysfunction exhibited an elevated risk of death (HR of 2.04; 95% CI [1.24-3.36]; p = 0.006), combined risk of death, and HF rehospitalization (HR of 2.03; 95% CI [1.34-3.05]; p = 0.005). Similarly, tubular dysfunction correlated with a higher death risk (HR of 1.72; 95% CI [1.04-2.82]; p = 0.03) and a higher combined risk (HR of 1.82; 95% CI [1.21-2.74]; p = 0.004). While renal metabolic dysfunction was linked to increased death risk (HR of 1.82; 95% CI [1.07-3.11]; p = 0.028), it was not associated with composite risk (HR of 1.37; 95% CI [0.88-2.15]; p = 0.174). Multivariate analysis revealed a direct association between the renal dysfunction score and death risk (HR of 1.92 per 1 point; 95% CI [1.47-2.52]; p < 0.0001) and the combined risk of death and HF rehospitalization (HR of 1.78 per 1 point; 95% CI [1.43-2.22]; p < 0.0001). Conclusions: Renal dysfunction is common, with varied overlaps. Glomerular, tubular, and metabolic dysfunctions predict adverse outcomes in AHF. The established renal score may aid patient stratification and prognosis.
Collapse
Affiliation(s)
- Gracjan Iwanek
- Institute of Heart Diseases, Wroclaw Medical University, 50-367 Wrocław, Poland; (M.G.); (R.Z.); (K.A.); (J.B.)
| | - Barbara Ponikowska
- Student Scientific Organization, Wroclaw Medical University, 50-367 Wrocław, Poland;
| | - Husam Salah
- Department of Medicine, Duke University, Durham, NC 27708, USA; (H.S.); (M.F.)
| | - Marat Fudim
- Department of Medicine, Duke University, Durham, NC 27708, USA; (H.S.); (M.F.)
- Duke Clinical Research Institute, Durham, NC 27701, USA
| | - Mateusz Guzik
- Institute of Heart Diseases, Wroclaw Medical University, 50-367 Wrocław, Poland; (M.G.); (R.Z.); (K.A.); (J.B.)
| | - Robert Zymliński
- Institute of Heart Diseases, Wroclaw Medical University, 50-367 Wrocław, Poland; (M.G.); (R.Z.); (K.A.); (J.B.)
| | - Krzysztof Aleksandrowicz
- Institute of Heart Diseases, Wroclaw Medical University, 50-367 Wrocław, Poland; (M.G.); (R.Z.); (K.A.); (J.B.)
| | - Beata Ponikowska
- Department of Physiology and Pathophysiology, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Jan Biegus
- Institute of Heart Diseases, Wroclaw Medical University, 50-367 Wrocław, Poland; (M.G.); (R.Z.); (K.A.); (J.B.)
| |
Collapse
|
4
|
Xu Z, Chen Y, Zeng D, Shi X, Zheng T, Zhang C, Feng X, Yan L, Zhao G, Jie H. Preliminary exploration of the musk biosynthetic mechanism by transcriptomic sequencing in muskrats. Sci Rep 2024; 14:29041. [PMID: 39580543 PMCID: PMC11585609 DOI: 10.1038/s41598-024-80080-3] [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: 03/26/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024] Open
Abstract
Musk, secreted by adult male forest musk deer, is a kind of precious Chinese traditional medicine for treating cardiovascular, cerebrovascular and neurogenic diseases. However, a lack of knowledge on musk biosynthetic mechanism and limited musk deer population have seriously hindered the development of the musk industry. Fortunately, given that muskrat musk has similar constituents and pharmacological action with deer musk, muskrat is an ideal model animal for exploring musk biosynthetic mechanism. To explore the biosynthetic mechanism of muskrat musk, in the current study, transcriptomic analysis in the liver, kidney and musk glands of male muskrats between musk secreting and non-musk secreting stages was conducted. The findings indicated that the role of muskrat liver on musk biosynthesis was altering sugar, lipid and amino acid metabolism as well as producing basic resources to support musk glands. Moreover, Tigar, Slc11a2, Gpt, Hmgcr, Slc27a4, and Elovl1 were identified as candidate genes for musk biosynthesis via a remotely controlled process. Expression of the Tigar, Slc11a2, and Gpt genes in the liver are downregulated to support the production of musk in muskrat musk gland. And the Hmgcr, Slc27a4, and Elovl1 genes in the musk gland participate in muskrat musk synthesis by influencing lipid metabolism in the musk secreting period. This study provided novel insights into the musk biosynthetic pathway in muskrat by transcriptomic analysis and preliminarily suggested the remote control of metabolism from the liver to musk gland during musk biosynthesis, which was useful to further understanding the musk biosynthetic process and improve musk production in the future.
Collapse
Affiliation(s)
- Zhongxian Xu
- Bio-Resource Research and Utilization Joint Key Laboratory of Sichuan and Chongqing, Chongqing Institute of Medicinal Plant Cultivation, Chongqing, China
- Sichuan Wildlife Rehabilitation and Breeding Research Center, Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong, China
| | - Yinglian Chen
- Bio-Resource Research and Utilization Joint Key Laboratory of Sichuan and Chongqing, Chongqing Institute of Medicinal Plant Cultivation, Chongqing, China
| | - Dejun Zeng
- Bio-Resource Research and Utilization Joint Key Laboratory of Sichuan and Chongqing, Chongqing Institute of Medicinal Plant Cultivation, Chongqing, China
| | - Xin Shi
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Tingting Zheng
- Sichuan Wildlife Rehabilitation and Breeding Research Center, Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong, China
| | - Chenglu Zhang
- Bio-Resource Research and Utilization Joint Key Laboratory of Sichuan and Chongqing, Chongqing Institute of Medicinal Plant Cultivation, Chongqing, China
| | - Xiaolan Feng
- Bio-Resource Research and Utilization Joint Key Laboratory of Sichuan and Chongqing, Chongqing Institute of Medicinal Plant Cultivation, Chongqing, China
| | - Linbo Yan
- Sichuan Wildlife Rehabilitation and Breeding Research Center, Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong, China
| | - Guijun Zhao
- Bio-Resource Research and Utilization Joint Key Laboratory of Sichuan and Chongqing, Chongqing Institute of Medicinal Plant Cultivation, Chongqing, China
| | - Hang Jie
- Bio-Resource Research and Utilization Joint Key Laboratory of Sichuan and Chongqing, Chongqing Institute of Medicinal Plant Cultivation, Chongqing, China.
| |
Collapse
|
5
|
Wang W, Dai R, Cheng M, Chen Y, Gao Y, Hong X, Zhang W, Wang Y, Zhang L. Metabolic reprogramming and renal fibrosis: what role might Chinese medicine play? Chin Med 2024; 19:148. [PMID: 39465434 PMCID: PMC11514863 DOI: 10.1186/s13020-024-01004-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/15/2024] [Indexed: 10/29/2024] Open
Abstract
Metabolic reprogramming is a pivotal biological process in which cellular metabolic patterns change to meet the energy demands of increased cell growth and proliferation. In this review, we explore metabolic reprogramming and its impact on fibrotic diseases, providing a detailed overview of the key processes involved in the metabolic reprogramming of renal fibrosis, including fatty acid decomposition and synthesis, glycolysis, and amino acid catabolism. In addition, we report that Chinese medicine ameliorates renal inflammation, oxidative stress, and apoptosis in chronic kidney disease by regulating metabolic processes, thereby inhibiting renal fibrosis. Furthermore, we reveal that multiple targets and signaling pathways contribute to the metabolic regulatory effects of Chinese medicine. In summary, this review aims to elucidate the mechanisms by which Chinese medicine inhibits renal fibrosis through the remodeling of renal cell metabolic processes, with the goal of discovering new therapeutic drugs for treating renal fibrosis.
Collapse
Affiliation(s)
- Weili Wang
- First Clinical Medical College, Anhui University of Chinese Medicine, Hefei, China
| | - Rong Dai
- Department of Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Meishan Road 117, Shushang District, Hefei, 230031, China
| | - Meng Cheng
- Department of Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Meishan Road 117, Shushang District, Hefei, 230031, China
| | - Yizhen Chen
- First Clinical Medical College, Anhui University of Chinese Medicine, Hefei, China
| | - Yilin Gao
- First Clinical Medical College, Anhui University of Chinese Medicine, Hefei, China
| | - Xin Hong
- First Clinical Medical College, Anhui University of Chinese Medicine, Hefei, China
| | - Wei Zhang
- First Clinical Medical College, Anhui University of Chinese Medicine, Hefei, China
| | - Yiping Wang
- Department of Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Meishan Road 117, Shushang District, Hefei, 230031, China.
| | - Lei Zhang
- Department of Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Meishan Road 117, Shushang District, Hefei, 230031, China.
| |
Collapse
|
6
|
Ahmadi A, Yu J, Loza JE, Howard BC, Palma I, Goussous N, Sageshima J, Roshanravan B, Perez RV. Deceased donor kidney function and branched chain amino acid metabolism during ex vivo normothermic perfusion. Kidney Int 2024; 106:712-722. [PMID: 39074554 DOI: 10.1016/j.kint.2024.06.026] [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: 11/27/2023] [Revised: 05/03/2024] [Accepted: 06/25/2024] [Indexed: 07/31/2024]
Abstract
Current kidney perfusion protocols are not optimized for addressing the ex vivo physiological and metabolic needs of the kidney. Ex vivo normothermic perfusion may be utilized to distinguish high-risk kidneys to determine suitability for transplantation. Here, we assessed the association of tissue metabolic changes with changes in a kidney injury biomarker and functional parameters in eight deceased donor kidneys deemed unsuitable for transplantation during a 12-hour ex vivo normothermic perfusion. The kidneys were grouped into good and poor performers based on blood flow and urine output. The mean age of the deceased kidney donors was 43 years with an average cold ischemia time of 37 hours. Urine output and creatinine clearance progressively increased and peaked at six hours post-perfusion among good performers. Poor performers had 71 ng/ml greater (95% confidence interval 1.5, 140) urinary neutrophil gelatinase-associated lipocalin at six hours compared to good performers corresponding to peak functional differences. Organ performance was distinguished by tissue metabolic differences in branched chain amino acid metabolism and that their tissue levels negatively correlated with urine output among all kidneys at six hours. Tissue lipid profiling showed poor performers were highlighted by the accumulation of membrane structure components including glycerolipids and sphingolipids at early perfusion time points. Thus, we showed that six hours is needed for kidney function recovery during ex vivo normothermic perfusion and that branched chain amino acid metabolism may be a major determinant of organ function and resilience.
Collapse
Affiliation(s)
- Armin Ahmadi
- Department of Medicine, Division of Nephrology, University of California, Davis, California, USA
| | - Jacquelyn Yu
- Department of Surgery, Division of Transplant, University of California Davis Health, Sacramento, California, USA
| | - Jennifer E Loza
- Department of Surgery, Division of Transplant, University of California Davis Health, Sacramento, California, USA
| | - Brian C Howard
- Department of Surgery, Division of Transplant, University of California Davis Health, Sacramento, California, USA
| | - Ivonne Palma
- Department of Surgery, Division of Transplant, University of California Davis Health, Sacramento, California, USA
| | - Naeem Goussous
- Department of Surgery, Division of Transplant, University of California Davis Health, Sacramento, California, USA
| | - Junichiro Sageshima
- Department of Surgery, Division of Transplant, University of California Davis Health, Sacramento, California, USA
| | - Baback Roshanravan
- Department of Medicine, Division of Nephrology, University of California, Davis, California, USA.
| | - Richard V Perez
- Department of Surgery, Division of Transplant, University of California Davis Health, Sacramento, California, USA.
| |
Collapse
|
7
|
Kuypers DRJ, Kamphorst JJ, de Loor H, O'Day EM. Perspective: metabolomics has the potential to change the landscape of kidney transplantation diagnostics. Biomark Med 2024; 18:787-794. [PMID: 39234983 PMCID: PMC11457662 DOI: 10.1080/17520363.2024.2394383] [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: 05/17/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024] Open
Abstract
Kidney transplantation is the most efficient renal replacement therapy. Current diagnostics for monitoring graft health are either invasive or lack precision. Metabolomics is an emerging discipline focused on the analysis of the small molecules involved in metabolism. Given the kidneys' central role in metabolic homeostasis and previous observations of altered metabolites correlating with restricted kidney graft function, metabolomics is highly promising for the discovery of novel biomarkers and the development of novel diagnostics. In this perspective, we summarize the known metabolic roles for the kidney, discuss biomarkers of graft health and immune status emerging from metabolomics research, and provide our perspective on how these and future findings can be integrated in clinical practice to enable precision diagnostics.
Collapse
Affiliation(s)
- Dirk R J Kuypers
- Department of Nephrology & Renal Transplantation, University Hospitals Leuven, Belgium
- Department of Microbiology, Immunology & Transplantation, Nephrology & Renal Transplantation Research Group, KU Leuven, Belgium
| | | | - Henriette de Loor
- Department of Nephrology & Renal Transplantation, University Hospitals Leuven, Belgium
| | | |
Collapse
|
8
|
Song G, Liu X, Lei K, Li T, Li W, Chen D. ExpoNano: A Strategy Based on Hyper-Cross-Linked Polymers Achieves Urinary Exposome Assessment for Biomonitoring. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024. [PMID: 39096285 DOI: 10.1021/acs.est.4c01146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/05/2024]
Abstract
Urinary analysis of exogenous and endogenous molecules constitutes an efficient, noninvasive approach to evaluate human health status. However, the exposome characterization of urinary molecules remains extremely challenging with current techniques. Herein, we develop an ExpoNano strategy based on hyper-cross-linked polymers (HCPs) to achieve ultrahigh-throughput measurement of exo/endogenous molecules in urine. The strategy includes a simple trapping-detrapping procedure (15 min) with HCPs in enzymatically treated urine, followed by mass spectrometer determination. Molecules that can be determined by ExpoNano have a wide range of molecular weight (75-837 Da) and Log Kow (octanol-water partition coefficient; -9.86 to 10.56). The HCPs can be repeatedly used five times without decreasing the trapping efficiency. Application of ExpoNano in a biomonitoring study revealed a total of 63 environmental chemicals detected in >50% of the urine pools collected from Chinese adults living in 13 cities, with a median concentration of 0.026-47 ng/mL, while nontargeted analysis detected an additional 243 exogenous molecules. Targeted and nontargeted analysis also detected 926 endogenous molecules in pooled urine. Collectively, the ExpoNano strategy demonstrates unique advantages over traditional urine analysis approaches, including a wide range of analytes, satisfactory trapping efficiency, high simplicity and reusability, and extremely reduced time demand and financial cost.
Collapse
Affiliation(s)
- Guixian Song
- College of Environment and Climate, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, China
| | - Xiaotu Liu
- College of Environment and Climate, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, China
| | - Kunxiang Lei
- College of Environment and Climate, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, China
| | - Tiantian Li
- China CDC Key Laboratory of Environment and Public Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Wanbin Li
- College of Environment and Climate, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, China
| | - Da Chen
- College of Environment and Climate, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, China
| |
Collapse
|
9
|
Sims-Lucas S, Goetzman ES, Kleyman TR. Cystic fibrosis-related metabolic defects: crosstalk between ion channels and organs. J Clin Invest 2024; 134:e182329. [PMID: 38949023 PMCID: PMC11213462 DOI: 10.1172/jci182329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024] Open
Abstract
Cystic fibrosis is a debilitating disease characterized by a poor medical prognosis due to devastating lung injury. Recent medical advances targeting the major genetic mutation ΔF508 of the cystic fibrosis transmembrane conductance regulator (CFTR) protein have dramatically increased the lifespan of patients with this mutation. This development has led to major changes in the field and has pushed research beyond the ion transport nature of cystic fibrosis and toward multiorgan physiological reprogramming. In this issue of the JCI, Bae, Kim, and colleagues utilized a large animal pig model prior to the onset of disease. They revealed metabolic reprogramming and organ crosstalk that occurred prior to disease progression. These findings provide paradigm-shifting insight into this complex disease.
Collapse
Affiliation(s)
| | | | - Thomas R. Kleyman
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
10
|
Dorrani M, Zhao J, Bekhti N, Trimigno A, Min S, Ha J, Han A, O’Day E, Kamphorst JJ. Olaris Global Panel (OGP): A Highly Accurate and Reproducible Triple Quadrupole Mass Spectrometry-Based Metabolomics Method for Clinical Biomarker Discovery. Metabolites 2024; 14:280. [PMID: 38786757 PMCID: PMC11123370 DOI: 10.3390/metabo14050280] [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: 04/11/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
Mass spectrometry (MS)-based clinical metabolomics is very promising for the discovery of new biomarkers and diagnostics. However, poor data accuracy and reproducibility limit its true potential, especially when performing data analysis across multiple sample sets. While high-resolution mass spectrometry has gained considerable popularity for discovery metabolomics, triple quadrupole (QqQ) instruments offer several benefits for the measurement of known metabolites in clinical samples. These benefits include high sensitivity and a wide dynamic range. Here, we present the Olaris Global Panel (OGP), a HILIC LC-QqQ MS method for the comprehensive analysis of ~250 metabolites from all major metabolic pathways in clinical samples. For the development of this method, multiple HILIC columns and mobile phase conditions were compared, the robustness of the leading LC method assessed, and MS acquisition settings optimized for optimal data quality. Next, the effect of U-13C metabolite yeast extract spike-ins was assessed based on data accuracy and precision. The use of these U-13C-metabolites as internal standards improved the goodness of fit to a linear calibration curve from r2 < 0.75 for raw data to >0.90 for most metabolites across the entire clinical concentration range of urine samples. Median within-batch CVs for all metabolite ratios to internal standards were consistently lower than 7% and less than 10% across batches that were acquired over a six-month period. Finally, the robustness of the OGP method, and its ability to identify biomarkers, was confirmed using a large sample set.
Collapse
Affiliation(s)
- Masoumeh Dorrani
- Olaris, Inc., 175 Crossing Boulevard Suite 410, Framingham, MA 01702, USA; (M.D.); (J.Z.); (N.B.); (A.T.); (E.O.)
| | - Jifang Zhao
- Olaris, Inc., 175 Crossing Boulevard Suite 410, Framingham, MA 01702, USA; (M.D.); (J.Z.); (N.B.); (A.T.); (E.O.)
| | - Nihel Bekhti
- Olaris, Inc., 175 Crossing Boulevard Suite 410, Framingham, MA 01702, USA; (M.D.); (J.Z.); (N.B.); (A.T.); (E.O.)
| | - Alessia Trimigno
- Olaris, Inc., 175 Crossing Boulevard Suite 410, Framingham, MA 01702, USA; (M.D.); (J.Z.); (N.B.); (A.T.); (E.O.)
| | - Sangil Min
- Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; (S.M.); (J.H.); (A.H.)
| | - Jongwon Ha
- Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; (S.M.); (J.H.); (A.H.)
| | - Ahram Han
- Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; (S.M.); (J.H.); (A.H.)
| | - Elizabeth O’Day
- Olaris, Inc., 175 Crossing Boulevard Suite 410, Framingham, MA 01702, USA; (M.D.); (J.Z.); (N.B.); (A.T.); (E.O.)
| | - Jurre J. Kamphorst
- Olaris, Inc., 175 Crossing Boulevard Suite 410, Framingham, MA 01702, USA; (M.D.); (J.Z.); (N.B.); (A.T.); (E.O.)
| |
Collapse
|
11
|
Pfeffer T, Krug SM, Kracke T, Schürfeld R, Colbatzky F, Kirschner P, Medert R, Freichel M, Schumacher D, Bartosova M, Zarogiannis SG, Muckenthaler MU, Altamura S, Pezer S, Volk N, Schwab C, Duensing S, Fleming T, Heidenreich E, Zschocke J, Hell R, Poschet G, Schmitt CP, Peters V. Knock-out of dipeptidase CN2 in human proximal tubular cells disrupts dipeptide and amino acid homeostasis and para- and transcellular solute transport. Acta Physiol (Oxf) 2024; 240:e14126. [PMID: 38517248 DOI: 10.1111/apha.14126] [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: 08/24/2023] [Revised: 02/15/2024] [Accepted: 02/21/2024] [Indexed: 03/23/2024]
Abstract
AIM Although of potential biomedical relevance, dipeptide metabolism has hardly been studied. We found the dipeptidase carnosinase-2 (CN2) to be abundant in human proximal tubules, which regulate water and solute homeostasis. We therefore hypothesized, that CN2 has a key metabolic role, impacting proximal tubular transport function. METHODS A knockout of the CN2 gene (CNDP2-KO) was generated in human proximal tubule cells and characterized by metabolomics, RNA-seq analysis, paracellular permeability analysis and ion transport. RESULTS CNDP2-KO in human proximal tubule cells resulted in the accumulation of cellular dipeptides, reduction of amino acids and imbalance of related metabolic pathways, and of energy supply. RNA-seq analyses indicated altered protein metabolism and ion transport. Detailed functional studies demonstrated lower CNDP2-KO cell viability and proliferation, and altered ion and macromolecule transport via trans- and paracellular pathways. Regulatory and transport protein abundance was disturbed, either as a consequence of the metabolic imbalance or the resulting functional disequilibrium. CONCLUSION CN2 function has a major impact on intracellular amino acid and dipeptide metabolism and is essential for key metabolic and regulatory functions of proximal tubular cells. These findings deserve in vivo analysis of the relevance of CN2 for nephron function and regulation of body homeostasis.
Collapse
Affiliation(s)
- Tilman Pfeffer
- Medical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
- Tissue Bank of the German Center for Infection Research (DZIF), Partner Site Heidelberg, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Susanne M Krug
- Clinical Physiology/Nutritional Medicine, Charité-Universitätsmedizin Berlin, CBF, Berlin, Germany
| | - Tamara Kracke
- Medical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| | - Robin Schürfeld
- Medical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| | - Florian Colbatzky
- Medical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| | - Philip Kirschner
- Medical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| | - Rebekka Medert
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Dagmar Schumacher
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Maria Bartosova
- Medical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| | - Sotiris G Zarogiannis
- Medical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| | - Martina U Muckenthaler
- Department of Pediatric Oncology, Hematology and Immunology and Hopp Children Cancer Center (KiTZ), University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), EMBL and University of Heidelberg, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Sandro Altamura
- Department of Pediatric Oncology, Hematology and Immunology and Hopp Children Cancer Center (KiTZ), University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), EMBL and University of Heidelberg, Heidelberg, Germany
| | - Silvia Pezer
- Medical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| | - Nadine Volk
- Tissue Bank of the National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Constantin Schwab
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Duensing
- Department of Urology, University Hospital Heidelberg and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Thomas Fleming
- Internal Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
| | - Elena Heidenreich
- Centre for Organismal Studies (COS), University of Heidelberg, Heidelberg, Germany
| | - Johannes Zschocke
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Rüdiger Hell
- Centre for Organismal Studies (COS), University of Heidelberg, Heidelberg, Germany
| | - Gernot Poschet
- Centre for Organismal Studies (COS), University of Heidelberg, Heidelberg, Germany
| | - Claus P Schmitt
- Medical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| | - Verena Peters
- Medical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
12
|
Liang B, Shi X, Wang X, Ma C, Leslie WD, Lix LM, Shi X, Kan B, Yang S. Association between amino acids and recent osteoporotic fracture: a matched incident case-control study. Front Nutr 2024; 11:1360959. [PMID: 38567247 PMCID: PMC10985241 DOI: 10.3389/fnut.2024.1360959] [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: 12/24/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Context Osteoporotic fracture is a major public health issue globally. Human research on the association between amino acids (AAs) and fracture is still lacking. Objective To examine the association between AAs and recent osteoporotic fractures. Methods This age and sex matched incident case-control study identified 44 recent x-ray confirmed fracture cases in the Second Hospital of Jilin University and 88 community-based healthy controls aged 50+ years. Plasma AAs were measured by high performance liquid chromatography coupled with mass spectrometry. After adjusting for covariates (i.e., body mass index, milk intake >1 time/week, falls and physical activity), we conducted conditional logistical regression models to test the association between AAs and fracture. Results Among cases there were 23 (52.3%) hip fractures and 21 (47.7%) non-hip fractures. Total, essential, and non-essential AAs were significantly lower in cases than in controls. In the multivariable conditional logistic regression models, after adjusting for covariates, each standard deviation increase in the total (odds ratio [OR]: 0.304; 95% confidence interval [CI]: 0.117-0.794), essential (OR: 0.408; 95% CI: 0.181-0.923) and non-essential AAs (OR: 0.290; 95%CI: 0.107-0.782) was negatively associated with recent fracture. These inverse associations were mainly found for hip fracture, rather than non-hip fractures. Among these AAs, lysine, alanine, arginine, glutamine, histidine and piperamide showed the significantly negative associations with fracture. Conclusion There was a negative relationship between AAs and recent osteoporotic fracture; such relationship appeared to be more obvious for hip fracture.
Collapse
Affiliation(s)
- Bing Liang
- Department of Endocrinology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiao tong University, Xi’an, China
| | - Xinyan Shi
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Xinwei Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Chao Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - William D. Leslie
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Lisa M. Lix
- Department of Community Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Bo Kan
- Department of Clinical Laboratory, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Shuman Yang
- Department of Endocrinology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| |
Collapse
|
13
|
Jia PP, Li Y, Zhang LC, Wu MF, Li TY, Pei DS. Metabolome evidence of CKDu risks after chronic exposure to simulated Sri Lanka drinking water in zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 273:116149. [PMID: 38412632 DOI: 10.1016/j.ecoenv.2024.116149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/10/2024] [Accepted: 02/22/2024] [Indexed: 02/29/2024]
Abstract
It is still a serious public health issue that chronic kidney disease of uncertain etiology (CKDu) in Sri Lanka poses challenges in identification, prevention, and treatment. What environmental factors in drinking water cause kidney damage remains unclear. This study aimed to investigate the risks of various environmental factors that may induce CKDu, including water hardness, fluoride (HF), heavy metals (HM), microcystin-LR (MC-LR), and their combined exposure (HFMM). The research focused on comprehensive metabolome analysis, and correlation with transcriptomic and gut microbiota changes. Results revealed that chronic exposure led to kidney damage and pancreatic toxicity in adult zebrafish. Metabolomics profiling showed significant alterations in biochemical processes, with enriched metabolic pathways of oxidative phosphorylation, folate biosynthesis, arachidonic acid metabolism, FoxO signaling pathway, lysosome, pyruvate metabolism, and purine metabolism. The network analysis revealed significant changes in metabolites associated with renal function and diseases, including 20-Hydroxy-LTE4, PS(18:0/22:2(13Z,16Z)), Neuromedin N, 20-Oxo-Leukotriene E4, and phenol sulfate, which are involved in the fatty acyls and glycerophospholipids class. These metabolites were closely associated with the disrupted gut bacteria of g_ZOR0006, g_Pseudomonas, g_Tsukamurella, g_Cetobacterium, g_Flavobacterium, which belonged to dominant phyla of Firmicutes and Proteobacteria, etc., and differentially expressed genes (DEGs) such as egln3, ca2, jun, slc2a1b, and gls2b in zebrafish. Exploratory omics analyses revealed the shared significantly changed pathways in transcriptome and metabolome like calcium signaling and necroptosis, suggesting potential biomarkers for assessing kidney disease.
Collapse
Affiliation(s)
- Pan-Pan Jia
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Yan Li
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Lan-Chen Zhang
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Ming-Fei Wu
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Tian-Yun Li
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - De-Sheng Pei
- School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
14
|
Wang MY, Zhang Z, Zhao S, Onodera T, Sun XN, Zhu Q, Li C, Li N, Chen S, Paredes M, Gautron L, Charron MJ, Marciano DK, Gordillo R, Drucker DJ, Scherer PE. Downregulation of the kidney glucagon receptor, essential for renal function and systemic homeostasis, contributes to chronic kidney disease. Cell Metab 2024; 36:575-597.e7. [PMID: 38237602 PMCID: PMC10932880 DOI: 10.1016/j.cmet.2023.12.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 09/10/2023] [Accepted: 12/19/2023] [Indexed: 02/12/2024]
Abstract
The glucagon receptor (GCGR) in the kidney is expressed in nephron tubules. In humans and animal models with chronic kidney disease, renal GCGR expression is reduced. However, the role of kidney GCGR in normal renal function and in disease development has not been addressed. Here, we examined its role by analyzing mice with constitutive or conditional kidney-specific loss of the Gcgr. Adult renal Gcgr knockout mice exhibit metabolic dysregulation and a functional impairment of the kidneys. These mice exhibit hyperaminoacidemia associated with reduced kidney glucose output, oxidative stress, enhanced inflammasome activity, and excess lipid accumulation in the kidney. Upon a lipid challenge, they display maladaptive responses with acute hypertriglyceridemia and chronic proinflammatory and profibrotic activation. In aged mice, kidney Gcgr ablation elicits widespread renal deposition of collagen and fibronectin, indicative of fibrosis. Taken together, our findings demonstrate an essential role of the renal GCGR in normal kidney metabolic and homeostatic functions. Importantly, mice deficient for kidney Gcgr recapitulate some of the key pathophysiological features of chronic kidney disease.
Collapse
Affiliation(s)
- May-Yun Wang
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhuzhen Zhang
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shangang Zhao
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Sam and Ann Barshop Institute for Longevity and Aging Studies, Division of Endocrinology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Toshiharu Onodera
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xue-Nan Sun
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Qingzhang Zhu
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chao Li
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Na Li
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shiuhwei Chen
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Megan Paredes
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Laurent Gautron
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Maureen J Charron
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Denise K Marciano
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ruth Gordillo
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daniel J Drucker
- Lunenfeld-TanenbaumResearchInstitute, Mt. Sinai Hospital, Toronto, ON M5G1X5, Canada; Department of Medicine, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
15
|
Hu Y, Jiang W. Mannose and glycine: Metabolites with potentially causal implications in chronic kidney disease pathogenesis. PLoS One 2024; 19:e0298729. [PMID: 38354117 PMCID: PMC10866514 DOI: 10.1371/journal.pone.0298729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/29/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Chronic Kidney Disease (CKD) represents a global health challenge, with its etiology and underlying mechanisms yet to be fully elucidated. Integrating genomics with metabolomics can offer insights into the putatively causal relationships between serum metabolites and CKD. METHODS Utilizing bidirectional Mendelian Randomization (MR), we assessed the putatively causal associations between 486 serum metabolites and CKD. Genetic data for these metabolites were sourced from comprehensive genome-wide association studies, and CKD data were obtained from the CKDGen Consortium. RESULTS Our analysis identified four metabolites with a robust association with CKD risk, of which mannose and glycine showed the most reliable causal relationships. Pathway analysis spotlighted five significant metabolic pathways, notably including "Methionine Metabolism" and "Arginine and Proline Metabolism", as key contributors to CKD pathogenesis. CONCLUSION This study underscores the potential of certain serum metabolites as biomarkers for CKD and illuminates pivotal metabolic pathways in CKD's pathogenesis. Our findings lay the groundwork for potential therapeutic interventions and warrant further research for validation.
Collapse
Affiliation(s)
- Yongzheng Hu
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wei Jiang
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
16
|
Li T, Ihanus A, Ohukainen P, Järvelin MR, Kähönen M, Kettunen J, Raitakari OT, Lehtimäki T, Mäkinen VP, Tynkkynen T, Ala-Korpela M. Clinical and biochemical associations of urinary metabolites: quantitative epidemiological approach on renal-cardiometabolic biomarkers. Int J Epidemiol 2024; 53:dyad162. [PMID: 38030573 PMCID: PMC10859141 DOI: 10.1093/ije/dyad162] [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: 09/04/2022] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Urinary metabolomics has demonstrated considerable potential to assess kidney function and its metabolic corollaries in health and disease. However, applications in epidemiology remain sparse due to technical challenges. METHODS We added 17 metabolites to an open-access urinary nuclear magnetic resonance metabolomics platform, extending the panel to 61 metabolites (n = 994). We also introduced automated quantification for 11 metabolites, extending the panel to 12 metabolites (+creatinine). Epidemiological associations between these 12 metabolites and 49 clinical measures were studied in three independent cohorts (up to 5989 participants). Detailed regression analyses with various confounding factors are presented for body mass index (BMI) and smoking. RESULTS Sex-specific population reference concentrations and distributions are provided for 61 urinary metabolites (419 men and 575 women), together with methodological intra-assay metabolite variations as well as the biological intra-individual and epidemiological population variations. For the 12 metabolites, 362 associations were found. These are mostly novel and reflect potential molecular proxies to estimate kidney function, as the associations cannot be simply explained by estimated glomerular filtration rate. Unspecific renal excretion results in leakage of amino acids (and glucose) to urine in all individuals. Seven urinary metabolites associated with smoking, providing questionnaire-independent proxy measures of smoking status in epidemiological studies. Common confounders did not affect metabolite associations with smoking, but insulin had a clear effect on most associations with BMI, including strong effects on 2-hydroxyisobutyrate, valine, alanine, trigonelline and hippurate. CONCLUSIONS Urinary metabolomics provides new insight on kidney function and related biomarkers on the renal-cardiometabolic system, supporting large-scale applications in epidemiology.
Collapse
Affiliation(s)
- Tianqi Li
- Systems Epidemiology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Andrei Ihanus
- Systems Epidemiology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Pauli Ohukainen
- Systems Epidemiology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Marjo-Riitta Järvelin
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, and Finnish Cardiovascular Research Center Tampere, Tampere University, Tampere, Finland
| | - Johannes Kettunen
- Systems Epidemiology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center Tampere, Tampere University, Tampere, Finland
| | - Ville-Petteri Mäkinen
- Systems Epidemiology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Tuulia Tynkkynen
- Systems Epidemiology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Mika Ala-Korpela
- Systems Epidemiology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
17
|
Zhang Q, Yin R, Guan G, Liu H, Song G. Renal clearable magnetic nanoparticles for magnetic resonance imaging and guided therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1929. [PMID: 37752407 DOI: 10.1002/wnan.1929] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/10/2023] [Accepted: 08/19/2023] [Indexed: 09/28/2023]
Abstract
Magnetic resonance imaging (MRI) is a non-invasive, radiation-free imaging technique widely used for disease detection and therapeutic evaluation due to its infinite penetration depth. Magnetic nanoparticles (MNPs) have unique magnetic and physicochemical properties, making them ideal as contrast agents for MRI. However, the in vivo toxicity of MNPs, resulting from metal ion leakage and long-term accumulation in the reticuloendothelial system (RES), limits their clinical application. To overcome these challenges, there is considerable interest in the development of renal-clearable MNPs that can be completely cleared through the kidney, reducing retention time and potential toxic risks. In this review, we provide an overview of recent advancements in the development of renal-clearable MNPs for disease imaging and treatment. We discuss the factors influencing renal clearance, summarize the types of renal-clearable MNPs, their synthesis methods, and biomedical applications. This review aims to offer comprehensive information for the design and clinical translation of renal-clearable MNPs. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Diagnostic Tools > Biosensing.
Collapse
Affiliation(s)
- Qinpeng Zhang
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan, China
| | - Rui Yin
- College of Chemistry, Xiangtan University, Xiangtan, Hunan, China
| | - Guoqiang Guan
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan, China
| | - Huiyi Liu
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan, China
| | - Guosheng Song
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan, China
| |
Collapse
|
18
|
Park S, Jin HS. Association of IGF1R polymorphisms with kidney function-related traits. Genes Genomics 2023; 45:1509-1515. [PMID: 37477779 DOI: 10.1007/s13258-023-01430-5] [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: 08/29/2022] [Accepted: 07/11/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Unimpaired kidney function is important for maintaining a healthy state of homeostasis in the body. A genome-wide association study (GWAS) conducted on kidney function-associated traits and novel loci of Japanese subjects found that insulin-like-growth factor 1 receptor (IGF1R) gene variants associated with replication were responsible for all three kinds of kidney functions and satisfied the Bonferroni significance level. OBJECTIVE This study aimed to investigate whether a comparable relationship exists in the Korean population. METHOD This study replicated three SNPs (rs28657002, rs10794486, and rs4966025) and conducted a linear regression analysis between 46 SNPs in the IGF1R gene and three kidney function-related traits, namely blood urea nitrogen (BUN), creatinine, and estimated glomerular filtration rate (eGFR) in Koreans. RESULTS The IGF1R gene was found to be replicated with a significant correlation in Koreans and was extended to the entire gene region to confirm its association with kidney-related functions. Three SNPs in IGF1R were replicated (rs28657002, BUN, P = 3.39 × 10-4; rs10794486, creatinine, P = 5.79 × 10-6; rs4966025, eGFR, P = 1.57 × 10-5) and five SNPs (rs28657002, rs10794486, rs4966025, rs12439557, and rs11247372) showed common significance among the three traits. Additionally, two significant SNPs (rs11857366 and rs28657002) showed the potential to affect IGF1R expression. CONCLUSIONS The results suggest that genetic polymorphisms in the IGF1R replicated previous studies and could affect kidney function. The results of this study will further enhance our understanding of how genetic differences in individuals affect kidney function-related traits.
Collapse
Affiliation(s)
- Sangjung Park
- Department of Biomedical Laboratory Science, College of Life and Health Sciences, Hoseo University, 20 Hoseo-Ro 79 Beon-Gil, Asan-Si, Chungcheongnam-Do, 31499, Republic of Korea
| | - Hyun-Seok Jin
- Department of Biomedical Laboratory Science, College of Life and Health Sciences, Hoseo University, 20 Hoseo-Ro 79 Beon-Gil, Asan-Si, Chungcheongnam-Do, 31499, Republic of Korea.
| |
Collapse
|
19
|
Xu B, Li W, Zhang Y, Chen Y, Feng J, Song X. Untargeted and spatial-resolved metabolomics characterize serum and tissue-specific metabolic reprogramming in acute kidney injury. Heliyon 2023; 9:e21171. [PMID: 38027662 PMCID: PMC10660029 DOI: 10.1016/j.heliyon.2023.e21171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Background Acute kidney injury (AKI) is one of the most common clinical emergencies characterized by rapid progression, difficulty in early diagnosis, and high mortality. Currently, there are no effective AKI early diagnostic methods and treatments. Therefore, identifying new mechanisms of AKI have become urgent for development new targets for early diagnosis and treatment of AKI in the current clinical setting. Methods In this study, systematic analysis and comparison of serum metabolic profiles of clinical AKI patients, chronic kidney disease (CKD) patients, and healthy subjects were performed using untargeted metabolomics. Moreover, the first spatial metabolomic analysis of kidney tissues in an AKI mouse model using MALDI-TOF MS technology was conducted. Differentially expressed metabolites were identified using a comprehensive, publicly available database. The metabolic data obtained were evaluated using principal component analysis, (orthogonal) partial least squares discriminant analysis, and metabolic pathway analysis to explore the unique serum metabolic profile of the patients, as well as to characterize the spatial distribution of differential metabolites in the kidneys of AKI mice. Results Significant changes in the metabolite levels of amino acids, carnitine, and lipids were observed in the AKI and CKD groups versus the healthy population, suggesting that kidney injury may lead to abnormalities in various metabolic pathways, such as amino acids, fatty acids, and lipids. The significant difference between the AKI and CKD groups were found for the first time in these indexes including amino acid, carnitine, fatty acid, and lipid levels. Additionally, spatial metabolomics results revealed that amino acid, carnitine, organic acid, and fatty acid metabolites were more likely significantly altered in the renal cortex, while lipid metabolites were both differentially distributed in the cortex and medulla of the AKI group. Conclusion Abnormalities in the serum metabolism of amino acids, carnitine, and lipids in patients with kidney diseases, such as AKI and CKD, are closely associated with the physiological dysfunction of kidney injury. Metabolic differences between patients with AKI and CKD were compared for the first time, showing that fatty acid oxidative inhibition was more severe in patients with AKI. Furthermore, spatial metabolomics has revealed metabolic reprogramming with tissue heterogeneity in AKI mice model. Our study provides valuable information in the molecular pathological features of AKI in the kidney tissues.
Collapse
Affiliation(s)
- Bei Xu
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Wanyi Li
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Yamei Zhang
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Yan Chen
- Department of Clinical Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiafu Feng
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Xiangrong Song
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
20
|
Hissen KL, He W, Wu G, Criscitiello MF. Immunonutrition: facilitating mucosal immune response in teleost intestine with amino acids through oxidant-antioxidant balance. Front Immunol 2023; 14:1241615. [PMID: 37841275 PMCID: PMC10570457 DOI: 10.3389/fimmu.2023.1241615] [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: 06/19/2023] [Accepted: 08/24/2023] [Indexed: 10/17/2023] Open
Abstract
Comparative animal models generate fundamental scientific knowledge of immune responses. However, these studies typically are conducted in mammals because of their biochemical and physiological similarity to humans. Presently, there has been an interest in using teleost fish models to study intestinal immunology, particularly intestinal mucosa immune response. Instead of targeting the pathogen itself, a preferred approach for managing fish health is through nutrient supplementation, as it is noninvasive and less labor intensive than vaccine administrations while still modulating immune properties. Amino acids (AAs) regulate metabolic processes, oxidant-antioxidant balance, and physiological requirements to improve immune response. Thus, nutritionists can develop sustainable aquafeeds through AA supplementation to promote specific immune responses, including the intestinal mucosa immune system. We propose the use of dietary supplementation with functional AAs to improve immune response by discussing teleost fish immunology within the intestine and explore how oxidative burst is used as an immune defense mechanism. We evaluate immune components and immune responses in the intestine that use oxidant-antioxidant balance through potential selection of AAs and their metabolites to improve mucosal immune capacity and gut integrity. AAs are effective modulators of teleost gut immunity through oxidant-antioxidant balance. To incorporate nutrition as an immunoregulatory means in teleost, we must obtain more tools including genomic, proteomic, nutrition, immunology, and macrobiotic and metabonomic analyses, so that future studies can provide a more holistic understanding of the mucosal immune system in fish.
Collapse
Affiliation(s)
- Karina L. Hissen
- Comparative Immunogenetics Laboratory Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| | - Wenliang He
- Amino Acid Laboratory, Department of Animal Science, Texas A&M University, College Station, TX, United States
| | - Guoyao Wu
- Amino Acid Laboratory, Department of Animal Science, Texas A&M University, College Station, TX, United States
| | - Michael F. Criscitiello
- Comparative Immunogenetics Laboratory Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
21
|
Xian L, Tian J, Long Y, Ma H, Tian M, Liu X, Yin G, Wang L. Metabolomics and transcriptomics analyses provide new insights into the nutritional quality during the endosperm development of different ploidy rice. FRONTIERS IN PLANT SCIENCE 2023; 14:1210134. [PMID: 37409294 PMCID: PMC10319422 DOI: 10.3389/fpls.2023.1210134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 05/30/2023] [Indexed: 07/07/2023]
Abstract
Autotetraploid rice is developed from diploid rice by doubling the chromosomes, leading to higher nutritional quality. Nevertheless, there is little information about the abundances of different metabolites and their changes during endosperm development in autotetraploid rice. In this research, two different kinds of rice, autotetraploid rice (AJNT-4x) and diploid rice (AJNT-2x), were subjected to experiments at various time points during endosperm development. A total of 422 differential metabolites, were identified by applying a widely used metabolomics technique based on LC-MS/MS. KEGG classification and enrichment analysis showed the differences in metabolites were primarily related to biosynthesis of secondary metabolites, microbial metabolism in diverse environments, biosynthesis of cofactors, and so on. Twenty common differential metabolites were found at three developmental stages of 10, 15 and 20 DAFs, which were considered the key metabolites. To identify the regulatory genes of metabolites, the experimental material was subjected to transcriptome sequencing. The DEGs were mainly enriched in starch and sucrose metabolism at 10 DAF, and in ribosome and biosynthesis of amino acids at 15 DAF, and in biosynthesis of secondary metabolites at 20 DAF. The numbers of enriched pathways and the DEGs gradually increased with endosperm development of rice. The related metabolic pathways of rice nutritional quality are cysteine and methionine metabolism, tryptophan metabolism, lysine biosynthesis and histidine metabolism, and so on. The expression level of the genes regulating lysine content was higher in AJNT-4x than in AJNT-2x. By applying CRISPR/Cas9 gene-editing technology, we identified two novel genes, OsLC4 and OsLC3, negatively regulated lysine content. These findings offer novel insight into dynamic metabolites and genes expression variations during endosperm development of different ploidy rice, which will aid in the creation of rice varieties with better grain nutritional quality.
Collapse
Affiliation(s)
- Lin Xian
- College of Agriculture, South China Agricultural University, Guangzhou, China
- Guizhou Academy of Tobacco Science, Guiyang, China
- State Key Laboratory of Agricultural Genomics, BGI-Shenzhen, Shenzhen, China
| | - Jiaqi Tian
- College of Agriculture, South China Agricultural University, Guangzhou, China
| | - Yanxi Long
- College of Agriculture, South China Agricultural University, Guangzhou, China
| | - Huijin Ma
- College of Agriculture, South China Agricultural University, Guangzhou, China
| | - Min Tian
- College of Agriculture, South China Agricultural University, Guangzhou, China
| | - Xiangdong Liu
- College of Agriculture, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Plant Molecular Breeding, College of Agriculture, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Guoying Yin
- College of Agriculture, South China Agricultural University, Guangzhou, China
- Guizhou Academy of Tobacco Science, Guiyang, China
| | - Lan Wang
- College of Agriculture, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Plant Molecular Breeding, College of Agriculture, South China Agricultural University, Guangzhou, China
| |
Collapse
|
22
|
Cuevas-Delgado P, Miguel V, Rupérez FJ, Lamas S, Barbas C. Impact of renal tubular Cpt1a overexpression on the kidney metabolome in the folic acid-induced fibrosis mouse model. Front Mol Biosci 2023; 10:1161036. [PMID: 37377862 PMCID: PMC10291237 DOI: 10.3389/fmolb.2023.1161036] [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: 02/07/2023] [Accepted: 04/24/2023] [Indexed: 06/29/2023] Open
Abstract
Background: Chronic kidney disease (CKD) is characterized by the progressive and irreversible deterioration of kidney function and structure with the appearance of renal fibrosis. A significant decrease in mitochondrial metabolism, specifically a reduction in fatty acid oxidation (FAO) in tubular cells, is observed in tubulointerstitial fibrosis, whereas FAO enhancement provides protection. Untargeted metabolomics offers the potential to provide a comprehensive analysis of the renal metabolome in the context of kidney injury. Methodology: Renal tissue from a carnitine palmitoyl transferase 1a (Cpt1a) overexpressing mouse model, which displays enhanced FAO in the renal tubule, subjected to folic acid nephropathy (FAN) was studied through a multiplatform untargeted metabolomics approach based on LC-MS, CE-MS and GC-MS analysis to achieve the highest coverage of the metabolome and lipidome affected by fibrosis. The expression of genes related to the biochemical routes showing significant changes was also evaluated. Results: By combining different tools for signal processing, statistical analysis and feature annotation, we were able to identify variations in 194 metabolites and lipids involved in many metabolic routes: TCA cycle, polyamines, one-carbon metabolism, amino acid metabolism, purine metabolism, FAO, glycerolipids and glycerophospholipids synthesis and degradation, glycosphingolipids interconversion, and sterol metabolism. We found several metabolites strongly altered by FAN, with no reversion induced by Cpt1a overexpression (v.g. citric acid), whereas other metabolites were influenced by CPT1A-induced FAO (v.g. glycine-betaine). Conclusion: It was implemented a successful multiplatform metabolomics approach for renal tissue analysis. Profound metabolic changes accompany CKD-associated fibrosis, some associated with tubular FAO failure. These results highlight the importance of addressing the crosstalk between metabolism and fibrosis when undertaking studies attempting to elucidate the mechanism of CKD progression.
Collapse
Affiliation(s)
- Paula Cuevas-Delgado
- Centre for Metabolomics and Bioanalysis (CEMBIO), School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Madrid, Spain
| | - Verónica Miguel
- Program of Physiological and Pathological Processes, Centro de Biología Molecular “Severo Ochoa” (CBMSO, CSIC-UAM), Madrid, Spain
| | - Francisco J. Rupérez
- Centre for Metabolomics and Bioanalysis (CEMBIO), School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Madrid, Spain
| | - Santiago Lamas
- Program of Physiological and Pathological Processes, Centro de Biología Molecular “Severo Ochoa” (CBMSO, CSIC-UAM), Madrid, Spain
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Madrid, Spain
| |
Collapse
|
23
|
Zhou Z, Feng D, Shi D, Gao P, Wang L, Wu Z. Untargeted and targeted metabolomics reveal bile acid profile changes in rats with ethylene glycol-induced calcium oxalate nephrolithiasis. Chem Biol Interact 2023; 381:110570. [PMID: 37244400 DOI: 10.1016/j.cbi.2023.110570] [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: 04/03/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023]
Abstract
Calcium oxalate (CaOx) nephrolithiasis is a prevalent disorder linked to metabolism. Examining metabolic alterations could potentially give an initial understanding of the origins of CaOx nephrolithiasis. This study aims to determine gut metabolic biomarkers differentiating CaOx nephrolithiasis utilizing untargeted and targeted metabolomics. CaOx nephrolithiasis model rats were built by 1% ethylene glycol administration. Histologic staining and renal function measurement revealed the presence of crystals in the lumen of the renal tubules, the renal injury and interstitial fibrosis in CaOx rats, demonstrating that the models of CaOx were established successfully. Hematoxylin & eosin (H&E) staining showed that CaOx group had inflammation and damage in the ileal tissue. Immunofluorescence and PCR results displayed that the tight junction proteins, ZO-1 and Occludin levels were decreased in the ileal tissues of the CaOx group. The untargeted metabolomic analysis revealed that 269 gut metabolites were differentially expressed between the CaOx group and the control group. Meanwhile, bile secretion, the main metabolic pathway in CaOx nephrolithiasis, was identified. Following, five significant bile acid metabolites were selected utilizing the targeted bile acid metabolomics, including Hyodeoxycholic acid (HDCA), Glycohyodeoxycholic acid (GHDCA), Nor-Deoxycholic Acid, omega-muricholic acid, and Taurolithocholic acid. Among these metabolites, HDCA and GHDCA presented the highest predictive accuracy with AUC = 1 to distinguish the CaOx group from the control group. As a result of network pharmacology, target genes of HDCA and GHDCA in CaOx nephrolithiasis were enriched in oxidative stress and apoptosis pathways. Conclusively, our study provides insight into bile acids metabolic changes related to CaOx nephrolithiasis. Although alterations in biochemical pathways indicate a complex pathology in CaOx rats, bile acid changes may serve as biomarkers of CaOx nephrolithiasis.
Collapse
Affiliation(s)
- Zijian Zhou
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
| | - Dexiang Feng
- Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, 215123, PR China
| | - Donghui Shi
- Department of Urology, Suzhou Wu Zhong People's Hospital, Suzhou, 215100, PR China
| | - Peng Gao
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040, PR China; Clinical Research Center of Urolithiasis, Shanghai Medical College, Fudan University, Shanghai, 200040, PR China
| | - Lujia Wang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040, PR China; Clinical Research Center of Urolithiasis, Shanghai Medical College, Fudan University, Shanghai, 200040, PR China.
| | - Zhong Wu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040, PR China; Clinical Research Center of Urolithiasis, Shanghai Medical College, Fudan University, Shanghai, 200040, PR China.
| |
Collapse
|
24
|
Yang Y, Li T, Jing W, Yan Z, Li X, Fu W, Zhang R. Dual-modality and Noninvasive Diagnostic of MNP-PEG-Mn Nanoprobe for Renal Fibrosis Based on Photoacoustic and Magnetic Resonance Imaging. ACS APPLIED MATERIALS & INTERFACES 2023; 15:12797-12808. [PMID: 36866785 DOI: 10.1021/acsami.2c22512] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
To date, imaging-guided multimodality therapy is important to improve the accuracy of the diagnosis of renal fibrosis, and nanoplatforms for imaging-guided multimodality diagnosis are gaining more and more attention. There are many limitations and deficiencies in clinical use for early-stage diagnosis of renal fibrosis, and multimodal imaging can contribute more thoroughly and provide in-detail information for effective clinical diagnosis. Melanin is an endogenous biomaterial, and we developed an ultrasmall particle size melanin nanoprobe (MNP-PEG-Mn) based on photoacoustic (PA) and magnetic resonance (MR) dual-modal imaging. MNP-PEG-Mn nanoprobe, with the average diameter about 2.7 nm, can be passively targeted for accumulation in the kidney, and it has excellent free radical scavenging and antioxidant abilities without further exacerbating renal fibrosis. Using the normal group signal as a control, the dual-modal imaging results showed that the MR imaging (MAI) and PA imaging (PAI) signals reached the strongest at 6 h when MNP-PEG-Mn entered the 7 day renal fibrosis group via the left vein of the tail end of the mice; however, the strength of the dual-modal imaging signal and the gradient of signal change were significantly weaker in the 28 day renal fibrosis group than in the 7 day renal fibrosis group and normal group. The phenomenon preliminarily indicates that as a PAI/MRI dual-modality contrast medium candidate, MNP-PEG-Mn has outstanding ability in clinical application potential.
Collapse
Affiliation(s)
- Yilin Yang
- Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Tingting Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Wenyu Jing
- Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Zirui Yan
- Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Xueqi Li
- Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Weihua Fu
- Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Ruiping Zhang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
25
|
Identification of AGXT2, SHMT1, and ACO2 as important biomarkers of acute kidney injury by WGCNA. PLoS One 2023; 18:e0281439. [PMID: 36735737 PMCID: PMC9897545 DOI: 10.1371/journal.pone.0281439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/23/2023] [Indexed: 02/04/2023] Open
Abstract
Acute kidney injury (AKI) is a serious and frequently observed disease associated with high morbidity and mortality. Weighted gene co-expression network analysis (WGCNA) is a research method that converts the relationship between tens of thousands of genes and phenotypes into the association between several gene sets and phenotypes. We screened potential target genes related to AKI through WGCNA to provide a reference for the diagnosis and treatment of AKI. Key biomolecules of AKI were investigated based on transcriptome analysis. RNA sequencing data from 39 kidney biopsy specimens of AKI patients and 9 normal subjects were downloaded from the GEO database. By WGCNA, the top 20% of mRNAs with the largest variance in the data matrix were used to construct a gene co-expression network with a p-value < 0.01 as a screening condition, showing that the blue module was most closely associated with AKI. Thirty-two candidate biomarker genes were screened according to the threshold values of |MM|≥0.86 and |GS|≥0.4, and PPI and enrichment analyses were performed. The top three genes with the most connected nodes, alanine-glyoxylate aminotransferase 2(AGXT2), serine hydroxymethyltransferase 1(SHMT1) and aconitase 2(ACO2), were selected as the central genes based on the PPI network. A rat AKI model was constructed, and the mRNA and protein expression levels of the central genes in the model and control groups were verified by PCR and immunohistochemistry experiments. The results showed that the relative mRNA expression and protein levels of AGXT2, SHMT1 and ACO2 showed a decrease in the model group. In conclusion, we inferred that there is a close association between AGXT2, SHMT1 and ACO2 genes and the development of AKI, and the down-regulation of their expression levels may induce AKI.
Collapse
|
26
|
Wu T, Wang M, Ning F, Zhou S, Hu X, Xin H, Reilly S, Zhang X. Emerging role for branched-chain amino acids metabolism in fibrosis. Pharmacol Res 2023; 187:106604. [PMID: 36503000 DOI: 10.1016/j.phrs.2022.106604] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/24/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
Fibrosis is a common pathological feature of organ diseases resulting from excessive production of extracellular matrix, which accounts for significant morbidity and mortality. However, there is currently no effective treatment targeting fibrogenesis. Recently, metabolic alterations are increasingly considered as essential factors underlying fibrogenesis, and especially research on metabolic regulation of amino acids is flourishing. Among them, branched-chain amino acids (BCAAs) are the most abundant essential amino acids, including leucine, isoleucine and valine, which play significant roles in the substance and energy metabolism and their regulation. Dysregulation of BCAAs metabolism has been proven to contribute to numerous diseases. In this review, we summarize the metabolic regulation of fibrosis and the changes in BCAAs metabolism secondary to fibrosis. We also review the effects and mechanisms of the BCAAs intervention, and its therapeutic targeting in hepatic, renal and cardiac fibrosis, with a focus on the fibrosis in liver and associated hepatocellular carcinoma.
Collapse
Affiliation(s)
- Tiangang Wu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mengling Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Fengling Ning
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shilin Zhou
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xuetao Hu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Hong Xin
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China; Shanghai Zhangjiang Institute of Medical Innovation, Shanghai 201204, China.
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
27
|
Vigers T, Vinovskis C, Li LP, Prasad P, Heerspink H, D'Alessandro A, Reisz JA, Piani F, Cherney DZ, van Raalte DH, Nadeau KJ, Pavkov ME, Nelson RG, Pyle L, Bjornstad P. Plasma levels of carboxylic acids are markers of early kidney dysfunction in young people with type 1 diabetes. Pediatr Nephrol 2023; 38:193-202. [PMID: 35507146 PMCID: PMC10182875 DOI: 10.1007/s00467-022-05531-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/25/2022] [Accepted: 03/07/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND We compared plasma metabolites of amino acid oxidation and the tricarboxylic acid (TCA) cycle in youth with and without type 1 diabetes mellitus (T1DM) and related the metabolites to glomerular filtration rate (GFR), renal plasma flow (RPF), and albuminuria. Metabolites associated with impaired kidney function may warrant future study as potential biomarkers or even future interventions to improve kidney bioenergetics. METHODS Metabolomic profiling of fasting plasma samples using a targeted panel of 644 metabolites and an untargeted panel of 19,777 metabolites was performed in 50 youth with T1DM ≤ 10 years and 20 controls. GFR and RPF were ascertained by iohexol and p-aminohippurate clearance, and albuminuria calculated as urine albumin to creatinine ratio. Sparse partial least squares discriminant analysis and moderated t tests were used to identify metabolites associated with GFR and RPF. RESULTS Adolescents with and without T1DM were similar in age (16.1 ± 3.0 vs. 16.1 ± 2.9 years) and BMI (23.4 ± 5.1 vs. 22.7 ± 3.7 kg/m2), but those with T1DM had higher GFR (189 ± 40 vs. 136 ± 22 ml/min) and RPF (820 ± 125 vs. 615 ± 65 ml/min). Metabolites of amino acid oxidation and the TCA cycle were significantly lower in adolescents with T1DM vs. controls, and the measured metabolites were able to discriminate diabetes status with an AUC of 0.82 (95% CI: 0.71, 0.93) and error rate of 0.21. Lower glycine (r:-0.33, q = 0.01), histidine (r:-0.45, q < 0.001), methionine (r: -0.29, q = 0.02), phenylalanine (r: -0.29, q = 0.01), serine (r: -0.42, q < 0.001), threonine (r: -0.28, q = 0.02), citrate (r: -0.35, q = 0.003), fumarate (r: -0.24, q = 0.04), and malate (r: -0.29, q = 0.02) correlated with higher GFR. Lower glycine (r: -0.28, q = 0.04), phenylalanine (r:-0.3, q = 0.03), fumarate (r: -0.29, q = 0.04), and malate (r: -0.5, q < 0.001) correlated with higher RPF. Lower histidine (r: -0.28, q = 0.02) was correlated with higher mean ACR. CONCLUSIONS In conclusion, adolescents with relatively short T1DM duration exhibited lower plasma levels of carboxylic acids that associated with hyperfiltration and hyperperfusion. TRIAL REGISTRATION ClinicalTrials.gov NCT03618420 and NCT03584217 A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Timothy Vigers
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA.
- Department of Biostatistics and Informatics, Colorado School of Public Health, 13123 E 16th Ave, A036-B265, Aurora, CO, 80045, USA.
| | - Carissa Vinovskis
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lu-Ping Li
- Department of Radiology, NorthShore University HealthSystem, Evanston, IL, USA
| | - Pottumarthi Prasad
- Department of Radiology, NorthShore University HealthSystem, Evanston, IL, USA
| | - Hiddo Heerspink
- Department Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Federica Piani
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA
| | - David Z Cherney
- Department of Medicine, Division of Nephrology, University of Toronto School of Medicine, Toronto, Ontario, Canada
| | - Daniel H van Raalte
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, location VUmc, Amsterdam, the Netherlands
| | - Kristen J Nadeau
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Meda E Pavkov
- Division of Diabetes Translation, Center for Disease Control and Prevention, Atlanta, GA, USA
| | - Robert G Nelson
- Chronic Kidney Disease Section, Phoenix Epidemiology and Clinical Research Branch, NIDDK, Phoenix, AZ, USA
| | - Laura Pyle
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, 13123 E 16th Ave, A036-B265, Aurora, CO, 80045, USA
| | - Petter Bjornstad
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Medicine, Division of Nephrology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
28
|
Perinatal Oxidative Stress and Kidney Health: Bridging the Gap between Animal Models and Clinical Reality. Antioxidants (Basel) 2022; 12:antiox12010013. [PMID: 36670875 PMCID: PMC9855228 DOI: 10.3390/antiox12010013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/02/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Oxidative stress arises when the generation of reactive oxygen species or reactive nitrogen species overwhelms antioxidant systems. Developing kidneys are vulnerable to oxidative stress, resulting in adult kidney disease. Oxidative stress in fetuses and neonates can be evaluated by assessing various biomarkers. Using animal models, our knowledge of oxidative-stress-related renal programming, the molecular mechanisms underlying renal programming, and preventive interventions to avert kidney disease has grown enormously. This comprehensive review provides an overview of the impact of perinatal oxidative stress on renal programming, the implications of antioxidant strategies on the prevention of kidney disease, and the gap between animal models and clinical reality.
Collapse
|
29
|
Zou J, Zhou X, Chen X, Ma Y, Yu R. Shenkang Injection for Treating Renal Fibrosis-Metabonomics and Regulation of E3 Ubiquitin Ligase Smurfs on TGF-β/Smads Signal Transduction. Front Pharmacol 2022; 13:849832. [PMID: 35721120 PMCID: PMC9201572 DOI: 10.3389/fphar.2022.849832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/25/2022] [Indexed: 12/31/2022] Open
Abstract
At present, TGF-β is the most critical fibrogenic factor known. Smad ubiquitin ligase Smurfs play an important role in the regulation of the TGF-/Smads signaling pathway, which is linked to metabolite changes in renal fibrosis. Previous studies have shown that Shenkang injection can prevent and treat chronic kidney disease through multiple channels of action. However, the precise relationship between Shenkang injection and the regulation of the TGF-/Smads signaling pathway in the treatment of chronic kidney disease is unknown. Here, we evaluated the pharmacological effects of Shenkang injection on ubiquitination and metabolic changes of the TGF-β/Smads signaling pathway in UUO mice using pathology-related indicators, immunoprecipitation, subcellular co-location, and metabonomics analysis. Our findings indicate that Shenkang injection can promote nuclear translocation of Smurf1 and Smurf2 to TGF- membrane receptors TR-I and Smad2 and ubiquitinated degradation of these proteins. Furthermore, the formation of TβR-I/TβR-II, TβR-I/Smad2, and TβR-I/Smad3 complexes was inhibited to negatively regulate the TGF-β/Smad signaling pathway induced renal tubular epithelial transdifferentiation (EMT). The EMT process is not very relevant in vivo, although it is clear that TGF-β induces EMT in cultured cells, which has been demonstrated by numerous teams around the world. However, this is not the case with the in vivo models of kidney fibrosis, especially UUO. In addition, Shenkang injection can improve amino acid metabolism, purine metabolism, and fatty acid metabolism disorders.
Collapse
Affiliation(s)
- Junju Zou
- Hunan Provincial Key Laboratory of Translational Research in TCM Prescriptions and Zheng, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaotao Zhou
- School of Basic Medicine, Chengdu University of Chinese Medicine, Chengdu, China
| | - Xian Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuerong Ma
- School of Basic Medicine, Chengdu University of Chinese Medicine, Chengdu, China
| | - Rong Yu
- Hunan Provincial Key Laboratory of Translational Research in TCM Prescriptions and Zheng, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
30
|
Artemether Alleviates Diabetic Kidney Disease by Modulating Amino Acid Metabolism. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7339611. [PMID: 35601149 PMCID: PMC9117059 DOI: 10.1155/2022/7339611] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 04/08/2022] [Indexed: 11/17/2022]
Abstract
Diabetes is a worldwide metabolic disease with rapid growing incidence, characterized by hyperglycemia. Diabetic kidney disease (DKD), the leading cause of chronic kidney disease (CKD), has a high morbidity according to the constantly increasing diabetic patients and always develops irreversible deterioration of renal function. Though different in pathogenesis, clinical manifestations, and therapies, both type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM) can evolve into DKD. Since amino acids are both biomarkers and causal agents, rarely report has been made about its metabolism which lies in T1DM- and T2DM-related kidney disease. This study was designed to investigate artemether in adjusting renal amino acid metabolism in T1DM and T2DM mice. Artemether was applied as treatment in streptozotocin (STZ) induced T1DM mice and db/db T2DM mice, respectively. Artemether-treated mice showed lower FBG and HbA1c and reduced urinary albumin excretion, as well as urinary NAG. Both types of diabetic mice showed enlarged kidneys, as confirmed by increased kidney weight and the ratio of kidney weight to body weight. Artemether normalized kidney size and thus attenuated renal hypertrophy. Kidney tissue UPLC-MS analysis showed that branched-chain amino acids (BCAAs) and citrulline were upregulated in diabetic mice without treatment and downregulated after being treated with artemether. Expressions of glutamine, glutamic acid, aspartic acid, ornithine, glycine, histidine, phenylalanine and threonine were decreased in both types of diabetic mice whereas they increased after artemether treatment. The study demonstrates the initial evidence that artemether exerted renal protection in DKD by modulating amino acid metabolism.
Collapse
|
31
|
Losartan ameliorates renal interstitial fibrosis through metabolic pathway and Smurfs-TGF-β/Smad. Biomed Pharmacother 2022; 149:112931. [PMID: 36068784 DOI: 10.1016/j.biopha.2022.112931] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/27/2022] [Accepted: 04/05/2022] [Indexed: 11/22/2022] Open
Abstract
The genesis and development of renal fibrosis involve a variety of pathways closely related to inflammation, cytokines, oxidative stress and metabolic abnormalities. Renal fibrosis is the result of a complex combination of a variety of lesions. Epithelial-mesenchymal transdifferentiation (EMT) of renal tubular epithelial cells is considered the key to renal fibrosis. Losartan is a typical Angiotensin II (ANG II) receptor antagonist and relaxes blood vessels. In this study, we investigated the effects of losartan on Unilateral Ureteral Obstruction (UUO) model mice by studying the changes in the TGF-β/Smad and metabolomics. Male C57BL/6 J mice were intervened with the UUO model and given losartan (10, 20, 30 mg/kg/d) for 28 consecutive days. The results showed that losartan could reduce UUO-induced abnormal serum metabolic spectrum and renal function. It could also improve renal tubular-interstitial injury and fibrosis by reducing tubulointerstitial dilation and collagen deposition. In addition, losartan promoted the expression of Smurf2 and Smurf1, i.e., Smad7 and E3 ubiquitin-linked enzymes, in the nucleus to degrade the type I receptor of TGF-β1 (TβR-I) and P-Smad2/3 to inhibit renal tubular epithelial cells EMT. In summary, these findings indicated that losartan could regulate the TGF-β/Smad and metabolic pathway in UUO model mice through ubiquitination to reduce renal fibrosis.
Collapse
|
32
|
Wang J, Zha M, Zhao H, Yue W, Wu D, Li K. Detection of Kidney Dysfunction through In Vivo Magnetic Resonance Imaging with Renal-Clearable Gadolinium Nanoprobes. Anal Chem 2022; 94:4005-4011. [PMID: 35188754 DOI: 10.1021/acs.analchem.1c05140] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Kidney dysfunction is a clinical syndrome that can subsequently result in lethal kidney failure. The exploration of emerging bioimaging contrast agents with translational potential is highly challenging for a feasible diagnosis of kidney dysfunction. Herein, a class of renal-clearable gadolinium nanoparticles (Gd@PEG NPs) with an ultrasmall size of ∼5 nm, good monodispersity, and T1 relaxivity are synthesized using mesoporous silica nanoparticles as the template. Assisted by such renal-clearable Gd@PEG NPs, the diagnosis of kidney dysfunction in a mice model with a damaged kidney has been achieved through in vivo noninvasive magnetic resonance imaging. As a result, this work paves the way to synthesize monodispersible ultrasmall Gd contrast agents, facilitating the exploration of translational strategies for an in vivo analysis of kidney dysfunction.
Collapse
Affiliation(s)
- Jun Wang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen 518055, China
| | - Menglei Zha
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen 518055, China
| | - Hui Zhao
- Department of MRI Diagnosis, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Wentong Yue
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen 518055, China
| | - Decheng Wu
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen 518055, China
| | - Kai Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen 518055, China
| |
Collapse
|
33
|
Xu S, Jia P, Fang Y, Jin J, Sun Z, Zhou W, Li J, Zhang Y, Wang X, Ren T, Zou Z, Ding X. Nuclear farnesoid X receptor attenuates acute kidney injury through fatty acid oxidation. Kidney Int 2022; 101:987-1002. [DOI: 10.1016/j.kint.2022.01.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 12/28/2021] [Accepted: 01/11/2022] [Indexed: 12/14/2022]
|
34
|
Stenhouse C, Suva LJ, Gaddy D, Wu G, Bazer FW. Phosphate, Calcium, and Vitamin D: Key Regulators of Fetal and Placental Development in Mammals. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1354:77-107. [PMID: 34807438 DOI: 10.1007/978-3-030-85686-1_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Normal calcium and bone homeostasis in the adult is virtually fully explained by the interactions of several key regulatory hormones, including parathyroid hormone, 1,25 dihydroxy vitamin D3, fibroblast growth factor-23, calcitonin, and sex steroids (estradiol and testosterone). In utero, bone and mineral metabolism is regulated differently from the adult. During development, it is the placenta and not the fetal kidneys, intestines, or skeleton that is the primary source of minerals for the fetus. The placenta is able to meet the almost inexhaustible needs of the fetus for minerals by actively driving the transport of calcium and phosphorus from the maternal circulation to the growing fetus. These fundamentally important minerals are maintained in the fetal circulation at higher concentrations than those in maternal blood. Maintenance of these inordinately higher fetal levels is necessary for the developing skeleton to accrue sufficient minerals by term. Importantly, in livestock species, prenatal mineralization of the skeleton is crucial for the high levels of offspring activity soon after birth. Calcium is required for mineralization, as well as a plethora of other physiological functions. Placental calcium and phosphate transport are regulated by several mechanisms that are discussed in this review. It is clear that phosphate and calcium metabolism is intimately interrelated and, therefore, placental transport of these minerals cannot be considered in isolation.
Collapse
Affiliation(s)
- Claire Stenhouse
- Departments of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Larry J Suva
- Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843, USA
| | - Dana Gaddy
- Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Guoyao Wu
- Departments of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Fuller W Bazer
- Department of Animal Science, Kleberg Center, Texas A&M University, College Station, TX, 77843-2471, USA.
| |
Collapse
|
35
|
Moses RM, Kramer AC, Seo H, Wu G, Johnson GA, Bazer FW. A Role for Fructose Metabolism in Development of Sheep and Pig Conceptuses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1354:49-62. [PMID: 34807436 DOI: 10.1007/978-3-030-85686-1_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The period of conceptus (embryo and extraembryonic membrane) development between fertilization and implantation in mammalian species is critical as it sets the stage for placental and fetal development. The trophectoderm and endoderm of pre-implantation ovine and porcine conceptuses undergo elongation, which requires rapid proliferation, migration, and morphological modification of the trophectoderm cells. These complex events occur in a hypoxic intrauterine environment and are supported through the transport of secretions from maternal endometrial glands to the conceptus required for the biochemical processes of cell proliferation, migration, and differentiation. The conceptus utilizes glucose provided by the mother to initiate metabolic pathways that provide energy and substrates for other metabolic pathways. Fructose, however, is in much greater abundance than glucose in amniotic and allantoic fluids, and fetal blood during pregnancy. Despite this, the role(s) of fructose is largely unknown even though a switch to fructosedriven metabolism in subterranean rodents and some cancers are key to their adaptation to hypoxic environments.
Collapse
Affiliation(s)
- Robyn M Moses
- Departments of Animal Science and Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Avery C Kramer
- Departments of Animal Science and Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Heewon Seo
- Departments of Animal Science and Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Guoyao Wu
- Departments of Animal Science and Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Gregory A Johnson
- Departments of Animal Science and Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Fuller W Bazer
- Departments of Animal Science and Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
36
|
Nutrition and Metabolism: Foundations for Animal Growth, Development, Reproduction, and Health. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1354:1-24. [PMID: 34807434 DOI: 10.1007/978-3-030-85686-1_1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Consumption of high-quality animal protein plays an important role in improving human nutrition, growth, development, and health. With an exponential growth of the global population, demands for animal-sourced protein are expected to increase by 60% between 2021 and 2050. In addition to the production of food protein and fiber (wool), animals are useful models for biomedical research to prevent and treat human diseases and serve as bioreactors to produce therapeutic proteins. For a high efficiency to transform low-quality feedstuffs and forages into high-quality protein and highly bioavailable essential minerals in diets of humans, farm animals have dietary requirements for energy, amino acids, lipids, carbohydrates, minerals, vitamins, and water in their life cycles. All nutrients interact with each other to influence the growth, development, and health of mammals, birds, fish, and crustaceans, and adequate nutrition is crucial for preventing and treating their metabolic disorders (including metabolic diseases) and infectious diseases. At the organ level, the small intestine is not only the terminal site for nutrient digestion and absorption, but also intimately interacts with a diverse community of intestinal antigens and bacteria to influence gut and whole-body health. Understanding the species and metabolism of intestinal microbes, as well as their interactions with the intestinal immune systems and the host intestinal epithelium can help to mitigate antimicrobial resistance and develop prebiotic and probiotic alternatives to in-feed antibiotics in animal production. As abundant sources of amino acids, bioactive peptides, energy, and highly bioavailable minerals and vitamins, animal by-product feedstuffs are effective for improving the growth, development, health, feed efficiency, and survival of livestock and poultry, as well as companion and aquatic animals. The new knowledge covered in this and related volumes of Adv Exp Med Biol is essential to ensure sufficient provision of animal protein for humans, while helping reduce greenhouse gas emissions, minimize the urinary and fecal excretion of nitrogenous and other wastes to the environment, and sustain animal agriculture (including aquaculture).
Collapse
|
37
|
Hepatic Glucose Metabolism and Its Disorders in Fish. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1354:207-236. [PMID: 34807444 DOI: 10.1007/978-3-030-85686-1_11] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Carbohydrate, which is the most abundant nutrient in plant-sourced feedstuffs, is an economically indispensable component in commercial compound feeds for fish. This nutrient can enhance the physical quality of diets and allow for pellet expansion during extrusion. There is compelling evidence that an excess dietary intake of starch causes hepatic disorders, thereby further reducing the overall food consumption and growth performance of fish species. Among the severe metabolic disturbances are glycogenic hepatopathy (hepatomegaly caused by the excessive accumulation of glycogen in hepatocytes) and hepatic steatosis (the accumulation of large vacuoles of triacylglycerols in hepatocytes). The development of those disorders is mainly due to the limited ability of fish to oxidize glucose and control blood glucose concentration. The prolonged elevations of blood glucose increase glucose intake by the liver, and excess glucose is stored either as glycogen through glycogenesis in hepatocytes or as triglycerides via lipogenesis in tissues, depending on the species. In some fish species (e.g., largemouth bass), the liver has a low ability to regulate glycolysis, gluconeogenesis, and glycogen breakdown in response to high starch intake. For most species of fish, the liver size increases with lipid or glycogen accumulation when they have a high starch intake. It is a challenge to develop the same set of diagnostic criteria for all fish species as their physiology or metabolic patterns differ. Although glycogenic hepatopathy appears to be a common disease in carnivorous fish, it has been under-recognized in many studies. As a result, understanding these diseases and their pathogeneses in different fish species is crucial for manufacturing cost-effective pellet diets to promote the health, growth, survival, and feed efficiency of fish in future.
Collapse
|
38
|
Jia S, Li X, He W, Wu G. Protein-Sourced Feedstuffs for Aquatic Animals in Nutrition Research and Aquaculture. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1354:237-261. [PMID: 34807445 DOI: 10.1007/978-3-030-85686-1_12] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Aquatic animals have particularly high requirements for dietary amino acids (AAs) for health, survival, growth, development, and reproduction. These nutrients are usually provided from ingested proteins and may also be derived from supplemental crystalline AA. AAs are the building blocks of protein (a major component of tissue growth) and, therefore, are the determinants of the growth performance and feed efficiency of farmed fish. Because protein is generally the most expensive ingredient in aqua feeds, much attention has been directed to ensure that dietary protein feedstuff is of high quality and cost-effective for feeding fish, crustaceans, and other aquatic animals worldwide. Due to the rapid development of aquaculture worldwide and a limited source of fishmeal (the traditionally sole or primary source of AAs for aquatic animals), alternative protein sources must be identified to feed aquatic animals. Plant-sourced feedstuffs for aquatic animals include soybean meal, extruded soybean meal, fermented soybean meal, soybean protein concentrates, soybean protein isolates, leaf meal, hydrolyzed plant protein, wheat, wheat hydrolyzed protein, canola meal, cottonseed meal, peanut meal, sunflower meal, peas, rice, dried brewers grains, and dried distillers grains. Animal-sourced feedstuffs include fishmeal, fish paste, bone meal, meat and bone meal, poultry by-product meal, chicken by-product meal, chicken visceral digest, spray-dried poultry plasma, spray-dried egg product, hydrolyzed feather meal, intestine-mucosa product, peptones, blood meal (bovine or poultry), whey powder with high protein content, cheese powder, and insect meal. Microbial sources of protein feedstuffs include yeast protein and single-cell microbial protein (e.g., algae); they have more balanced AA profiles than most plant proteins for animal feeding. Animal-sourced ingredients can be used as a single source of dietary protein or in complementary combinations with plant and microbial sources of proteins. All protein feedstuffs must adequately provide functional AAs for aquatic animals.
Collapse
Affiliation(s)
- Sichao Jia
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Xinyu Li
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Wenliang He
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
39
|
Furukawa K, He W, Bailey CA, Bazer FW, Toyomizu M, Wu G. Polyamine synthesis from arginine and proline in tissues of developing chickens. Amino Acids 2021; 53:1739-1748. [PMID: 34613458 DOI: 10.1007/s00726-021-03084-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/23/2021] [Indexed: 12/15/2022]
Abstract
Polyamines (putrescine, spermidine, and spermine) are synthesized primarily from ornithine via ornithine decarboxylase (ODC) in mammals. Although avian tissues contain ODC activity, little is known about intracellular sources of ornithine for their polyamine synthesis. This study tested the hypothesis that arginase and proline oxidase contribute to polyamine synthesis in chickens. Kidney, jejunum, leg muscle, and liver from 0-, 7-, 14- and 21-day-old broiler chickens were assayed for the activities of arginase, proline oxidase (POX), ornithine aminotransferase (OAT), and ornithine decarboxylase (ODC). Kidney slices were also used to determine 14C-polyamine synthesis from [U-14C]arginine and [U-14C]proline. Furthermore, these tissues and plasma were analyzed for polyamines. Results indicate that all tissues contained OAT (mitochondrial) and ODC (cytosolic) activities, but arginase and POX activities were only detected in the mitochondria of chicken kidneys. Renal POX and arginase activities were greater at 7 days of age compared to newly hatched birds, and declined by Day 14. Renal arginase activity was greater at 21 days compared to 14 days of age, but there was no change in renal POX activity during that same period. Concentrations of polyamines in the kidneys and plasma were greater on Day 7 compared to Day 0 and decreased thereafter on Days 14 and 21. Kidney slices readily converted arginine and proline into polyamines, with peak rates being on Day 7. Concentrations of putrescine, spermidine and spermine in the plasma of chickens were about 20- to 100-fold greater than those in mammals. Our results indicate that polyamines are synthesized from arginine and proline in avian kidneys. Unlike mammals, polyamines released from the kidneys are likely the major source of polyamines in the blood and other extra-renal tissues in chickens.
Collapse
Affiliation(s)
- Kyohei Furukawa
- Department of Animal Science, Texas A&M University, College Station, TX, 77843-2471, USA.,Animal Nutrition, Graduate School of Agricultural Science, Tohoku University, Sendai, 980-8572, Japan.,International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, 980-8572, Japan
| | - Wenliang He
- Department of Animal Science, Texas A&M University, College Station, TX, 77843-2471, USA
| | - Christopher A Bailey
- Department of Poultry Science, Texas A&M University, College Station, TX, 77843-2471, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX, 77843-2471, USA
| | - Masaaki Toyomizu
- Animal Nutrition, Graduate School of Agricultural Science, Tohoku University, Sendai, 980-8572, Japan.,International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, 980-8572, Japan
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, 77843-2471, USA.
| |
Collapse
|
40
|
Grima-Reyes M, Martinez-Turtos A, Abramovich I, Gottlieb E, Chiche J, Ricci JE. Physiological impact of in vivo stable isotope tracing on cancer metabolism. Mol Metab 2021; 53:101294. [PMID: 34256164 PMCID: PMC8358691 DOI: 10.1016/j.molmet.2021.101294] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/30/2021] [Accepted: 07/08/2021] [Indexed: 11/25/2022] Open
Abstract
Background There is growing interest in the analysis of tumor metabolism to identify cancer-specific metabolic vulnerabilities and therapeutic targets. Finding of such candidate metabolic pathways mainly relies on the highly sensitive identification and quantitation of numerous metabolites and metabolic fluxes using metabolomics and isotope tracing analyses. However, nutritional requirements and metabolic routes used by cancer cells cultivated in vitro do not always reflect the metabolic demands of malignant cells within the tumor milieu. Therefore, to understand how the metabolism of tumor cells in its physiological environment differs from that of normal cells, these analyses must be performed in vivo. Scope of Review This review covers the physiological impact of the exogenous administration of a stable isotope tracer into cancer animal models. We discuss specific aspects of in vivo isotope tracing protocols based on discrete bolus injections of a labeled metabolite: the tracer administration per se and the fasting period prior to it. In addition, we illustrate the complex physiological scenarios that arise when studying tumor metabolism – by isotopic labeling in animal models fed with a specific amino acid restricted diet. Finally, we provide strategies to minimize these limitations. Major Conclusions There is growing evidence that metabolic dependencies in cancers are influenced by tissue environment, cancer lineage, and genetic events. An increasing number of studies describe discrepancies in tumor metabolic dependencies when studied in in vitro settings or in vivo models, including cancer patients. Therefore, in-depth in vivo profiling of tumor metabolic routes within the appropriate pathophysiological environment will be key to identify relevant alterations that contribute to cancer onset and progression. In vivo isotope tracing is the state-of-the-art approach to study tumor metabolism. In vivo tracer administration challenges the physiological metabolism of mice. Interorgan conversion of the tracer might confound tumor labeling patterns. Mouse fasting before in vivo tracing impacts on systemic and tumor metabolism. Optimization is key to minimize physiological alterations linked to in vivo tracing.
Collapse
Affiliation(s)
- Manuel Grima-Reyes
- Université Côte d'Azur, INSERM, C3M, Nice, France; Equipe labellisée LIGUE Contre le Cancer, Nice, France
| | - Adriana Martinez-Turtos
- Université Côte d'Azur, INSERM, C3M, Nice, France; Equipe labellisée LIGUE Contre le Cancer, Nice, France
| | - Ifat Abramovich
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Eyal Gottlieb
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Johanna Chiche
- Université Côte d'Azur, INSERM, C3M, Nice, France; Equipe labellisée LIGUE Contre le Cancer, Nice, France
| | - Jean-Ehrland Ricci
- Université Côte d'Azur, INSERM, C3M, Nice, France; Equipe labellisée LIGUE Contre le Cancer, Nice, France.
| |
Collapse
|
41
|
Liu J, Su X, Lu J, Ning J, Lin M, Zhou H. PM 2.5 induces intestinal damage by affecting gut microbiota and metabolites of rats fed a high-carbohydrate diet. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 279:116849. [PMID: 33773181 DOI: 10.1016/j.envpol.2021.116849] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/04/2021] [Accepted: 02/23/2021] [Indexed: 06/12/2023]
Abstract
PM2.5 has a major impact on the gastrointestinal system, but the specific mechanism behind this action is not fully understood. Current studies have focused on the relationship between PM2.5 and intestinal flora disorder, while ignoring the important influence of diet on gut microbes. In this study, SD rats were fed either a normal, high-fat, or high-carbohydrate diet for two months and exposed to PM2.5 (7 mg/kg b.w.) by intratracheal instillation. The results showed that the body and kidney weights of the rats in the high-fat diet group were significantly increased relative to those with a normal diet, and changes in the intestinal microbes and metabolites induced by PM2.5 were observed. Rats in the high-carbohydrate diet group had a significant response, and the diversity and richness indices of the flora were reduced (p < 0.05); additionally, intestinal Biffidobacterium and Lactobacillus were enriched, while many endogenous metabolites were found. Some amino acids derivatives and long-chain fatty acids were increased (p < 0.05). Both diet structure and PM2.5 exposure can affect the composition of gut microbiota, and intestinal metabolites may be associated with cell membrane damage when a high-carbohydrate diet interacts with PM2.5. This study considers multiple dietary factors to further supplement the evidence of intestinal damage via PM2.5.
Collapse
Affiliation(s)
- Jinhua Liu
- School of Chemistry and Chemical Engineering, Shihezi University, Key Laboratory of Environmental Monitoring and Pollutant Control of Xinjiang Bingtuan, Xinjiang, 832003, China
| | - Xianghui Su
- Department of Endocrinology, Changji Branch, First Affiliated Hospital of Xinjiang Medical University, Xinjiang, 831100, China
| | - Jianjiang Lu
- School of Chemistry and Chemical Engineering, Shihezi University, Key Laboratory of Environmental Monitoring and Pollutant Control of Xinjiang Bingtuan, Xinjiang, 832003, China.
| | - Jianying Ning
- The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832003, China
| | - Meng Lin
- School of Chemistry and Chemical Engineering, Shihezi University, Key Laboratory of Environmental Monitoring and Pollutant Control of Xinjiang Bingtuan, Xinjiang, 832003, China
| | - Hongjuan Zhou
- School of Chemistry and Chemical Engineering, Shihezi University, Key Laboratory of Environmental Monitoring and Pollutant Control of Xinjiang Bingtuan, Xinjiang, 832003, China
| |
Collapse
|
42
|
Che D, Nyingwa PS, Ralinala KM, Maswanganye GMT, Wu G. Amino Acids in the Nutrition, Metabolism, and Health of Domestic Cats. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1285:217-231. [PMID: 33770409 DOI: 10.1007/978-3-030-54462-1_11] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Domestic cats (carnivores) require high amounts of dietary amino acids (AAs) for normal growth, development, and reproduction. Amino acids had been traditionally categorised as nutritionally essential (EAAs) or nonessential (NEAAs), depending on whether they are synthesized de novo in the body. This review will focus on AA nutrition and metabolism in cats. Like other mammals, cats do not synthesize the carbon skeletons of twelve proteinogenic AAs: Arg, Cys, His, Ile, Leu, Lys, Met, Phe, Thr, Trp, Tyr, and Val. Like other feline carnivores but unlike many mammals, cats do not synthesize citrulline and have a very limited ability to produce taurine from Cys. Except for Leu and Lys that are strictly ketogenic AAs, most EAAs are both glucogenic and ketogenic AAs. All the EAAs (including taurine) must be provided in diets for cats. These animals are sensitive to dietary deficiencies of Arg and taurine, which rapidly result in life-threatening hyperammonemia and retinal damage, respectively. Although the National Research Council (NCR, Nutrient requirements of dogs and cats. National Academies Press, Washington, DC, 2006) does not recommend dietary requirements of cats for NEAAs, much attention should be directed to this critical issue of nutrition. Cats can synthesize de novo eight proteinogenic AAs: Ala, Asn, Asp, Gln, Glu, Gly, Pro, and Ser, as well as some nonproteinogenic AAs, such as γ-aminobutyrate, ornithine, and β-alanine with important physiological functions. Some of these AAs (e.g., Gln, Glu, Pro, and Gly) are crucial for intestinal integrity and health. Except for Gln, AAs in the arterial blood of cats may not be available to the mucosa of the small intestine. Plant-source foodstuffs lack taurine and generally contain inadequate Met and Cys and, therefore, should not be fed to cats in any age group. Besides meat, animal-source foodstuffs (including ruminant meat & bone meal, poultry by-product meal, porcine mucosal protein, and chicken visceral digest) are good sources of proteinogenic AAs and taurine for cats. Meeting dietary requirements for both EAAs and NEAAs in proper amounts and balances is crucial for improving the health, wellbeing, longevity, and reproduction of cats.
Collapse
Affiliation(s)
- Dongsheng Che
- College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, and Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Jilin Agricultural University, Changchun, China
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Pakama S Nyingwa
- College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, and Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Khakhathi M Ralinala
- College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, and Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Gwen M T Maswanganye
- College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, and Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
43
|
Interorgan Metabolism of Amino Acids in Human Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1332:129-149. [PMID: 34251642 DOI: 10.1007/978-3-030-74180-8_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Amino acids are integral for human health, influencing an array of physiological processes from gene expression to vasodilation to the immune response. In accordance with this expansive range of unique functions, the tissues of the body engage in a complex interplay of amino acid exchange and metabolism to respond to the organism's dynamic needs for a range of nitrogenous products. Interorgan amino acid metabolism is required for numerous metabolic pathways, including the synthesis of functional amino acids like arginine, glutamate, glutamine, and glycine. This physiological process requires the cooperative handling of amino acids by organs (e.g., the small intestine, skeletal muscle, kidneys, and liver), as well as the complete catabolism of nutritionally essential amino acids such as the BCAAs, with their α-ketoacids shuttled from muscle to liver. These exchanges are made possible by several mechanisms, including organ location, as well as the functional zonation of enzymes and the cell-specific expression of amino acid transporters. The cooperative handling of amino acids between the various organs does not appear to be under the control of any centralized regulation, but is instead influenced by factors such as fluctuations in nutrient availability, hormones, changes associated with development, and altered environmental factors. While the normal function of these pathways is associated with health and homeostasis, affected by physical activity, diet and body composition, dysregulation is observed in numerous disease states, including cardiovascular disease and cancer cachexia, presenting potential avenues for the manipulation of amino acid consumption as part of the therapeutic approach to these conditions in individuals.
Collapse
|
44
|
Oxidation of Energy Substrates in Tissues of Fish: Metabolic Significance and Implications for Gene Expression and Carcinogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1332:67-83. [PMID: 34251639 DOI: 10.1007/978-3-030-74180-8_5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Fish are useful animal models for studying effects of nutrients and environmental factors on gene expression (including epigenetics), toxicology, and carcinogenesis. To optimize the response of the animals to substances of interest (including toxins and carcinogens), water pollution, or climate changes, it is imperative to understand their fundamental biochemical processes. One of these processes concerns energy metabolism for growth, development, and survival. We have recently shown that tissues of hybrid striped bass (HSB), zebrafish, and largemouth bass (LMB) use amino acids (AAs; such as glutamate, glutamine, aspartate, alanine, and leucine) as major energy sources. AAs contribute to about 80% of ATP production in the liver, proximal intestine, kidney, and skeletal muscle tissue of the fish. Thus, as for mammals (including humans), AAs are the primary metabolic fuels in the proximal intestine of fish. In contrast, glucose and fatty acids are only minor metabolic fuels in the fish. Fish tissues have high activities of glutamate dehydrogenase, glutamate-oxaloacetate transaminase, and glutamate-pyruvate transaminase, as well as high rates of glutamate uptake. In contrast, the activities of hexokinase, pyruvate dehydrogenase, and carnitine palmitoyltransferase 1 in all the tissues are relatively low. Furthermore, unlike mammals, the skeletal muscle (the largest tissue) of HSB and LMB has a limited uptake of long-chain fatty acids and barely oxidizes fatty acids. Our findings explain differences in the metabolic patterns of AAs, glucose, and lipids among various tissues in fish. These new findings have important implications for understanding metabolic significance of the tissue-specific oxidation of AAs (particularly glutamate and glutamine) in gene expression (including epigenetics), nutrition, and health, as well as carcinogenesis in fish, mammals (including humans), and other animals.
Collapse
|