1
|
Zhang Y, Yan Z, Nan N, Li S, Qin G. Ozone exposure induced kidney damage in diabetic mice: The key role of lipid metabolism and water-electrolyte homeostasis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 372:125963. [PMID: 40037426 DOI: 10.1016/j.envpol.2025.125963] [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: 09/12/2024] [Revised: 02/18/2025] [Accepted: 03/01/2025] [Indexed: 03/06/2025]
Abstract
Ozone (O3) is an important environmental pollutant that has garnered growing public concern. Epidemiological studies indicate that exposure to O3 is associated with an elevated risk of kidney disease, a common complication of diabetes. However, the harmful effects of O3 on the kidneys remain unconfirmed. Herein, we established models of non-diabetic and diabetic mice exposed to 0.5 ppm O3 for 28 days (4 h/day). We evaluated O3-induced renal injury and potential mechanisms through analyzing biochemical markers related to renal function, along with histopathology and transcriptomic sequencing of the kidneys. The results showed that O3 exposure caused glomerular hypertrophy in both non-diabetic and diabetic mice, with mesangial hypercellularity and kidney function impairment specifically in diabetic mice. Furthermore, renal levels of free fatty acids and cholesterol were significantly elevated in O3-exposed diabetic mice. The important roles of lipid and water-electrolyte metabolism related pathways in O3-induced kidney damage were found by transcriptome sequencing analysis. The mRNA and/or protein expressions of some genes involved in β-ENaC and AQP2 pathways, which are related to renal water and sodium retention, were changed in diabetic mice following O3 exposure by real-time quantitative PCR, immunofluorescence staining, and Western blotting. Overall, diabetic mice exhibited a higher vulnerability to adverse effects in the kidney after O3 exposure than non-diabetic mice. Dysregulation of lipid metabolism and imbalance in water-electrolyte homeostasis have been discovered as key contributing mechanisms. This study offers valuable insights into mechanisms through which ambient O3 poses renal health risks to both the general subjects and susceptible individuals.
Collapse
Affiliation(s)
- Yaru Zhang
- Shanxi Key Laboratory of Coal-based Emerging Pollutant Identification and Risk Control, Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Zhipeng Yan
- Shanxi Key Laboratory of Coal-based Emerging Pollutant Identification and Risk Control, Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Nan Nan
- Shanxi Key Laboratory of Coal-based Emerging Pollutant Identification and Risk Control, Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Shiya Li
- Shanxi Key Laboratory of Coal-based Emerging Pollutant Identification and Risk Control, Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Guohua Qin
- Shanxi Key Laboratory of Coal-based Emerging Pollutant Identification and Risk Control, Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan, Shanxi, 030006, People's Republic of China.
| |
Collapse
|
2
|
Cui X, Qiao R, Wang B, Hu Y, Sun G, Hu W, Luan Z, Ren H, Xu H, Guan Y, Zhang X. Uric acid reduces the expression of aquaporins in renal collecting ducts to increase urine output in hyperuricemia. Front Physiol 2025; 16:1504328. [PMID: 40271210 PMCID: PMC12014756 DOI: 10.3389/fphys.2025.1504328] [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: 09/30/2024] [Accepted: 03/13/2025] [Indexed: 04/25/2025] Open
Abstract
Background Hyperuricemia (HUA) has attracted wide attention due to its close relationship with gout, hypertension, hypertriglyceridemia, obesity, atherosclerotic heart disease, type 2 diabetes and chronic kidney disease. Clinical observations suggest that people with high levels of serum uric acid (sUA) exhibits impaired urine concentration. We speculate that UA may regulate the expression of AQPs through inflammatory pathways, resulting in impaired renal urine concentration. Methods and results We revealed that patients and mice with HUA had a polyuria phenotype and found that the expression of aquaporin 2 (AQP2), AQP3 and AQP4 were significantly reduced in the kidneys of mice with HUA. Similarly, uric acid (UA) treatment markedly suppressed the expression of AQP2, AQP3 and AQP4 in cultured inner medullary collecting duct cells (IMCDs). We observed an increased expression of NF-κB in the kidneys of mice with HUA and in the IMCD cells treated with UA. Blockade of NF-κB by its inhibitor Bay 11-7082 dramatically attenuated UA-suppressed expression of AQP2, AQP3 and AQP4. Furthermore, the luciferase reporter, CHIP and EMSA assays showed that NF-κB can directly bind to the promoter regions of AQP2, AQP3 and AQP4 genes to suppress their transcription. Conclusion Our findings demonstrate that UA reduces the expression of AQP2, AQP3 and AQP4 in an NFκB-dependent manner, which contributes to the polyuria phenotype in the subjects with HUA.
Collapse
Affiliation(s)
- Xiaohui Cui
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Rongfang Qiao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Bing Wang
- Department of Endocrinology and Metabolism, The Central hospital of Dalian University of Technology, Dalian, China
| | - Yitong Hu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Guoying Sun
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Wenjuan Hu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Zhilin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Huiwen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Hu Xu
- Kidney Health Institute, East China Normal University, Shanghai, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaoyan Zhang
- Kidney Health Institute, East China Normal University, Shanghai, China
| |
Collapse
|
3
|
Melchior M, Van Eycken M, Nicaise C, Duquesne T, Longueville L, Collin A, Decaestecker C, Salmon I, Delporte C, Soyfoo M. Decreased Expression of Aquaporins as a Feature of Tubular Damage in Lupus Nephritis. Cells 2025; 14:380. [PMID: 40072108 PMCID: PMC11899336 DOI: 10.3390/cells14050380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/17/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Background: Tubulointerstitial hypoxia is a key factor for lupus nephritis progression to end-stage renal disease. Numerous aquaporins (AQPs) are expressed by renal tubules and are essential for their proper functioning. The aim of this study is to characterize the tubular expression of AQP1, AQP2 and AQP3, which could provide a better understanding of tubulointerstitial stress during lupus nephritis. Methods: This retrospective monocentric study was conducted at Erasme-HUB Hospital. We included 37 lupus nephritis samples and 9 healthy samples collected between 2000 and 2020, obtained from the pathology department. Immunohistochemistry was performed to target AQP1, AQP2 and AQP3 and followed by digital analysis. Results: No difference in AQP1, AQP2 and AQP3 staining location was found between healthy and lupus nephritis samples. However, we observed significant differences between these two groups, with a decrease in AQP1 expression in the renal cortex and in AQP3 expression in the cortex and medulla. In the subgroup of proliferative glomerulonephritis (class III/IV), this decrease in AQPs expression was more pronounced, particularly for AQP3. In addition, within this subgroup, we detected lower AQP2 expression in patients with higher interstitial inflammation score and lower AQP3 expression when higher interstitial fibrosis and tubular atrophy were present. Conclusions: We identified significant differences in the expression of aquaporins 1, 2, and 3 in patients with lupus nephritis. These findings strongly suggest that decreased AQP expression could serve as an indicator of tubular injury. Further research is warranted to evaluate AQP1, AQP2, and AQP3 as prognostic markers in both urinary and histological assessments of lupus nephritis.
Collapse
Affiliation(s)
- Maxime Melchior
- Departement of Rheumatology, Erasme-HUB Hospital, Université Libre de Bruxelles, 1070 Brussels, Belgium;
| | - Marie Van Eycken
- Department of Pathology, Erasme-HUB Hospital, Université Libre de Bruxelles, 1050 Brussels, Belgium; (M.V.E.); (I.S.)
| | - Charles Nicaise
- URPhyM, NARILIS, Université de Namur, 5000 Namur, Belgium; (C.N.); (T.D.); (L.L.)
| | - Thomas Duquesne
- URPhyM, NARILIS, Université de Namur, 5000 Namur, Belgium; (C.N.); (T.D.); (L.L.)
| | - Léa Longueville
- URPhyM, NARILIS, Université de Namur, 5000 Namur, Belgium; (C.N.); (T.D.); (L.L.)
| | - Amandine Collin
- DIAPath, Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles, 6041 Gosselies, Belgium; (A.C.); (C.D.)
| | - Christine Decaestecker
- DIAPath, Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles, 6041 Gosselies, Belgium; (A.C.); (C.D.)
- Laboratory of Image Synthesis and Analysis, Université Libre de Bruxelles, 1050 Brussels, Belgium
| | - Isabelle Salmon
- Department of Pathology, Erasme-HUB Hospital, Université Libre de Bruxelles, 1050 Brussels, Belgium; (M.V.E.); (I.S.)
- DIAPath, Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles, 6041 Gosselies, Belgium; (A.C.); (C.D.)
- Centre Universitaire Inter Régional D’expertise en Anatomie Pathologique Hospitalière, 6040 Jumet, Belgium
| | - Christine Delporte
- Laboratory of Pathophysiological and Nutritional Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 1070 Brussels, Belgium;
| | - Muhammad Soyfoo
- Departement of Rheumatology, Erasme-HUB Hospital, Université Libre de Bruxelles, 1070 Brussels, Belgium;
| |
Collapse
|
4
|
Speranza E, Sorrentino I, Boletta A, Sitia R. Exquisite sensitivity of Polycystin-1 to H 2O 2 concentration in the endoplasmic reticulum. Redox Biol 2025; 80:103486. [PMID: 39787899 PMCID: PMC11763840 DOI: 10.1016/j.redox.2024.103486] [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: 10/29/2024] [Revised: 12/09/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Aquaporin11 (AQP11) is an endoplasmic reticulum (ER) resident peroxiporin. It allows H2O2 transport from the lumen to the cytosol, guaranteeing redox homeostasis and signaling in and between the two organelles. Interestingly, Aqp11-/- mice develop a fatal, early onset polycystic kidney disease (PKD) similar to Autosomal Dominant PKD, a condition frequently associated with mutations of polycystin-1 (PC-1) in human patients. Here we investigated the molecular mechanisms of AQP11-associated PKD. Using different cell models, we show that transient downregulation of AQP11 selectively prevents the biogenesis of overexpressed PC-1. Expression of catalase in the ER lumen rescues the phenotype, demonstrating a direct role of (H2O2)ER in controlling the complex maturation of PC-1. Analysis of endogenous Pc-1 revealed an additional regulatory role at the pre-translational level. Taken together, our results show that AQP11 controls the complex biogenesis of PC-1 at multiple levels governing H2O2 intra and inter-organellar fluxes, with important implications in the pathogenesis and onset of PKD.
Collapse
Affiliation(s)
- Elisa Speranza
- Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, 20132, Milan, Italy; Università Vita-Salute San Raffaele, 20132, Milan, Italy
| | - Ilaria Sorrentino
- Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, 20132, Milan, Italy; Università Vita-Salute San Raffaele, 20132, Milan, Italy
| | - Alessandra Boletta
- Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, 20132, Milan, Italy
| | - Roberto Sitia
- Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, 20132, Milan, Italy; Università Vita-Salute San Raffaele, 20132, Milan, Italy.
| |
Collapse
|
5
|
Ray A, Yang C, Stelloh C, Tutaj M, Liu P, Liu Y, Qiu Q, Auer PL, Lin CW, Widlansky ME, Geurts AM, Cowley AW, Liang M, Kwitek AE, Greene AS, Rao S. Chromatin State Maps of Blood Pressure-Relevant Renal Segments Reveal Potential Regulatory Role for SNPs. Hypertension 2025; 82:476-488. [PMID: 39723540 DOI: 10.1161/hypertensionaha.124.23873] [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: 08/21/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Hypertension or elevated blood pressure (BP) is a worldwide clinical challenge and the leading primary risk factor for kidney dysfunctions, heart failure, and cerebrovascular disease. The kidney is a central regulator of BP by maintaining sodium-water balance. Multiple genome-wide association studies revealed that BP is a heritable quantitative trait, modulated by several genetic, epigenetic, and environmental factors. The SNPs identified in genome-wide association studies predominantly (>95%) reside within noncoding genomic regions, making it difficult to understand how they regulate BP. Given the central role of the kidney in regulating BP, we hypothesized that chromatin-accessible regions in renal tissue would be enriched for BP-associated single nucleotide polymorphisms. METHODS We manually dissected 2 important kidney segments that maintain the sodium-water balance: proximal tubules and medullary thick ascending limbs from the human and rat kidneys. To delineate their chromatin and transcriptomic profiles, we performed the assay for transposase-accessible chromatin and RNA sequencing, respectively. RESULTS The chromatin accessibility maps revealed the shared and unique cis-regulatory elements that modulate the chromatin accessibility in proximal tubule and medullary thick ascending limbs of humans and rats. We developed a visualization tool to compare the cross-species epigenomic maps to identify potential regulatory targets for hypertension pathogenesis. We also identified a significant enrichment of BP-associated single nucleotide polymorphisms (1064 for human proximal tubule and 1172 for human medullary thick ascending limbs) within accessible chromatin regions of both segments, including rs1173771 and rs1421811 at the NPR3 locus and rs1800470 at the TGFb1 locus. CONCLUSIONS Collectively, this study lays a foundation for interrogating how intergenic single nucleotide polymorphisms may regulate polygenic traits such as BP.
Collapse
Affiliation(s)
- Atrayee Ray
- Versiti Blood Research Institute, Milwaukee, WI (A.R., C.S., S.R.)
| | - Chun Yang
- Department of Physiology (C.Y., M.T., A.M.G., A.W.C., A.E.K.), Medical College of Wisconsin, Milwaukee
| | - Cary Stelloh
- Versiti Blood Research Institute, Milwaukee, WI (A.R., C.S., S.R.)
| | - Monika Tutaj
- Department of Physiology (C.Y., M.T., A.M.G., A.W.C., A.E.K.), Medical College of Wisconsin, Milwaukee
| | - Pengyuan Liu
- Department of Physiology, University of Arizona, Tucson (P.L., Y.L., Q.Q., M.L.)
| | - Yong Liu
- Department of Physiology, University of Arizona, Tucson (P.L., Y.L., Q.Q., M.L.)
| | - Qiongzi Qiu
- Department of Physiology, University of Arizona, Tucson (P.L., Y.L., Q.Q., M.L.)
| | - Paul L Auer
- The Institute for Health and Equity (P.L.A.), Medical College of Wisconsin, Milwaukee
| | - Chien-Wei Lin
- Division of Biostatistics, Data Science Institute (C.-W.L.), Medical College of Wisconsin, Milwaukee
| | | | - Aron M Geurts
- Department of Physiology (C.Y., M.T., A.M.G., A.W.C., A.E.K.), Medical College of Wisconsin, Milwaukee
| | - Allen W Cowley
- Department of Physiology (C.Y., M.T., A.M.G., A.W.C., A.E.K.), Medical College of Wisconsin, Milwaukee
| | - Mingyu Liang
- Department of Physiology, University of Arizona, Tucson (P.L., Y.L., Q.Q., M.L.)
| | - Anne E Kwitek
- Department of Physiology (C.Y., M.T., A.M.G., A.W.C., A.E.K.), Medical College of Wisconsin, Milwaukee
| | | | - Sridhar Rao
- Versiti Blood Research Institute, Milwaukee, WI (A.R., C.S., S.R.)
- Department of Pediatrics, Section of Hematology/Oncology/Transplantation (S.R.), Medical College of Wisconsin, Milwaukee
- Department of Cell Biology, Neurobiology, and Anatomy (S.R.), Medical College of Wisconsin, Milwaukee
| |
Collapse
|
6
|
Fang L, Yi X, Shen J, Deng N, Peng X. Gut-brain axis mediated by intestinal content microbiota was associated with Zhishi Daozhi decoction on constipation. Front Cell Infect Microbiol 2025; 15:1539277. [PMID: 39963403 PMCID: PMC11830728 DOI: 10.3389/fcimb.2025.1539277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
Background Constipation is a common digestive system disorder, which is closely related to the intestinal flora. Zhishi Daozhi decoction (ZDD) is a traditional Chinese medicine prescription used to treat constipation caused by indigestion. This study is to evaluate the efficacy of ZDD in treating constipation and to elucidate the underlying mechanism. Methods In this study, Kunming mice were administered a high-protein diet (HFHPD) and loperamide hydrochloride injections to induce constipation. The mice then received varying doses (2.4, 4.7, and 9.4 mg/kg) of ZDD for seven days. Following the sampling process, we measured fecal microbial activity. The levels of 5-hydroxytryptamine (5-HT), vasoactive intestinal peptide (VIP), and aquaporin-3 (AQP3) were quantified using enzyme-linked immunosorbent assay. Changes in the gut microbiota were evaluated through 16S rRNA gene sequencing. Additionally, we investigated the correlation between specific microbiota features and the levels of 5-HT, VIP, and AQP3. Results The fecal surface of the mice in the model group (CMM) was rough and dry. The stool of mice in the low-dose ZDD group (CLD), medium-dose ZDD group (CMD), and high-dose ZDD group (CHD) exhibited a smoother texture, closely resembling that of the normal group (CNM). 5-HT levels in the CMM group were significantly lower than in the CNM, CLD, and CHD. VIP levels in the CMD were lower than in the other four groups, and AQP3 levels in CMM showed a decreasing trend. The fecal microbial activity of the CMM group was significantly higher than that of the other groups. Diversity analysis indicated that CMD and CHD treatments were more effective in restoring the intestinal microbiota structure. Potential pathogenic bacteria, including Clostridium, Aerococcus, Jeotgalicoccus, and Staphylococcus were enriched in CMM. In contrast, beneficial bacteria such as Faecalibacterium, Bacillaceae, and Bacillus were more prevalent in the CLD, CMD, and CHD. Correlation analysis revealed that Streptococcus and Enterococcus were positively correlated with VIP, while Succinivibrio showed a negative correlation with 5-HT. Conclusions Constipation induced by HFHPD and loperamide hydrochloride disrupts the structure of the intestinal microbiota. ZDD appears to alleviate constipation, potentially through mechanisms linked to the brain-gut axis and its interaction with the intestinal microbiota. Among the treatment groups, the medium dose of ZDD demonstrated the most effective results.
Collapse
Affiliation(s)
- Leyao Fang
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xin Yi
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Junxi Shen
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Na Deng
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xinxin Peng
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
7
|
Porter AW, Vorndran HE, Marciszyn A, Mutchler SM, Subramanya AR, Kleyman TR, Hendershot LM, Brodsky JL, Buck TM. Excess dietary sodium restores electrolyte and water homeostasis caused by loss of the endoplasmic reticulum molecular chaperone, GRP170, in the mouse nephron. Am J Physiol Renal Physiol 2025; 328:F173-F189. [PMID: 39556479 DOI: 10.1152/ajprenal.00192.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/15/2024] [Accepted: 11/06/2024] [Indexed: 01/16/2025] Open
Abstract
The maintenance of fluid and electrolyte homeostasis by the kidney requires proper folding and trafficking of ion channels and transporters in kidney epithelia. Each of these processes requires a specific subset of a diverse class of proteins termed molecular chaperones. One such chaperone is GRP170, which is an Hsp70-like, endoplasmic reticulum (ER)-localized chaperone that plays roles in protein quality control and protein folding in the ER. We previously determined that loss of GRP170 in the mouse nephron leads to hypovolemia, electrolyte imbalance, and rapid weight loss. In addition, GRP170-deficient mice develop an acute kidney injury (AKI)-like phenotype, typified by tubular injury, elevation of kidney injury markers, and induction of the unfolded protein response (UPR). By using an inducible GRP170 knockout cellular model, we confirmed that GRP170 depletion induces the UPR, triggers apoptosis, and disrupts protein homeostasis. Based on these data, we hypothesized that UPR induction underlies hyponatremia and volume depletion in these rodents and that these and other phenotypes might be rectified by sodium supplementation. To test this hypothesis, control and GRP170 tubule-specific knockout mice were provided a diet containing 8% sodium chloride. We discovered that sodium supplementation improved electrolyte imbalance and kidney injury markers in a sex-specific manner but was unable to restore weight or tubule integrity. These results are consistent with UPR induction contributing to the kidney injury phenotype in the nephron-specific GR170 knockout model and indicate that GRP170 function in kidney epithelia is essential to both maintain electrolyte balance and ER homeostasis.NEW & NOTEWORTHY Loss of the endoplasmic reticulum chaperone, GRP170, results in widespread kidney injury and induction of the unfolded protein response (UPR). We now show that sodium supplementation is able to at least partially restore electrolyte imbalance and reduce kidney injury markers in a sex-dependent manner.
Collapse
Affiliation(s)
- Aidan W Porter
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Division of Pediatric Nephrology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Hannah E Vorndran
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Allison Marciszyn
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Stephanie M Mutchler
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Arohan R Subramanya
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Linda M Hendershot
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Teresa M Buck
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
8
|
Vrettou CS, Issaris V, Kokkoris S, Poupouzas G, Keskinidou C, Lotsios NS, Kotanidou A, Orfanos SE, Dimopoulou I, Vassiliou AG. Exploring Aquaporins in Human Studies: Mechanisms and Therapeutic Potential in Critical Illness. Life (Basel) 2024; 14:1688. [PMID: 39768394 PMCID: PMC11676363 DOI: 10.3390/life14121688] [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: 12/02/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Aquaporins (AQPs) are membrane proteins facilitating water and other small solutes to be transported across cell membranes. They are crucial in maintaining cellular homeostasis by regulating water permeability in various tissues. Moreover, they regulate cell migration, signaling pathways, inflammation, tumor growth, and metastasis. In critically ill patients, such as trauma, sepsis, and patients with acute respiratory distress syndrome (ARDS), which are frequently encountered in intensive care units (ICUs), water transport regulation is crucial for maintaining homeostasis, as dysregulation can lead to edema or dehydration, with the latter also implicating hemodynamic compromise. Indeed, AQPs are involved in fluid transport in various organs, including the lungs, kidneys, and brain, where their dysfunction can exacerbate conditions like ARDS, acute kidney injury (AKI), or cerebral edema. In this review, we discuss the implication of AQPs in the clinical entities frequently encountered in ICUs, such as systemic inflammation and sepsis, ARDS, AKI, and brain edema due to different types of primary brain injury from a clinical perspective. Current and possible future therapeutic implications are also considered.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Alice G. Vassiliou
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (C.S.V.); (V.I.); (S.K.); (G.P.); (C.K.); (N.S.L.); (A.K.); (S.E.O.); (I.D.)
| |
Collapse
|
9
|
Salmon-Cabrales IS, de la Garza-Kalife DA, García-González G, Estrada-Rodríguez AE, Jiménez-Gutiérrez MA, Santoyo-Suárez MG, Rodríguez-Núñez O, Garza-Treviño EN, Benítez-Chao DF, Padilla-Rivas GR, Islas JF. Exploring the Functionality of the Krüppel-like Factors in Kidney Development, Metabolism, and Diseases. Life (Basel) 2024; 14:1671. [PMID: 39768378 PMCID: PMC11728015 DOI: 10.3390/life14121671] [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: 10/07/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
The kidneys contribute to the overall health of an organism by maintaining systemic homeostasis. This process involves various biological mechanisms, in which the Krüppel-like factors (KLFs), a family of transcription factors, are essential for regulating development, differentiation, proliferation, and cellular apoptosis. They also play a role in the metabolic regulation of essential nutrients, such as glucose and lipids. The dysregulation of these transcription factors is associated with the development of various pathologies, which can ultimately lead to renal fibrosis, severely compromising kidney function. In this context, the present article provides a comprehensive review of the existing literature, offering an enriching analysis of the findings related to the role of KLFs in nephrology, while also highlighting their potential therapeutic role in the treatment of renal diseases.
Collapse
Affiliation(s)
- Itzel S. Salmon-Cabrales
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - David A. de la Garza-Kalife
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Gabriel García-González
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Ana E. Estrada-Rodríguez
- Departmento de Ciencias Básicas, Vicerrectoría de Ciencias de la Salud, Universidad de Monterrey, Ignacio Morones Prieto 4500, Jesus M. Garza, San Pedro Garza García 66238, Nuevo León, Mexico;
| | - Marco Antonio Jiménez-Gutiérrez
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Michelle G. Santoyo-Suárez
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Oscar Rodríguez-Núñez
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Elsa N. Garza-Treviño
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Diego F. Benítez-Chao
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Gerardo R. Padilla-Rivas
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Jose Francisco Islas
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| |
Collapse
|
10
|
Parvez RK, Csipán RL, Liu J, Gevorgyan A, Rutledge EA, Guo J, Kim DK, McMahon AP. Developmental and Cell Fate Analyses Support a Postnatal Origin for the Cortical Collecting System in the Mouse Kidney. J Am Soc Nephrol 2024:00001751-990000000-00509. [PMID: 39665296 DOI: 10.1681/asn.0000000579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024] Open
Abstract
Key Points
An adult-like corticomedullary organization underlying kidney function is established 10 days after birth in the mouse kidney.Genetic lineage tracing demonstrates the cortical collecting duct network is generated from progenitors after birth.Mature cell types of the nephron progenitor–derived connecting tubule and ureteric progenitor–derived collecting epithelium are established by P15.
Background
Structure and function in the mammalian kidney are organized along a radial axis highlighted by the corticomedullary organization and regional patterning of the collecting system. The arborized collecting epithelium arises through controlled growth, branching, and commitment of Wnt11+ ureteric progenitor cells within cortically localized branch tips until postnatal day 3.
Methods
We applied in situ hybridization and immunofluorescence to key markers of collecting duct cell types to examine their distribution in the embryonic and postnatal mouse kidneys. To address the contribution of ureteric progenitor cells at a given time to cell diversity and spatial organization in the adult mouse kidney, we performed genetic lineage tracing of Wnt11
+
cells in the embryonic and early postnatal mouse kidney.
Results
Cell fate analyses showed much of the cortical collecting duct network was established postnatally. Furthermore, epithelial reorganization, regional differentiation, and functional maturation of key cell types to an adult-like collecting epithelium was not complete until around 2 weeks after birth in both ureteric progenitor cell–derived collecting system and structurally homologous nephron progenitor cell–derived connecting tubule.
Conclusions
These studies underline the importance of the relatively understudied early postnatal period to the development of a functional mammalian kidney.
Collapse
Affiliation(s)
- Riana K Parvez
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
| | - Réka L Csipán
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
| | - Jing Liu
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Penn Transplant Institute, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ara Gevorgyan
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
| | - Elisabeth A Rutledge
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
- Amgen, Inc., Thousand Oaks, California
| | - Jinjin Guo
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
| | - Doh Kyung Kim
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
| | - Andrew P McMahon
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
- Division of Biology and Biological Engineering, California Institute for Technology, Pasadena, California
| |
Collapse
|
11
|
Noel S, Kapoor R, Rabb H. New approaches to acute kidney injury. Clin Kidney J 2024; 17:65-81. [PMID: 39583139 PMCID: PMC11581771 DOI: 10.1093/ckj/sfae265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Indexed: 11/26/2024] Open
Abstract
Acute kidney injury (AKI) is a common and serious clinical syndrome that involves complex interplay between different cellular, molecular, metabolic and immunologic mechanisms. Elucidating these pathophysiologic mechanisms is crucial to identify novel biomarkers and therapies. Recent innovative methodologies and the advancement of existing technologies has accelerated our understanding of AKI and led to unexpected new therapeutic candidates. The aim of this review is to introduce and update the reader about recent developments applying novel technologies in omics, imaging, nanomedicine and artificial intelligence to AKI research, plus to provide examples where this can be translated to improve patient care.
Collapse
Affiliation(s)
- Sanjeev Noel
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Radhika Kapoor
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Hamid Rabb
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
12
|
Ariyo OW, Kwakye J, Sovi S, Aryal B, Hartono E, Ghareeb AFA, Milfort MC, Fuller AL, Rekaya R, Aggrey SE. mRNA expression of kidney aquaporins and blood composition of meat-type chickens raised with or without glucose supplementation under cyclic heat or thermoneutral condition. J Therm Biol 2024; 126:104003. [PMID: 39637608 DOI: 10.1016/j.jtherbio.2024.104003] [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: 06/05/2024] [Revised: 09/11/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024]
Abstract
Heat stress (HS) disrupts water homeostasis in broiler chickens. Kidney aquaporins (AQPs) facilitate water permeability in the renal tubules, thereby maintaining homeostasis of body water and metabolites. We evaluated the mRNA expression of kidney AQPs 1, 2, 3 and 4, and the blood composition of broilers raised under thermoneutral (TN) or cyclic HS condition with or without glucose supplementation. The treatments were TN+0% glucose (TN0), TN+6% glucose (TN6), HS+0% glucose (HS0), and HS+6% glucose (HS6). Each treatment had 6 replicates of 19 birds each. Groups with glucose (Glu) supplementation continuously received 6% Glu in water, and HS groups were exposed to a cyclic HS regime involving 35 °C from 8am to 8pm, and subsequently retuned to 25 °C (thermoneutral) from 8pm to 8am. Both heat and glucose supplementation were applied from d28 to d35. Blood and kidney were collected from 1 bird/replicate on d35. AQP-1, -2, and -3 were highly expressed (p < 0.05) in HS0 relative to TN0, TN6, and HS6. Birds in the TN6 and HS6 had a lower expression of AQP2 while HS0 and HS6 had a lower expression of AQP4. Birds in the HS0 group had a higher water consumption relative to TN0, TN6 and HS6. HS0 and HS6 had a higher water conversion ratio relative to TN0 and TN6. Heat stress lowered (p < 0.05) blood PCO2, TCO2, and HCO3. The blood pH, Na, K, and Cl composition were not affected (p > 0.05) with heat or glucose supplementation. Cyclic HS (p < 0.0001) and glucose supplementation (p = 0.0090) raised the blood glucose level. Kidney aquaporins 1, 2, and 3 are important indicators of water utilization in HS broiler chickens.
Collapse
Affiliation(s)
- Oluwatomide W Ariyo
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602, USA
| | - Josephine Kwakye
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602, USA
| | - Selorm Sovi
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602, USA
| | - Bikash Aryal
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602, USA
| | - Evan Hartono
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602, USA
| | - Ahmed F A Ghareeb
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602, USA
| | - Marie C Milfort
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602, USA
| | - Alberta L Fuller
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602, USA
| | - Romdhane Rekaya
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, 30602, USA
| | - Samuel E Aggrey
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
13
|
Qiao R, Cui X, Hu Y, Wei H, Xu H, Zhang C, Du C, Chang J, Li Y, Ming W, Qi Y, Guan Y, Zhang X. Hypoxia Reduces Mouse Urine Output via HIF1α-Mediated Upregulation of Renal AQP1. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:504-518. [PMID: 39664329 PMCID: PMC11631171 DOI: 10.1159/000542087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 10/14/2024] [Indexed: 12/13/2024]
Abstract
Introduction Patients with acute mountain sickness (AMS) due to hypoxia at high altitudes often exhibit abnormal water metabolism. Hypoxia-inducible factors (HIFs) are major regulators of adaptive responses to hypoxia. As transcription factors, HIFs are involved in the regulation of erythropoiesis, iron metabolism, angiogenesis, energy metabolism, and cell survival by promoting the transcriptional expression of hundreds of target genes. Roxadustat, a novel drug for the treatment of anemia associated with chronic kidney disease (CKD), acts by inhibiting the degradation of HIFs to increase their protein levels. However, the clinical use of roxadustat is frequently associated with peripheral edema, suggesting the involvement of HIFs in regulating the body's water balance possibly by modulating water reabsorption in the kidney. Methods We first evaluated the effect of hypoxia (8% O2) on mouse urine output. We then performed in vitro experiments using hypoxia (1% O2) and roxadustat on mouse primary proximal tubular cells (mPTCs). The quantitative polymerase chain reaction, Western blot, and immunofluorescence were used to assess AQP1 mRNA and protein expression levels. Luciferase, Chromatin immunoprecipitation (ChIP), and electrophoretic mobility shift assay (EMSA) were used to investigate the transcriptional regulation of AQP1 by HIF1α. Results We found that mice exposed to hypoxia (8% O2) had significantly reduced urine volume compared to mice exposed to normoxia (21% O2). Hypoxia significantly elevated AQP1 expression at both mRNA and protein levels. In vitro experiments using mouse primary cultured proximal tubular cells (mPTCs) revealed that both hypoxia and roxadustat increased AQP1 expression. Mechanistically, overexpression of HIF1α, but not HIF2α, markedly increased AQP1 protein expression. Furthermore, the upregulation of AQP1 by hypoxia and roxadustat can be blocked by the HIF1α inhibitor PX-478 in mPTCs. Finally, we found that the AQP1 gene promoter contains a putative hypoxia response element and confirmed that AQP1 is a target gene of HIF1α using Luciferase reporter, ChIP, and EMSA assays. Conclusion This study demonstrates that hypoxia can reduce the urine volume of mice via upregulating AQP1 expression by HIF1α in the proximal tubular epithelial cells. Our findings also suggest a potential mechanism involved in water metabolism disorders in patients with AMS and in patients with CKD receiving roxadustat treatment.
Collapse
Affiliation(s)
- Rongfang Qiao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaohui Cui
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Yitong Hu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Haoqing Wei
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Hu Xu
- Kidney Health Institute, Health Science Center, East China Normal University, Shanghai, China
| | - Cong Zhang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Chunxiu Du
- Kidney Health Institute, Health Science Center, East China Normal University, Shanghai, China
| | - Jiazhen Chang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Yaqing Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Wenhua Ming
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Yinghui Qi
- Department of Nephrology, Pudong New District Punan Hospital, Shanghai, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaoyan Zhang
- Kidney Health Institute, Health Science Center, East China Normal University, Shanghai, China
| |
Collapse
|
14
|
Carty JS, Bessho R, Zuchowski Y, Trapani JB, Davidoff O, Kobayashi H, Roland JT, Watts JA, Terker AS, Bock F, Arroyo JP, Haase VH. Disruption of mitochondrial electron transport impairs urinary concentration via AMPK-dependent suppression of aquaporin 2. JCI Insight 2024; 9:e182087. [PMID: 39361429 PMCID: PMC11601893 DOI: 10.1172/jci.insight.182087] [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: 04/18/2024] [Accepted: 10/02/2024] [Indexed: 10/05/2024] Open
Abstract
Urinary concentration is an energy-dependent process that minimizes body water loss by increasing aquaporin 2 (AQP2) expression in collecting duct (CD) principal cells. To investigate the role of mitochondrial (mt) ATP production in renal water clearance, we disrupted mt electron transport in CD cells by targeting ubiquinone (Q) binding protein QPC (UQCRQ), a subunit of mt complex III essential for oxidative phosphorylation. QPC-deficient mice produced less concentrated urine than controls, both at baseline and after type 2 vasopressin receptor stimulation with desmopressin. Impaired urinary concentration in QPC-deficient mice was associated with reduced total AQP2 protein levels in CD tubules, while AQP2 phosphorylation and membrane trafficking remained unaffected. In cultured inner medullary CD cells treated with mt complex III inhibitor antimycin A, the reduction in AQP2 abundance was associated with activation of 5' adenosine monophosphate-activated protein kinase (AMPK) and was reversed by treatment with AMPK inhibitor SBI-0206965. In summary, our studies demonstrated that the physiological regulation of AQP2 abundance in principal CD cells was dependent on mt electron transport. Furthermore, our data suggested that oxidative phosphorylation in CD cells was dispensable for maintaining water homeostasis under baseline conditions, but necessary for maximal stimulation of AQP2 expression and urinary concentration.
Collapse
Affiliation(s)
- Joshua S. Carty
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Ryoichi Bessho
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Yvonne Zuchowski
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jonathan B. Trapani
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Olena Davidoff
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Research and Medical Services, Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Hanako Kobayashi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Research and Medical Services, Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Joseph T. Roland
- Section of Surgical Sciences, Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jason A. Watts
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, USA
| | - Andrew S. Terker
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Fabian Bock
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Juan Pablo Arroyo
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Volker H. Haase
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Research and Medical Services, Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
15
|
Reyer H, Honerlagen H, Oster M, Ponsuksili S, Kuhla B, Wimmers K. Multi-tissue gene expression profiling of cows with a genetic predisposition for low and high milk urea levels. Anim Biotechnol 2024; 35:2322542. [PMID: 38426941 DOI: 10.1080/10495398.2024.2322542] [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] [Indexed: 03/02/2024]
Abstract
Milk urea (MU) concentration is proposed as an indicator trait for breeding toward reduced nitrogen (N) emissions and leaching in dairy. We selected 20 German Holstein cows based on MU breeding values, with 10 cows each having low (LMUg) and high (HMUg) MU genetic predisposition. Using RNA-seq, we characterized these cows to unravel molecular pathways governing post-absorptive body N pools focusing on renal filtration and reabsorption of nitrogenous compounds, hepatic urea formation and mammary gland N excretion. While we observed minor adjustments in cellular energy metabolism in different tissues associated with different MU levels, no transcriptional differences in liver ammonia detoxification were detected, despite significant differences in MU between the groups. Differential expression of AQP3 and SLC38A2 in the kidney provides evidence for higher urea concentration in the collecting duct of LMU cows than HMU cows. The mammary gland exhibited the most significant differences, particularly in tricarboxylic acid (TCA) cycle genes, amino acid transport, tRNA binding, and casein synthesis. These findings suggest that selecting for lower MU could lead to altered urinary urea (UU) handling and changes in milk protein synthesis. However, given the genetic variability in N metabolism components, the long-term effectiveness of MU-based selection in reducing N emissions remains uncertain.
Collapse
Affiliation(s)
- Henry Reyer
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Hanne Honerlagen
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
- Animal Breeding and Genomics, Wageningen University and Research, Wageningen, The Netherlands
| | - Michael Oster
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Siriluck Ponsuksili
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Björn Kuhla
- Institute of Nutritional Physiology 'Oskar Kellner', Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Klaus Wimmers
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
- Faculty of Agriculture and Environmental Sciences, Professorship of Animal Breeding and Genetics, University of Rostock, Rostock, Germany
| |
Collapse
|
16
|
Rump K, Adamzik M. Aquaporins in sepsis- an update. Front Immunol 2024; 15:1495206. [PMID: 39544938 PMCID: PMC11560437 DOI: 10.3389/fimmu.2024.1495206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 10/14/2024] [Indexed: 11/17/2024] Open
Abstract
Aquaporins (AQPs), a family of membrane proteins that facilitate the transport of water and small solutes, have garnered increasing attention for their role in sepsis, not only in fluid balance but also in immune modulation and metabolic regulation. Sepsis, characterized by an excessive and dysregulated immune response to infection, leads to widespread organ dysfunction and significant mortality. This review focuses on the emerging roles of aquaporins in immune metabolism and their potential as therapeutic targets in sepsis, with particular attention to the modulation of inflammatory responses and organ protection. Additionally, it explores the diverse roles of aquaporins across various organ systems, highlighting their contributions to renal function, pulmonary gas exchange, cardiac protection, and gastrointestinal barrier integrity in the context of sepsis. Recent studies suggest that AQPs, particularly aquaglyceroporins like AQP3, AQP7, AQP9, and AQP10, play pivotal roles in immune cell metabolism and offer new therapeutic avenues for sepsis treatment. In the context of sepsis, immune cells undergo metabolic shifts to meet the heightened energy demands of the inflammatory response. A key adaptation is the shift from oxidative phosphorylation (OXPHOS) to aerobic glycolysis, where pyruvate is converted to lactate, enabling faster ATP production. AQPs, particularly aquaglyceroporins, may facilitate this process by transporting glycerol, a substrate that fuels glycolysis. AQP3, for example, enhances glucose metabolism by transporting glycerol and complementing glucose uptake via GLUT1, while also regulating O-GlcNAcylation, a post-translational modification that boosts glycolytic flux. AQP7 could further contributes to immune cell energy production by influencing lipid metabolism and promoting glycolysis through p38 signaling. These mechanisms could be crucial for maintaining the energy supply needed for an effective immune response during sepsis. Beyond metabolism, AQPs also regulate key immune functions. AQP9, highly expressed in septic patients, is essential for neutrophil migration and activation, both of which are critical for controlling infection. AQP3, on the other hand, modulates inflammation through the Toll-like receptor 4 (TLR4) pathway, while AQP1 plays a role in immune responses by activating the PI3K pathway, promoting macrophage polarization, and protecting against lipopolysaccharide (LPS)-induced acute kidney injury (AKI). These insights into the immunoregulatory roles of AQPs suggest their potential as therapeutic targets to modulate inflammation in sepsis. Therapeutically, AQPs present promising targets for reducing organ damage and improving survival in sepsis. For instance, inhibition of AQP9 with compounds like HTS13286 or RG100204 has been shown to reduce inflammation and improve survival by modulating NF-κB signaling and decreasing oxidative stress in animal models. AQP5 inhibition with methazolamide and furosemide has demonstrated efficacy in reducing immune cell migration and lung injury, suggesting its potential in treating acute lung injury (ALI) in sepsis. Additionally, the regulation of AQP1 through non-coding RNAs (lncRNAs and miRNAs) may offer new strategies to mitigate organ damage and inflammatory responses. Moreover, AQPs have emerged as potential biomarkers for sepsis progression and outcomes. Altered expression of AQPs, such as AQP1, AQP3, and AQP5, correlates with sepsis severity, and polymorphisms in AQP5 have been linked to better survival rates and improved outcomes in sepsis-related acute respiratory distress syndrome (ARDS). This suggests that AQP expression could be used to stratify patients and tailor treatments based on individual AQP profiles. In conclusion, AQPs play a multifaceted role in the pathophysiology of sepsis, extending beyond fluid balance to crucial involvement in immune metabolism and inflammation. Targeting AQPs offers novel therapeutic strategies to mitigate sepsis-induced organ damage and improve patient survival. Continued research into the metabolic and immune functions of AQPs will be essential for developing targeted therapies that can be translated into clinical practice.
Collapse
Affiliation(s)
- Katharina Rump
- Klinik für Anästhesiologie Intensivmedizin und Schmerztherapie Universitätsklinikum Knappschaftskrankenhaus Bochum, University Clinic of Ruhr University Bochum, Bochum, Germany
| | | |
Collapse
|
17
|
Polonsky M, Gerhardt LMS, Yun J, Koppitch K, Colón KL, Amrhein H, Wold B, Zheng S, Yuan GC, Thomson M, Cai L, McMahon AP. Spatial transcriptomics defines injury specific microenvironments and cellular interactions in kidney regeneration and disease. Nat Commun 2024; 15:7010. [PMID: 39237549 PMCID: PMC11377535 DOI: 10.1038/s41467-024-51186-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 08/01/2024] [Indexed: 09/07/2024] Open
Abstract
Kidney injury disrupts the intricate renal architecture and triggers limited regeneration, together with injury-invoked inflammation and fibrosis. Deciphering the molecular pathways and cellular interactions driving these processes is challenging due to the complex tissue structure. Here, we apply single cell spatial transcriptomics to examine ischemia-reperfusion injury in the mouse kidney. Spatial transcriptomics reveals injury-specific and spatially-dependent gene expression patterns in distinct cellular microenvironments within the kidney and predicts Clcf1-Crfl1 in a molecular interplay between persistently injured proximal tubule cells and their neighboring fibroblasts. Immune cell types play a critical role in organ repair. Spatial analysis identifies cellular microenvironments resembling early tertiary lymphoid structures and associated molecular pathways. Collectively, this study supports a focus on molecular interactions in cellular microenvironments to enhance understanding of injury, repair and disease.
Collapse
Affiliation(s)
- Michal Polonsky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Louisa M S Gerhardt
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
- Fifth Department of Medicine, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Jina Yun
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Kari Koppitch
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Katsuya Lex Colón
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Henry Amrhein
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Barbara Wold
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Shiwei Zheng
- Department of Genetics and Genomic Sciences and Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences and Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Long Cai
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
18
|
Lv W, Liao J, Li C, Liu D, Luo X, Diao R, Wang Y, Jin Y. Aquaporin 1 is renoprotective in septic acute kidney injury by attenuating inflammation, apoptosis and fibrosis through inhibition of P53 expression. Front Immunol 2024; 15:1443108. [PMID: 39238634 PMCID: PMC11374652 DOI: 10.3389/fimmu.2024.1443108] [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: 06/03/2024] [Accepted: 07/29/2024] [Indexed: 09/07/2024] Open
Abstract
Sepsis associated Acute kidney injury (AKI) is a common clinical syndrome characterized by suddenly decreased in renal function and urinary volume. This study was designed to investigate the role of Aquaporin 1 (AQP1) and P53 in the development of sepsis-induced AKI and their potential regulatory mechanisms. Firstly, transcriptome sequencing analysis of mice kidney showed AQP1 expression was reduced and P53 expression was elevated in Cecal ligation and puncture (CLP)-induced AKI compared with controls. Bioinformatics confirmed that AQP1 expression was remarkably decreased and P53 expression was obviously elevated in renal tissues or peripheral blood of septic AKI patients. Moreover, we found in vivo experiments that AQP1 mRNA levels were dramatically decreased and P53 mRNA significantly increased following the increased expression of inflammation, apoptosis, fibrosis, NGAL and KIM-1 at various periods in septic AKI. Meanwhile, AQP1 and P53 protein levels increased significantly first and then decreased gradually in kidney tissue and serum of rats in different stages of septic AKI. Most importantly, in vivo and vitro experiments demonstrated that silencing of AQP1 greatly exacerbates renal or cellular injury by up-regulating P53 expression promoting inflammatory response, apoptosis and fibrosis. Overexpression of AQP1 prevented the elevation of inflammation, apoptosis and fibrosis by down-regulating P53 expression in Lipopolysaccharide (LPS)-induced AKI or HK-2 cells. Therefore, our results suggested that AQP1 plays a protective role in modulating AKI and can attenuate inflammatory response, apoptosis and fibrosis via downregulating P53 in septic AKI or LPS-induced HK-2cells. The pharmacological targeting of AQP1 mediated P53 expression might be identified as potential targets for the early treatment of septic AKI.
Collapse
Affiliation(s)
- Wuyang Lv
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Department of Clinical Laboratory, Shangluo Central Hospital, Shangluo, Shaanxi, China
| | - Jia Liao
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Cuicui Li
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Dongyang Liu
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaoxiao Luo
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - RuXue Diao
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - YuChen Wang
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yingyu Jin
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
19
|
Soltani-Fard E, Taghvimi S, Karimi F, Vahedi F, Khatami SH, Behrooj H, Deylami Hayati M, Movahedpour A, Ghasemi H. Urinary biomarkers in diabetic nephropathy. Clin Chim Acta 2024; 561:119762. [PMID: 38844018 DOI: 10.1016/j.cca.2024.119762] [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: 04/18/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Diabetic nephropathy (DN), a significant consequence of diabetes, is associated with adverse cardiovascular and renal disease as well as mortality. Although microalbuminuria is considered the best non-invasive marker for DN, better predictive markers are needed of sufficient sensitivity and specificity to detect disease in general and in early disease specifically. Even prior to appearance of microalbuminuria, urinary biomarkers increase in diabetics and can serve as accurate nephropathy biomarkers even in normoalbuminuria. In this review, a number of novel urine biomarkers including those reflecting kidney damage caused by glomerular/podocyte damage, tubular damage, oxidative stress, inflammation, and intrarenal renin-angiotensin system activation are discussed. Our review also includes emerging biomarkers such as urinary microRNAs. These short noncoding miRNAs regulate gene expression and could be utilized to identify potential novel biomarkers in DN development and progression. .
Collapse
Affiliation(s)
- Elahe Soltani-Fard
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran; Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sina Taghvimi
- Department of Biology, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | | | - Farzaneh Vahedi
- Biomedical and Microbial Advanced Technologies Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Seyyed Hossein Khatami
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | | | - Hassan Ghasemi
- Research Center for Environmental Contaminants (RCEC), Abadan University of Medical Sciences, Abadan, Iran.
| |
Collapse
|
20
|
Candan B, Ilhan I, Sarman E, Sevimli M. Irbesartan restored aquaporin-1 levels via inhibition of NF-kB expression in acute kidney injury model. Nefrologia 2024; 44:540-548. [PMID: 39216981 DOI: 10.1016/j.nefroe.2023.11.003] [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: 07/20/2023] [Revised: 11/23/2023] [Accepted: 11/26/2023] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION Acute kidney injury (AKI) is a serious pathology that progress with dysfunction of regulating blood pressure and fluid balance, concentrating urine due to decrement of aquaporin-1 (AQP) levels during the inflammation process. Irbesartan (IRN), angiotensin receptor blocker, is widely used in the treatment of hypertension, which also has anti-inflammatory, antioxidant and anti-apoptotic properties. The aim of this study is to investigate the protective effects of IRN in lipopolysaccharide (LPS)-induced kidney injury. MATERIAL AND METHODS Twenty-four rats divided into three groups as control, LPS and LPS+IRN group. After 6h of LPS administration, rats were sacrificed. Blood samples and half of the kidney tissues were collected for biochemical analysis and remaining tissues were taken for histopathological and immunohistochemical analysis. RESULTS In the LPS group, glomerular congestion and shrinkage, degeneration of distal tubules, mononuclear cell infiltration, cellular debris and intense proteinous accumulation in the tubules, increased expressions of Cas-3, nuclear factor kappa beta-p65 (NF-kB p65), levels of creatinin, TOS, OSI and decreased levels of TAS, AQP-1 were found significantly. IRN treatment reversed all these parameters. IRN's restorated AQP-1 levels by its anti-inflammatory, antioxidant and anti-apoptotic effects due to inhibiting NF-kB expression. CONCLUSION This study suggests that IRN can be used in conditions affecting the kidneys such as AKI. Further studies needed for detailed molecular investigation of IRN at different doses and durations.
Collapse
Affiliation(s)
- Busra Candan
- Alanya Alaaddin Keykubat University, School of Medicine, Department of Anatomy, Alanya, Turkey
| | - Ilter Ilhan
- Suleyman Demirel University, Faculty of Medicine, Department of Biochemistry, Isparta, Turkey
| | - Emine Sarman
- Afyonkarahisar Health Sciences University, Faculty of Medicine, Department of Histology and Embryology, Afyonkarahisar, Turkey.
| | - Murat Sevimli
- Suleyman Demirel University, Faculty of Medicine, Department of Histology and Embryology, Isparta, Turkey
| |
Collapse
|
21
|
Candan B, Ilhan I, Sarman E, Sevimli M. Irbesartan restored aquaporin-1 levels via inhibition of NF-kB expression in acute kidney injury model. Nefrologia 2024; 44:540-548. [DOI: 10.1016/j.nefro.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024] Open
|
22
|
Kharin A, Klussmann E. Many kinases for controlling the water channel aquaporin-2. J Physiol 2024; 602:3025-3039. [PMID: 37440212 DOI: 10.1113/jp284100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/26/2023] [Indexed: 07/14/2023] Open
Abstract
Aquaporin-2 (AQP2) is a member of the aquaporin water channel family. In the kidney, AQP2 is expressed in collecting duct principal cells where it facilitates water reabsorption in response to antidiuretic hormone (arginine vasopressin, AVP). AVP induces the redistribution of AQP2 from intracellular vesicles and its incorporation into the plasma membrane. The plasma membrane insertion of AQP2 represents the crucial step in AVP-mediated water reabsorption. Dysregulation of the system preventing the AQP2 plasma membrane insertion causes diabetes insipidus (DI), a disease characterised by an impaired urine concentrating ability and polydipsia. There is no satisfactory treatment of DI available. This review discusses kinases that control the localisation of AQP2 and points out potential kinase-directed targets for the treatment of DI.
Collapse
Affiliation(s)
- Andrii Kharin
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Enno Klussmann
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
23
|
Obi Y, Raimann JG, Kalantar-Zadeh K, Murea M. Residual Kidney Function in Hemodialysis: Its Importance and Contribution to Improved Patient Outcomes. Toxins (Basel) 2024; 16:298. [PMID: 39057938 PMCID: PMC11281084 DOI: 10.3390/toxins16070298] [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: 04/30/2024] [Revised: 05/27/2024] [Accepted: 06/11/2024] [Indexed: 07/28/2024] Open
Abstract
Individuals afflicted with advanced kidney dysfunction who require dialysis for medical management exhibit different degrees of native kidney function, called residual kidney function (RKF), ranging from nil to appreciable levels. The primary focus of this manuscript is to delve into the concept of RKF, a pivotal yet under-represented topic in nephrology. To begin, we unpack the definition and intrinsic nature of RKF. We then juxtapose the efficiency of RKF against that of hemodialysis in preserving homeostatic equilibrium and facilitating physiological functions. Given the complex interplay of RKF and overall patient health, we shed light on the extent of its influence on patient outcomes, particularly in those living with advanced kidney dysfunction and on dialysis. This manuscript subsequently presents methodologies and measures to assess RKF, concluding with the potential benefits of targeted interventions aimed at preserving RKF.
Collapse
Affiliation(s)
- Yoshitsugu Obi
- Division of Nephrology, Department of Medicine, The University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Jochen G. Raimann
- Renal Research Institute, New York, NY 10065, USA;
- Katz School of Science and Health, Yeshiva University, New York, NY 10033, USA
| | - Kamyar Kalantar-Zadeh
- Tibor Rubin Veterans Affairs Long Beach Healthcare System, Long Beach, CA 90822, USA;
- The Lundquist Institute at Harbor, UCLA Medical Center, Torrance, CA 90502, USA
- Division of Nephrology, Hypertension, and Kidney Transplantation, University of California Irvine, Orange, CA 92868, USA
| | - Mariana Murea
- Department of Internal Medicine, Section on Nephrology, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|
24
|
Guo Y, Qiao R, Xie G, Yao Y, Du C, Shao Y, Guan Y, Zhang X. Activation of TGR5 Increases Urine Concentration by Inducing AQP2 and AQP3 Expression in Renal Medullary Collecting Ducts. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:181-192. [PMID: 38835402 PMCID: PMC11149995 DOI: 10.1159/000538107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 02/26/2024] [Indexed: 06/06/2024]
Abstract
Introduction G protein-coupled bile acid receptor (TGR5), the first G protein-coupled receptor for bile acids identified, is capable of activating a variety of intracellular signaling pathways after interacting with bile acids. TGR5 plays an important role in multiple physiological processes and is considered to be a potential target for the treatment of various metabolic diseases, including type 2 diabetes. Evidence has emerged that genetic deletion of TGR5 results in an increase in basal urine output, suggesting that it may play a critical role in renal water and salt reabsorption. The present study aims to elucidate the effect and mechanism of TGR5 activation on urine concentration. Methods Mice were treated with TGR5 agonists (LCA and INT-777) for 3 days. The 24-h urine of mice was collected and analyzed for urine biochemical parameters. The mRNA expressions were detected by real-time PCR, and the protein expressions were detected by western blot. Immunohistochemistry and immunofluorescence were performed to examine the cellular location of proteins. The cultured primary medullary collecting duct cells were pretreated with H89 (a PKA inhibitor) for 1 h, followed by 12-h treatment of LCA and INT-777. Luciferase reporter assays were used to detect the effect of CREB on the gene transcription of AQPs. Gel electrophoretic mobility shift assays were used to analyze DNA-protein interactions. Results Treatment of mice with the TGR5 agonist LCA and INT-777 markedly reduced urine output and increased urine osmolality, accompanied by a marked increase in AQP2 and AQP3 protein expression and membrane translocation. In cultured primary medullary collecting duct cells, LCA and INT-777 dose-dependently upregulated AQP2 and AQP3 expression in a cAMP/PKA-dependent manner. Mechanistically, both AQP2 and AQP3 gene promoter contains a putative CREB-binding site, which can be bound and activated by CREB as assessed by both gene promoter-driven luciferase and gel shift assays. Conclusion Collectively, our findings demonstrate that activation of TGR5 can promote urine concentration by upregulation of AQP2 and AQP3 expression in renal collecting ducts. TGR5 may represent an attractive target for the treatment of patients with urine concentration defect.
Collapse
Affiliation(s)
- Yanlin Guo
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, China
- Health Science Center, East China Normal University, Shanghai, China
| | - Rongfang Qiao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Guixiang Xie
- Health Science Center, East China Normal University, Shanghai, China
| | - Yao Yao
- Division of Nephrology, Affiliated Hospital and Medical School, Nantong University, Nantong, China
| | - Chunxiu Du
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, China
- Health Science Center, East China Normal University, Shanghai, China
| | - Yunxia Shao
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaoyan Zhang
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, China
- Health Science Center, East China Normal University, Shanghai, China
| |
Collapse
|
25
|
Lan Q, Li J, Zhang H, Zhou Z, Fang Y, Yang B. Mechanistic complement of autosomal dominant polycystic kidney disease: the role of aquaporins. J Mol Med (Berl) 2024; 102:773-785. [PMID: 38668786 DOI: 10.1007/s00109-024-02446-4] [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: 01/09/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 05/21/2024]
Abstract
Autosomal dominant polycystic kidney disease is a genetic kidney disease caused by mutations in the genes PKD1 or PKD2. Its course is characterized by the formation of progressively enlarged cysts in the renal tubules bilaterally. The basic genetic explanation for autosomal dominant polycystic kidney disease is the double-hit theory, and many of its mechanistic issues can be explained by the cilia doctrine. However, the precise molecular mechanisms underpinning this condition's occurrence are still not completely understood. Experimental evidence suggests that aquaporins, a class of transmembrane channel proteins, including aquaporin-1, aquaporin-2, aquaporin-3, and aquaporin-11, are involved in the mechanism of autosomal dominant polycystic kidney disease. Aquaporins are either a potential new target for the treatment of autosomal dominant polycystic kidney disease, and further study into the physiopathological role of aquaporins in autosomal dominant polycystic kidney disease will assist to clarify the disease's pathophysiology and increase the pool of potential treatment options. We primarily cover pertinent findings on aquaporins in autosomal dominant polycystic kidney disease in this review.
Collapse
Affiliation(s)
- Qiumei Lan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Jie Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Hanqing Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Zijun Zhou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Yaxuan Fang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Bo Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
- Department of Nephrology, The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, No.88, Changling Road, Xiqing District, Tianjin, 300193, China.
| |
Collapse
|
26
|
Dai M, Yang J, Wang Z, Xue F, Wang Y, Hu E, Gong Y, Routledge MN, Qiao B. Aquaporins alteration revealed kidney damages in cerebral ischemia/reperfusion rats. Heliyon 2024; 10:e31532. [PMID: 38807874 PMCID: PMC11130722 DOI: 10.1016/j.heliyon.2024.e31532] [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: 01/22/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024] Open
Abstract
Background Restoration of blood supply is a desired goal for the treatment of acute ischemic stroke. However, the restoration often leads to cerebral ischemia-reperfusion injury (CIR/I), which greatly increases the risk of non-neural organ damage. In particular, the acute kidney injury might be one of the most common complications. Aims The study aimed to understand the damage occurred and the potential molecular mechanisms. Methods The study was explored on the CIR/I rats generated by performing middle cerebral artery occlusion/reperfusion (MCAO/Reperfusion). The rats were evaluated with injury on the brains, followed by the non-neural organs including kidneys, livers, colons and stomachs. They were examined further with histopathological changes, and gene expression alterations by using RT-qPCR of ten aquaporins (Aqps) subtypes including Aqp1~Aqp9 and Aqp11. Furthermore, the Aqps expression profiles were constructed for each organ and analyzed by performing Principle Component Analysis. In addition, immunohistochemistry was explored to look at the protein expression of Aqp1, Aqp2, Aqp3 and Aqp4 in the rat kidneys. Results There was a prominent down-regulation profile in the MCAO/Reperfusion rat kidneys. The protein expression of Aqp1, Aqp2, Aqp3 and Aqp4 was decreased in the kidneys of the MCAO/Reperfusion rats. We suggested that the kidney was in the highest risk to be damaged following the CIR/I. Down-regulation of Aqp2, Aqp3 and Aqp4 was involved in the acute kidney injury induced by the CIR/I.
Collapse
Affiliation(s)
- Meng Dai
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Ministry of Education, Northwest University, No. 229 TaiBai North Road, Xi'an, Shaanxi Province, 710069, PR China
- Shaanxi Traditional Chinese Medicine Innovation Engineering Technology Research Center, No. 229 Taibai North Road, Xi'an, Shaanxi Province, 710069, PR China
| | - Jinglei Yang
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Ministry of Education, Northwest University, No. 229 TaiBai North Road, Xi'an, Shaanxi Province, 710069, PR China
- Shaanxi Traditional Chinese Medicine Innovation Engineering Technology Research Center, No. 229 Taibai North Road, Xi'an, Shaanxi Province, 710069, PR China
| | - Zhaoyang Wang
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Ministry of Education, Northwest University, No. 229 TaiBai North Road, Xi'an, Shaanxi Province, 710069, PR China
- Shaanxi Traditional Chinese Medicine Innovation Engineering Technology Research Center, No. 229 Taibai North Road, Xi'an, Shaanxi Province, 710069, PR China
| | - Fangli Xue
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Ministry of Education, Northwest University, No. 229 TaiBai North Road, Xi'an, Shaanxi Province, 710069, PR China
- Shaanxi Traditional Chinese Medicine Innovation Engineering Technology Research Center, No. 229 Taibai North Road, Xi'an, Shaanxi Province, 710069, PR China
| | - Yourui Wang
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Ministry of Education, Northwest University, No. 229 TaiBai North Road, Xi'an, Shaanxi Province, 710069, PR China
- Shaanxi Traditional Chinese Medicine Innovation Engineering Technology Research Center, No. 229 Taibai North Road, Xi'an, Shaanxi Province, 710069, PR China
| | - Enjie Hu
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Ministry of Education, Northwest University, No. 229 TaiBai North Road, Xi'an, Shaanxi Province, 710069, PR China
- Shaanxi Traditional Chinese Medicine Innovation Engineering Technology Research Center, No. 229 Taibai North Road, Xi'an, Shaanxi Province, 710069, PR China
| | - Yunyun Gong
- School of Medicine, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Michael N. Routledge
- School of Medicine, University of Leicester, Leicester, LE1 7RH, United Kingdom
- Jiangsu University, Sch Food & Biol Engn, Zhenjiang, 212013, PR China
| | - Boling Qiao
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Ministry of Education, Northwest University, No. 229 TaiBai North Road, Xi'an, Shaanxi Province, 710069, PR China
- Shaanxi Traditional Chinese Medicine Innovation Engineering Technology Research Center, No. 229 Taibai North Road, Xi'an, Shaanxi Province, 710069, PR China
| |
Collapse
|
27
|
Li H, Li D, Ledru N, Xuanyuan Q, Wu H, Asthana A, Byers LN, Tullius SG, Orlando G, Waikar SS, Humphreys BD. Transcriptomic, epigenomic, and spatial metabolomic cell profiling redefines regional human kidney anatomy. Cell Metab 2024; 36:1105-1125.e10. [PMID: 38513647 PMCID: PMC11081846 DOI: 10.1016/j.cmet.2024.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/20/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024]
Abstract
A large-scale multimodal atlas that includes major kidney regions is lacking. Here, we employed simultaneous high-throughput single-cell ATAC/RNA sequencing (SHARE-seq) and spatially resolved metabolomics to profile 54 human samples from distinct kidney anatomical regions. We generated transcriptomes of 446,267 cells and chromatin accessibility profiles of 401,875 cells and developed a package to analyze 408,218 spatially resolved metabolomes. We find that the same cell type, including thin limb, thick ascending limb loop of Henle and principal cells, display distinct transcriptomic, chromatin accessibility, and metabolomic signatures, depending on anatomic location. Surveying metabolism-associated gene profiles revealed non-overlapping metabolic signatures between nephron segments and dysregulated lipid metabolism in diseased proximal tubule (PT) cells. Integrating multimodal omics with clinical data identified PLEKHA1 as a disease marker, and its in vitro knockdown increased gene expression in PT differentiation, suggesting possible pathogenic roles. This study highlights previously underrepresented cellular heterogeneity underlying the human kidney anatomy.
Collapse
Affiliation(s)
- Haikuo Li
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Dian Li
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Nicolas Ledru
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Qiao Xuanyuan
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Haojia Wu
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Amish Asthana
- Department of Surgery, Atrium Health Wake Forest Baptist, Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Lori N Byers
- Department of Surgery, Atrium Health Wake Forest Baptist, Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Stefan G Tullius
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Giuseppe Orlando
- Department of Surgery, Atrium Health Wake Forest Baptist, Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Sushrut S Waikar
- Section of Nephrology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA; Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
28
|
Dalghi MG, DuRie E, Ruiz WG, Clayton DR, Montalbetti N, Mutchler SB, Satlin LM, Kleyman TR, Carattino MD, Shi YS, Apodaca G. Expression and localization of the mechanosensitive/osmosensitive ion channel TMEM63B in the mouse urinary tract. Physiol Rep 2024; 12:e16043. [PMID: 38724885 PMCID: PMC11082094 DOI: 10.14814/phy2.16043] [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: 03/04/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/12/2024] Open
Abstract
The epithelial cells that line the kidneys and lower urinary tract are exposed to mechanical forces including shear stress and wall tension; however, the mechanosensors that detect and respond to these stimuli remain obscure. Candidates include the OSCA/TMEM63 family of ion channels, which can function as mechanosensors and osmosensors. Using Tmem63bHA-fl/HA-fl reporter mice, we assessed the localization of HA-tagged-TMEM63B within the urinary tract by immunofluorescence coupled with confocal microscopy. In the kidneys, HA-TMEM63B was expressed by proximal tubule epithelial cells, by the intercalated cells of the collecting duct, and by the epithelial cells lining the thick ascending limb of the medulla. In the urinary tract, HA-TMEM63B was expressed by the urothelium lining the renal pelvis, ureters, bladder, and urethra. HA-TMEM63B was also expressed in closely allied organs including the epithelial cells lining the seminal vesicles, vas deferens, and lateral prostate glands of male mice and the vaginal epithelium of female mice. Our studies reveal that TMEM63B is expressed by subsets of kidney and lower urinary tract epithelial cells, which we hypothesize are sites of TMEM63B mechanosensation or osmosensation, or both.
Collapse
Affiliation(s)
- Marianela G. Dalghi
- Department of Medicine and George M. O'Brien Pittsburgh Center for Kidney ResearchUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Ella DuRie
- Department of Medicine and George M. O'Brien Pittsburgh Center for Kidney ResearchUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Wily G. Ruiz
- Department of Medicine and George M. O'Brien Pittsburgh Center for Kidney ResearchUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Dennis R. Clayton
- Department of Medicine and George M. O'Brien Pittsburgh Center for Kidney ResearchUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Nicolas Montalbetti
- Department of Medicine and George M. O'Brien Pittsburgh Center for Kidney ResearchUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Stephanie B. Mutchler
- Department of Medicine and George M. O'Brien Pittsburgh Center for Kidney ResearchUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Lisa M. Satlin
- Department of PediatricsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Thomas R. Kleyman
- Department of Medicine and George M. O'Brien Pittsburgh Center for Kidney ResearchUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Chemical Biology & PharmacologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Marcelo D. Carattino
- Department of Medicine and George M. O'Brien Pittsburgh Center for Kidney ResearchUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Yun Stone Shi
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical SchoolNanjing UniversityNanjingChina
| | - Gerard Apodaca
- Department of Medicine and George M. O'Brien Pittsburgh Center for Kidney ResearchUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
29
|
Yao X, Mu Y, Zhang L, Chen L, Zou S, Chen X, Lu K, Dong H. AtPIP1;4 and AtPIP2;4 Cooperatively Mediate H 2O 2 Transport to Regulate Plant Growth and Disease Resistance. PLANTS (BASEL, SWITZERLAND) 2024; 13:1018. [PMID: 38611547 PMCID: PMC11013698 DOI: 10.3390/plants13071018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/22/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024]
Abstract
The rapid production of hydrogen peroxide (H2O2) is a hallmark of plants' successful recognition of pathogen infection and plays a crucial role in innate immune signaling. Aquaporins (AQPs) are membrane channels that facilitate the transport of small molecular compounds across cell membranes. In plants, AQPs from the plasma membrane intrinsic protein (PIP) family are utilized for the transport of H2O2, thereby regulating various biological processes. Plants contain two PIP families, PIP1s and PIP2s. However, the specific functions and relationships between these subfamilies in plant growth and immunity remain largely unknown. In this study, we explore the synergistic role of AtPIP1;4 and AtPIP2;4 in regulating plant growth and disease resistance in Arabidopsis. We found that in plant cells treated with H2O2, AtPIP1;4 and AtPIP2;4 act as facilitators of H2O2 across membranes and the translocation of externally applied H2O2 from the apoplast to the cytoplasm. Moreover, AtPIP1;4 and AtPIP2;4 collaborate to transport bacterial pathogens and flg22-induced apoplastic H2O2 into the cytoplasm, leading to increased callose deposition and enhanced defense gene expression to strengthen immunity. These findings suggest that AtPIP1;4 and AtPIP2;4 cooperatively mediate H2O2 transport to regulate plant growth and immunity.
Collapse
Affiliation(s)
- Xiaohui Yao
- National Key Laboratory of Wheat Improvement, College of Plant Protection, Shandong Agricultural University, Taian 271018, China
| | - Yanjie Mu
- National Key Laboratory of Wheat Improvement, College of Plant Protection, Shandong Agricultural University, Taian 271018, China
- Qingdao King Agroot Crop Science, Qingdao 266071, China
| | - Liyuan Zhang
- National Key Laboratory of Wheat Improvement, College of Plant Protection, Shandong Agricultural University, Taian 271018, China
| | - Lei Chen
- National Key Laboratory of Wheat Improvement, College of Plant Protection, Shandong Agricultural University, Taian 271018, China
| | - Shenshen Zou
- National Key Laboratory of Wheat Improvement, College of Plant Protection, Shandong Agricultural University, Taian 271018, China
| | - Xiaochen Chen
- National Key Laboratory of Wheat Improvement, College of Plant Protection, Shandong Agricultural University, Taian 271018, China
| | - Kai Lu
- National Key Laboratory of Wheat Improvement, College of Plant Protection, Shandong Agricultural University, Taian 271018, China
| | - Hansong Dong
- National Key Laboratory of Wheat Improvement, College of Plant Protection, Shandong Agricultural University, Taian 271018, China
| |
Collapse
|
30
|
Loukelis K, Koutsomarkos N, Mikos AG, Chatzinikolaidou M. Advances in 3D bioprinting for regenerative medicine applications. Regen Biomater 2024; 11:rbae033. [PMID: 38845855 PMCID: PMC11153344 DOI: 10.1093/rb/rbae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/13/2024] [Accepted: 03/17/2024] [Indexed: 06/09/2024] Open
Abstract
Biofabrication techniques allow for the construction of biocompatible and biofunctional structures composed from biomaterials, cells and biomolecules. Bioprinting is an emerging 3D printing method which utilizes biomaterial-based mixtures with cells and other biological constituents into printable suspensions known as bioinks. Coupled with automated design protocols and based on different modes for droplet deposition, 3D bioprinters are able to fabricate hydrogel-based objects with specific architecture and geometrical properties, providing the necessary environment that promotes cell growth and directs cell differentiation towards application-related lineages. For the preparation of such bioinks, various water-soluble biomaterials have been employed, including natural and synthetic biopolymers, and inorganic materials. Bioprinted constructs are considered to be one of the most promising avenues in regenerative medicine due to their native organ biomimicry. For a successful application, the bioprinted constructs should meet particular criteria such as optimal biological response, mechanical properties similar to the target tissue, high levels of reproducibility and printing fidelity, but also increased upscaling capability. In this review, we highlight the most recent advances in bioprinting, focusing on the regeneration of various tissues including bone, cartilage, cardiovascular, neural, skin and other organs such as liver, kidney, pancreas and lungs. We discuss the rapidly developing co-culture bioprinting systems used to resemble the complexity of tissues and organs and the crosstalk between various cell populations towards regeneration. Moreover, we report on the basic physical principles governing 3D bioprinting, and the ideal bioink properties based on the biomaterials' regenerative potential. We examine and critically discuss the present status of 3D bioprinting regarding its applicability and current limitations that need to be overcome to establish it at the forefront of artificial organ production and transplantation.
Collapse
Affiliation(s)
- Konstantinos Loukelis
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Nikos Koutsomarkos
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Maria Chatzinikolaidou
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), Heraklion 70013, Greece
| |
Collapse
|
31
|
Xu X, Khunsriraksakul C, Eales JM, Rubin S, Scannali D, Saluja S, Talavera D, Markus H, Wang L, Drzal M, Maan A, Lay AC, Prestes PR, Regan J, Diwadkar AR, Denniff M, Rempega G, Ryszawy J, Król R, Dormer JP, Szulinska M, Walczak M, Antczak A, Matías-García PR, Waldenberger M, Woolf AS, Keavney B, Zukowska-Szczechowska E, Wystrychowski W, Zywiec J, Bogdanski P, Danser AHJ, Samani NJ, Guzik TJ, Morris AP, Liu DJ, Charchar FJ, Tomaszewski M. Genetic imputation of kidney transcriptome, proteome and multi-omics illuminates new blood pressure and hypertension targets. Nat Commun 2024; 15:2359. [PMID: 38504097 PMCID: PMC10950894 DOI: 10.1038/s41467-024-46132-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
Genetic mechanisms of blood pressure (BP) regulation remain poorly defined. Using kidney-specific epigenomic annotations and 3D genome information we generated and validated gene expression prediction models for the purpose of transcriptome-wide association studies in 700 human kidneys. We identified 889 kidney genes associated with BP of which 399 were prioritised as contributors to BP regulation. Imputation of kidney proteome and microRNAome uncovered 97 renal proteins and 11 miRNAs associated with BP. Integration with plasma proteomics and metabolomics illuminated circulating levels of myo-inositol, 4-guanidinobutanoate and angiotensinogen as downstream effectors of several kidney BP genes (SLC5A11, AGMAT, AGT, respectively). We showed that genetically determined reduction in renal expression may mimic the effects of rare loss-of-function variants on kidney mRNA/protein and lead to an increase in BP (e.g., ENPEP). We demonstrated a strong correlation (r = 0.81) in expression of protein-coding genes between cells harvested from urine and the kidney highlighting a diagnostic potential of urinary cell transcriptomics. We uncovered adenylyl cyclase activators as a repurposing opportunity for hypertension and illustrated examples of BP-elevating effects of anticancer drugs (e.g. tubulin polymerisation inhibitors). Collectively, our studies provide new biological insights into genetic regulation of BP with potential to drive clinical translation in hypertension.
Collapse
Affiliation(s)
- Xiaoguang Xu
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | | | - James M Eales
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Sebastien Rubin
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - David Scannali
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Sushant Saluja
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - David Talavera
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Havell Markus
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Lida Wang
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Maciej Drzal
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Akhlaq Maan
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Abigail C Lay
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Priscilla R Prestes
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Australia
| | - Jeniece Regan
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Avantika R Diwadkar
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Matthew Denniff
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Grzegorz Rempega
- Department of Urology, Medical University of Silesia, Katowice, Poland
| | - Jakub Ryszawy
- Department of Urology, Medical University of Silesia, Katowice, Poland
| | - Robert Król
- Department of General, Vascular and Transplant Surgery, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - John P Dormer
- Department of Cellular Pathology, University Hospitals of Leicester, Leicester, UK
| | - Monika Szulinska
- Department of Obesity, Metabolic Disorders Treatment and Clinical Dietetics, Karol Marcinkowski University of Medical Sciences, Poznan, Poland
| | - Marta Walczak
- Department of Internal Diseases, Metabolic Disorders and Arterial Hypertension, Poznan University of Medical Sciences, Poznan, Poland
| | - Andrzej Antczak
- Department of Urology and Uro-oncology, Karol Marcinkowski University of Medical Sciences, Poznan, Poland
| | - Pamela R Matías-García
- Institute of Epidemiology, Helmholtz Center Munich, Neuherberg, Germany
- Research Unit Molecular Epidemiology, Helmholtz Center Munich, Neuherberg, Germany
- German Research Center for Cardiovascular Disease (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Melanie Waldenberger
- Institute of Epidemiology, Helmholtz Center Munich, Neuherberg, Germany
- Research Unit Molecular Epidemiology, Helmholtz Center Munich, Neuherberg, Germany
- German Research Center for Cardiovascular Disease (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Royal Manchester Children's Hospital and Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Bernard Keavney
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust Manchester, Manchester Royal Infirmary, Manchester, UK
| | | | - Wojciech Wystrychowski
- Department of General, Vascular and Transplant Surgery, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Joanna Zywiec
- Department of Internal Medicine, Diabetology and Nephrology, Zabrze, Medical University of Silesia, Katowice, Poland
| | - Pawel Bogdanski
- Department of Obesity, Metabolic Disorders Treatment and Clinical Dietetics, Karol Marcinkowski University of Medical Sciences, Poznan, Poland
| | - A H Jan Danser
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Tomasz J Guzik
- Department of Internal Medicine, Jagiellonian University Medical College, Kraków, Poland
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Kraków, Poland
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal & Dermatological Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Dajiang J Liu
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Fadi J Charchar
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Australia
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- Department of Physiology, University of Melbourne, Melbourne, Australia
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK.
- Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust Manchester, Manchester Royal Infirmary, Manchester, UK.
| |
Collapse
|
32
|
Liu J, Livingston MJ, Dong G, Wei Q, Zhang M, Mei S, Zhu J, Zhang C, Dong Z. HIF-1 contributes to autophagy activation via BNIP3 to facilitate renal fibrosis in hypoxia in vitro and UUO in vivo. Am J Physiol Cell Physiol 2024; 326:C935-C947. [PMID: 38284121 PMCID: PMC11193486 DOI: 10.1152/ajpcell.00458.2023] [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: 09/18/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 01/30/2024]
Abstract
The molecular basis of renal interstitial fibrosis, a major pathological feature of progressive kidney diseases, remains poorly understood. Autophagy has been implicated in renal fibrosis, but whether it promotes or inhibits fibrosis remains controversial. Moreover, it is unclear how autophagy is activated and sustained in renal fibrosis. The present study was designed to address these questions using the in vivo mouse model of unilateral ureteral obstruction and the in vitro model of hypoxia in renal tubular cells. Both models showed the activation of hypoxia-inducible factor-1 (HIF-1) and autophagy along with fibrotic changes. Inhibition of autophagy with chloroquine reduced renal fibrosis in unilateral ureteral obstruction model, whereas chloroquine and autophagy-related gene 7 knockdown decreased fibrotic changes in cultured renal proximal tubular cells, supporting a profibrotic role of autophagy. Notably, pharmacological and genetic inhibition of HIF-1 led to the suppression of autophagy and renal fibrosis in these models. Mechanistically, knock down of BCL2 and adenovirus E1B 19-kDa-interacting protein 3 (BNIP3), a downstream target gene of HIF, decreased autophagy and fibrotic changes during hypoxia in BUMPT cells. Together, these results suggest that HIF-1 may activate autophagy via BNIP3 in renal tubular cells to facilitate the development of renal interstitial fibrosis.NEW & NOTEWORTHY Autophagy has been reported to participate in renal fibrosis, but its role and underlying activation mechanism is unclear. In this study, we report the role of HIF-1 in autophagy activation in models of renal fibrosis and further investigate the underlying mechanism.
Collapse
Affiliation(s)
- Jing Liu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - Man J Livingston
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - Guie Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - Ming Zhang
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - Shuqin Mei
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, United States
- Department of Nephrology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jiefu Zhu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| |
Collapse
|
33
|
Martos-Benítez FD, Burgos-Aragüez D, García-Mesa L, Orama-Requejo V, Cárdenas-González RC, Michelena-Piedra JC, Izquierdo-Castañeda J, Sánchez-de-la-Rosa E, Corrales-González O. Fluid balance, biomarkers of renal function and mortality in critically ill patients with AKI diagnosed before, or within 24 h of intensive care unit admission: a prospective study. J Nephrol 2024; 37:439-449. [PMID: 38189864 DOI: 10.1007/s40620-023-01829-z] [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: 07/17/2023] [Accepted: 11/08/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND To evaluate fluid balance, biomarkers of renal function and its relation to mortality in patients with acute kidney injury (AKI) diagnosed before, or within 24 h of intensive care unit admission. METHODS A prospective cohort study considered 773 critically ill patients observed over six years. Pre-intensive care unit-onset AKI was defined as AKI diagnosed before, or within 24 h of intensive care unit admission. Body weight-adjusted fluid balance and fluid balance-adjusted biomarkers of renal function were measured daily for the first three days of intensive care unit admission. Primary outcome was mortality in the intensive care unit. RESULTS Prevalence of pre-intensive care unit-onset AKI was 55.1%, of which 55.6% of cases were hospital-acquired and 44.4% were community-acquired. Fluid balance was higher in AKI patients than in non-AKI patients (p < 0.001) and had a negative correlation with urine output (p < 0.01). Positive fluid balance and biomarkers of renal function were independently related to mortality. Multivariate analysis identified the following AKI-related variables associated with increased mortality: (1) In AKI patients: type 1 cardiorenal syndrome (OR 2.00), intra-abdominal hypertension (OR 1.71), AKI stage 3 (OR 2.15) and increase in AKI stage (OR 4.99); 2) In patients with community-acquired AKI: type 1 cardiorenal syndrome (OR 5.16), AKI stage 2 (OR 2.72), AKI stage 3 (OR 4.95) and renal replacement therapy (OR 3.05); and 3) In patients with hospital-acquired AKI: intra-abdominal hypertension (OR 2.31) and increase in AKI stage (OR 4.51). CONCLUSIONS In patients with pre-intensive care unit-onset AKI, positive fluid balance is associated with worse renal outcomes. Positive fluid balance and decline in biomarkers of renal function are related to increased mortality, thus in this subpopulation of critically ill patients, positive fluid balance is not recommended and renal function must be closely monitored.
Collapse
Affiliation(s)
- Frank Daniel Martos-Benítez
- Intensive Care Unit, National Institute of Neurology and Neurosurgery, 29 St. and D St., Vedado, Plaza, 10400, Havana, Cuba.
| | - Dailé Burgos-Aragüez
- Intensive Care Unit-8, Hermanos Ameijeiras Hospital, San Lázaro St., Centro Havana, 10200, Havana, Cuba
| | - Liselotte García-Mesa
- Intensive Care Unit-8, Hermanos Ameijeiras Hospital, San Lázaro St., Centro Havana, 10200, Havana, Cuba
| | | | | | - Juan Carlos Michelena-Piedra
- Intensive Care Unit, National Institute of Neurology and Neurosurgery, 29 St. and D St., Vedado, Plaza, 10400, Havana, Cuba
| | - Judet Izquierdo-Castañeda
- Intensive Care Unit, National Institute of Neurology and Neurosurgery, 29 St. and D St., Vedado, Plaza, 10400, Havana, Cuba
| | - Ernesto Sánchez-de-la-Rosa
- Intensive Care Unit, National Institute of Neurology and Neurosurgery, 29 St. and D St., Vedado, Plaza, 10400, Havana, Cuba
| | - Olivia Corrales-González
- Intensive Care Unit, National Institute of Neurology and Neurosurgery, 29 St. and D St., Vedado, Plaza, 10400, Havana, Cuba
| |
Collapse
|
34
|
Liu Y, Hong X, Liu L, Li X, Huang S, Luo Q, Huang Q, Qiu J, Qiu P, Li C. Shen Qi Wan ameliorates nephritis in chronic kidney disease via AQP1 and DEFB1 regulation. Biomed Pharmacother 2024; 170:116027. [PMID: 38113630 DOI: 10.1016/j.biopha.2023.116027] [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: 10/06/2023] [Revised: 12/04/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023] Open
Abstract
Shen Qi Wan (SQW) has been proven to exert anti-inflammatory effects in the kidneys of CKD models accompanied by unclear therapeutic mechanisms. This study aims to evaluate the kidney-protective and anti-inflammatory effects of SQW and to elucidate its fundamental mechanisms for CKD treatment. Firstly, the main active components of SQW were identified by UPLC-Q-TOF/MS technique. Subsequently, we evaluated inflammatory factors, renal function and renal pathology changes following SQW treatment utilizing adenine-induced CKD mice and aquaporin 1 knockout (AQP1-/-) mice. Additionally, we conducted RNA-seq analysis and bioinformatics analysis to predict the SQW potential therapeutic targets and anti-nephritis pathways. Simultaneously, WGCNA analysis method and machine learning algorithms were used to perform a clinical prognostic analysis of potential biomarkers in CKD patients from the GEO database and validated through clinical samples. Lipopolysaccharide-induced HK-2 cells were further used to explore the mechanism. We found that renal collagen deposition was reduced, serum inflammatory cytokine levels decreased, and renal function was improved after SQW intervention. It can be inferred that β-defensin 1 (DEFB1) may be a pivotal target, as confirmed by serum and renal tissue samples from CKD patients. Furthermore, SQW assuages inflammatory responses by fostering AQP1-mediated DEFB1 expression was confirmed in in vitro and in vivo studies. Significantly, the renal-protective effect of SQW is to some extent attenuated after AQP1 gene knockout. SQW could reduce inflammatory responses by modulating AQP1 and DEFB1. These findings underscore the potential of SQW as a promising contender for novel prevention and treatment strategies within the ambit of CKD management.
Collapse
Affiliation(s)
- Yiming Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiao Hong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Liu Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xinyue Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shuo Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qihan Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qiaoyan Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiang Qiu
- Department of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Ping Qiu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Changyu Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
35
|
Huynh NV, Rehage C, Hyndman KA. Mild dehydration effects on the murine kidney single-nucleus transcriptome and chromatin accessibility. Am J Physiol Renal Physiol 2023; 325:F717-F732. [PMID: 37767569 PMCID: PMC11550884 DOI: 10.1152/ajprenal.00161.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/12/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Daily, we may experience mild dehydration with a rise in plasma osmolality that triggers the release of vasopressin. Although the effect of dehydration is well characterized in collecting duct principal cells (CDPCs), we hypothesized that mild dehydration (<12 h) results in many kidney cell-specific changes in transcriptomes and chromatin accessibility. Single-nucleus (sn) multiome (RNA-assay for transposase-accessible chromatin) sequencing and bulk RNA sequencing of kidneys from male and female mice that were mildly water deprived or not were compared. Water-deprived mice had a significant increase in plasma osmolality. sn-multiome-seq resulted in 19,837 nuclei that were annotated into 33 clusters. In CDPCs, aquaporin 2 (Aqp2) and aquaporin 3 (Apq3) were greater in dehydrated mice, but there were novel genes like gremlin 2 (Grem2; a cytokine) that were increased compared with ad libitum mice. The transcription factor cAMP-responsive element modulator (Crem) was greater in CDPCs of dehydrated mice, and the Crem DNA motif was more accessible. There were hundreds of sex- and dehydration-specific differentially expressed genes (DEGs) throughout the kidney, especially in the proximal tubules and thin limbs. In male mice, DEGs were enriched in pathways related to lipid metabolism, whereas female DEGs were enriched in organic acid metabolism. Many highly expressed genes had a positive correlation with increased chromatin accessibility, and mild dehydration exerted many transcriptional changes that we detected at the chromatin level. Even with a rise in plasma osmolality, male and female kidneys have distinct transcriptomes suggesting that there may be diverse mechanisms used to remain in fluid balance.NEW & NOTEWORTHY The kidney consists of >30 cell types that work collectively to maintain fluid-electrolyte balance. Kidney single-nucleus transcriptomes and chromatin accessibility profiles from male and female control (ad libitum water and food) or mildly dehydrated mice (ad libitum food, water deprivation) were determined. Mild dehydration caused hundreds of cell- and sex-specific transcriptomic changes, even though the kidney function to conserve water was the same.
Collapse
Affiliation(s)
- Nha Van Huynh
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Cassidy Rehage
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Kelly A Hyndman
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
36
|
Polonsky M, Gerhardt LMS, Yun J, Koppitch K, Colón KL, Amrhein H, Zheng S, Yuan GC, Thomson M, Cai L, McMahon AP. Spatial transcriptomics defines injury-specific microenvironments in the adult mouse kidney and novel cellular interactions in regeneration and disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.22.568217. [PMID: 38045285 PMCID: PMC10690238 DOI: 10.1101/2023.11.22.568217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Kidney injury disrupts the intricate renal architecture and triggers limited regeneration, and injury-invoked inflammation and fibrosis. Deciphering molecular pathways and cellular interactions driving these processes is challenging due to the complex renal architecture. Here, we applied single cell spatial transcriptomics to examine ischemia-reperfusion injury in the mouse kidney. Spatial transcriptomics revealed injury-specific and spatially-dependent gene expression patterns in distinct cellular microenvironments within the kidney and predicted Clcf1-Crfl1 in a molecular interplay between persistently injured proximal tubule cells and neighboring fibroblasts. Immune cell types play a critical role in organ repair. Spatial analysis revealed cellular microenvironments resembling early tertiary lymphoid structures and identified associated molecular pathways. Collectively, this study supports a focus on molecular interactions in cellular microenvironments to enhance understanding of injury, repair and disease.
Collapse
Affiliation(s)
- Michal Polonsky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Louisa M. S. Gerhardt
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Jina Yun
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Kari Koppitch
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Katsuya Lex Colón
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Henry Amrhein
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Shiwei Zheng
- Department of Genetics and Genomic Sciences and Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences and Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Long Cai
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Andrew P. McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, California
| |
Collapse
|
37
|
Jovanovic M, Sabovic M. Refractory drug-induced systemic small-vessel vasculitis with two varied extracutaneous manifestations: a case report and review of the literature. J Med Case Rep 2023; 17:470. [PMID: 37885023 PMCID: PMC10605860 DOI: 10.1186/s13256-023-04174-8] [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: 06/05/2023] [Accepted: 09/11/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Clopidogrel and ticagrelor are rarely reported to cause vasculitis via drug hypersensitivity reaction, largely mediated by T cells and immunoglobulin E (IgE). Despite therapeutic advances, the etiology of refractory vasculitides remains incompletely understood. Recently, (non)immunological mechanisms bypassing T cells and IgE have been proposed to explain resistance to standard immunosuppressants. Herein, we report a case of refractory drug-induced systemic small-vessel vasculitis with varied extracutaneous manifestations and incorporate multiple sources of data to provide detailed accounts of complex (non)immunological phenomena involved in this case. Study objectives are to provide an insight about rare presentations of commonly used drugs, upgrade the pathophysiological concepts of drug-induced vasculitis, raise need for further investigation to define causes and risk factors for refractory vasculitis, and discuss most of the current knowledge suggesting novel therapeutic approaches to treat this vasculitis. To our knowledge, this is the first case of the two flares of systemic small-vessel vasculitis in a single patient in response to clopidogrel and ticagrelor exposure, respectively. However, this report is limited by attribution/observer bias. CASE PRESENTATION We herein report a 24-year-old Caucasian male student with a medical history of mild seasonal allergic rhinoconjunctivitis, tension-type headaches, posttraumatic arterial stenosis, and previous exposure to ibuprofen, acetylsalicylic acid, and mRNA coronavirus disease 2019 (COVID-19) vaccine who suffered largely from acute urticaria and dyspnea after 20 days of acetylsalicylic acid and clopidogrel introduction. A skin punch biopsy confirmed leukocytoclastic vasculitis. Serologic antibody testing, complement analysis, microbiologic testing, and cancer biomarkers revealed no abnormalities. Regarding the patient's medical history, both acetylsalicylic acid and clopidogrel were exchanged for ticagrelor. Furthermore, the addition of naproxen, cyclosporine, bilastine, prednisolone, and montelukast resulted in complete recovery. After 7 days, diarrhea and hematuria occurred. Urinalysis and computed tomography showed reversible proteinuria with gross hematuria and hypodense changes in kidney medulla, respectively, associated with discontinuation of ticagrelor and naproxen. In addition, the patient recovered completely without any immunosuppression up-titration. CONCLUSIONS This case highlights the role of clopidogrel and ticagrelor as possible triggering agents for systemic small-vessel vasculitis and offers an insight into novel therapeutic strategies for refractory vasculitides. Further research is needed to build on the findings of a current report.
Collapse
Affiliation(s)
- Mark Jovanovic
- Department of Internal Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia.
| | - Miso Sabovic
- Department of Angiology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
38
|
Özden ES, Aşcı H, Büyükbayram Hİ, Sevük MA, İmeci OB, Doğan HK, Özmen Ö. Dexpanthenol protects against lipopolysaccharide-induced acute kidney injury by restoring aquaporin-2 levels via regulation of the silent information regulator 1 signaling pathway. Korean J Anesthesiol 2023; 76:501-509. [PMID: 37232072 PMCID: PMC10562075 DOI: 10.4097/kja.23207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/15/2023] [Accepted: 05/25/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a serious pathology that causes dysfunction in concentrating urine due to kidney damage, resulting in blood pressure dysregulation and increased levels of toxic metabolites. Dexpanthenol (DEX), a pantothenic acid analog, exhibits anti-inflammatory and anti-apoptotic properties in various tissues. This study investigated the protective effects of DEX against systemic inflammation-induced AKI. METHODS Thirty-two female rats were randomly assigned to the control, lipopolysaccharide (LPS), LPS+DEX, and DEX groups. LPS (5 mg/kg, single dose on the third day, 6 h before sacrifice) and DEX (500 mg/kg/d for 3 d) were administered intraperitoneally. After sacrifice, blood samples and kidney tissues were collected. Hematoxylin and eosin, caspase-3 (Cas-3), and tumor necrosis factor alpha (TNF-α) staining were performed on the kidney tissues. The total oxidant status (TOS) and total antioxidant status were measured using spectrophotometric methods. Aquaporin-2 (AQP-2), silent information regulator 1 (SIRT1), and interleukin-6 (IL-6) were detected using quantitative reverse transcription-polymerase chain reaction analysis. RESULTS Histopathological analysis revealed that DEX treatment ameliorated histopathological changes. In the LPS group, an increase in the blood urea nitrogen, creatinine, urea, IL-6, Cas-3, TNF-α, and TOS levels and oxidative stress index was observed compared with the control group, whereas AQP-2 and SIRT1 levels decreased. DEX treatment reversed these effects. CONCLUSIONS DEX was found to effectively prevent inflammation, oxidative stress, and apoptosis in the kidneys via the SIRT1 signaling pathway. These protective properties suggest DEX's potential as a therapeutic agent for the treatment of kidney pathologies.
Collapse
Affiliation(s)
- Eyyüp Sabri Özden
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Halil Aşcı
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | | | - Mehmet Abdulkadir Sevük
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Orhan Berk İmeci
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Hatice Kübra Doğan
- Department of Bioengineering, Institute of Science, Suleyman Demirel University, Isparta, Turkey
| | - Özlem Özmen
- Department of Pathology, Faculty of Veterinary Medicine, Mehmet Akif Ersoy University, Burdur, Turkey
| |
Collapse
|
39
|
Dugbartey GJ. Physiological role of hydrogen sulfide in the kidney and its therapeutic implications for kidney diseases. Biomed Pharmacother 2023; 166:115396. [PMID: 37647689 DOI: 10.1016/j.biopha.2023.115396] [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: 07/21/2023] [Revised: 08/21/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023] Open
Abstract
For over three centuries, hydrogen sulfide (H2S) has been known as a toxic and deadly gas at high concentrations, with a distinctive smell of rotten eggs. However, studies over the past two decades have shown that H2S has risen above its historically notorious label and has now received significant scientific attention as an endogenously produced gaseous signaling molecule that participates in cellular homeostasis and influences a myriad of physiological and pathological processes at low concentrations. Its endogenous production is enzymatically regulated, and when dysregulated, contributes to pathogenesis of renal diseases. In addition, exogenous H2S administration has been reported to exhibit important therapeutic characteristics that target multiple molecular pathways in common renal pathologies in which reduced levels of renal and plasma H2S were observed. This review highlights functional anatomy of the kidney and renal production of H2S. The review also discusses current understanding of H2S in renal physiology and seeks to lay the foundation as a new targeted therapeutic agent for renal pathologies such as hypertensive nephropathy, diabetic kidney disease and water balance disorders.
Collapse
Affiliation(s)
- George J Dugbartey
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Legon, Accra, Ghana; Accra College of Medicine, Magnolia St, JVX5+FX9, East Legon, Accra, Ghana.
| |
Collapse
|
40
|
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: 12] [Impact Index Per Article: 6.0] [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
|
41
|
Ishida Y, Nosaka M, Ishigami A, Kondo T. Forensic application of aquaporins. Leg Med (Tokyo) 2023; 63:102249. [PMID: 37060638 DOI: 10.1016/j.legalmed.2023.102249] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/04/2023] [Indexed: 04/03/2023]
Abstract
Aquaporins (AQPs) are a family of water channel proteins that primarily elicit the basic functions of water transport and osmotic homeostasis. To date, at least 17 mammalian AQPs have been identified, AQP-0 to -12 have been found in higher orders including human, and AQP-13 to -16 have been described in older lineages. Moreover, these proteins have recently been shown to regulate many biological processes through unique activities, such as cell proliferation, migration, apoptosis, and mitochondrial metabolism. Several studies have focused on the involvement of AQPs in cell biology aspect, showing that they are involved in a variety of physiological processes and pathophysiological conditions. Furthermore, in the field of forensic medicine, studies on whether AQPs can be a useful marker for diagnosing various causes of death have been conducted using autopsy samples and animal experiments, which have produced interesting results. Herein, we review certain observations regarding AQPs and discuss their potential to contribute to the future practice of forensic research.
Collapse
|
42
|
Azırak S. Prevention of nephrotoxicity induced by amikacin: The role of misoprostol, A prostaglandin E1 analogue. Prostaglandins Other Lipid Mediat 2023; 164:106682. [PMID: 36349661 DOI: 10.1016/j.prostaglandins.2022.106682] [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: 09/19/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/05/2022]
Abstract
Amikacin (AK) is an aminoglycoside that is widely used to treat life-threatening Gram-negative infections, especially in intensive care units. Despite its wide clinical indications, AK causes serious side effects such as kidney toxicity. AK was found to lead to tissue damage primarily through apoptosis and oxidative stress. Therefore, it was investigated whether misoprostol (MP), which has antioxidant and antiapoptotic properties, had a beneficial effect on kidney damage caused by AK. It was observed that kidney injury molecule-1 (KIM-1) mRNA, blood urea nitrogen (BUN), creatinine (Cr), NADPH oxidase-4 (NOX-4) and Caspase-3 (CAS-3) levels increased in the AK-treated group in comparison with the control group, while uric acid, albumin, and total protein levels were decreased. In rats that were treated with AK+MP, the levels of KIM-1 mRNA, BUN, Cr, NOX-4 and CAS-3 were significantly decreased in comparison with the AK group, while uric acid, albumin and total protein levels increased. According to the obtained results, MP was found to be quite effective in the protection of kidneys from the toxic effects of AK.
Collapse
Affiliation(s)
- Sebile Azırak
- Vocational School of Health Services, University of Adıyaman, Adıyaman, Turkey.
| |
Collapse
|
43
|
Price ME, Fishler KP, Muff-Luett M, Mauch TJ, Brunelli L, Euteneuer JC. Variants in AQP11 may result in autosomal recessive bilateral cystic renal dysgenesis. Am J Med Genet A 2023; 191:612-616. [PMID: 36420936 DOI: 10.1002/ajmg.a.63056] [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: 10/07/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/25/2022]
Abstract
Congenital renal cystic dysplasia is a rare disease that occurs in approximately 1 in 4000 children and is often discovered in the antenatal period by ultrasound. It is commonly associated with oligohydramnios in utero and/or renal insufficiency or failure in the postnatal period. Aquaporins are membrane proteins that serve as transport channels in the transfer of water or small solutes across cell membranes. They play a role in the development of renal cysts. Aquaporin 11 (AQP11) deficient mice develop polycystic kidney disease in utero due to disruption of polycystin-1. Here we describe a case of bilateral cystic kidney disease in a patient with novel compound heterozygous variants in AQP11: c.780G>T (p. Trp260Cys) and c.472C>T (p.Pro158Ser) (NM_173039.2) identified by whole genome sequencing. These findings suggest, for the first time, the potential role of AQP11 in congenital renal cystic dysplasia.
Collapse
Affiliation(s)
- Michael E Price
- University of Nebraska Medical Center, College of Medicine, Omaha, Nebraska, USA
| | - Kristen P Fishler
- Munroe-Meyer Institute of Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Melissa Muff-Luett
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Teri J Mauch
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
| | - Luca Brunelli
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
| | - Joshua C Euteneuer
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
44
|
Jung SC, Zhou T, Ko EA. Age-dependent expression of ion channel genes in rat. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2023; 27:85-94. [PMID: 36575936 PMCID: PMC9806634 DOI: 10.4196/kjpp.2023.27.1.85] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 12/29/2022]
Abstract
Ion channels regulate a large number of cellular functions and their functional role in many diseases makes them potential therapeutic targets. Given their diverse distribution across multiple organs, the roles of ion channels, particularly in age-associated transcriptomic changes in specific organs, are yet to be fully revealed. Using RNA-seq data, we investigated the rat transcriptomic profiles of ion channel genes across 11 organs/tissues and 4 developmental stages in both sexes of Fischer 344 rats and identify tissue-specific and age-dependent changes in ion channel gene expression. Organ-enriched ion channel genes were identified. In particular, the brain showed higher tissue-specificity of ion channel genes, including Gabrd, Gabra6, Gabrg2, Grin2a, and Grin2b. Notably, age-dependent changes in ion channel gene expression were prominently observed in the thymus, including in Aqp1, Clcn4, Hvcn1, Itpr1, Kcng2, Kcnj11, Kcnn3, and Trpm2. Our comprehensive study of ion channel gene expression will serve as a primary resource for biological studies of aging-related diseases caused by abnormal ion channel functions.
Collapse
Affiliation(s)
- Sung-Cherl Jung
- Department of Physiology, School of Medicine, Jeju National University, Jeju 63243, Korea
| | - Tong Zhou
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Eun-A Ko
- Department of Physiology, School of Medicine, Jeju National University, Jeju 63243, Korea,Correspondence Eun-A Ko, E-mail:
| |
Collapse
|
45
|
Hu J, Ananth D, Sethi SK, Taliwal N, Govindan S, Raina R. Neonatal AKI: An update. J Neonatal Perinatal Med 2023; 16:361-373. [PMID: 37718869 DOI: 10.3233/npm-230120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Neonatal acute kidney injury (AKI) is a common complication, especially in the neonatal intensive care unit, that is associated with long term consequences and poor outcomes. Early detection and treatment is critical. Currently, neonatal AKI is defined with urinary markers and serum creatinine, with limitations on early detection and individual treatment. There have been numerous biomarkers and risk factor scores that have been studied for their ability to predict neonatal AKI. To move towards personalized medicine, neonatal AKI must be categorized into phenotypes and subphenotypes that fully encapsulate the diverse causes and specific treatments. This review aims to advance our understanding of neonatal AKI detection through the use of biomarkers, subphenotypes, and phenotypes to move towards personalized treatment strategies.
Collapse
Affiliation(s)
- J Hu
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH, USA
| | - D Ananth
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH, USA
| | - S K Sethi
- Pediatric Nephrology & Pediatric Kidney Transplantation, Kidney and Urology Institute, Medanta, The Medicity Hospital, Gurgaon, India
| | - N Taliwal
- Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, OH, USA
| | - S Govindan
- Department of Pediatric Nephrology, Dr. Mehta's Hospitals, Chetpet and Vellapanchavadi, Chennai, India
| | - R Raina
- Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, OH, USA
- Department of Nephrology, Akron Children's Hospital, Akron, OH, USA
| |
Collapse
|
46
|
Sandhya P, Akaishi T, Fujihara K, Aoki M. A novel association of osmotic demyelination in Sjögren's syndrome prompts revisiting role of aquaporins in CNS demyelinating diseases: A literature review. Mult Scler Relat Disord 2023; 69:104466. [PMID: 36584554 DOI: 10.1016/j.msard.2022.104466] [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: 09/30/2022] [Revised: 11/21/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Primary Sjögren's syndrome (SS) is a chronic systemic autoimmune disease with varied neurological manifestations. SS is associated with anti-aquaporin-4 antibody (AQP4-IgG)-positive neuromyelitis optica spectrum disorder (NMOSD), a demyelinating autoimmune disorder of the central nervous system (CNS). Intriguingly, there are reports of osmotic demyelinating syndrome (ODS), a supposedly non-inflammatory disorder, in the context of SS and renal tubular acidosis (RTA), both of which are not yet established risk factors for ODS. METHODS A literature search was undertaken to identify case reports of ODS in patients with SS. Details of the clinical and laboratory features of these patients were compiled. Additionally, we searched for NMOSD in patients with SS. We looked for co-existing RTA in patients with SS-ODS as well as SS-NMOSD. We also screened for reports of ODS in RTA without underlying SS. RESULTS & DISCUSSION We identified 15 patients (all women, median age 40 years) with ODS in SS, and all of these patients had comorbid RTA. There were only three reported cases of ODS in RTA without underlying SS. We identified a total of 67 patients with SS-NMOSD, of whom only 3 (4.5%) had RTA. Hence, unlike NMOSD, the development of ODS in SS requires a prolonged osmotic or electrolyte abnormality caused by the comorbid RTA. The 15 patients with ODS and SS -RTA, showed heterogeneous clinical manifestations and outcomes. The most common symptom was quadriparesis, seen in 14 of the 15 patients. Eleven of the 15 patients had one of the following features, either alone or in combination: worsening of the sensorium, extensor plantar response, dysphagia/dysarthria, and facial palsy. The latter four manifestations were present at the onset in 7 patients and later in the course of the illness in the remaining 4 patients. Ocular palsy was seen in only four of the 15 patients and was a late manifestation. One patient who had extensive long-segment myelitis and subsequent ODS died, but most patients recovered without significant sequelae. None had hyponatremia, while all patients had hypokalemia and/or hypernatremia. Hypokalemia causing nephrogenic diabetes insipidus (NDI) followed by rapid rise in sodium and the resultant osmotic stress could potentially explain the occurrence of ODS in SS-RTA. Aquaporin (AQP) in astrocytes is implicated in ODS, and renal AQP is downregulated in NDI. Antibodies against AQPs are present in some patients with SS. Defective AQP is therefore a common link underlying all the connected diseases, namely SS, NDI, and ODS, raising the possibility of immune-mediated AQP dysfunction in the pathogenesis. CONCLUSION The hitherto unreported association between SS-RTA and ODS may implicate SS and/or RTA in the development of ODS. In the setting of SS-RTA, ODS must be suspected when a patient with flaccid quadriparesis does not respond to the correction of potassium or develops additional neurological features along with a rise in sodium. Defective functions of AQPs may be a possible mechanism linking demyelinating CNS lesions, SS, and RTA. Studies evaluating AQP functions and serum antibodies against AQPs in these conditions are warranted.
Collapse
Affiliation(s)
- Pulukool Sandhya
- Department of Rheumatology, St Stephen's Hospital, Delhi-110054, India.
| | - Tetsuya Akaishi
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Kazuo Fujihara
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Masashi Aoki
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
47
|
Costa IPD, Hautem N, Schiano G, Uchida S, Nishino T, Devuyst O. Fasting influences aquaporin expression, water transport and adipocyte metabolism in the peritoneal membrane. Nephrol Dial Transplant 2022; 38:1408-1420. [PMID: 36520078 DOI: 10.1093/ndt/gfac318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The water channels AQP1 and AQP7 are abundantly expressed in the peritoneal membrane. While AQP1 facilitates water transport during peritoneal dialysis (PD), the role of AQP7, which mediates glycerol transport during fasting, remains unknown. METHODS We investigated the distribution of AQP7 and AQP1 and used a mouse model of PD to investigate the role of AQP7 in the peritoneal membrane at baseline and after fasting. Results. Single nucleus RNA-sequencing revealed that AQP7 was mostly detected in mature adipocytes, whereas AQP1 was essentially expressed in endothelial cells. Fasting induced significant decreases in whole body fat, plasma glucose, insulin, and triglycerides, as well as higher plasma glycerol and corticosterone levels in mice, paralleled by major decreases in adipocyte size and levels of fatty acid synthase and leptin, and increased levels of hormone sensitive lipase mRNAs in the peritoneum. Mechanistically, fasting upregulated the expression of AQP1 and AQP7 in the peritoneum, with increased ultrafiltration but no change in small solute transport. Studies based on Aqp1 and Aqp7 knockout mice and RU-486 inhibition demonstrated that the glucocorticoid induction of AQP1 mediates the increase in ultrafiltration whereas AQP7 regulates the size of adipocytes in the peritoneum. CONCLUSIONS Fasting induces a coordinated regulation of lipolytic and lipogenic factors and aqua(glycero)porins in the peritoneum, driving structural and functional changes. These data yield novel information on the specific roles of aquaporins in the peritoneal membrane and indicate that fasting improves fluid removal in a mouse model of PD.
Collapse
Affiliation(s)
| | | | - Gugliemo Schiano
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Shinichi Uchida
- Department of Nephrology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomoya Nishino
- IREC, UCLouvain, Brussels, Belgium.,Department of Nephrology, Nagasaki University Hospital, Nagasaki, Japan
| | - Olivier Devuyst
- IREC, UCLouvain, Brussels, Belgium.,Institute of Physiology, University of Zurich, Zurich, Switzerland.,Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
48
|
El-Agawy MSED, Badawy AMM, Rabei MR, Elshaer MMA, El Nashar EM, Alghamdi MA, Alshehri MA, Elsayed HRH. Methotrexate-Induced Alteration of Renal Aquaporins 1 and 2, Oxidative Stress and Tubular Apoptosis Can Be Attenuated by Omega-3 Fatty Acids Supplementation. Int J Mol Sci 2022; 23:12794. [PMID: 36361584 PMCID: PMC9653681 DOI: 10.3390/ijms232112794] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/08/2022] [Accepted: 10/19/2022] [Indexed: 09/10/2023] Open
Abstract
Methotrexate (MTX) is a potent anti-cancer drug, commonly associated with nephrotoxicity via the induction of oxidative stress and apoptosis with alteration of renal water channel proteins, namely aquaporins (AQPs). Omega-3 long-chain polyunsaturated fatty acids (LC-PUFA) have shown cytoprotective effects through their anti-oxidant and antiapoptotic activities. The present study aims for the first time to explore the role of LC-PUFA against MTX-induced nephrotoxicity. Rats were divided into the following groups: saline control, LC-PUFA control, MTX, MTX + LC-PUFA (150 mg/kg), or MTX + LC-PUFA (300 mg/kg). Then, H&E staining and immunohistochemical staining for the anti-apoptosis marker B-cell lymphoma 2 (BCL-2), the apoptosis marker BCL2-Associated X Protein (BAX), the proinflammatory marker Nuclear factor kappa B (NF-kB), AQPs 1 and 2 were performed in kidney sections with an assessment of renal oxidative stress. The MTX caused a renal histopathological alteration, upregulated renal BAX and NF-kB, downregulated Bcl-2 and AQP1, altered the distribution of AQP2, and caused oxidative stress. The LC-PUFA attenuated the pathological changes and decreased renal BAX and NF-kB, increased BCL-2 and AQP1, restored the normal distribution of AQP2, and decreased the oxidative stress. Therefore, LC-PUFA is a good adjuvant to MTX to prevent its adverse effects on kidneys through its antiapoptotic, antioxidant, and anti-inflammatory effect and its role in the restoration of the expression of AQPs 1 and 2.
Collapse
Affiliation(s)
- Mosaab Salah El-din El-Agawy
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
- Department of Anatomy, Faculty of Medicine, New Mansoura University, New Mansoura 35712, Egypt
| | | | - Mohammed R. Rabei
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
- Department of Physiology, Faculty of Medicine, King Salman International University, South Sinai 46511, Egypt
| | - Mohamed Mahmoud Abdelraheem Elshaer
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
- Department of Clinical Pharmacology, Faculty of Medicine, King Salman International University, South Sinai 46511, Egypt
| | - Eman Mohamad El Nashar
- Department of Anatomy, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha 13511, Egypt
| | - Mansour A. Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
- Genomics and Personalized Medicine Unit, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
| | - Mohammed A. Alshehri
- Nephrology Section, Internal Medicine Department, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
| | - Hassan Reda Hassan Elsayed
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
- Department of Anatomy, Faculty of Medicine, New Mansoura University, New Mansoura 35712, Egypt
| |
Collapse
|
49
|
Wang B, Wang Y, Wen Y, Zhang YL, Ni WJ, Tang TT, Cao JY, Yin Q, Jiang W, Yin D, Li ZL, Lv LL, Liu BC. Tubular-specific CDK12 knockout causes a defect in urine concentration due to premature cleavage of the slc12a1 gene. Mol Ther 2022; 30:3300-3312. [PMID: 35581939 PMCID: PMC9552909 DOI: 10.1016/j.ymthe.2022.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/21/2022] [Accepted: 05/11/2022] [Indexed: 11/27/2022] Open
Abstract
Cyclin-dependent kinase 12 (CDK12) plays a critical role in regulating gene transcription. CDK12 inhibition is a potential anticancer therapeutic strategy. However, several clinical trials have shown that CDK inhibitors might cause renal dysfunction and electrolyte disorders. CDK12 is abundant in renal tubular epithelial cells (RTECs), but the exact role of CDK12 in renal physiology remains unclear. Genetic knockout of CDK12 in mouse RTECs causes polydipsia, polyuria, and hydronephrosis. This phenotype is caused by defects in water reabsorption that are the result of reduced Na-K-2Cl cotransporter 2 (NKCC2) levels in the kidney. In addition, CKD12 knockout causes an increase in Slc12a1 (which encodes NKCC2) intronic polyadenylation events, which results in Slc12a1 truncated transcript production and NKCC2 downregulation. These findings provide novel insight into CDK12 being necessary for maintaining renal homeostasis by regulating NKCC2 transcription, which explains the critical water and electrolyte disturbance that occurs during the application of CDK12 inhibitors for cancer treatment. Therefore, there are safety concerns about the clinical use of these new anticancer drugs.
Collapse
Affiliation(s)
- Bin Wang
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Yao Wang
- Nanjing Medical University, Nanjing, Jiangsu, China; Department of Nephrology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yi Wen
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China.
| | - Yi-Lin Zhang
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Wei-Jie Ni
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Tao-Tao Tang
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Jing-Yuan Cao
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Qing Yin
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Wei Jiang
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Di Yin
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Lin-Li Lv
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China; Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
50
|
Mishra M, Nichols L, Dave AA, Pittman EH, Cheek JP, Caroland AJV, Lotwala P, Drummond J, Bridges CC. Molecular Mechanisms of Cellular Injury and Role of Toxic Heavy Metals in Chronic Kidney Disease. Int J Mol Sci 2022; 23:11105. [PMID: 36232403 PMCID: PMC9569673 DOI: 10.3390/ijms231911105] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 01/10/2023] Open
Abstract
Chronic kidney disease (CKD) is a progressive disease that affects millions of adults every year. Major risk factors include diabetes, hypertension, and obesity, which affect millions of adults worldwide. CKD is characterized by cellular injury followed by permanent loss of functional nephrons. As injured cells die and nephrons become sclerotic, remaining healthy nephrons attempt to compensate by undergoing various structural, molecular, and functional changes. While these changes are designed to maintain appropriate renal function, they may lead to additional cellular injury and progression of disease. As CKD progresses and filtration decreases, the ability to eliminate metabolic wastes and environmental toxicants declines. The inability to eliminate environmental toxicants such as arsenic, cadmium, and mercury may contribute to cellular injury and enhance the progression of CKD. The present review describes major molecular alterations that contribute to the pathogenesis of CKD and the effects of arsenic, cadmium, and mercury on the progression of CKD.
Collapse
Affiliation(s)
- Manish Mishra
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Larry Nichols
- Department of Pathology and Clinical Sciences Education, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Aditi A. Dave
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Elizabeth H Pittman
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - John P. Cheek
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Anasalea J. V. Caroland
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Purva Lotwala
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - James Drummond
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Christy C. Bridges
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| |
Collapse
|