1
|
Shu L, Quan L, Wang Y, Chen Y, Yong C, Tian F, Gao K, Zhou E. Suyin Detoxification Prescription Regulates the Klotho and ERK/NF-κB Signaling Pathways to Alleviate Renal Injury. Cell Biochem Biophys 2025:10.1007/s12013-025-01695-5. [PMID: 39966333 DOI: 10.1007/s12013-025-01695-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2025] [Indexed: 02/20/2025]
Abstract
Chronic Kidney Disease (CKD) is an irreversible pathological condition resulting from the gradual accumulation of nephrotoxic substances. The application of the "Kidney Toxicity" theory in CKD treatment holds significant research potential and promising clinical prospects. We established a murine model of unilateral ureteral obstruction (UUO) and administered the traditional Chinese medicine prescription Suyin detoxification prescription (SDP). The efficacy of SDP was assessed by comparing the expression levels of Klotho, NF-κB, pNF-κB, VEGF, α-SMA, pERK1/2, and ERK1/2 proteins with those in the Sham operation group using immunohistochemistry, RT-qPCR, and Western Blot techniques. The study revealed a significant reduction in the expression of Klotho and VEGF proteins during the progression of renal fibrosis in mice, while there was a marked increase in the levels of pNF-κB, α-SMA, and pERK1/2 proteins, demonstrating statistical significance (p < 0.05). In UUO mice treated with a high dose of SDP, these proteins exhibited an opposite expression trend compared to that observed in pure operated model mice, with statistically significant differences (p < 0.05). Subsequently, we investigated the relationship between Klotho protein and the ERK/NF-κB signaling pathway-related proteins in human umbilical vein endothelial cells (HUVECs). Knockdown of Klotho protein in HUVECs confirmed its potential as a target for SDP's renal protective effects in vivo by regulating ERK/NF-κB signaling pathway-related proteins to some extent. The renoprotective effect of SDP is mediated through modulation of Klotho protein expression in renal tissues, thereby influencing the ERK/NF-κB signaling pathway and ameliorating the inflammatory processes associated with renal fibrosis. The present study has significantly contributed to the advancement and refinement of the pathogenesis of "Kidney Toxicity" as well as the therapeutic approach of "Kidney Detoxification".
Collapse
Affiliation(s)
- Lianghui Shu
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
- Department of Nephrology, Wuxi Ninth People's Hospital Affiliated of Soochow University, Wuxi, People's Republic of China
| | - Li Quan
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Yanping Wang
- Department of Nephrology, Afffliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Yanlin Chen
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Chen Yong
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People's Republic of China
| | - Fang Tian
- Research Center of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People's Republic of China
| | - Kun Gao
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People's Republic of China
- Nanjing University of Chinese Medicine, Jiangsu University Key Laboratory of Tonifying Kidney and Anti-senescenc, Nanjing, People's Republic of China
| | - Enchao Zhou
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People's Republic of China.
- Nanjing University of Chinese Medicine, Jiangsu University Key Laboratory of Tonifying Kidney and Anti-senescenc, Nanjing, People's Republic of China.
| |
Collapse
|
2
|
Addario G, Moroni L, Mota C. Kidney Fibrosis In Vitro and In Vivo Models: Path Toward Physiologically Relevant Humanized Models. Adv Healthc Mater 2025:e2403230. [PMID: 39906010 DOI: 10.1002/adhm.202403230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/14/2025] [Indexed: 02/06/2025]
Abstract
Chronic kidney disease (CKD) affects over 10% of the global population and is a leading cause of mortality. Kidney fibrosis, a key endpoint of CKD, disrupts nephron tubule anatomy and filtration function, and disease pathomechanisms are not fully understood. Kidney fibrosis is currently investigated with in vivo models, that gradually support the identification of possible mechanisms of fibrosis, but with limited translational research, as they do not fully recapitulate human kidney physiology, metabolism, and molecular pathways. In vitro 2D cell culture models are currently used, as a starting point in disease modeling and pharmacology, however, they lack the 3D kidney architecture complexity and functions. The failure of several therapies and drugs in clinical trials highlights the urgent need for advanced 3D in vitro models. This review discusses the urinary system's anatomy, associated diseases, and diagnostic methods, including biomarker analysis and tissue biopsy. It evaluates 2D and in vivo models, highlighting their limitations. The review explores the state-of-the-art 3D-humanized in vitro models, such as 3D cell aggregates, on-chip models, biofabrication techniques, and hybrid models, which aim to mimic kidney morphogenesis and functions. These advanced models hold promise for translating new therapies and drugs for kidney fibrosis into clinics.
Collapse
Affiliation(s)
- Gabriele Addario
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, ER Maastricht, 6229, The Netherlands
| | - Lorenzo Moroni
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, ER Maastricht, 6229, The Netherlands
| | - Carlos Mota
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, ER Maastricht, 6229, The Netherlands
| |
Collapse
|
3
|
Giuliani KTK, Nag P, Adams BC, Wang X, Hong S, Grivei A, Johnston RL, Waddell N, Ho KKC, Tian Y, Khan MA, Kim CS, Ng MSY, Gobe G, Ungerer JPJ, Forbes JM, Healy HG, Kassianos AJ. Human proximal tubular epithelial cell interleukin-1 receptor signalling triggers G2/M arrest and cellular senescence during hypoxic kidney injury. Cell Death Dis 2025; 16:61. [PMID: 39890773 PMCID: PMC11785723 DOI: 10.1038/s41419-025-07386-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/12/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
Hypoxia and interleukin (IL)-1β are independent mediators of tubulointerstitial fibrosis, the histological hallmark of chronic kidney disease (CKD). Here, we examine how hypoxia and IL-1β act in synergy to augment maladaptive proximal tubular epithelial cell (PTEC) repair in human CKD. Ex vivo patient-derived PTECs were cultured under normoxic (21% O2) or hypoxic (1% O2) conditions in the absence or presence of IL-1β and examined for maladaptive repair signatures. Hypoxic PTECs incubated with IL-1β displayed a discrete transcriptomic profile distinct from PTECs cultured under hypoxia alone, IL-1β alone or under normoxia. Hypoxia+IL-1β-treated PTECs had 692 'unique' differentially expressed genes (DEGs) compared to normoxic PTECs, with 'cell cycle' the most significantly enriched KEGG pathway based on 'unique' down-regulated DEGs (including CCNA2, CCNB1 and CCNB2). Hypoxia+IL-1β-treated PTECs displayed signatures of cellular senescence, with reduced proliferation, G2/M cell cycle arrest, increased p21 expression, elevated senescence-associated β-galactosidase (SA-β-gal) activity and increased production of pro-inflammatory/fibrotic senescence-associated secretory phenotype (SASP) factors compared to normoxic conditions. Treatment of Hypoxia+IL-1β-treated PTECs with either a type I IL-1 receptor (IL-1RI) neutralizing antibody or a senolytic drug combination, quercetin+dasatinib, attenuated senescent cell burden. In vitro findings were validated in human CKD bio-specimens (kidney tissue, urine), with elevated PTEC IL-1RI expression and senescence (SA-β-gal activity) detected in fibrotic kidneys and numbers of senescent (SA-β-gal+) urinary PTECs correlating with urinary IL-1β levels and severity of interstitial fibrosis. Our data identify a mechanism whereby hypoxia in combination with IL-1β/IL-1RI signalling trigger PTEC senescence, providing novel therapeutic and diagnostic check-points for restoring tubular regeneration in human CKD.
Collapse
Affiliation(s)
- Kurt T K Giuliani
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Purba Nag
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Benjamin C Adams
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Xiangju Wang
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Seokchan Hong
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Centre, Seoul, Republic of Korea
| | - Anca Grivei
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | | | - Nicola Waddell
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Kenneth K C Ho
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Yilin Tian
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Muhammad Ali Khan
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Kidney Disease Research Collaborative, Princess Alexandra Hospital and University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Dhaka, Bangladesh
| | - Chang Seong Kim
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Department of Internal Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Monica S Y Ng
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Glenda Gobe
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Kidney Disease Research Collaborative, Princess Alexandra Hospital and University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Jacobus P J Ungerer
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Josephine M Forbes
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Mater Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Helen G Healy
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Andrew J Kassianos
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia.
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia.
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
- Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.
| |
Collapse
|
4
|
Qian L, Hu W, Wang Y, Waheed YA, Hu S, Sun D, Li S. LncRNA TUG1 mitigates chronic kidney disease through miR-542-3p/HIF-1α/VEGF axis. Heliyon 2025; 11:e40891. [PMID: 39811365 PMCID: PMC11730199 DOI: 10.1016/j.heliyon.2024.e40891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/29/2024] [Accepted: 12/02/2024] [Indexed: 01/16/2025] Open
Abstract
Renal interstitial fibrosis (RIF) is a common pathway in chronic kidney disease (CKD) that ultimately leads to end-stage renal failure, worsening both glomerulosclerosis and interstitial fibrosis. Ten percent of the adult population in the world suffers from CKD, and as the ageing population continues to rise, it is increasingly regarded as a global threat-a silent epidemic. CKD has been discovered to be closely associated with both long noncoding RNAs (lncRNAs) and microRNAs (miRNAs), while the precise molecular processes behind this relationship are still unclear. This study evaluated the impact of miR-542-3p and lncRNA TUG1 on renal fibrosis, along with the underlying regulatory mechanisms. Through in vitro tube formation assays, research demonstrated that knocking down lncRNA TUG1 may enhance angiogenesis and repair damaged endothelial cell-cell connections. We used Western blot and qRT-PCR methods in the unilateral ureteral obstruction (UUO) model to identify tissue hypoxia and fibrotic lesions. Additionally, a cutting-edge method known as fluorescence microangiography (FMA) was employed to detect damage to the peritubular capillaries (PTCs), with MATLAB software utilised for data evaluation. Furthermore, the coexpression of CD31 and α-SMA helped identify cells in the obstructed kidney that were transitioning from endothelium to myofibroblasts. On the contrary, lncRNA TUG1 downregulation showed a protective effect against the transition from endothelial cells to myofibroblasts. Additionally, knocking down lncRNA TUG1 has been shown to reduce the expression of fibrotic markers by alleviating tissue hypoxia. This effect was significantly counteracted by the inhibition of miR-542-3p. Collectively, our findings offer fresh perspectives on how lncRNA TUG1 and the miR-542-3p/HIF-1α/VEGF axis are regulated as renal fibrosis advances.
Collapse
Affiliation(s)
- Luoxiang Qian
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 221002, China
- Department of Internal Medicine, Weinan Maternal and Child Health Hospital, Weinan, 714000, China
| | - Wanru Hu
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 221002, China
| | - Yanping Wang
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 221002, China
| | | | - Shuqun Hu
- Laboratory of Emergency Medicine, Second Clinical Medical College of Xuzhou Medical University, Xuzhou, 221002, China
| | - Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 221002, China
- Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, 221002, China
| | - Shulin Li
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 221002, China
| |
Collapse
|
5
|
Nast CC. Shape, Size, and Spatial Relationships: Peritubular Capillary Features in Kidney Fibrosis and Disease Progression. Clin J Am Soc Nephrol 2025; 20:01277230-990000000-00530. [PMID: 39792456 PMCID: PMC11835152 DOI: 10.2215/cjn.0000000647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Affiliation(s)
- Cynthia C Nast
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
6
|
Huang Y, Cong A, Li J, Zhou Z, Zhou H, Su C, Hu Z, Hou FF, Cao W. Glycolysis in Peritubular Endothelial Cells and Microvascular Rarefaction in CKD. J Am Soc Nephrol 2025; 36:19-33. [PMID: 39226371 PMCID: PMC11706556 DOI: 10.1681/asn.0000000000000488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024] Open
Abstract
Key Points Peritubular endothelial cells have a hypoglycolytic metabolism in CKD. Restoration of glycolysis in CKD peritubular endothelial cells by overexpressing 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase attenuates microvascular rarefaction and kidney fibrosis. Strategies targeting the metabolic defect in glycolysis in peritubular endothelial cells may be effective in the treatment of CKD. Background Peritubular endothelial cell dropout leading to microvascular rarefaction is a common manifestation of CKD. The role of metabolism reprogramming in peritubular endothelial cell loss in CKD is undetermined. Methods Single-cell sequencing and metabolic analysis were used to characterize the metabolic profile of peritubular endothelial cells from patients with CKD and from CKD mouse models. In vivo and in vitro models demonstrated metabolic reprogramming in peritubular endothelial cells in conditions of CKD and its contribution to microvascular rarefaction. Results In this study, we identified glycolysis as a top dysregulated metabolic pathway in peritubular endothelial cells from patients with CKD. Specifically, CKD peritubular endothelial cells were hypoglycolytic while displaying an antiangiogenic response with decreased proliferation and increased apoptosis. The hypoglycolytic phenotype of peritubular endothelial cells was recapitulated in CKD mouse models and in peritubular endothelial cells stimulated by hydrogen peroxide. Mechanically, oxidative stress, through activating a redox sensor kruppel-like transcription factor 9, downregulated the glycolytic activator 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase expression, thereby reprogramming peritubular endothelial cells toward a hypoglycolytic phenotype. 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase overexpression in peritubular endothelial cells restored hydrogen peroxide–induced reduction in glycolysis and cellular ATP levels and enhanced the G1/S cell cycle transition, enabling peritubular endothelial cells to improve proliferation and reduce apoptosis. Consistently, restoration of peritubular endothelial cell glycolysis in CKD mice, by overexpressing endothelial Pfkfb3, reversed the antiangiogenic response in peritubular endothelial cells and protected the kidney from microvascular rarefaction and fibrosis. By contrast, suppression of glycolysis by endothelial Pfkfb3 deletion exacerbated microvascular rarefaction and fibrosis in CKD mice. Conclusions Our study revealed a disrupted regulation of glycolysis in peritubular endothelial cells as an initiator of microvascular rarefaction in CKD. Restoration of peritubular endothelial cell glycolysis in CKD kidney improved microvascular rarefaction and ameliorated fibrotic lesions.
Collapse
Affiliation(s)
- Yujie Huang
- Division of Nephrology, Nanfang Hospital, Southern Medical University; State Key Laboratory of Organ Failure Research; National Clinical Research Center for Kidney Disease; Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Furusho T, Das R, Hakui H, Sairavi A, Adachi K, Galbraith-Liss MS, Rajagopal P, Horikawa M, Luo S, Li L, Yamada K, Andeen N, Dissen GA, Nakai H. Enhancing gene transfer to renal tubules and podocytes by context-dependent selection of AAV capsids. Nat Commun 2024; 15:10728. [PMID: 39737896 PMCID: PMC11685967 DOI: 10.1038/s41467-024-54475-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 11/13/2024] [Indexed: 01/01/2025] Open
Abstract
AAV vectors show promise for gene therapy; however, kidney gene transfer remains challenging. Here we conduct a barcode-seq-based comparison of 47 AAV capsids administered through different routes in mice, followed by individual validation. We find that local delivery of AAV-KP1, but not AAV9, via the renal vein or pelvis effectively transduces proximal tubules with minimal off-target liver transduction, while systemic AAV9, but not AAV-KP1, enhances proximal tubule and podocyte transduction in chronic kidney disease. We demonstrate that these contrasting observations are partly due to differences in their pharmacokinetics. Importantly, we show that renal pelvis injection overcomes pre-existing immunity, leading to robust and exclusive proximal tubule transduction, in non-human primates (NHPs). In addition, we highlight drastic differences in renal transduction profiles between mice and NHPs. Thus, this study provides mechanistic insights and underscores importance of context-dependent selection of AAV capsids to overcome challenges in gene delivery to the kidney.
Collapse
Affiliation(s)
- Taisuke Furusho
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Ranjan Das
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Hideyuki Hakui
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Anusha Sairavi
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Kei Adachi
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Mia S Galbraith-Liss
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Pratheppa Rajagopal
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Masahiro Horikawa
- Dotter Department of Interventional Radiology, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Shuhua Luo
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Lena Li
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Kentaro Yamada
- Dotter Department of Interventional Radiology, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Nicole Andeen
- Department of Pathology, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Gregory A Dissen
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
- Molecular Virology Core, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Hiroyuki Nakai
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, Portland, OR, USA.
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA.
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University School of Medicine, Portland, OR, USA.
| |
Collapse
|
8
|
Lee K, Jang HR, Rabb H. Lymphocytes and innate immune cells in acute kidney injury and repair. Nat Rev Nephrol 2024; 20:789-805. [PMID: 39095505 DOI: 10.1038/s41581-024-00875-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 08/04/2024]
Abstract
Acute kidney injury (AKI) is a common and serious disease entity that affects native kidneys and allografts but for which no specific treatments exist. Complex intrarenal inflammatory processes driven by lymphocytes and innate immune cells have key roles in the development and progression of AKI. Many studies have focused on prevention of early injury in AKI. However, most patients with AKI present after injury is already established. Increasing research is therefore focusing on mechanisms of renal repair following AKI and prevention of progression from AKI to chronic kidney disease. CD4+ and CD8+ T cells, B cells and neutrophils are probably involved in the development and progression of AKI, whereas regulatory T cells, double-negative T cells and type 2 innate lymphoid cells have protective roles. Several immune cells, such as macrophages and natural killer T cells, can have both deleterious and protective effects, depending on their subtype and/or the stage of AKI. The immune system not only participates in injury and repair processes during AKI but also has a role in mediating AKI-induced distant organ dysfunction. Targeted manipulation of immune cells is a promising therapeutic strategy to improve AKI outcomes.
Collapse
Affiliation(s)
- Kyungho Lee
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Cell and Gene Therapy Institute, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Nephrology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hye Ryoun Jang
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Cell and Gene Therapy Institute, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hamid Rabb
- Nephrology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
9
|
Xiao B, Huang J, Chen L, Lin Y, Luo J, Chen H, Fu L, Tang F, Ouyang W, Wu Y. Ultra-processed food consumption and the risk of incident chronic kidney disease: a systematic review and meta-analysis of cohort studies. Ren Fail 2024; 46:2306224. [PMID: 38345016 PMCID: PMC10863522 DOI: 10.1080/0886022x.2024.2306224] [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: 07/19/2023] [Accepted: 01/11/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Recent individual studies have indicated that ultra-processed food (UPF) consumption may be associated with the incidence of chronic kidney disease (CKD). We conducted a systematic review and meta-analysis based on those longitudinal studies evaluating the relationship between UPF consumption and the risk of incident CKD, and synthesizing the results. METHOD PubMed, Embase, The Cochrane Library, Web of Science, and Scopus were searched from inception through 22 March 2023. Any longitudinal studies evaluating the relationship between UPF consumption and the risk of incident CKD were included. Two researchers independently conducted the literature screening and data extraction. RR and its 95% CI were regarded as the effect size. The Newcastle-Ottawa Scale (NOS) was applied to assess the quality of the studies included, and the effect of UPF consumption on the risk of incident CKD was analyzed with STATA version 15.1. This study's protocol was registered in PROSPERO (CRD42023411951). RESULTS Four cohort studies with a total of 219,132 participants were included after screening. The results of the meta-analysis suggested that the highest UPF intake was associated with an increased risk of incident CKD (RR = 1.25; 95% CI: 1.18-1.33). CONCLUSIONS High-dose UPF intake was associated with an increased risk of incident CKD. However, the underlying mechanisms remain unknown. Thus, more standardized clinical studies and further exploration of the mechanisms are needed in the future.
Collapse
Affiliation(s)
- Bingjie Xiao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinxian Huang
- The Fourth Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Linyi Chen
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yujie Lin
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianghong Luo
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huifen Chen
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lizhe Fu
- Chronic Disease Management Outpatient, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Fang Tang
- Chronic Disease Management Outpatient, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Wenwei Ouyang
- Key Unit of Methodology in Clinical Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Department of Global Public Health, Karolinska Institute, Stockholm, Sweden
| | - Yifan Wu
- Chronic Disease Management Outpatient, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| |
Collapse
|
10
|
Wang B, Xu M, Fu S, Wang Y, Ling H, Li Y, Li B, Liu X, Ouyang Q, Zhang X, Li A, Zhang X, Liu M. Tiny clue reveals the general trend: a bibliometric and visualized analysis of renal microcirculation. Ren Fail 2024; 46:2329249. [PMID: 38482598 PMCID: PMC10946277 DOI: 10.1080/0886022x.2024.2329249] [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: 12/14/2023] [Accepted: 03/06/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Renal microcirculation plays a pivotal role in kidney function by maintaining structural and functional integrity, facilitating oxygen and nutrient delivery, and waste removal. However, a thorough bibliometric analysis in this area remains lacking. Therefore, we aim to provide valuable insights through a bibliometric analysis of renal microcirculation literature using the Web of Science database. METHODS We collected renal microcirculation-related publications from the Web of Science database from January 01, 1990, to December 31, 2022. The co-authorship of authors, organizations, and countries/regions was analyzed with VOSviewer1.6.18. The co-occurrence of keywords and co-cited references were analyzed using CiteSpace6.1.R6 software to generate visualization maps. Additionally, burst detection was applied to keywords and cited references to forecast research hotspots and future trends. RESULTS Our search yielded 7462 publications, with the American Journal of Physiology-Renal Physiology contributing the most articles. The United States, Mayo Clinic, and Lerman Lilach O emerged with the highest publication count, indicating their active collaborations. 'Type 2 diabetes' was the most significant keyword cluster, and 'diabetic kidney disease' was the largest cluster of cited references. 'Cardiovascular outcome' and 'diabetic kidney diseases' were identified as keywords in their burst period over the past three years. CONCLUSION Our bibliometric analysis illuminates the contours of nephrology and microcirculation research, revealing a landscape ripe for challenges and the seeds of future scientific innovation. While the trends discerned from the literature emerging opportunities in diagnostic innovation, renal microcirculation research, and precision medicine interventions, their translation to clinical practice is anticipated to be a deliberate process.
Collapse
Affiliation(s)
- Bing Wang
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Mengting Xu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Sunjing Fu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yingyu Wang
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Hao Ling
- Department of Radiology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Yuan Li
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Bingwei Li
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xueting Liu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Qin Ouyang
- Department of Pathology, Wangjing Hospital, China Academy of Chinese Medical Science, Beijing, China
| | - Xiaoyan Zhang
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Ailing Li
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xu Zhang
- Laboratory of Electron Microscopy, Ultrastructural Pathology Center, Peking University First Hospital, Beijing, China
| | - Mingming Liu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Diabetes Research Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
11
|
Chen S, He Q, Yang H, Huang H. Endothelial Birc3 promotes renal fibrosis through modulating Drp1-mediated mitochondrial fission via MAPK/PI3K/Akt pathway. Biochem Pharmacol 2024; 229:116477. [PMID: 39128586 DOI: 10.1016/j.bcp.2024.116477] [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: 04/14/2024] [Revised: 07/12/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Renal fibrosis serves as the shared pathway in chronic kidney disease (CKD) progression towards end-stage renal disease (ESRD). Endothelial-mesenchymal transition (EndMT) is a vital mechanism leading to the generation of myofibroblasts, thereby contributing to the advancement of fibrogenesis. Baculoviral IAP Repeat Containing 3(Birc3) was identified as a crucial inhibitor of cell death and a significant mediator in inflammatory signaling and immunity. However, its involvement in the development of renal interstitial fibrosis via EndMT still needs to be clarified. Herein, elevated levels of Birc3 expression along with EndMT-associated alterations, including increased α-smooth muscle actin (α-SMA) levels and decreased CD31 expression, were observed in fibrotic kidneys of Unilateral Ureteral Obstruction (UUO)-induced mouse models and transforming growth factor-β (TGF-β)-induced EndMT in Human Umbilical Vein Endothelial Cells (HUVECs). Functionally, Birc3 knockdown inhibited EndMT and mitochondrial fission mediated by dynamin-related protein 1 (Drp1) both in vivo and in vitro. Mechanistically, endothelial Birc3 exacerbated Drp-1-induced mitochondrial fission through the MAPK/PI3K/Akt signaling pathway in endothelial cell models stimulated TGF-β. Collectively, these findings illuminate the mechanisms and indicate that targeting Birc3 could offer a promising therapeutic strategy to improve endothelial cell survival and mitigate the progression of CKD.
Collapse
Affiliation(s)
- Shuai Chen
- Department of Urology, Zhongshan People's Hospital, Zhongshan 528400, China
| | - Qingqing He
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huaiyu Yang
- Administrative Office, Zhongshan People's Hospital, Zhongshan 528400, China
| | - Hongxing Huang
- Department of Urology, Zhongshan People's Hospital, Zhongshan 528400, China.
| |
Collapse
|
12
|
Zhou W, Xu C, Niu J, Xiong Y, He Z, Xu H, Zhang M, Wang H, Xu Q, Wang X, Wang Z. Inhibitory effects of Eplerenone on angiogenesis via modulating SGK1/TGF-β pathway in contralateral kidney of CKD pregnancy rats. Cell Signal 2024; 122:111346. [PMID: 39147296 DOI: 10.1016/j.cellsig.2024.111346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/07/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Eplerenone is a selective aldosterone receptor blocker that is effective in preventing the progression of chroinic kidney disease (CKD). However, its mechanism and role in CKD pregnancy still remain uncertain. The aim of this study was to evaluate whether eplerenone could attenuated the fibrosis of unilateral ureteral obstruction (UUO) pregnant rats' contralateral kidney, improved pregnancy outcome and explore its therapeutic mechanisms. METHODS A pregnancy rat model of UUO established, female Wistar rats were randomly assigned into sham-operated group (Sham group),sham-operated combined pregnancy group (SP group), unilateral ureteral obstruction combined pregnancy group (UUO + Pregnancy group), unilateral ureteral obstruction combined pregnancy, administered eplerenone (UUO + Pregnancy+Eplerenone group). On the 18th day of pregnancy, the rats were placed in a metabolic cage, 24 h urine was collected and stored at -80 °C. Next day, all animals were euthanized, and serum was collected by centrifugation and stored at -20 °C. Then the right kidney was extracted, a part of the kidney was placed in 4% paraformaldehyde for morphology, immunohistochemical staining, and immunofluorescence staining, and the other part was placed in a - 80 °C refrigerator for RNA and protein extraction. In vitro, HUVECs was treated with aldosterone, progesterone and estradiol, VEGFA and its receptor blocker bevacizumab. The ability of proliferation, migration and tubularization of HUVECs was detected by CCK-8, scratch wound assay and endothelial tube formation assay. And the co-expression of CD34 and α-SMA of HUVECs was detected by Flow cytometry. RESULTS Immunofluorescence results showed that the co-expression of CD34 and α-SMA increased in the UUO + Pregnancy group was significantly increased. The expression of SGK-1, TGFβ-1, Smad2, Smad3, VEGF-A, VEGFR2, CD34, α-SMA and Collagen I was significantly higher in the kidneys of the UUO + Pregnancy group compared to the Sham group and SP group. Eplerenone inhibited the expression of those results. In vitro, the ability of proliferation, migration and tubularization was increased after treated with aldosterone, aldosterone with progesterone and estradiol or VEGFA. Similarly, the expression of α-SMA on the surface of HUVECs treated with aldosterone, aldosterone with progesterone and estradiol were increased, while eplerenone supressed its expression. CONCLUSION Eplerenone inhibits renal angiogenesis by blocking the SGK-1/TGFβ signal transduction pathway, thereby inhibiting the phenotypic transformation of endothelial cells, slowing down renal fibrosis, and reducing kidney damage caused by pregnancy.
Collapse
Affiliation(s)
- Wenping Zhou
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Chang Xu
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Jieqi Niu
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yunzhao Xiong
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Zhen He
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Hepeng Xu
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Mengjuan Zhang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Hongshuang Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Qingyou Xu
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xiangting Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China.
| | - Zheng Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China.
| |
Collapse
|
13
|
Zhang Q, Qin Z, Wang Q, Lu L, Wang J, Lu M, Wang P, Liu D, Zhou C, Liu Z. Pharmacokinetic profiling of ZCL-278, a cdc42 inhibitor, and its effectiveness against chronic kidney disease. Biomed Pharmacother 2024; 179:117329. [PMID: 39180793 DOI: 10.1016/j.biopha.2024.117329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/09/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024] Open
Abstract
ZCL-278 is a selective small molecule specifically inhibiting the Cdc42-intersectin interaction, yet its in-vivo pharmacokinetic and pharmacodynamic properties against renal diseases had not been determined. Thus, our study explored the absorption, distribution and excretion of ZCL-278 as well as its pharmacological efficacy against chronic kidney disease (CKD). With the optimized detection method, absolute oral bioavailability of ZCL-278 was determined as 10.99 % in male and 17.34 % in female rats. ZCL-278 was rapidly and abundantly distributed in various tissues, especially the kidney and heart, while few excreted through urine and feces. In the adenine-induced CKD mice, the increased plasma creatinine and urea, the decreased body weight as well as the renal pathological alterations, including vacuolization of renal tubular epithelial cells, granular degeneration, cell flattening, luminal dilation, and cylindruria, were significantly ameliorated after ZCL-278 administration. Moreover, ZCL-278 could also reverse the increased intensities of renal inflammation and fibrosis in the CKD mice. These results clarified the pharmacokinetics of ZCL-278 in rats and preliminarily indicated that ZCL-278 has favorable pharmacodynamic properties for CKD primed for lead development and optimization, warranting further drug development.
Collapse
Affiliation(s)
- Qing Zhang
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Province Research Center for Kidney Disease, Zhengzhou, China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhiying Qin
- Department of Pharmacy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China; Henan Engineering Research Center for Application and Translation of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Qiang Wang
- Department of Nephrology, The Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Liqian Lu
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Province Research Center for Kidney Disease, Zhengzhou, China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Jiao Wang
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Province Research Center for Kidney Disease, Zhengzhou, China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Manman Lu
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Province Research Center for Kidney Disease, Zhengzhou, China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Pei Wang
- Blood Purification Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongwei Liu
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Province Research Center for Kidney Disease, Zhengzhou, China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Chunyu Zhou
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Province Research Center for Kidney Disease, Zhengzhou, China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China; Blood Purification Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Zhangsuo Liu
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Province Research Center for Kidney Disease, Zhengzhou, China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China.
| |
Collapse
|
14
|
Gazzard SE, Cullen-McEwen LA, Nikulina M, Clever AB, Gardner BS, Smith DW, Lee CJ, Nyengaard JR, Evans RG, Bertram JF. Alterations to peritubular capillary structure in a rat model of kidney interstitial fibrosis: Implications for oxygen diffusion. Anat Rec (Hoboken) 2024. [PMID: 39238265 DOI: 10.1002/ar.25576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/19/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
Fibrosis and loss of functional capillary surface area may contribute to renal tissue hypoxia in a range of kidney diseases. However, there is limited quantitative information on the impact of kidney disease on the barriers to oxygen diffusion from cortical peritubular capillaries (PTCs) to kidney epithelial tubules. Here, we used stereological methods to quantify changes in total cortical PTC length and surface area, PTC length and surface densities, and diffusion distances between PTCs and kidney tubules in adenine-induced kidney injury. After 7 days of oral gavage of adenine (100 mg), plasma creatinine was 3.5-fold greater than in vehicle-treated rats, while total kidney weight was 83% greater. The total length of PTCs was similar in adenine-treated (1.47 ± 0.23 km (mean ± standard deviation)) to vehicle-treated (1.24 ± 0.24 km) rats, as was the surface density of PTCs (0.025 ± 0.002 vs. 0.024 ± 0.004 μm2/μm3). The total surface area of PTCs was 69% greater in adenine-treated than vehicle-treated rats. However, the length density of PTCs was 28% less in adenine-treated than vehicle-treated rats. Diffusion distances, from PTCs to the basal membrane of the nearest renal tubule (108%), and to the mid-point of the cytoplasmic height of the nearest tubular epithelial cell (57%), were markedly increased. These findings indicate that, in adenine-induced kidney injury, expansion of the renal cortical interstitium increases the distance required for diffusion of oxygen from PTCs to tubules, rendering the kidney cortex susceptible to hypoxia.
Collapse
Affiliation(s)
- Sarah E Gazzard
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Luise A Cullen-McEwen
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Marina Nikulina
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Arnold B Clever
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Bruce S Gardner
- School of Mathematics, Statistics, Chemistry and Physics, Murdoch University, Perth, Western Australia, Australia
| | - David W Smith
- Faculty of Engineering and Mathematical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Chang-Joon Lee
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Jens R Nyengaard
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University; and Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Roger G Evans
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Pre-Clinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - John F Bertram
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
15
|
Hu Y, Lei Y, Yu M, Zhang Y, Huang X, Zhang G, Deng Q. Ultrasound super-resolution imaging for the assessment of renal allograft dysfunction: A pilot study. Heliyon 2024; 10:e36515. [PMID: 39247269 PMCID: PMC11380004 DOI: 10.1016/j.heliyon.2024.e36515] [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: 03/26/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 09/10/2024] Open
Abstract
Background The purpose of this study was to examine the feasibility and practical application of ultrasound (US) super-resolution imaging (SRI) in evaluating microvasculature and measuring renal allograft function. Methods Sixteen consecutive patients who received kidney transplants were prospectively enrolled. The patients were assigned as: normal allograft function (n = 6), and allograft malfunction (n = 10). Localizing each potential contrast signal resulted in super-resolution images (SRI). SRI was utilized to assess micro-vessel density (MVD) and microvascular flow rate, whereas contrast-enhanced (CE) US images were statistically processed to get the time to peak (TTP) and peak intensity. Logistic regression was utilized to evaluate their relationship. Results US SRI may be utilized effectively on allografts to show microvasculature with significantly higher resolution than typical color Doppler flow and CEUS pictures. In the multivariate analysis, MVD and TTP were significant US markers of renal allograft failure (p = 0.031 and p = 0.045). The combination of MVD and TTP produced an AUC of 0.783 (p < 0.05) for allograft dysfunction. Conclusions SRI can accurately portray the microvasculature of renal allografts, while MVD and TTP are appropriate US markers for assessing renal allograft failure.
Collapse
Affiliation(s)
- Yugang Hu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430061, China
| | - Yumeng Lei
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Meihui Yu
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Yao Zhang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430061, China
| | - Xingyue Huang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430061, China
| | - Ge Zhang
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Qing Deng
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430061, China
| |
Collapse
|
16
|
Wang B, Xiong Y, Deng X, Wang Y, Gong S, Yang S, Yang B, Yang Y, Leng Y, Li W, Li W. The role of intercellular communication in diabetic nephropathy. Front Immunol 2024; 15:1423784. [PMID: 39238645 PMCID: PMC11374600 DOI: 10.3389/fimmu.2024.1423784] [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: 04/26/2024] [Accepted: 08/01/2024] [Indexed: 09/07/2024] Open
Abstract
Diabetic nephropathy, a common and severe complication of diabetes, is the leading cause of end-stage renal disease, ultimately leading to renal failure and significantly affecting the prognosis and lives of diabetics worldwide. However, the complexity of its developmental mechanisms makes treating diabetic nephropathy a challenging task, necessitating the search for improved therapeutic targets. Intercellular communication underlies the direct and indirect influence and interaction among various cells within a tissue. Recently, studies have shown that beyond traditional communication methods, tunnel nanotubes, exosomes, filopodial tip vesicles, and the fibrogenic niche can influence pathophysiological changes in diabetic nephropathy by disrupting intercellular communication. Therefore, this paper aims to review the varied roles of intercellular communication in diabetic nephropathy, focusing on recent advances in this area.
Collapse
Affiliation(s)
- Bihan Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yonghong Xiong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinqi Deng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yunhao Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Siyuan Gong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Songyuan Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Baichuan Yang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuhang Yang
- The First Clinical College of Wuhan University, Wuhan, China
| | - Yan Leng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenyuan Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Chen Q, Hazra R, Crosby D, Lenhart D, Lenhart SC, Mondal P, Zhang Y, Nouraie SM, Tan RJ, Esmon CT, Rao LVM, Kim K, Ghosh S. Heme-induced loss of renovascular endothelial protein C receptor promotes chronic kidney disease in sickle mice. Blood 2024; 144:552-564. [PMID: 38820589 PMCID: PMC11307268 DOI: 10.1182/blood.2023023528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/30/2024] [Accepted: 05/18/2024] [Indexed: 06/02/2024] Open
Abstract
ABSTRACT Chronic kidney disease (CKD) is a major contributor to morbidity and mortality in sickle cell disease (SCD). Anemia, induced by chronic persistent hemolysis, is associated with the progressive deterioration of renal health, resulting in CKD. Moreover, patients with SCD experience acute kidney injury (AKI), a risk factor for CKD, often during vaso-occlusive crisis associated with acute intravascular hemolysis. However, the mechanisms of hemolysis-driven pathogenesis of the AKI-to-CKD transition in SCD remain elusive. Here, we investigated the role of increased renovascular rarefaction and the resulting substantial loss of the vascular endothelial protein C receptor (EPCR) in the progressive deterioration of renal function in transgenic SCD mice. Multiple hemolytic events raised circulating levels of soluble EPCR (sEPCR), indicating loss of EPCR from the cell surface. Using bone marrow transplantation and super-resolution ultrasound imaging, we demonstrated that SCD mice overexpressing EPCR were protective against heme-induced CKD development. In a cohort of patients with SCD, plasma sEPCR was significantly higher in individuals with CKD than in those without CKD. This study concludes that multiple hemolytic events may trigger CKD in SCD through the gradual loss of renovascular EPCR. Thus, the restoration of EPCR may be a therapeutic target, and plasma sEPCR can be developed as a prognostic marker for sickle CKD.
Collapse
MESH Headings
- Animals
- Anemia, Sickle Cell/complications
- Anemia, Sickle Cell/pathology
- Anemia, Sickle Cell/metabolism
- Anemia, Sickle Cell/blood
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/blood
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/etiology
- Endothelial Protein C Receptor/metabolism
- Endothelial Protein C Receptor/genetics
- Mice
- Heme/metabolism
- Humans
- Mice, Transgenic
- Male
- Female
- Hemolysis
- Kidney/metabolism
- Kidney/pathology
Collapse
Affiliation(s)
- Qiyang Chen
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Rimi Hazra
- Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
| | - Danielle Crosby
- Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
| | - Diane Lenhart
- Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
| | - Shane C. Lenhart
- Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
| | - Paritosh Mondal
- Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
| | - Yingze Zhang
- Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
- Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Seyed M. Nouraie
- Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
- Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Roderick J. Tan
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Charles T. Esmon
- Coagulation Biology Laboratory, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - L. Vijay Mohan Rao
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX
| | - Kang Kim
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
- Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA
| | - Samit Ghosh
- Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
18
|
Song Y, Zhang J, Luo Z, Wu L, Cai Z, Zhong X, Zeng X, Cao T, Chen HE, Xu S, Wang CY. Association between dietary branched-chain amino acids and multiple chronic conditions among older adults in Chinese communities. Nutr Metab (Lond) 2024; 21:56. [PMID: 39080679 PMCID: PMC11290232 DOI: 10.1186/s12986-024-00825-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/08/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND The association of BCAAs (isoleucine, leucine, and valine) with cardiovascular and cerebrovascular diseases has been widely recognized by researchers, but there is limited evidence to support the relationship between BCAAs and multiple chronic conditions (MCCs) in older adults. This study aimed to explore the correlation between BCAA levels in the diets of older adults and MCCs. METHODS Based on a health management cohort project in Nanshan District of Shenzhen, 4278 individuals over 65 years old were selected as participants via multi-stage stratified sampling from May 2018 to December 2019. Data were collected using a validated semi-quantitative food frequency questionnaire, as well as anthropometric and chronic disease reports. MCC was defined as the coexistence of two or more chronic diseases, namely, hypertension, dyslipidemia, diabetes, CAD, stroke, CKD, and CLD. Multivariate unconditional logistic regression analysis was used to analyze the relationship between dietary BCAAs and MCCs in older adults, and then, gender stratification analysis was performed. A restricted cubic spline model (a fitted smooth curve) was used to determine the dose-response relationship of isoleucine with MCCs. RESULTS A total of 4278 older adults aged 65 and above were included in this study, with an average age of 72.73 ± 5.49 years. The cohort included 1861 males (43.50%). Regardless of whether confounding factors were corrected, isoleucine was a risk factor for MCCs (OR = 3.388, 95%CI:1.415,8.109). After gender stratification, the relationships between dietary isoleucine and MCCs (OR = 6.902, 95%CI:1.875,25.402) and between leucine (OR = 0.506,95%CI:0.309,0.830) and MCCs were significant in women, but not in men. No significant association between valine and MCCs was observed. In addition, isoleucine was a risk factor for MCCs when its intake was greater than 4.297 g/d. CONCLUSION Isoleucine may play an important role in regulating age-related diseases. BCAAs such as isoleucine can be used as risk markers for MCCs in older adults.
Collapse
Affiliation(s)
- Yuanfeng Song
- Department of Public Health and Preventive Medicine, School of Public Health, Zunyi Medical University, Zunyi, 563000, China
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, Guangdong, 518000, China
| | - Ji Zhang
- Department of Public Health and Preventive Medicine, School of Public Health, Zunyi Medical University, Zunyi, 563000, China
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, Guangdong, 518000, China
| | - Ziqiang Luo
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, Guangdong, 518000, China
| | - Lanlan Wu
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, Guangdong, 518000, China
| | - Zhaopei Cai
- Department of Public Health and Preventive Medicine, School of Public Health, Zunyi Medical University, Zunyi, 563000, China
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, Guangdong, 518000, China
| | - Xiaoqi Zhong
- Department of Public Health and Preventive Medicine, School of Public Health, Zunyi Medical University, Zunyi, 563000, China
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, Guangdong, 518000, China
| | - Xiaoxue Zeng
- Department of Public Health and Preventive Medicine, School of Public Health, Zunyi Medical University, Zunyi, 563000, China
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, Guangdong, 518000, China
| | - Tingxi Cao
- Department of Public Health and Preventive Medicine, School of Public Health, Zunyi Medical University, Zunyi, 563000, China
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, Guangdong, 518000, China
| | - Hong-En Chen
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, Guangdong, 518000, China
| | - Shan Xu
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, Guangdong, 518000, China
| | - Chang-Yi Wang
- Department of Public Health and Preventive Medicine, School of Public Health, Zunyi Medical University, Zunyi, 563000, China.
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, Guangdong, 518000, China.
| |
Collapse
|
19
|
Acevedo Cintrón JA, Hunter DA, Schellhardt L, Pan D, Mackinnon SE, Wood MD. Limited Nerve Regeneration across Acellular Nerve Allografts (ANAs) Coincides with Changes in Blood Vessel Morphology and the Development of a Pro-Inflammatory Microenvironment. Int J Mol Sci 2024; 25:6413. [PMID: 38928119 PMCID: PMC11204013 DOI: 10.3390/ijms25126413] [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: 05/14/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
The use of acellular nerve allografts (ANAs) to reconstruct long nerve gaps (>3 cm) is associated with limited axon regeneration. To understand why ANA length might limit regeneration, we focused on identifying differences in the regenerative and vascular microenvironment that develop within ANAs based on their length. A rat sciatic nerve gap model was repaired with either short (2 cm) or long (4 cm) ANAs, and histomorphometry was used to measure myelinated axon regeneration and blood vessel morphology at various timepoints (2-, 4- and 8-weeks). Both groups demonstrated robust axonal regeneration within the proximal graft region, which continued across the mid-distal graft of short ANAs as time progressed. By 8 weeks, long ANAs had limited regeneration across the ANA and into the distal nerve (98 vs. 7583 axons in short ANAs). Interestingly, blood vessels within the mid-distal graft of long ANAs underwent morphological changes characteristic of an inflammatory pathology by 8 weeks post surgery. Gene expression analysis revealed an increased expression of pro-inflammatory cytokines within the mid-distal graft region of long vs. short ANAs, which coincided with pathological changes in blood vessels. Our data show evidence of limited axonal regeneration and the development of a pro-inflammatory environment within long ANAs.
Collapse
Affiliation(s)
| | | | | | | | | | - Matthew D. Wood
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (J.A.A.C.); (D.A.H.); (L.S.); (D.P.); (S.E.M.)
| |
Collapse
|
20
|
Dong Y, Gai Z, Han M, Xu J, Zou K. Reduction in Serum Concentrations of Uremic Toxins Driven by Bifidobacterium Longum Subsp. Longum BL21 is Associated with Gut Microbiota Changes in a Rat Model of Chronic Kidney Disease. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10293-5. [PMID: 38829564 DOI: 10.1007/s12602-024-10293-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 06/05/2024]
Abstract
Gut microbiota dysbiosis and consequent impairment of gut barrier function, culminating in elevated levels of uremic toxins, are prevalent in chronic kidney disease (CKD) patients. These toxins, notably indoxyl sulphate (IS), indole-3-acetic acid (IAA), and trimethylamine oxide (TMAO), are implicated in a spectrum of CKD-related complications, including cardiovascular disease, bone and mineral disorders, and inflammation. The specific impacts of various probiotics on these CKD manifestations remain unexplored. This study delved into the potential of dietary probiotic interventions, particularly Bifidobacterium longum subsp. longum BL21, to modulate gut microbiota and mitigate metabolic disorders in a CKD rat model. Over a six-week period, we administered a dietary regimen of BL21 and conducted comprehensive analyses, including serum uremic toxin quantification and 16S rRNA gene sequencing, to systematically profile gut microbial alterations at the phylogenetic level. Our findings reveal that BL21 intervention significantly ameliorated CKD-induced disruptions in gut microbial populations, enhancing both microbial richness and the relative abundance of key taxa. Importantly, BL21 appeared to exert its beneficial effects by modulating the abundance of crucial species such as Barnesiella and Helicobacter. Functionally, the intervention markedly normalized serum levels of IS, IAA, and TMAO, while potentially attenuating p-cresol sulphate (PCS) and p-cresol glucuronide (PCG) concentrations. Consequently, BL21 demonstrated efficacy in regulating gut microbiota and curtailing the accumulation of uremic toxins. Our results advocate for the utilization of BL21 as a dietary intervention to diminish serum uremic toxins and re-establish gut microbiota equilibrium at the phylogenetic level, underscoring the promise of probiotic strategies in the management of CKD.
Collapse
Affiliation(s)
- Yao Dong
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zhonghui Gai
- Department of Research and Development, Wecare Probiotics Co., Ltd, Suzhou, 215200, China
| | - Mei Han
- Department of Food Quality and Safety, Shanghai Business School, Shanghai, 200235, China
| | - Jiaqi Xu
- Department of Research and Development, Wecare Probiotics Co., Ltd, Suzhou, 215200, China
| | - Kang Zou
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
21
|
Jiang L, Hu X, Feng Y, Wang Z, Tang H, Lin Q, Shen Y, Zhu Y, Xu Q, Li X. Reduction of renal interstitial fibrosis by targeting Tie2 in vascular endothelial cells. Pediatr Res 2024; 95:959-965. [PMID: 38012310 PMCID: PMC10920200 DOI: 10.1038/s41390-023-02893-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 10/08/2023] [Accepted: 10/30/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Tie2, a functional angiopoietin receptor, is expressed in vascular endothelial cells and plays an important role in angiogenesis and vascular stability. This study aimed to evaluate the effects of an agonistic Tie2 signal on renal interstitial fibrosis (RIF) and elucidate the underlying mechanisms. METHODS We established an in vivo mouse model of folic acid-induced nephropathy (FAN) and an in vitro model of lipopolysaccharide-stimulated endothelial cell injury, then an agonistic Tie2 monoclonal antibody (Tie2 mAb) was used to intervent these processes. The degree of tubulointerstitial lesions and related molecular mechanisms were determined by histological assessment, immunohistochemistry, western blotting, and qPCR. RESULTS Tie2 mAb attenuated RIF and reduced the level of fibroblast-specific protein 1 (FSP1). Further, it suppressed vascular cell adhesion molecule-1 (VCAM-1) and increased CD31 density in FAN. In the in vitro model, Tie2 mAb was found to decrease the expression of VCAM-1, Bax, and α-smooth muscle actin (α-SMA). CONCLUSIONS The present findings indicate that the agonistic Tie2 mAb exerted vascular protective effects and ameliorated RIF via inhibition of vascular inflammation, apoptosis, and fibrosis. Therefore, Tie2 may be a potential target for the treatment of this disease. IMPACT This is the first report to confirm that an agonistic Tie2 monoclonal antibody can reduce renal interstitial fibrosis in folic acid-induced nephropathy in mice. This mechanism possibly involves vascular protective effects brought about by inhibition of vascular inflammation, apoptosis and fibrosis. Our data show that Tie2 signal may be a novel, endothelium-specific target for the treatment of tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Lu Jiang
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Xiaohan Hu
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Yajun Feng
- Department of Pediatrics, Jiangyin People's Hospital, Jiangyin, 214400, China
| | - Zhen Wang
- Department of Pediatrics, Zibo Maternal and Child Health Care Hospital, Zibo, 255000, China
| | - Hanyun Tang
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Qiang Lin
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Yunyan Shen
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Yun Zhu
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Qinying Xu
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Xiaozhong Li
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, 215003, China.
| |
Collapse
|
22
|
Ghallab A, González D, Strängberg E, Hofmann U, Myllys M, Hassan R, Hobloss Z, Brackhagen L, Begher-Tibbe B, Duda JC, Drenda C, Kappenberg F, Reinders J, Friebel A, Vucur M, Turajski M, Seddek AL, Abbas T, Abdelmageed N, Morad SAF, Morad W, Hamdy A, Albrecht W, Kittana N, Assali M, Vartak N, van Thriel C, Sous A, Nell P, Villar-Fernandez M, Cadenas C, Genc E, Marchan R, Luedde T, Åkerblad P, Mattsson J, Marschall HU, Hoehme S, Stirnimann G, Schwab M, Boor P, Amann K, Schmitz J, Bräsen JH, Rahnenführer J, Edlund K, Karpen SJ, Simbrunner B, Reiberger T, Mandorfer M, Trauner M, Dawson PA, Lindström E, Hengstler JG. Inhibition of the renal apical sodium dependent bile acid transporter prevents cholemic nephropathy in mice with obstructive cholestasis. J Hepatol 2024; 80:268-281. [PMID: 37939855 PMCID: PMC10849134 DOI: 10.1016/j.jhep.2023.10.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 10/06/2023] [Accepted: 10/23/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND & AIMS Cholemic nephropathy (CN) is a severe complication of cholestatic liver diseases for which there is no specific treatment. We revisited its pathophysiology with the aim of identifying novel therapeutic strategies. METHODS Cholestasis was induced by bile duct ligation (BDL) in mice. Bile flux in kidneys and livers was visualized by intravital imaging, supported by MALDI mass spectrometry imaging and liquid chromatography-tandem mass spectrometry. The effect of AS0369, a systemically bioavailable apical sodium-dependent bile acid transporter (ASBT) inhibitor, was evaluated by intravital imaging, RNA-sequencing, histological, blood, and urine analyses. Translational relevance was assessed in kidney biopsies from patients with CN, mice with a humanized bile acid (BA) spectrum, and via analysis of serum BAs and KIM-1 (kidney injury molecule 1) in patients with liver disease and hyperbilirubinemia. RESULTS Proximal tubular epithelial cells (TECs) reabsorbed and enriched BAs, leading to oxidative stress and death of proximal TECs, casts in distal tubules and collecting ducts, peritubular capillary leakiness, and glomerular cysts. Renal ASBT inhibition by AS0369 blocked BA uptake into TECs and prevented kidney injury up to 6 weeks after BDL. Similar results were obtained in mice with humanized BA composition. In patients with advanced liver disease, serum BAs were the main determinant of KIM-1 levels. ASBT expression in TECs was preserved in biopsies from patients with CN, further highlighting the translational potential of targeting ASBT to treat CN. CONCLUSIONS BA enrichment in proximal TECs followed by oxidative stress and cell death is a key early event in CN. Inhibiting renal ASBT and consequently BA enrichment in TECs prevents CN and systemically decreases BA concentrations. IMPACT AND IMPLICATIONS Cholemic nephropathy (CN) is a severe complication of cholestasis and an unmet clinical need. We demonstrate that CN is triggered by the renal accumulation of bile acids (BAs) that are considerably increased in the systemic blood. Specifically, the proximal tubular epithelial cells of the kidney take up BAs via the apical sodium-dependent bile acid transporter (ASBT). We developed a therapeutic compound that blocks ASBT in the kidneys, prevents BA overload in tubular epithelial cells, and almost completely abolished all disease hallmarks in a CN mouse model. Renal ASBT inhibition represents a potential therapeutic strategy for patients with CN.
Collapse
Affiliation(s)
- Ahmed Ghallab
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany; Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, 83523 Qena, Egypt.
| | - Daniela González
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| | | | - Ute Hofmann
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Auerbachstr. 112, 70376 Stuttgart, Germany
| | - Maiju Myllys
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| | - Reham Hassan
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany; Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, 83523 Qena, Egypt
| | - Zaynab Hobloss
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| | - Lisa Brackhagen
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| | - Brigitte Begher-Tibbe
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| | - Julia C Duda
- Department of Statistics, TU Dortmund University, 44227 Dortmund, Germany
| | - Carolin Drenda
- Department of Statistics, TU Dortmund University, 44227 Dortmund, Germany
| | | | - Joerg Reinders
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| | - Adrian Friebel
- Institute of Computer Science & Saxonian Incubator for Clinical Research (SIKT), University of Leipzig, Haertelstraße 16-18, 04107 Leipzig, Germany
| | - Mihael Vucur
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty at Heinrich-Heine-University, 40225 Dusseldorf, Germany
| | - Monika Turajski
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| | - Abdel-Latief Seddek
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, 83523 Qena, Egypt
| | - Tahany Abbas
- Histology Department, Faculty of Medicine, South Valley University, 83523 Qena, Egypt
| | - Noha Abdelmageed
- Department of Pharmacology, Faculty of Veterinary Medicine, Sohag University, 82524 Sohag, Egypt
| | - Samy A F Morad
- Department of Pharmacology, Faculty of Veterinary Medicine, South Valley University, 83523 Qena, Egypt
| | - Walaa Morad
- Histology Department, Faculty of Medicine, South Valley University, 83523 Qena, Egypt
| | - Amira Hamdy
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, 83523 Qena, Egypt
| | - Wiebke Albrecht
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| | - Naim Kittana
- Department of Biomedical Sciences, An-Najah National University, P.O. Box 7 Nablus, Palestine, Israel
| | - Mohyeddin Assali
- Department of Pharmacy, An-Najah National University, P.O. Box 7 Nablus, Palestine, Israel
| | - Nachiket Vartak
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| | - Christoph van Thriel
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| | - Ansam Sous
- Department of Pharmacy, An-Najah National University, P.O. Box 7 Nablus, Palestine, Israel
| | - Patrick Nell
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| | - Maria Villar-Fernandez
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| | - Cristina Cadenas
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| | - Erhan Genc
- MRI Unit, Leibniz Research Centre for Working Environment and Human Factors, Department of Psychology and Neurosciences, Technical University Dortmund, 44139 Dortmund, Germany
| | - Rosemarie Marchan
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty at Heinrich-Heine-University, 40225 Dusseldorf, Germany
| | | | | | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Stefan Hoehme
- Institute of Computer Science & Saxonian Incubator for Clinical Research (SIKT), University of Leipzig, Haertelstraße 16-18, 04107 Leipzig, Germany
| | - Guido Stirnimann
- University Clinic for Visceral Surgery and Medicine, Inselspital University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Auerbachstr. 112, 70376 Stuttgart, Germany; Departments of Clinical Pharmacology, and of Biochemistry and Pharmacy, University Tuebingen, 72076 Tuebingen, Germany; Cluster of Excellence iFIT (EXC2180), Image-Guided and Functionally Instructed Tumor Therapies, University of Tuebingen, 69120 Tuebingen, Germany
| | - Peter Boor
- Institute of Pathology and Department of Nephrology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany
| | - Kerstin Amann
- Department of Nephropathology, Friedrich-Alexander-University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Jessica Schmitz
- Institute of Pathology, Nephropathology Unit, Hannover Medical School, 30625 Hannover, Germany
| | - Jan H Bräsen
- Institute of Pathology, Nephropathology Unit, Hannover Medical School, 30625 Hannover, Germany
| | - Jörg Rahnenführer
- Department of Statistics, TU Dortmund University, 44227 Dortmund, Germany
| | - Karolina Edlund
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| | - Saul J Karpen
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, GA 30322, United States
| | - Benedikt Simbrunner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria; Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria; Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Mattias Mandorfer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria; Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria; Hans Popper Laboratory of Molecular Hepatology, Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Paul A Dawson
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, GA 30322, United States
| | | | - Jan G Hengstler
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139 Dortmund, Germany.
| |
Collapse
|
23
|
Meijer EM, van Dijk CGM, Giles R, Gijsen K, Chrifi I, Verhaar MC, Cheng C. Induction of Fenestrae in Human Induced Pluripotent Stem Cell-Derived Endothelial Cells for Disease Modeling. Tissue Eng Part A 2024; 30:168-180. [PMID: 38126303 DOI: 10.1089/ten.tea.2023.0236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
The endothelial linings of capillaries, such as those in the kidney and small intestines, possess fenestrae that facilitate fluid and exchange of small molecules. Alterations in the size and number of endothelial fenestrae have been implicated in the pathogenesis of various diseases. The re-creation of fenestrated endothelium using human induced pluripotent stem cells (hiPSCs) provides a promising avenue to investigate the involvement of fenestrae in disease mechanisms and pharmacodynamics. In this project, we aim to induce the formation of fenestrae in nonfenestrated hiPSCs-derived endothelial cells (hiPSC-ECs). Vascular endothelial growth factor A (VEGFA) and phorbol myristate acetate (PMA) were used as inducers of fenestrae in hiPSC-ECs. The assessment of fenestrae formation included gene-expression analysis, scanning electron microscopy (SEM), transmission electron microscopy (TEM), and immunofluorescent staining. Endothelial monolayer functionality was evaluated by dextran permeability assays. Stimulation with VEGFA and PMA significantly induced expression of the diaphragmed fenestrae-associated marker, plasmalemmal vesicle-associated protein (PLVAP), in hiPSC-ECs at the mRNA, and protein levels. SEM analysis revealed VEGFA- and PMA-induced fenestrae structures on the cell membrane of hiPSC-ECs. The increased membrane localization of PLVAP visualized by TEM and immunofluorescent staining supported these findings. The induced fenestrated endothelium in hiPSC-ECs demonstrated selective passage of small solutes across a confluent monolayer with intact cell junctions, confirming functional competence. In conclusion, we present a novel methodology for inducing and regulating fenestrated endothelium in hiPSC-ECs. This innovative approach paves the way for the development of fenestrated microvasculature in human organ-on-a-chip systems, enabling complex disease modeling and physiologically relevant investigations of pharmacodynamics.
Collapse
Affiliation(s)
- Elana M Meijer
- Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Christian G M van Dijk
- Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rachel Giles
- Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Karlijn Gijsen
- Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ihsan Chrifi
- Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
- Experimental Cardiology, Department of Cardiology, Thoraxcenter Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marianne C Verhaar
- Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Caroline Cheng
- Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
- Experimental Cardiology, Department of Cardiology, Thoraxcenter Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
24
|
Steegh FMEG, Keijbeck AA, de Hoogt PA, Rademakers T, Houben AJHM, Reesink KD, Stehouwer CDA, Daemen MJAP, Peutz-Kootstra CJ. Capillary rarefaction: a missing link in renal and cardiovascular disease? Angiogenesis 2024; 27:23-35. [PMID: 37326760 DOI: 10.1007/s10456-023-09883-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/28/2023] [Indexed: 06/17/2023]
Abstract
Patients with chronic kidney disease (CKD) have an increased risk for cardiovascular morbidity and mortality. Capillary rarefaction may be both one of the causes as well as a consequence of CKD and cardiovascular disease. We reviewed the published literature on human biopsy studies and conclude that renal capillary rarefaction occurs independently of the cause of renal function decline. Moreover, glomerular hypertrophy may be an early sign of generalized endothelial dysfunction, while peritubular capillary loss occurs in advanced renal disease. Recent studies with non-invasive measurements show that capillary rarefaction is detected systemically (e.g., in the skin) in individuals with albuminuria, as sign of early CKD and/or generalized endothelial dysfunction. Decreased capillary density is found in omental fat, muscle and heart biopsies of patients with advanced CKD as well as in skin, fat, muscle, brain and heart biopsies of individuals with cardiovascular risk factors. No biopsy studies have yet been performed on capillary rarefaction in individuals with early CKD. At present it is unknown whether individuals with CKD and cardiovascular disease merely share the same risk factors for capillary rarefaction, or whether there is a causal relationship between rarefaction in renal and systemic capillaries. Further studies on renal and systemic capillary rarefaction, including their temporal relationship and underlying mechanisms are needed. This review stresses the importance of preserving and maintaining capillary integrity and homeostasis in the prevention and management of renal and cardiovascular disease.
Collapse
Affiliation(s)
- Floor M E G Steegh
- Department of Pathology, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Anke A Keijbeck
- Department of Pathology, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Patrick A de Hoogt
- Surgery, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Timo Rademakers
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Alfons J H M Houben
- Internal Medicine, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Koen D Reesink
- Biomedical Engineering, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Coen D A Stehouwer
- Internal Medicine, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Mat J A P Daemen
- Department of Pathology, UMC Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Carine J Peutz-Kootstra
- Department of Pathology, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.
- Department of Pathology, Gelre Ziekenhuizen, Apeldoorn, The Netherlands.
- , Porthoslaan 39, 6213 CN, Maastricht, The Netherlands.
| |
Collapse
|
25
|
Locatelli M, Rottoli D, Mahmoud R, Abbate M, Corna D, Cerullo D, Tomasoni S, Remuzzi G, Zoja C, Benigni A, Macconi D. Endothelial Glycocalyx of Peritubular Capillaries in Experimental Diabetic Nephropathy: A Target of ACE Inhibitor-Induced Kidney Microvascular Protection. Int J Mol Sci 2023; 24:16543. [PMID: 38003732 PMCID: PMC10671403 DOI: 10.3390/ijms242216543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Peritubular capillary rarefaction is a recurrent aspect of progressive nephropathies. We previously found that peritubular capillary density was reduced in BTBR ob/ob mice with type 2 diabetic nephropathy. In this model, we searched for abnormalities in the ultrastructure of peritubular capillaries, with a specific focus on the endothelial glycocalyx, and evaluated the impact of treatment with an angiotensin-converting enzyme inhibitor (ACEi). Mice were intracardially perfused with lanthanum to visualise the glycocalyx. Transmission electron microscopy analysis revealed endothelial cell abnormalities and basement membrane thickening in the peritubular capillaries of BTBR ob/ob mice compared to wild-type mice. Remodelling and focal loss of glycocalyx was observed in lanthanum-stained diabetic kidneys, associated with a reduction in glycocalyx components, including sialic acids, as detected through specific lectins. ACEi treatment preserved the endothelial glycocalyx and attenuated the ultrastructural abnormalities of peritubular capillaries. In diabetic mice, peritubular capillary damage was associated with an enhanced tubular expression of heparanase, which degrades heparan sulfate residues of the glycocalyx. Heparanase was also detected in renal interstitial macrophages that expressed tumor necrosis factor-α. All these abnormalities were mitigated by ACEi. Our findings suggest that, in experimental diabetic nephropathy, preserving the endothelial glycocalyx is important in order to protect peritubular capillaries from damage and loss.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, 24126 Bergamo, Italy; (M.L.); (D.R.); (R.M.); (M.A.); (D.C.); (D.C.); (S.T.); (G.R.); (C.Z.); (D.M.)
| | | |
Collapse
|
26
|
Gaupp C, Schmid B, Tripal P, Edwards A, Daniel C, Zimmermann S, Goppelt-Struebe M, Willam C, Rosen S, Schley G. Reconfiguration and loss of peritubular capillaries in chronic kidney disease. Sci Rep 2023; 13:19660. [PMID: 37952029 PMCID: PMC10640592 DOI: 10.1038/s41598-023-46146-4] [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: 02/03/2023] [Accepted: 10/27/2023] [Indexed: 11/14/2023] Open
Abstract
Functional and structural alterations of peritubular capillaries (PTCs) are a major determinant of chronic kidney disease (CKD). Using a software-based algorithm for semiautomatic segmentation and morphometric quantification, this study analyzes alterations of PTC shape associated with chronic tubulointerstitial injury in three mouse models and in human biopsies. In normal kidney tissue PTC shape was predominantly elongated, whereas the majority of PTCs associated with chronic tubulointerstitial injury had a rounder shape. This was reflected by significantly reduced PTC luminal area, perimeter and diameters as well as by significantly increased circularity and roundness. These morphological alterations were consistent in all mouse models and human kidney biopsies. The mean circularity of PTCs correlated significantly with categorized glomerular filtration rates and the degree of interstitial fibrosis and tubular atrophy (IFTA) and classified the presence of CKD or IFTA. 3D reconstruction of renal capillaries revealed not only a significant reduction, but more importantly a substantial simplification and reconfiguration of the renal microvasculature in mice with chronic tubulointerstitial injury. Computational modelling predicted that round PTCs can deliver oxygen more homogeneously to the surrounding tissue. Our findings indicate that alterations of PTC shape represent a common and uniform reaction to chronic tubulointerstitial injury independent of the underlying kidney disease.
Collapse
Affiliation(s)
- Charlotte Gaupp
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
| | - Benjamin Schmid
- Optical Imaging Center Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Philipp Tripal
- Optical Imaging Center Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Aurélie Edwards
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Christoph Daniel
- Department of Nephropathology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| | - Stefan Zimmermann
- Department of Computer Science, University of Applied Sciences Worms, Worms, Germany
| | - Margarete Goppelt-Struebe
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
| | - Carsten Willam
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
| | - Seymour Rosen
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Gunnar Schley
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Ulmenweg 18, 91054, Erlangen, Germany.
| |
Collapse
|
27
|
Zhao BH, Ruze A, Zhao L, Li QL, Tang J, Xiefukaiti N, Gai MT, Deng AX, Shan XF, Gao XM. The role and mechanisms of microvascular damage in the ischemic myocardium. Cell Mol Life Sci 2023; 80:341. [PMID: 37898977 PMCID: PMC11073328 DOI: 10.1007/s00018-023-04998-z] [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: 05/22/2023] [Revised: 09/08/2023] [Accepted: 10/02/2023] [Indexed: 10/31/2023]
Abstract
Following myocardial ischemic injury, the most effective clinical intervention is timely restoration of blood perfusion to ischemic but viable myocardium to reduce irreversible myocardial necrosis, limit infarct size, and prevent cardiac insufficiency. However, reperfusion itself may exacerbate cell death and myocardial injury, a process commonly referred to as ischemia/reperfusion (I/R) injury, which primarily involves cardiomyocytes and cardiac microvascular endothelial cells (CMECs) and is characterized by myocardial stunning, microvascular damage (MVD), reperfusion arrhythmia, and lethal reperfusion injury. MVD caused by I/R has been a neglected problem compared to myocardial injury. Clinically, the incidence of microvascular angina and/or no-reflow due to ineffective coronary perfusion accounts for 5-50% in patients after acute revascularization. MVD limiting drug diffusion into injured myocardium, is strongly associated with the development of heart failure. CMECs account for > 60% of the cardiac cellular components, and their role in myocardial I/R injury cannot be ignored. There are many studies on microvascular obstruction, but few studies on microvascular leakage, which may be mainly due to the lack of corresponding detection methods. In this review, we summarize the clinical manifestations, related mechanisms of MVD during myocardial I/R, laboratory and clinical examination means, as well as the research progress on potential therapies for MVD in recent years. Better understanding the characteristics and risk factors of MVD in patients after hemodynamic reconstruction is of great significance for managing MVD, preventing heart failure and improving patient prognosis.
Collapse
Affiliation(s)
- Bang-Hao Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Amanguli Ruze
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Ling Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Qiu-Lin Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Jing Tang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Nilupaer Xiefukaiti
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Min-Tao Gai
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - An-Xia Deng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Xue-Feng Shan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Xiao-Ming Gao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China.
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China.
| |
Collapse
|
28
|
Tiwari R, Sharma R, Rajendran G, Borkowski GS, An SY, Schonfeld M, O’Sullivan J, Schipma MJ, Zhou Y, Courbon G, David V, Quaggin SE, Thorp E, Chandel NS, Kapitsinou PP. Post-ischemic inactivation of HIF prolyl hydroxylases in endothelium promotes maladaptive kidney repair by inducing glycolysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.03.560700. [PMID: 37873349 PMCID: PMC10592920 DOI: 10.1101/2023.10.03.560700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Ischemic acute kidney injury (AKI) is common in hospitalized patients and increases the risk for chronic kidney disease (CKD). Impaired endothelial cell (EC) functions are thought to contribute in AKI to CKD transition, but the underlying mechanisms remain unclear. Here, we identify a critical role for endothelial oxygen sensing prolyl hydroxylase domain (PHD) enzymes 1-3 in regulating post-ischemic kidney repair. In renal endothelium, we observed compartment-specific differences in the expression of the three PHD isoforms in both mice and humans. We found that post-ischemic concurrent inactivation of endothelial PHD1, PHD2, and PHD3 but not PHD2 alone promoted maladaptive kidney repair characterized by exacerbated tissue injury, fibrosis, and inflammation. Single-cell RNA-seq analysis of the post-ischemic endothelial PHD1, PHD2 and PHD3 deficient (PHDTiEC) kidney revealed an endothelial glycolytic transcriptional signature, also observed in human kidneys with severe AKI. This metabolic program was coupled to upregulation of the SLC16A3 gene encoding the lactate exporter monocarboxylate transporter 4 (MCT4). Strikingly, treatment with the MCT4 inhibitor syrosingopine restored adaptive kidney repair in PHDTiEC mice. Mechanistically, MCT4 inhibition suppressed pro-inflammatory EC activation reducing monocyte-endothelial cell interaction. Our findings suggest avenues for halting AKI to CKD transition based on selectively targeting the endothelial hypoxia-driven glycolysis/MCT4 axis.
Collapse
Affiliation(s)
- Ratnakar Tiwari
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL
- Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rajni Sharma
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL
- Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ganeshkumar Rajendran
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gabriella S. Borkowski
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL
- Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Si Young An
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL
- Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Michael Schonfeld
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - James O’Sullivan
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL
- Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Matthew J. Schipma
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Yalu Zhou
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL
- Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Guillaume Courbon
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL
- Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Valentin David
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL
- Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Susan E. Quaggin
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL
- Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Edward Thorp
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Navdeep S. Chandel
- Robert H. Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Pinelopi P. Kapitsinou
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL
- Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
29
|
Habas E, Al Adab A, Arryes M, Alfitori G, Farfar K, Habas AM, Akbar RA, Rayani A, Habas E, Elzouki A. Anemia and Hypoxia Impact on Chronic Kidney Disease Onset and Progression: Review and Updates. Cureus 2023; 15:e46737. [PMID: 38022248 PMCID: PMC10631488 DOI: 10.7759/cureus.46737] [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] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Chronic kidney disease (CKD) is caused by hypoxia in the renal tissue, leading to inflammation and increased migration of pathogenic cells. Studies showed that leukocytes directly sense hypoxia and respond by initiating gene transcription, encoding the 2-integrin adhesion molecules. Moreover, other mechanisms participate in hypoxia, including anemia. CKD-associated anemia is common, which induces and worsens hypoxia, contributing to CKD progression. Anemia correction can slow CKD progression, but it should be cautiously approached. In this comprehensive review, the underlying pathophysiology mechanisms and the impact of renal tissue hypoxia and anemia in CKD onset and progression will be reviewed and discussed in detail. Searching for the latest updates in PubMed Central, Medline, PubMed database, Google Scholar, and Google search engines were conducted for original studies, including cross-sectional studies, cohort studies, clinical trials, and review articles using different keywords, phrases, and texts such as "CKD progression, anemia in CKD, CKD, anemia effect on CKD progression, anemia effect on CKD progression, and hypoxia and CKD progression". Kidney tissue hypoxia and anemia have an impact on CKD onset and progression. Hypoxia causes nephron cell death, enhancing fibrosis by increasing interstitium protein deposition, inflammatory cell activation, and apoptosis. Severe anemia correction improves life quality and may delay CKD progression. Detection and avoidance of the risk factors of hypoxia prevent recurrent acute kidney injury (AKI) and reduce the CKD rate. A better understanding of kidney hypoxia would prevent AKI and CKD and lead to new therapeutic strategies.
Collapse
Affiliation(s)
| | - Aisha Al Adab
- Internal Medicine, Hamad General Hospital, Doha, QAT
| | - Mehdi Arryes
- Internal Medicine, Hamad General Hospital, Doha, QAT
| | | | | | - Ala M Habas
- Internal Medicine, Tripoli University, Tripoli, LBY
| | - Raza A Akbar
- Internal Medicine, Hamad General Hospital, Doha, QAT
| | - Amnna Rayani
- Hemat-oncology Department, Pediatric Tripoli Hospital, Tripoli University, Tripoli, LBY
| | - Eshrak Habas
- Internal Medicine, Tripoli University, Tripoli, LBY
| | | |
Collapse
|
30
|
Rey-Serra C, Tituaña J, Lin T, Herrero JI, Miguel V, Barbas C, Meseguer A, Ramos R, Chaix A, Panda S, Lamas S. Reciprocal regulation between the molecular clock and kidney injury. Life Sci Alliance 2023; 6:e202201886. [PMID: 37487638 PMCID: PMC10366531 DOI: 10.26508/lsa.202201886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/02/2023] [Accepted: 07/03/2023] [Indexed: 07/26/2023] Open
Abstract
Tubulointerstitial fibrosis is the common pathological substrate for many etiologies leading to chronic kidney disease. Although perturbations in the circadian rhythm have been associated with renal disease, the role of the molecular clock in the pathogenesis of fibrosis remains incompletely understood. We investigated the relationship between the molecular clock and renal damage in experimental models of injury and fibrosis (unilateral ureteral obstruction, folic acid, and adenine nephrotoxicity), using genetically modified mice with selective deficiencies of the clock components Bmal1, Clock, and Cry We found that the molecular clock pathway was enriched in damaged tubular epithelial cells with marked metabolic alterations. In human tubular epithelial cells, TGFβ significantly altered the expression of clock components. Although Clock played a role in the macrophage-mediated inflammatory response, the combined absence of Cry1 and Cry2 was critical for the recruitment of neutrophils, correlating with a worsening of fibrosis and with a major shift in the expression of metabolism-related genes. These results support that renal damage disrupts the kidney peripheral molecular clock, which in turn promotes metabolic derangement linked to inflammatory and fibrotic responses.
Collapse
Affiliation(s)
- Carlos Rey-Serra
- Program of Physiological and Pathological Processes, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Jessica Tituaña
- Program of Physiological and Pathological Processes, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Terry Lin
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - J Ignacio Herrero
- Program of Physiological and Pathological Processes, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Verónica Miguel
- Program of Physiological and Pathological Processes, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, Madrid, Spain
| | - Anna Meseguer
- Renal Physiopathology Group, Vall d'Hebron Research Institute (VHIR)-CIBBIM Nanomedicine, Barcelona, Spain
| | - Ricardo Ramos
- Genomic Facility, Fundación Parque Científico de Madrid, Madrid, Spain
| | - Amandine Chaix
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Satchidananda Panda
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Santiago Lamas
- Program of Physiological and Pathological Processes, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| |
Collapse
|
31
|
Gu H, Li J, Ni Y. Sinomenine improves renal fibrosis by regulating mesenchymal stem cell-derived exosomes and affecting autophagy levels. ENVIRONMENTAL TOXICOLOGY 2023; 38:2524-2537. [PMID: 37436133 DOI: 10.1002/tox.23890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 06/12/2023] [Accepted: 06/29/2023] [Indexed: 07/13/2023]
Abstract
BACKGROUND This study attempts to investigate the therapeutic effect of sinomenine on renal fibrosis and its mechanism. METHODS The 8-week-old C57BL/6 male mice were randomly divided into sham group, UUO model group, UUO sinomenine group (UUO + Sino 50), UUO + sinomenine group (UUO + Sino 100), UUO + exosome group (exo), and UUO + exo-inhibitor. The pathological changes of kidney were observed by H&E staining, the degree of renal interstitial fibrosis was detected by MASSON and Sirius red staining, and the expressions of fibrosis and autophagy markers were detected by real-time fluorescence quantitative PCR and WB. NTA and electron microscopy were used to analyze exo secretion after sinomenine treatment. RESULTS Sinomenine could improve the progression of renal fibrosis without causing tissue damage including heart, lungs and liver. Sinomenine could promote autophagosome formation. It could promote the secretion of exosomes from bone marrow mesenchymal stem cells (BMSCs). Sinomine regulates the PI3K-AKT pathway through BMSC-exo carrying miR-204-5p, affecting autophagy level and alleviating the process of renal fibrosis. CONCLUSION Our study suggests that sinomine could improve the progression of renal fibrosis by influencing the expression of miR-204-5p in BMSC-exo and regulating the PI3K-AKT pathway.
Collapse
Affiliation(s)
- Hongping Gu
- Department of Internal Medicine, Yuyao Traditional Chinese Medicine Hospital, Yuyao, Zhejiang, China
| | - Jinrong Li
- Department of Encephalopathy, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Yuehan Ni
- Department of Internal Medicine, Yuyao Traditional Chinese Medicine Hospital, Yuyao, Zhejiang, China
| |
Collapse
|
32
|
Jin J, Yang X, Gong H, Li X. Time- and Gender-Dependent Alterations in Mice during the Aging Process. Int J Mol Sci 2023; 24:12790. [PMID: 37628974 PMCID: PMC10454612 DOI: 10.3390/ijms241612790] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/01/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Compared to young people and adults, there are differences in the ability of elderly people to resist diseases or injuries, with some noticeable features being gender-dependent. However, gender differences in age-related viscera alterations are not clear. To evaluate a potential possibility of gender differences during the natural aging process, we used three age groups to investigate the impact on spleens, kidneys, and adrenal glands. The immunofluorescence results showed that male-specific p21 proteins were concentrated in the renal tubule epithelial cells of the kidney. Histological staining revealed an increase in the frequencies of fat vacuoles located in the renal tubule epithelial cells of the cortex, under the renal capsule in the kidneys of male mice with age. In female mice, we found that the width of the globular zone in the adrenal gland cortex was unchanged with age. On the contrary, the male displayed a reduction in width. Compared to females, the content of epinephrine in adrenal gland tissue according to ELISA analysis was higher in adults, and a greater decline was observed in aged males particularly. These data confirmed the age-dependent differences between female and male mice; therefore, gender should be considered one of the major factors for personalized treatment in clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Jing Jin
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China (H.G.)
| | - Xiaoquan Yang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China (H.G.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, HUST-Suzhou Institute for Brainsmatics, JITRI, Chinese Academy of Medical Sciences, Suzhou 215004, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China (H.G.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, HUST-Suzhou Institute for Brainsmatics, JITRI, Chinese Academy of Medical Sciences, Suzhou 215004, China
| | - Xiangning Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China (H.G.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, HUST-Suzhou Institute for Brainsmatics, JITRI, Chinese Academy of Medical Sciences, Suzhou 215004, China
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou 570228, China
| |
Collapse
|
33
|
Dencks S, Schmitz G. Ultrasound localization microscopy. Z Med Phys 2023; 33:292-308. [PMID: 37328329 PMCID: PMC10517400 DOI: 10.1016/j.zemedi.2023.02.004] [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: 08/11/2022] [Revised: 01/24/2023] [Accepted: 02/10/2023] [Indexed: 06/18/2023]
Abstract
Ultrasound Localization Microscopy (ULM) is an emerging technique that provides impressive super-resolved images of microvasculature, i.e., images with much better resolution than the conventional diffraction-limited ultrasound techniques and is already taking its first steps from preclinical to clinical applications. In comparison to the established perfusion or flow measurement methods, namely contrast-enhanced ultrasound (CEUS) and Doppler techniques, ULM allows imaging and flow measurements even down to the capillary level. As ULM can be realized as a post-processing method, conventional ultrasound systems can be used for. ULM relies on the localization of single microbubbles (MB) of commercial, clinically approved contrast agents. In general, these very small and strong scatterers with typical radii of 1-3 µm are imaged much larger in ultrasound images than they actually are due to the point spread function of the imaging system. However, by applying appropriate methods, these MBs can be localized with sub-pixel precision. Then, by tracking MBs over successive frames of image sequences, not only the morphology of vascular trees but also functional information such as flow velocities or directions can be obtained and visualized. In addition, quantitative parameters can be derived to describe pathological and physiological changes in the microvasculature. In this review, the general concept of ULM and conditions for its applicability to microvessel imaging are explained. Based on this, various aspects of the different processing steps for a concrete implementation are discussed. The trade-off between complete reconstruction of the microvasculature and the necessary measurement time as well as the implementation in 3D are reviewed in more detail, as they are the focus of current research. Through an overview of potential or already realized preclinical and clinical applications - pathologic angiogenesis or degeneration of vessels, physiological angiogenesis, or the general understanding of organ or tissue function - the great potential of ULM is demonstrated.
Collapse
Affiliation(s)
- Stefanie Dencks
- Lehrstuhl für Medizintechnik, Fakultät für Elektrotechnik und Informationstechnik, Ruhr-Universität Bochum, Bochum, Germany.
| | - Georg Schmitz
- Lehrstuhl für Medizintechnik, Fakultät für Elektrotechnik und Informationstechnik, Ruhr-Universität Bochum, Bochum, Germany
| |
Collapse
|
34
|
Trucas M, Burattini S, Porcu S, Simbula M, Ristaldi MS, Kowalik MA, Serra MP, Gobbi P, Battistelli M, Perra A, Quartu M. Multi-Organ Morphological Findings in a Humanized Murine Model of Sickle Cell Trait. Int J Mol Sci 2023; 24:10452. [PMID: 37445630 DOI: 10.3390/ijms241310452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Sickle cell disease (SCD) is caused by the homozygous beta-globin gene mutation that can lead to ischemic multi-organ damage and consequently reduce life expectancy. On the other hand, sickle cell trait (SCT), the heterozygous beta-globin gene mutation, is still considered a benign condition. Although the mechanisms are not well understood, clinical evidence has recently shown that specific pathological symptoms can also be recognized in SCT carriers. So far, there are still scant data regarding the morphological modifications referable to possible multi-organ damage in the SCT condition. Therefore, after genotypic and hematological characterization, by conventional light microscopy and transmission electron microscopy (TEM), we investigated the presence of tissue alterations in 13 heterozygous Townes mice, one of the best-known animal models that, up to now, was used only for the study of the homozygous condition. We found that endothelial alterations, as among which the thickening of vessel basal lamina, are ubiquitous in the lung, liver, kidney, and spleen of SCT carrier mice. The lung shows the most significant alterations, with a distortion of the general tissue architecture, while the heart is the least affected. Collectively, our findings contribute novel data to the histopathological modifications at microscopic and ultrastructural levels, underlying the heterozygous beta-globin gene mutation, and indicate the translational suitability of the Townes model to characterize the features of multiple organ involvement in the SCT carriers.
Collapse
Affiliation(s)
- Marcello Trucas
- Department of Biomedical Sciences, Section of Cytomorphology, University of Cagliari, Cittadella Universitaria di Monserrato, 09042 Monserrato, Italy
| | - Sabrina Burattini
- Department of Biomolecular Sciences, Campus Scientifico "Enrico Mattei", University of Urbino Carlo Bo, Via Ca' le Suore 2-Località Crocicchia, 61029 Urbino, Italy
| | - Susanna Porcu
- Italian National Research Council (CNR)-IRGB, Cittadella Universitaria Monserrato, 09042 Monserrato, Italy
| | - Michela Simbula
- Italian National Research Council (CNR)-IRGB, Cittadella Universitaria Monserrato, 09042 Monserrato, Italy
| | - Maria Serafina Ristaldi
- Italian National Research Council (CNR)-IRGB, Cittadella Universitaria Monserrato, 09042 Monserrato, Italy
| | - Marta Anna Kowalik
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cittadella Universitaria di Monserrato, 09042 Monserrato, Italy
| | - Maria Pina Serra
- Department of Biomedical Sciences, Section of Cytomorphology, University of Cagliari, Cittadella Universitaria di Monserrato, 09042 Monserrato, Italy
| | - Pietro Gobbi
- Department of Biomolecular Sciences, Campus Scientifico "Enrico Mattei", University of Urbino Carlo Bo, Via Ca' le Suore 2-Località Crocicchia, 61029 Urbino, Italy
| | - Michela Battistelli
- Department of Biomolecular Sciences, Campus Scientifico "Enrico Mattei", University of Urbino Carlo Bo, Via Ca' le Suore 2-Località Crocicchia, 61029 Urbino, Italy
| | - Andrea Perra
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cittadella Universitaria di Monserrato, 09042 Monserrato, Italy
| | - Marina Quartu
- Department of Biomedical Sciences, Section of Cytomorphology, University of Cagliari, Cittadella Universitaria di Monserrato, 09042 Monserrato, Italy
| |
Collapse
|
35
|
Aiello FB, Ranelletti FO, Liberatore M, Felaco P, De Luca G, Lamolinara A, Schena FP, Bonomini M. Independent Prognostic and Predictive Role of Interstitial Macrophages in Kidney Biopsies of IgA Nephropathy Patients. J Pers Med 2023; 13:935. [PMID: 37373924 DOI: 10.3390/jpm13060935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
A relevant percentage of IgAN patients experience a progressive decline in kidney function. According to the KDIGO guidelines, proteinuria and eGFR are the only validated prognostic markers. The role of interstitial macrophages in kidney biopsies of IgAN patients and the outcome of patients treated with renin-angiotensin system inhibitors (RASBs) alone or combined with glucocorticoids were evaluated. Clinical and laboratory records (age, gender, hypertension, hematuria, proteinuria, eGFR, serum creatinine, and therapy), MEST-C parameters of the Oxford classification, C4d deposition, peritubular capillaries, and glomerular and interstitial macrophages in 47 IgAN patients undergoing kidney biopsy consecutively between 2003 and 2016 were examined. A high number of interstitial macrophages significantly correlated with peritubular capillary rarefaction and impairment of kidney function. Cox's multivariable regression analysis revealed that a value > 19.5 macrophages/HPF behaved as an independent marker of an unfavorable outcome. Patients exhibiting > 19.5 macrophages/HPF treated at the time of diagnosis with RASBs combined with methylprednisolone had an estimated probability of a favorable outcome higher than patients treated with RASBs alone. Thus, a value > 19.5 macrophages/HPF in IgAN biopsies can predict an unfavorable outcome and endorse a well-timed administration of glucocorticoids. Studies evaluating urine biomarkers associated with peritubular capillary rarefaction in patients with marked macrophage infiltration may help personalized treatment decisions.
Collapse
Affiliation(s)
- Francesca Bianca Aiello
- Department of Medicine and Aging Sciences, University G. D'Annunzio, Chieti-Pescara, 66100 Chieti, Italy
| | | | | | - Paolo Felaco
- UOC Nephrology and Dialysis PO, 64100 Teramo, Italy
| | - Graziano De Luca
- Graziano De Luca UO Clinical Pathology, Val Vibrata Hospital, 64027 Sant'Omero, Italy
| | - Alessia Lamolinara
- Department of Neurosciences, Imaging and Clinical Sciences, University G. D'Annunzio, Chieti-Pescara, 66100 Chieti, Italy
| | - Francesco Paolo Schena
- Department of Emergency and Organ Transplantation, University of Bari, 70121 Bari, Italy
- Schena Foundation, Valenzano, 70010 Bari, Italy
| | - Mario Bonomini
- Department of Medicine and Aging Sciences, University G. D'Annunzio, Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
36
|
Kung VL, Nelson JW. Machine Learning Illuminates the Extraglomerular Microvasculature. KIDNEY360 2023; 4:578-579. [PMID: 37229727 PMCID: PMC10371299 DOI: 10.34067/kid.0000000000000111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Affiliation(s)
- Vanderlene L. Kung
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, Oregon
| | - Jonathan W. Nelson
- Division of Nephrology and Hypertension, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
37
|
Chen Y, Zee J, Janowczyk AR, Rubin J, Toro P, Lafata KJ, Mariani LH, Holzman LB, Hodgin JB, Madabhushi A, Barisoni L. Clinical Relevance of Computationally Derived Attributes of Peritubular Capillaries from Kidney Biopsies. KIDNEY360 2023; 4:648-658. [PMID: 37016482 PMCID: PMC10278770 DOI: 10.34067/kid.0000000000000116] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 02/13/2023] [Indexed: 04/06/2023]
Abstract
Key Points Computational image analysis allows for the extraction of new information from whole-slide images with potential clinical relevance. Peritubular capillary (PTC) density is decreased in areas of interstitial fibrosis and tubular atrophy when measured in interstitial fractional space. PTC shape (aspect ratio) is associated with clinical outcome in glomerular diseases. Background The association between peritubular capillary (PTC) density and disease progression has been studied in a variety of kidney diseases using immunohistochemistry. However, other PTC attributes, such as PTC shape, have not been explored yet. The recent development of computer vision techniques provides the opportunity for the quantification of PTC attributes using conventional stains and whole-slide images. Methods To explore the relationship between PTC characteristics and clinical outcome, n =280 periodic acid–Schiff-stained kidney biopsies (88 minimal change disease, 109 focal segmental glomerulosclerosis, 46 membranous nephropathy, and 37 IgA nephropathy) from the Nephrotic Syndrome Study Network digital pathology repository were computationally analyzed. A previously validated deep learning model was applied to segment cortical PTCs. Average PTC aspect ratio (PTC major to minor axis ratio), size (PTC pixels per PTC segmentation), and density (PTC pixels per unit cortical area) were computed for each biopsy. Cox proportional hazards models were used to assess associations between these PTC parameters and outcome (40% eGFR decline or kidney failure). Cortical PTC characteristics and interstitial fractional space PTC density were compared between areas of interstitial fibrosis and tubular atrophy (IFTA) and areas without IFTA. Results When normalized PTC aspect ratio was below 0.6, a 0.1, increase in normalized PTC aspect ratio was significantly associated with disease progression, with a hazard ratio (95% confidence interval) of 1.28 (1.04 to 1.59) (P = 0.019), while PTC density and size were not significantly associated with outcome. Interstitial fractional space PTC density was lower in areas of IFTA compared with non-IFTA areas. Conclusions Computational image analysis enables quantification of the status of the kidney microvasculature and the discovery of a previously unrecognized PTC biomarker (aspect ratio) of clinical outcome.
Collapse
Affiliation(s)
- Yijiang Chen
- Center for Computational Imaging and Personalized Diagnostics, Case Western Reserve University, Cleveland, Ohio
| | - Jarcy Zee
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Andrew R. Janowczyk
- Geneva University Hospitals, Pathology and Oncology Departments, Geneva, Switzerland
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, Georgia
| | - Jeremy Rubin
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Paula Toro
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio
| | - Kyle J. Lafata
- Department of Radiology, Duke University, Durham, North Carolina
- Department of Electrical and Computer Engineering, Duke University, Durham, North Carolina
- Department of Radiation Oncology, Duke University, Durham, North Carolina
| | - Laura H. Mariani
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan
| | - Lawrence B. Holzman
- Department of Medicine, Renal-Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jeffrey B. Hodgin
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Anant Madabhushi
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, Georgia
- Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - Laura Barisoni
- Department of Pathology, Division of AI and Computational Pathology, Duke University, Durham, North Carolina
- Department of Medicine, Division of Nephrology, Duke University, Durham, North Carolina
| |
Collapse
|
38
|
Lee B, Kang W, Oh SH, Cho S, Shin I, Oh EJ, Kim YJ, Ahn JS, Yook JM, Jung SJ, Lim JH, Kim YL, Cho JH, Oh WY. In vivo imaging of renal microvasculature in a murine ischemia-reperfusion injury model using optical coherence tomography angiography. Sci Rep 2023; 13:6396. [PMID: 37076541 PMCID: PMC10115874 DOI: 10.1038/s41598-023-33295-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/11/2023] [Indexed: 04/21/2023] Open
Abstract
Optical coherence tomography angiography (OCTA) provides three-dimensional structural and semiquantitative imaging of microvasculature in vivo. We developed an OCTA imaging protocol for a murine kidney ischemia-reperfusion injury (IRI) model to investigate the correlation between renal microvascular changes and ischemic damage. Mice were divided into mild and moderate IRI groups according to the duration of ischemia (10 and 35 mins, respectively). Each animal was imaged at baseline; during ischemia; and at 1, 15, 30, 45, and 60 mins after ischemia. Amplitude decorrelation OCTA images were constructed with 1.5-, 3.0-, and 5.8-ms interscan times, to calculate the semiquantitative flow index in the superficial (50-70 μm) and the deep (220-340 μm) capillaries of the renal cortex. The mild IRI group showed no significant flow index change in both the superfial and the deep layers. The moderate IRI group showed a significantly decreased flow index from 15 and 45 mins in the superficial and deep layers, respectively. Seven weeks after IRI induction, the moderate IRI group showed lower kidney function and higher collagen deposition than the mild IRI group. OCTA imaging of the murine IRI model revealed changes in superficial blood flow after ischemic injury. A more pronounced decrease in superficial blood flow than in deep blood flow was associated with sustained dysfunction after IRI. Further investigation on post-IRI renal microvascular response using OCTA may improve our understanding of the relationship between the degree of ischemic insult and kidney function.
Collapse
Affiliation(s)
- ByungKun Lee
- Department of Mechanical Engineering, KAIST, Daejeon, Republic of Korea
- KI for Health Science and Technology, KAIST, Daejeon, Republic of Korea
| | - Woojae Kang
- Department of Mechanical Engineering, KAIST, Daejeon, Republic of Korea
- KI for Health Science and Technology, KAIST, Daejeon, Republic of Korea
| | - Se-Hyun Oh
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Seungwan Cho
- Department of Mechanical Engineering, KAIST, Daejeon, Republic of Korea
- KI for Health Science and Technology, KAIST, Daejeon, Republic of Korea
| | - Inho Shin
- Department of Mechanical Engineering, KAIST, Daejeon, Republic of Korea
- KI for Health Science and Technology, KAIST, Daejeon, Republic of Korea
| | - Eun-Joo Oh
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - You-Jin Kim
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Ji-Sun Ahn
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Ju-Min Yook
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Soo-Jung Jung
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Jeong-Hoon Lim
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Yong-Lim Kim
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Jang-Hee Cho
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea.
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea.
| | - Wang-Yuhl Oh
- Department of Mechanical Engineering, KAIST, Daejeon, Republic of Korea.
- KI for Health Science and Technology, KAIST, Daejeon, Republic of Korea.
| |
Collapse
|
39
|
Maas SL, Donners MMPC, van der Vorst EPC. ADAM10 and ADAM17, Major Regulators of Chronic Kidney Disease Induced Atherosclerosis? Int J Mol Sci 2023; 24:ijms24087309. [PMID: 37108478 PMCID: PMC10139114 DOI: 10.3390/ijms24087309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Chronic kidney disease (CKD) is a major health problem, affecting millions of people worldwide, in particular hypertensive and diabetic patients. CKD patients suffer from significantly increased cardiovascular disease (CVD) morbidity and mortality, mainly due to accelerated atherosclerosis development. Indeed, CKD not only affects the kidneys, in which injury and maladaptive repair processes lead to local inflammation and fibrosis, but also causes systemic inflammation and altered mineral bone metabolism leading to vascular dysfunction, calcification, and thus, accelerated atherosclerosis. Although CKD and CVD individually have been extensively studied, relatively little research has studied the link between both diseases. This narrative review focuses on the role of a disintegrin and metalloproteases (ADAM) 10 and ADAM17 in CKD and CVD and will for the first time shed light on their role in CKD-induced CVD. By cleaving cell surface molecules, these enzymes regulate not only cellular sensitivity to their micro-environment (in case of receptor cleavage), but also release soluble ectodomains that can exert agonistic or antagonistic functions, both locally and systemically. Although the cell-specific roles of ADAM10 and ADAM17 in CVD, and to a lesser extent in CKD, have been explored, their impact on CKD-induced CVD is likely, yet remains to be elucidated.
Collapse
Affiliation(s)
- Sanne L Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| | - Marjo M P C Donners
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), 80336 Munich, Germany
| |
Collapse
|
40
|
Zhao ZB, Marschner JA, Iwakura T, Li C, Motrapu M, Kuang M, Popper B, Linkermann A, Klocke J, Enghard P, Muto Y, Humphreys BD, Harris HE, Romagnani P, Anders HJ. Tubular Epithelial Cell HMGB1 Promotes AKI-CKD Transition by Sensitizing Cycling Tubular Cells to Oxidative Stress: A Rationale for Targeting HMGB1 during AKI Recovery. J Am Soc Nephrol 2023; 34:394-411. [PMID: 36857499 PMCID: PMC10103235 DOI: 10.1681/asn.0000000000000024] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 10/22/2022] [Indexed: 01/22/2023] Open
Abstract
SIGNIFICANCE STATEMENT Cells undergoing necrosis release extracellular high mobility group box (HMGB)-1, which triggers sterile inflammation upon AKI in mice. Neither deletion of HMGB1 from tubular epithelial cells, nor HMGB1 antagonism with small molecules, affects initial ischemic tubular necrosis and immediate GFR loss upon unilateral ischemia/reperfusion injury (IRI). On the contrary, tubular cell-specific HMGB1 deficiency, and even late-onset pharmacological HMGB1 inhibition, increased functional and structural recovery from AKI, indicating that intracellular HMGB1 partially counters the effects of extracellular HMGB1. In vitro studies indicate that intracellular HMGB1 decreases resilience of tubular cells from prolonged ischemic stress, as in unilateral IRI. Intracellular HMGB1 is a potential target to enhance kidney regeneration and to improve long-term prognosis in AKI. BACKGROUND Late diagnosis is a hurdle for treatment of AKI, but targeting AKI-CKD transition may improve outcomes. High mobility group box-1 (HMGB1) is a nuclear regulator of transcription and a driver of necroinflammation in AKI. We hypothesized that HMGB1 would also modulate AKI-CKD transition in other ways. METHODS We conducted single-cell transcriptome analysis of human and mouse AKI and mouse in vivo and in vitro studies with tubular cell-specific depletion of Hmgb1 and HMGB1 antagonists. RESULTS HMGB1 was ubiquitously expressed in kidney cells. Preemptive HMGB1 antagonism with glycyrrhizic acid (Gly) and ethyl pyruvate (EP) did not affect postischemic AKI but attenuated AKI-CKD transition in a model of persistent kidney hypoxia. Consistently, tubular Hmgb1 depletion in Pax8 rtTA, TetO Cre, Hmgb1fl/fl mice did not protect from AKI, but from AKI-CKD transition. In vitro studies confirmed that absence of HMGB1 or HMGB1 inhibition with Gly and EP does not affect ischemic necrosis of growth-arrested differentiated tubular cells but increased the resilience of cycling tubular cells that survived the acute injury to oxidative stress. This effect persisted when neutralizing extracellular HMGB1 with 2G7. Consistently, late-onset HMGB1 blockade with EP started after the peak of ischemic AKI in mice prevented AKI-CKD transition, even when 2G7 blocked extracellular HMGB1. CONCLUSION Treatment of AKI could become feasible when ( 1 ) focusing on long-term outcomes of AKI; ( 2 ) targeting AKI-CKD transition with drugs initiated after the AKI peak; and ( 3 ) targeting with drugs that block HMGB1 in intracellular and extracellular compartments.
Collapse
Affiliation(s)
- Zhi Bo Zhao
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Munich, Germany
| | - Julian A. Marschner
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Munich, Germany
| | - Takamasa Iwakura
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Munich, Germany
| | - Chenyu Li
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Munich, Germany
| | - Manga Motrapu
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Munich, Germany
| | - Meisi Kuang
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Munich, Germany
| | - Bastian Popper
- Biomedical Center, Core Facility Animal Models, LMU München, Munich, Germany
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Jan Klocke
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp Enghard
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Yoshiharu Muto
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Benjamin D. Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, Missouri
| | - Helena Erlandsson Harris
- Departments of Rheumatology and of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Paola Romagnani
- Department of Experimental and Biomedical Sciences "Mario Serio" and Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, Italy
| | - Hans-Joachim Anders
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Munich, Germany
| |
Collapse
|
41
|
Association of MAFLD with end-stage kidney disease: a prospective study of 337,783 UK Biobank participants. Hepatol Int 2023; 17:595-605. [PMID: 36809487 DOI: 10.1007/s12072-023-10486-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/12/2023] [Indexed: 02/23/2023]
Abstract
INTRODUCTION Metabolic dysfunction-associated fatty liver (MAFLD) has been found to be associated with the prevalence of chronic kidney disease (CKD). However, it is unknown whether MAFLD is associated with CKD development and the incidence of end-stage kidney disease (ESKD). We aimed to clarify the association between MAFLD and incident ESKD in the prospective UK Biobank cohort. METHODS We analyzed the data of 337,783 UK Biobank participants and relative risks for the ESKD were calculated by using the Cox regression analysis. RESULTS Among 337,783 participants over a median duration of 12.8 years follow-up, a total of 618 ESKD cases were diagnosed. Participants with MAFLD were twice likely to develop ESKD (hazard ratio [HR] 2.03, 95% confidence interval [CI] 1.68-2.46, p < 0.001). The association of MAFLD with ESKD risk remained significant in both non-CKD and CKD participants. Our results also showed that there were graded associations between liver fibrosis scores and the risk of ESKD in MAFLD cases. Compared to non-MAFLD individuals, the adjusted HRs for incident ESKD in MAFLD patients with increasing levels of NAFLD fibrosis score were 1.23 (95% CI 0.96-1.58), 2.45 (1.98-3.03) and 7.67 (5.48-10.73), respectively. Furthermore, the risking alleles of PNPLA3 rs738409, TM6SF2 rs58542926, GCKR rs1260326 and MBOAT7 rs641738 amplified the MAFLD effect on ESKD risk. In conclusion, MAFLD is associated with incident ESKD. CONCLUSION MAFLD may help identify the subjects at high risk of ESKD development and MAFLD interventions should be encouraged to slow down CKD progression.
Collapse
|
42
|
Kwiatkowska E, Kwiatkowski S, Dziedziejko V, Tomasiewicz I, Domański L. Renal Microcirculation Injury as the Main Cause of Ischemic Acute Kidney Injury Development. BIOLOGY 2023; 12:biology12020327. [PMID: 36829602 PMCID: PMC9953191 DOI: 10.3390/biology12020327] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/04/2023] [Accepted: 02/09/2023] [Indexed: 02/22/2023]
Abstract
Acute kidney injury (AKI) can result from multiple factors. The main cause is reduced renal perfusion. Kidneys are susceptible to ischemia due to the anatomy of microcirculation that wraps around the renal tubules-peritubular capillary (PTC) network. Cortical and medullary superficial tubules have a large share in transport and require the supply of oxygen for ATP production, while it is the cortex that receives almost 100% of the blood flowing through the kidneys and the medulla only accounts for 5-10% of it. This difference makes the tubules present in the superficial layer of the medulla very susceptible to ischemia. Impaired blood flow causes damage to the endothelium, with an increase in its prothrombotic and pro-adhesive properties. This causes congestion in the microcirculation of the renal medulla. The next stage is the migration of pericytes with the disintegration of these vessels. The phenomenon of destruction of small vessels is called peritubular rarefaction, attributed as the main cause of further irreversible changes in the damaged kidney leading to the development of chronic kidney disease. In this article, we will present the characteristic structure of renal microcirculation, its regulation, and the mechanism of damage in acute ischemia, and we will try to find methods of prevention with particular emphasis on the inhibition of the renin-angiotensin-aldosterone system.
Collapse
Affiliation(s)
- Ewa Kwiatkowska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University in Szczecin, Powstańców Wlkp, 72, 70-204 Szczecin, Poland
- Correspondence:
| | - Sebastian Kwiatkowski
- Department of Obstetrician and Gynecology, Pomeranian Medical University in Szczecin, Powstańców Wlkp, 72, 70-204 Szczecin, Poland
| | - Violetta Dziedziejko
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp, 72, 70-204 Szczecin, Poland
| | - Izabela Tomasiewicz
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University in Szczecin, Powstańców Wlkp, 72, 70-204 Szczecin, Poland
| | - Leszek Domański
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University in Szczecin, Powstańców Wlkp, 72, 70-204 Szczecin, Poland
| |
Collapse
|
43
|
Ermert K, Buhl EM, Klinkhammer BM, Floege J, Boor P. Reduction of Endothelial Glycocalyx on Peritubular Capillaries in Chronic Kidney Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:138-147. [PMID: 36414084 DOI: 10.1016/j.ajpath.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/07/2022] [Accepted: 11/03/2022] [Indexed: 11/20/2022]
Abstract
In chronic kidney disease (CKD), peritubular capillaries undergo anatomic and functional alterations, such as rarefaction and increased permeability. The endothelial glycocalyx (EG) is a carbohydrate-rich gel-like mesh, which covers the luminal surface of endothelial cells. It is involved in many regulatory functions of the endothelium, including vascular permeability. Herein, we investigated ultrastructural alterations of the EG in different murine CKD models. Fluorescence staining using different lectins with high affinity to components of the renal glycocalyx revealed a reduced binding to the endothelium in CKD in the animal models, and there were similar finding in human kidney specimens. Lanthanum Dysprosium Glycosamino Glycan adhesion staining technique was used to visualize the ultrastructure of the glycocalyx in transmission electron microscopy. This also enabled quantitative analyses, showing a significant reduction of the EG thickness and density. In addition, mRNA expression of proteins involved in glycocalyx biology, synthesis, and turnover (ie, syndecan 1 and glypican 1), which are main components of the glycocalyx, and exostosin 2, involved in the synthesis of the glycocalyx, were significantly up-regulated in endothelial cells isolated from murine CKD models. Visualization of glycocalyx using specific transmission electron microscopy analyses allows qualitative and quantitative analyses and revealed significant pathologic alterations in peritubular capillaries in CKD.
Collapse
Affiliation(s)
- Katja Ermert
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany; Division of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Eva M Buhl
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany; Division of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany; Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany
| | - Barbara M Klinkhammer
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany; Division of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Jürgen Floege
- Division of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany; Division of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany; Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
44
|
Ke B, Shen W, Song J, Fang X. MG53: A potential therapeutic target for kidney disease. Pharmacol Res Perspect 2023; 11:e01049. [PMID: 36583464 PMCID: PMC9801490 DOI: 10.1002/prp2.1049] [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: 12/01/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022] Open
Abstract
Ensuring cell survival and tissue regeneration by maintaining cellular integrity is important to the pathophysiology of many human diseases, including kidney disease. Mitsugumin 53 (MG53) is a member of the tripartite motif-containing (TRIM) protein family that plays an essential role in repairing cell membrane injury and improving tissue regeneration. In recent years, an increasing number of studies have demonstrated that MG53 plays a renoprotective role in kidney diseases. Moreover, with the beneficial effects of the recombinant human MG53 (rhMG53) protein in the treatment of kidney diseases in different animal models, rhMG53 shows significant therapeutic potential in kidney disease. In this review, we elucidate the role of MG53 and its molecular mechanism in kidney disease to provide new approaches to the treatment of kidney disease.
Collapse
Affiliation(s)
- Ben Ke
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wen Shen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital to Nanchang University, Nanchang, China
| | - Jianling Song
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiangdong Fang
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
45
|
Hurtado KA, Janda J, Schnellmann RG. Lasmiditan promotes recovery from acute kidney injury through induction of mitochondrial biogenesis. Am J Physiol Renal Physiol 2023; 324:F56-F63. [PMID: 36326468 PMCID: PMC9762961 DOI: 10.1152/ajprenal.00249.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Acute kidney injury (AKI) involves rapid loss of renal function and occurs in 8-16% of hospitalized patients. AKI can be induced by drugs, sepsis, and ischemia-reperfusion (I/R). Hallmarks of AKI include mitochondrial and microvasculature dysfunction as well as renal tubular injury. There is currently no available therapeutic for AKI. Previously, our group identified that serotonin (5-HT)1F receptor agonism with lasmiditan accelerated endothelial cell recovery and induced mitochondrial biogenesis (MB) in vitro. We hypothesized that lasmiditan, a Federal Drug Administration-approved drug, would induce MB and improve microvascular and renal function in a mouse model of AKI. Male mice were subjected to renal I/R and treated with lasmiditan (0.3 mg/kg) or vehicle beginning 24 h after injury and then daily until euthanasia at 6 or 12 days. Serum creatinine was measured to estimate glomerular filtration rate. The renal cortex was assessed for mitochondrial density, vascular permeability and integrity, tubular damage, and interstitial fibrosis. Lasmiditan increased mitochondrial number (1.4-fold) in renal cortices. At 6 days, serum creatinine decreased 41% in the I/R group and 72% with lasmiditan. At 6 or 12 days, kidney injury molecule-1 increased in the I/R group and decreased 50% with lasmiditan. At 12 days, interstitial fibrosis decreased with lasmiditan by 50% and collagen type 1 by 38%. Evan's blue dye leakage increased 2.5-fold in the I/R group and was restored with lasmiditan. The tight junction proteins zonula occludens-1, claudin-2, and claudin-5 decreased in the I/R group and recovered with lasmiditan. At 6 or 12 days, peroxisome proliferator-activated receptor-γ coactivator-1α and electron transport chain complexes increased only with lasmiditan. In conclusion, lasmiditan treatment beginning AKI induces MB, attenuated vascular and tubular injury, decreased interstitial fibrosis, and lowered serum creatinine. Given that lasmiditan is a Federal Drug Administration-approved drug, these preclinical data support repurposing lasmiditan as a therapeutic for AKI.NEW & NOTEWORTHY AKI pathology involves a rapid decline in kidney function and occurs in 8-16% of hospitalized patients. There is currently no therapeutic for AKI. AKI results in mitochondria dysfunction, microvasculature injury, and loss of renal tubular function. In an I/R-induced AKI mouse model, treatment with the FDA-approved 5-HT1F receptor-selective agonist lasmiditan induced mitochondrial biogenesis, improved vascular integrity, reduced fibrosis, and reduced proximal tubule damage. These data support repurposing lasmiditan for the treatment of AKI.
Collapse
Affiliation(s)
- Kevin A Hurtado
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Jaroslav Janda
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
- Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona
- Southwest Environmental Health Science Center, University of Arizona, Tucson, Arizona
| |
Collapse
|
46
|
Liu S, Lv K, Wang Y, Lou P, Zhou P, Wang C, Li L, Liao G, Zhang Y, Chen Y, Cheng J, Lu Y, Liu J. Improving the circulation time and renal therapeutic potency of extracellular vesicles using an endogenous ligand binding strategy. J Control Release 2022; 352:1009-1023. [PMID: 36375619 DOI: 10.1016/j.jconrel.2022.11.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/19/2022] [Accepted: 11/07/2022] [Indexed: 11/17/2022]
Abstract
Kidney diseases are a serious health issue worldwide, and novel therapeutics are urgently needed. Extracellular vesicles (EVs) have emerged as potent drug delivery systems (DDSs), but their therapeutic potential is limited by short circulation times and insufficient renal retention. Here, we report that endogenous ligand (albumin, ALB) binding is an efficient modification strategy to improve the therapeutic potency of EV-based DDSs for kidney diseases. Surface albumin-binding peptide (ABP)-displayed EVs (ABP-EVs) were produced by transfecting parent cells with the ABP-Lamp2b fusion plasmid. Compared with unmodified EVs (NC-EVs), ABP-EVs showed increased binding to ALB in vitro and elevated circulation time and multiple organ retention in vivo after systemic (iv) injection. Moreover, ABP-EVs had higher renal retention than NC-EVs in mice with acute kidney injury through a complex mechanism involving microvascular injury and megalin-mediated endocytosis. As a result, delivery of small molecule drugs (e.g., curcumin) or proteins (e.g., hepatocyte growth factor) by ABP-EVs had superior therapeutic (e.g., anti-apoptotic, antioxidant, anti-inflammatory) effects in vitro and in vivo. This study highlights that ABP-EVs are versatile DDSs for kidney diseases and provides insights into the new strategies of engineering EVs for drug delivery.
Collapse
Affiliation(s)
- Shuyun Liu
- NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, China
| | - Ke Lv
- NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, China
| | - Yizhuo Wang
- NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, China
| | - Peng Lou
- NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, China
| | - Pingya Zhou
- NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, China
| | - Chengshi Wang
- NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, China; Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, China
| | - Lan Li
- NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, China
| | - Guangneng Liao
- Animal Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yong Zhang
- Institutes for Systems Genetics, West China Hospital of Sichuan University, Chengdu, China
| | - Younan Chen
- NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, China
| | - Jingqiu Cheng
- NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, China
| | - Yanrong Lu
- NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, China
| | - Jingping Liu
- NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
47
|
Dominguez JH, Xie D, Dominguez JM, Kelly KJ. Role of coagulation in persistent renal ischemia following reperfusion in an animal model. Am J Physiol Renal Physiol 2022; 323:F590-F601. [PMID: 36007891 PMCID: PMC9602917 DOI: 10.1152/ajprenal.00162.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 12/14/2022] Open
Abstract
Ischemic acute kidney injury is common, deadly, and accelerates the progression of chronic kidney disease, yet has no specific therapy. After ischemia, reperfusion is patchy with early and persistent impairment in regional renal blood flow and cellular injury. We tested the hypothesis that intrarenal coagulation results in sustained renal ischemia following reperfusion, using a well-characterized model. Markedly decreased, but heterogeneous, microvascular plasma flow with microthrombi was found postischemia by intravital microscopy. Widespread tissue factor expression and fibrin deposition were also apparent. Clotting was accompanied by complement activation and inflammation. Treatment with exosomes derived from renal tubular cells or with the fibrinolytic urokinase, given 24 h postischemia when renal failure was established, significantly improved microvascular flow, coagulation, serum creatinine, and histological evidence of injury. These data support the hypothesis that intrarenal clotting occurs early and the resultant sustained ischemia is a critical determinant of renal failure following ischemia; they demonstrate that the coagulation abnormalities are amenable to therapy and that therapy results in improvement in both function and postischemic inflammation.NEW & NOTEWORTHY Ischemic renal injury carries very high morbidity and mortality, yet has no specific therapy. We found markedly decreased, heterogeneous microvascular plasma flow, tissue factor induction, fibrin deposition, and microthrombi after renal ischemia-reperfusion using a well-characterized model. Renal exosomes or the fibrinolytic urokinase, administered after renal failure was established, improved microvascular flow, coagulation, renal function, and histology. Data demonstrate that intrarenal clotting results in sustained ischemia amenable to therapy that improves both function and postischemic inflammation.
Collapse
Affiliation(s)
- Jesus H. Dominguez
- Nephrology Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Roudebush Veterans Administration Hospital, Indianapolis, Indiana
| | - Danhui Xie
- Nephrology Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - James M. Dominguez
- Nephrology Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - K. J. Kelly
- Nephrology Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Roudebush Veterans Administration Hospital, Indianapolis, Indiana
| |
Collapse
|
48
|
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: 12] [Impact Index Per Article: 4.0] [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
|
49
|
VEGFA promotes the occurrence of PLA2R-associated idiopathic membranous nephropathy by angiogenesis via the PI3K/AKT signalling pathway. BMC Nephrol 2022; 23:313. [PMID: 36114523 PMCID: PMC9482157 DOI: 10.1186/s12882-022-02936-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/05/2022] [Indexed: 12/03/2022] Open
Abstract
Background The M-type phospholipase A2 receptor (PLA2R)-associated idiopathic membranous nephropathy (IMN) is a common immune-related disease in adults. Vascular endothelial growth factor A (VEGFA) is the key mediator of angiogenesis, which leads to numerous kidney diseases. However, the role of VEGFA in IMN is poorly understood. Methods In the present study, we downloaded the microarray data GSE115857 from Gene Expression Omnibus (GEO). The differentially expressed genes (DEGs) were identified with R software. The cytoHubba plug-in were used to identify hub genes from the protein–protein interaction network. Gene set enrichment analysis (GSEA) was used to identify signalling pathway in IMN. CCK8 was performed to assess the cell viability in human vascular endothelial cells (HVECs). Then, passive Heymann nephritis (PHN) was induced in rats by a single tail vein injection of anti-Fx1A antiserum. Animals treated with VEGFA inhibitor bevacizumab (BV), with saline as a positive control. Proteinuria was evaluated by biochemical measurements. Immunohistochemistry and immunofluorescence was used to evaluate relative proteins expression. Electron microscopy was performed to observe the thickness of the glomerular basement membrane (GBM). Results We revealed 3 hub genes, including one up-regulated gene VEGFA and two down-regulated genes JUN and FOS, which are closely related to the development of PLA2R-associated IMN. Pathway enrichment analysis found that the biological process induced by VEGFA is associated with PI3K/Akt signalling. GSEA showed that the signalling pathway of DEGs in GSE115857 was focused on angiogenesis, in which VEGFA acts as a core gene. We confirmed the high expression of VEGFA, PI3K, and AKT in IMN renal biopsy samples with immunohistochemistry. In HVECs, we found that BV suppresses cell viability in a time and dose dependent manner. In vivo, we found low dose of BV attenuates proteinuria via inhibiting VEGFA/PI3K/AKT signalling. Meanwhile, low dose of BV alleviates the thickening of the GBM. Conclusion VEGFA/PI3K/AKT signalling may play significant roles in the pathogenesis of IMN, which may provide new targets for the treatment of IMN. Supplementary Information The online version contains supplementary material available at 10.1186/s12882-022-02936-y.
Collapse
|
50
|
Giuliani KTK, Grivei A, Nag P, Wang X, Rist M, Kildey K, Law B, Ng MS, Wilkinson R, Ungerer J, Forbes JM, Healy H, Kassianos AJ. Hypoxic human proximal tubular epithelial cells undergo ferroptosis and elicit an NLRP3 inflammasome response in CD1c + dendritic cells. Cell Death Dis 2022; 13:739. [PMID: 36030251 PMCID: PMC9420140 DOI: 10.1038/s41419-022-05191-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 01/21/2023]
Abstract
Inflammasomes are multiprotein platforms responsible for the release of pro-inflammatory cytokines interleukin (IL)-1β and IL-18. Mouse studies have identified inflammasome activation within dendritic cells (DC) as pivotal for driving tubulointerstitial fibrosis and inflammation, the hallmarks of chronic kidney disease (CKD). However, translation of this work to human CKD remains limited. Here, we examined the complex tubular cell death pathways mediating inflammasome activation in human kidney DC and, thus, CKD progression. Ex vivo patient-derived proximal tubular epithelial cells (PTEC) cultured under hypoxic (1% O2) conditions modelling the CKD microenvironment showed characteristics of ferroptotic cell death, including mitochondrial dysfunction, reductions in the lipid repair enzyme glutathione peroxidase 4 (GPX4) and increases in lipid peroxidation by-product 4-hydroxynonenal (4-HNE) compared with normoxic PTEC. The addition of ferroptosis inhibitor, ferrostatin-1, significantly reduced hypoxic PTEC death. Human CD1c+ DC activated in the presence of hypoxic PTEC displayed significantly increased production of inflammasome-dependent cytokines IL-1β and IL-18. Treatment of co-cultures with VX-765 (caspase-1/4 inhibitor) and MCC950 (NLRP3 inflammasome inhibitor) significantly attenuated IL-1β/IL-18 levels, supporting an NLRP3 inflammasome-dependent DC response. In line with these in vitro findings, in situ immunolabelling of human fibrotic kidney tissue revealed a significant accumulation of tubulointerstitial CD1c+ DC containing active inflammasome (ASC) specks adjacent to ferroptotic PTEC. These data establish ferroptosis as the primary pattern of PTEC necrosis under the hypoxic conditions of CKD. Moreover, this study identifies NLRP3 inflammasome signalling driven by complex tubulointerstitial PTEC-DC interactions as a key checkpoint for therapeutic targeting in human CKD.
Collapse
Affiliation(s)
- Kurt T. K. Giuliani
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Faculty of Medicine, University of Queensland, Brisbane, QLD Australia
| | - Anca Grivei
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia
| | - Purba Nag
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia
| | - Xiangju Wang
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia
| | - Melissa Rist
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia
| | - Katrina Kildey
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia
| | - Becker Law
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia ,grid.1024.70000000089150953Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD Australia
| | - Monica S. Ng
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Faculty of Medicine, University of Queensland, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Institute of Molecular Biosciences, University of Queensland, Brisbane, QLD Australia ,grid.412744.00000 0004 0380 2017Department of Nephrology, Princess Alexandra Hospital, Brisbane, QLD Australia
| | - Ray Wilkinson
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Faculty of Medicine, University of Queensland, Brisbane, QLD Australia ,grid.1024.70000000089150953Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD Australia
| | - Jacobus Ungerer
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Faculty of Medicine, University of Queensland, Brisbane, QLD Australia
| | - Josephine M. Forbes
- grid.1003.20000 0000 9320 7537Faculty of Medicine, University of Queensland, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Mater Research Institute, University of Queensland, Brisbane, QLD Australia
| | - Helen Healy
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Faculty of Medicine, University of Queensland, Brisbane, QLD Australia
| | - Andrew J. Kassianos
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Faculty of Medicine, University of Queensland, Brisbane, QLD Australia ,grid.1024.70000000089150953Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD Australia
| |
Collapse
|