1
|
Rabelink TJ, Wang G, van der Vlag J, van den Berg BM. The roles of hyaluronan in kidney development, physiology and disease. Nat Rev Nephrol 2024; 20:822-832. [PMID: 39191935 DOI: 10.1038/s41581-024-00883-5] [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: 07/30/2024] [Indexed: 08/29/2024]
Abstract
The hyaluronan (HA) matrix in the tissue microenvironment is crucial for maintaining homeostasis by regulating inflammatory signalling, endothelial-mesenchymal transition and cell migration. During development, covalent modifications and osmotic swelling of HA create mechanical forces that initiate midgut rotation, vascular patterning and branching morphogenesis. Together with its main cell surface receptor, CD44, HA establishes a physicochemical scaffold at the cell surface that facilitates the interaction and clustering of growth factors and receptors that is required for normal physiology. High-molecular-weight HA, tumour necrosis factor-stimulated gene 6, pentraxin 3 and CD44 form a stable pericellular matrix that promotes tissue regeneration and reduces inflammation. By contrast, breakdown of high-molecular-weight HA into depolymerized fragments by hyaluronidases triggers inflammatory signalling, leukocyte migration and angiogenesis, contributing to tissue damage and fibrosis in kidney disease. Targeting HA metabolism is challenging owing to its dynamic regulation and tissue-specific functions. Nonetheless, modulating HA matrix functions by targeting its binding partners holds promise as a therapeutic strategy for restoring tissue homeostasis and mitigating pathological processes. Further research in this area is warranted to enable the development of novel therapeutic approaches for kidney and other diseases characterized by dysregulated HA metabolism.
Collapse
Affiliation(s)
- Ton J Rabelink
- Department of Internal Medicine (Nephrology) & Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands.
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands.
| | - Gangqi Wang
- Department of Internal Medicine (Nephrology) & Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Johan van der Vlag
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bernard M van den Berg
- Department of Internal Medicine (Nephrology) & Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
Qi B, Lou Y, Zhu Y, Chen Y, Yang S, Meng F, Pan Z, Liu S, Yan G, Lu X, Huang LH. Elevated RHAMM as a biomarker for predicting diabetic kidney disease in patients with type 2 diabetes. Clin Kidney J 2024; 17:sfae196. [PMID: 39050866 PMCID: PMC11267228 DOI: 10.1093/ckj/sfae196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Indexed: 07/27/2024] Open
Abstract
Background Diabetic kidney disease (DKD) poses a significant challenge globally as a complication of diabetes. Hyaluronan (HA), a critical non-sulfated glycosaminoglycan in the extracellular matrix, plays a pivotal role in the progression of DKD. This study assesses the predictive significance of HA's corresponding receptor, RHAMM (receptor for HA-mediated motility), in DKD pathogenesis in type 2 diabetes (T2DM) patients. Methods Enzyme-linked immunosorbent assays were utilized to measure plasma and urine levels of HA, CD44 and RHAMM in 99 diabetic patients. Immunohistochemistry staining was employed to examine HA deposition, CD44 and RHAMM expressions from 18 biopsy-proven DKD patients. Spearman correlation analysis, linear regression and receiver operating characteristic (ROC) analysis were conducted to establish associations between plasma HA, CD44 and RHAMM levels, and clinical parameters in DKD patients with T2DM. Results Elevated plasma and urine HA, CD44 and RHAMM levels were notably observed in the severe renal dysfunction group. Plasma RHAMM exhibited positive correlations with HA (r = 0.616, P < .001) and CD44 (r = 0.220, P < .001), and a negative correlation with estimated glomerular filtration rate (eGFR) (r = -0.618, P < .001). After adjusting for other potential predictors, plasma RHAMM emerged as an independent predictor of declining eGFR (β = -0.160, P < .05). Increased HA, CD44 and RHAMM levels in kidney biopsies of DKD patients were closely associated with heightened kidney injury. The ROC curve analysis highlighted an area under the curve (AUC) of 0.876 for plasma RHAMM, indicating superior diagnostic efficacy compared to CD44 in predicting DKD pathogenesis. The combined AUC of 0.968 for plasma RHAMM, HA and CD44 also suggested even greater diagnostic potential for DKD pathogenesis. Conclusion These findings provide initial evidence that elevated RHAMM levels predict DKD pathogenesis in T2DM patients. The formation of a triple complex involving HA, CD44 and RHAMM on the cell surface shows promise as a targetable biomarker for early intervention to mitigate severe renal dysfunctions.
Collapse
Affiliation(s)
- Bingxue Qi
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
- Department of Endocrinology, Jilin Province People's Hospital, Changchun, China
| | - Yan Lou
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Yongyue Zhu
- Clinical Medicine College, Changchun University of Chinese Medicine, Changchun, China
| | - Yang Chen
- Clinical Medicine College, Changchun University of Chinese Medicine, Changchun, China
| | - Shixin Yang
- Department of Endocrinology, Jilin Province People's Hospital, Changchun, China
| | - Fanjie Meng
- Department of Endocrinology, Jilin Province People's Hospital, Changchun, China
| | - Zhuo Pan
- Precision Molecular Medicine Center, Jilin Province People's Hospital, Changchun, China
| | - Shuangshuang Liu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guanchi Yan
- Department of Endocrinology, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xiaodan Lu
- Precision Molecular Medicine Center, Jilin Province People's Hospital, Changchun, China
| | - Li-Hao Huang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Shang S, Li X, Wang H, Zhou Y, Pang K, Li P, Liu X, Zhang M, Li W, Li Q, Chen X. Targeted therapy of kidney disease with nanoparticle drug delivery materials. Bioact Mater 2024; 37:206-221. [PMID: 38560369 PMCID: PMC10979125 DOI: 10.1016/j.bioactmat.2024.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/09/2024] [Accepted: 03/10/2024] [Indexed: 04/04/2024] Open
Abstract
With the development of nanomedicine, nanomaterials have been widely used, offering specific drug delivery to target sites, minimal side effects, and significant therapeutic effects. The kidneys have filtration and reabsorption functions, with various potential target cell types and a complex structural environment, making the strategies for kidney function protection and recovery after injury complex. This also lays the foundation for the application of nanomedicine in kidney diseases. Currently, evidence in preclinical and clinical settings supports the feasibility of targeted therapy for kidney diseases using drug delivery based on nanomaterials. The prerequisite for nanomedicine in treating kidney diseases is the use of carriers with good biocompatibility, including nanoparticles, hydrogels, liposomes, micelles, dendrimer polymers, adenoviruses, lysozymes, and elastin-like polypeptides. These carriers have precise renal uptake, longer half-life, and targeted organ distribution, protecting and improving the efficacy of the drugs they carry. Additionally, attention should also be paid to the toxicity and solubility of the carriers. While the carriers mentioned above have been used in preclinical studies for targeted therapy of kidney diseases both in vivo and in vitro, extensive clinical trials are still needed to ensure the short-term and long-term effects of nano drugs in the human body. This review will discuss the advantages and limitations of nanoscale drug carrier materials in treating kidney diseases, provide a more comprehensive catalog of nanocarrier materials, and offer prospects for their drug-loading efficacy and clinical applications.
Collapse
Affiliation(s)
- Shunlai Shang
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Xiangmeng Li
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
- Key Laboratory of Bone Metabolism and Physiology in Chronic Kidney Disease of Hebei Province, China
- Peking Union Medical College, Beijing, China
| | - Haoran Wang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Yena Zhou
- School of Medicine, Nankai University, Tianjin, China
| | - Keying Pang
- College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, China
| | - Ping Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Xiaomin Liu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Min Zhang
- Department of Nephrology, Affiliated Beijing Chaoyang Hospital of Capital Medical University, Beijing, China
| | - Wenge Li
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Qinggang Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| |
Collapse
|
4
|
Prionas A, Hamaoui K, Vanezis K, Reebye V, Habib N, Papalois V. The Effect of Interleukin-10 Immunotherapy on Renal Ischemia-Reperfusion Injury: A Systematic Review and Meta-Analysis of Preclinical Studies. Int J Mol Sci 2024; 25:6231. [PMID: 38892418 PMCID: PMC11172821 DOI: 10.3390/ijms25116231] [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: 04/17/2024] [Revised: 05/31/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Renal ischemia-reperfusion is a common cause of acute kidney injury leading to significant morbidity and mortality. There are no effective treatments available in clinical practice. This meta-analysis aims to assess the effect of IL-10 immunotherapy on renal ischemia-reperfusion injury. Medline, Embase, Cochrane-library, Google Scholar and clinicaltrials.gov were searched up to 31 March 2023. Preclinical and clinical interventional studies investigating IL-10 immunotherapy for renal ischemia-reperfusion were eligible for inclusion. The primary endpoint was renal function (serum creatinine) following ischemia-reperfusion. The secondary endpoints included mitochondrial integrity, cellular proliferation, regulated cell death (TUNEL assay), expression of inflammatory cytokines (TNF-α, IL-6 and IL-1β), M1/M2 macrophage polarization, tissue integrity (tubular injury score), long-term kidney fibrosis (fibrotic area %) and adverse events (pulmonary toxicity, cardiotoxicity hepatotoxicity). The search returned 861 records. From these, 16 full texts were screened and subsequently, seven animal studies, corresponding to a population of 268 mice/rats, were included. Compared to the control treatment, IL-10 immunotherapy reduced serum creatinine more effectively within 24 h of administration (95% CI: -9.177, -5.601, I2 = 22.42%). IL-10 immunotherapy promoted mitochondrial integrity and cellular proliferation and reduced regulated cell death (95% CI: -11.000, -4.184, I2 = 74.94%). It decreased the expression of TNF-α, IL-6 and IL-1β, led to M2 polarization of the local macrophages, reduced tubular injury score (95% CI: -8.917, -5.755, I2 = 22.71%), and long-term kidney fibrosis (95% CI: -6.963, -3.438, I2 = 0%). No adverse outcomes were captured. In Conclusion, IL-10 immunotherapy safely improves outcomes in animal models of renal ischemia-reperfusion; the translational potential of IL-10 immunotherapy needs to be further investigated in clinical trials.
Collapse
Affiliation(s)
- Apostolos Prionas
- Department of Surgery and Cancer, Imperial College London, London W12 0HS, UK
| | - Karim Hamaoui
- Department of Surgery and Cancer, Imperial College London, London W12 0HS, UK
| | | | - Vikash Reebye
- Department of Surgery and Cancer, Imperial College London, London W12 0HS, UK
| | - Nagy Habib
- Department of Surgery and Cancer, Imperial College London, London W12 0HS, UK
| | - Vassilios Papalois
- Department of Surgery and Cancer, Imperial College London, London W12 0HS, UK
| |
Collapse
|
5
|
Zhang Y, Wang X, Ji Y, Hong H, Geng X, Zhang K, Fu Z, Cai G, Chen X, Li P, Hong Q. All-trans retinoic acid pretreatment of mesenchymal stem cells enhances the therapeutic effect on acute kidney injury. Cell Commun Signal 2024; 22:291. [PMID: 38802835 PMCID: PMC11129434 DOI: 10.1186/s12964-024-01671-1] [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: 02/21/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
A promising new therapy option for acute kidney injury (AKI) is mesenchymal stem cells (MSCs). However, there are several limitations to the use of MSCs, such as low rates of survival, limited homing capacity, and unclear differentiation. In search of better therapeutic strategies, we explored all-trans retinoic acid (ATRA) pretreatment of MSCs to observe whether it could improve the therapeutic efficacy of AKI. We established a renal ischemia/reperfusion injury model and treated mice with ATRA-pretreated MSCs via tail vein injection. We found that AKI mice treated with ATRA-MSCs significantly improved renal function compared with DMSO-MSCs treatment. RNA sequencing screened that hyaluronic acid (HA) production from MSCs promoted by ATRA. Further validation by chromatin immunoprecipitation experiments verified that retinoic acid receptor RARα/RXRγ was a potential transcription factor for hyaluronic acid synthase 2. Additionally, an in vitro hypoxia/reoxygenation model was established using human proximal tubular epithelial cells (HK-2). After co-culturing HK-2 cells with ATRA-pretreated MSCs, we observed that HA binds to cluster determinant 44 (CD44) and activates the PI3K/AKT pathway, which enhances the anti-inflammatory, anti-apoptotic, and proliferative repair effects of MSCs in AKI. Inhibition of the HA/CD44 axis effectively reverses the renal repair effect of ATRA-pretreated MSCs. Taken together, our study suggests that ATRA pretreatment promotes HA production by MSCs and activates the PI3K/AKT pathway in renal tubular epithelial cells, thereby enhancing the efficacy of MSCs against AKI.
Collapse
Affiliation(s)
- Yifan Zhang
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, National Clinical Research Center for Kidney Diseases, No.28 Fuxing Road, Beijing, 100853, China
| | - Xiaochen Wang
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, National Clinical Research Center for Kidney Diseases, No.28 Fuxing Road, Beijing, 100853, China
| | - Yuwei Ji
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, National Clinical Research Center for Kidney Diseases, No.28 Fuxing Road, Beijing, 100853, China
| | - Haijuan Hong
- Songjiang District Central Hospital, Shanghai, China
| | - Xiaodong Geng
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, National Clinical Research Center for Kidney Diseases, No.28 Fuxing Road, Beijing, 100853, China
- Health Care Office of the Service Bureau of Agency for Offices Administration of the Central Military Commission, Beijing, China
| | - Keying Zhang
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, National Clinical Research Center for Kidney Diseases, No.28 Fuxing Road, Beijing, 100853, China
| | - Zhangning Fu
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, National Clinical Research Center for Kidney Diseases, No.28 Fuxing Road, Beijing, 100853, China
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, National Clinical Research Center for Kidney Diseases, No.28 Fuxing Road, Beijing, 100853, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, National Clinical Research Center for Kidney Diseases, No.28 Fuxing Road, Beijing, 100853, China
| | - Ping Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, National Clinical Research Center for Kidney Diseases, No.28 Fuxing Road, Beijing, 100853, China.
| | - Quan Hong
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, National Clinical Research Center for Kidney Diseases, No.28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
6
|
Kocurkova A, Kerberova M, Nesporova K, Lehka K, Sandanusova M, Simek M, Velebny V, Kubala L, Ambrozova G. Endogenously produced hyaluronan contributes to the regulation of peritoneal adhesion development. Biofactors 2023; 49:940-955. [PMID: 37154260 DOI: 10.1002/biof.1957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/05/2023] [Indexed: 05/10/2023]
Abstract
Peritoneal adhesions are postsurgical fibrotic complications connected to peritoneal inflammation. The exact mechanism of development is unknown; however, an important role is attributed to activated mesothelial cells (MCs) overproducing macromolecules of extracellular matrix (ECM), including hyaluronic acid (HA). It was suggested that endogenously-produced HA contributes to the regulation of different fibrosis-related pathologies. However, little is known about the role of altered HA production in peritoneal fibrosis. We focused on the consequences of the increased turnover of HA in the murine model of peritoneal adhesions. Changes of HA metabolism were observed in early phases of peritoneal adhesion development in vivo. To study the mechanism, human MCs MeT-5A and murine MCs isolated from the peritoneum of healthy mice were pro-fibrotically activated by transforming growth factor β (TGFβ), and the production of HA was attenuated by two modulators of carbohydrate metabolism, 4-methylumbelliferone (4-MU) and 2-deoxyglucose (2-DG). The attenuation of HA production was mediated by upregulation of HAS2 and downregulation of HYAL2 and connected to the lower expression of pro-fibrotic markers, including fibronectin and α-smooth muscle actin (αSMA). Moreover, the inclination of MCs to form fibrotic clusters was also downregulated, particularly in 2-DG-treated cells. The effects of 2-DG, but not 4-MU, were connected to changes in cellular metabolism. Importantly, the inhibition of AKT phosphorylation was observed after the use of both HA production inhibitors. In summary, we identified endogenous HA as an important regulator of peritoneal fibrosis, not just a passive player during this pathological process.
Collapse
Affiliation(s)
- Anna Kocurkova
- Department of Experimental Biology, Faculty of Sciences, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Michaela Kerberova
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | | | | | - Miriam Sandanusova
- Department of Experimental Biology, Faculty of Sciences, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Matej Simek
- Contipro a.s., Dolni Dobrouc, Czech Republic
| | | | - Lukas Kubala
- Department of Experimental Biology, Faculty of Sciences, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Gabriela Ambrozova
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| |
Collapse
|
7
|
Lung Hyaluronasome: Involvement of Low Molecular Weight Ha (Lmw-Ha) in Innate Immunity. Biomolecules 2022; 12:biom12050658. [PMID: 35625586 PMCID: PMC9138743 DOI: 10.3390/biom12050658] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/01/2023] Open
Abstract
Hyaluronic acid (HA) is a major component of the extracellular matrix. It is synthesized by hyaluronan synthases (HAS) into high-molecular-weight chains (HMW-HA) that exhibit anti-inflammatory and immunomodulatory functions. In damaged, infected, and/or inflamed tissues, HMW-HA are degraded by hyaluronidases (HYAL) or reactive oxygen species (ROS) to give rise to low-molecular-weight HAs (LMW-HAs) that are potent pro-inflammatory molecules. Therefore, the size of HA regulates the balance of anti- or pro-inflammatory functions. The activities of HA depend also on its interactions with hyaladherins. HA synthesis, degradation, and activities through HA/receptors interactions define the hyaluronasome. In this review, a short overview of the role of high and low-molecular-weight HA polymers in the lungs is provided. The involvement of LMW-HA in pulmonary innate immunity via the activation of neutrophils, macrophages, dendritic cells, and epithelial cells is described to highlight LMW-HA as a therapeutic target in inflammatory respiratory diseases. Finally, the possibilities to counter LMW-HA’s deleterious effects in the lungs are discussed.
Collapse
|
8
|
Kaul A, Singampalli KL, Parikh UM, Yu L, Keswani SG, Wang X. Hyaluronan, a double-edged sword in kidney diseases. Pediatr Nephrol 2022; 37:735-744. [PMID: 34009465 PMCID: PMC8960635 DOI: 10.1007/s00467-021-05113-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/31/2021] [Accepted: 04/30/2021] [Indexed: 12/20/2022]
Abstract
Over the years, hyaluronic acid (HA) has emerged as an important molecule in nephrological and urological studies involving extracellular matrix (ECM) organization, inflammation, tissue regeneration, and viral sensing. During this time, many have noted the perplexing double-edged nature of the molecule, at times promoting pro-fibrotic events and at other times promoting anti-fibrotic events. Different molecular weights of HA can be attributed to these disparities, though most studies have yet to focus on this subtlety. With regard to the kidney, HA is induced in the initial response phase of injury and is subsequently decreased during disease progression of AKI, CKD, and diabetic nephropathy. These and other kidney diseases force patients, particularly pediatric patients, to face dialysis, surgical procedures, and ultimately, transplant. To summarize the current literature for researchers and pediatric nephrologists, this review aims to expound HA and elucidate its paradoxical effects in multiple kidney diseases using studies that emphasize HA molecular weight when available.
Collapse
Affiliation(s)
- Aditya Kaul
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Texas Children's Hospital/Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kavya L Singampalli
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Texas Children's Hospital/Baylor College of Medicine, Houston, TX, 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, 77030, TX, USA
- Department of Bioengineering, Rice University, Houston, 77030, TX, USA
| | - Umang M Parikh
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Texas Children's Hospital/Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ling Yu
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Texas Children's Hospital/Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sundeep G Keswani
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Texas Children's Hospital/Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xinyi Wang
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Texas Children's Hospital/Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
9
|
Kocurkova A, Nesporova K, Sandanusova M, Kerberova M, Lehka K, Velebny V, Kubala L, Ambrozova G. Endogenously-Produced Hyaluronan and Its Potential to Regulate the Development of Peritoneal Adhesions. Biomolecules 2021; 12:biom12010045. [PMID: 35053193 PMCID: PMC8773905 DOI: 10.3390/biom12010045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/17/2021] [Accepted: 12/24/2021] [Indexed: 12/23/2022] Open
Abstract
Formation of peritoneal adhesions (PA) is one of the major complications following intra-abdominal surgery. It is primarily caused by activation of the mesothelial layer and underlying tissues in the peritoneal membrane resulting in the transition of mesothelial cells (MCs) and fibroblasts to a pro-fibrotic phenotype. Pro-fibrotic transition of MCs—mesothelial-to-mesenchymal transition (MMT), and fibroblasts activation to myofibroblasts are interconnected to changes in cellular metabolism and culminate in the deposition of extracellular matrix (ECM) in the form of fibrotic tissue between injured sides in the abdominal cavity. However, ECM is not only a mechanical scaffold of the newly synthetized tissue but reciprocally affects fibrosis development. Hyaluronan (HA), an important component of ECM, is a non-sulfated glycosaminoglycan consisting of N-acetyl-D-glucosamine (GlcNAc) and D-glucuronic acid (GlcUA) that can affect the majority of processes involved in PA formation. This review considers the role of endogenously produced HA in the context of different fibrosis-related pathologies and its overlap in the development of PA.
Collapse
Affiliation(s)
- Anna Kocurkova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, 612 65 Brno, Czech Republic; (A.K.); (M.S.); (M.K.); (L.K.)
- Institute of Experimental Biology, Faculty of Science, Masaryk University, 611 37 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic
| | - Kristina Nesporova
- Contipro a.s., Dolní Dobrouč 401, 561 02 Dolní Dobrouč, Czech Republic; (K.N.); (K.L.); (V.V.)
| | - Miriam Sandanusova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, 612 65 Brno, Czech Republic; (A.K.); (M.S.); (M.K.); (L.K.)
- Institute of Experimental Biology, Faculty of Science, Masaryk University, 611 37 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic
| | - Michaela Kerberova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, 612 65 Brno, Czech Republic; (A.K.); (M.S.); (M.K.); (L.K.)
| | - Katerina Lehka
- Contipro a.s., Dolní Dobrouč 401, 561 02 Dolní Dobrouč, Czech Republic; (K.N.); (K.L.); (V.V.)
| | - Vladimir Velebny
- Contipro a.s., Dolní Dobrouč 401, 561 02 Dolní Dobrouč, Czech Republic; (K.N.); (K.L.); (V.V.)
| | - Lukas Kubala
- Institute of Biophysics, Academy of Sciences of the Czech Republic, 612 65 Brno, Czech Republic; (A.K.); (M.S.); (M.K.); (L.K.)
- Institute of Experimental Biology, Faculty of Science, Masaryk University, 611 37 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic
| | - Gabriela Ambrozova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, 612 65 Brno, Czech Republic; (A.K.); (M.S.); (M.K.); (L.K.)
- Correspondence:
| |
Collapse
|
10
|
Zhang F, Zhou X, Zou H, Liu L, Li X, Ruan Y, Xie Y, Shi M, Xiao Y, Wang Y, Zhou Y, Wu Y, Guo B. SAA1 is transcriptionally activated by STAT3 and accelerates renal interstitial fibrosis by inducing endoplasmic reticulum stress. Exp Cell Res 2021; 408:112856. [PMID: 34597680 DOI: 10.1016/j.yexcr.2021.112856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 02/07/2023]
Abstract
Renal interstitial fibrosis (RIF) is the common irreversible pathway by which chronic kidney disease (CKD) progresses to the end stage. The transforming growth factor-β (TGF-β)/signal transducer and activator of transcription 3 (STAT3) signaling pathway is a common factor leading to inflammation-mediated RIF, but its downstream regulatory mechanism is still unclear. Bioinformatics analysis predicted that serum amyloid A protein 1 (SAA1) was one of the target genes for transcriptional activation of STAT3 signaling. As an acute phase reaction protein, SAA1 plays an important role in many inflammatory reactions, and research has suggested that SAA1 is significantly elevated in the serum of patients with CKD. In this research, multiple experiments were performed to investigate the role of SAA1 in the process of RIF. SAA1 was abnormally highly expressed in kidney tissue from individuals who underwent unilateral ureteral obstruction (UUO) and TGF-β-induced HK2 cells, and the abnormal expression was directly related to the transcriptional activation of STAT3. Additionally, SAA1 can directly target and bind valosin-containing protein (VCP)-interacting membrane selenoprotein (VIMP) to inhibit the function of the Derlin-1/VCP/VIMP complex, preventing the transportation and degradation of the misfolded protein, resulting in endoplasmic reticulum (ER) stress characterized by an increase in glucose-regulated protein 78 (GRP78) levels and ultimately promoting the occurrence and development of RIF.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Xingcheng Zhou
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Huimei Zou
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China; School of Nursing, Guizhou Medical University, Guiyang, 550025, China
| | - Lirong Liu
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550025, China; Department of Clinical Hematology, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550025, China
| | - Xiaoying Li
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China; Department of Nephrology, Guiyang First People's Hospital, Guiyang, 550025, China
| | - Yuanyuan Ruan
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Ying Xie
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Mingjun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Ying Xiao
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Yuanyuan Wang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Yuxia Zhou
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Yuansheng Wu
- Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China.
| |
Collapse
|
11
|
Short WD, Wang X, Li H, Yu L, Kaul A, Calderon GA, Gilley J, Bollyky PL, Balaji S, Keswani SG. Interleukin-10 Producing T Lymphocytes Attenuate Dermal Scarring. Ann Surg 2021; 274:627-636. [PMID: 34506318 PMCID: PMC8428868 DOI: 10.1097/sla.0000000000004984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Demonstrate the impact of IL-10 producing T lymphocytes on mediating dermal scarring. SUMMARY BACKGROUND DATA We demonstrated that CD4+ cells are essential to improving postinjury wound healing and preventing fibrosis. CD4+ subsets secrete differential cytokine and growth factor profiles, though their role in fibrosis is not known. IL-10, a key anti-inflammatory cytokine shown to promote regenerative wound healing, is secreted by some CD4+ subsets. We, therefore, hypothesize that IL-10 producing CD4+ T lymphocyte subsets selectively attenuate dermal wound fibrosis. METHODS IL-10-/- and wild-type murine splenocytes were enriched for CD4+ lymphocytes and adoptively transferred into severe combined immunodeficient (SCID) mice that received full-thickness wounds which were analyzed at days 7 and 28 for inflammation and collagen content. We then sorted CD4+CD44int/lowFoxP3-CD62L+ T cells (Tnaive) or CD4+CD44HiFoxP3- type 1 regulatory (Tr1) T cell subsets from 10BiT murine splenocytes, activated them, and transferred them into wounds. In vitro, dermal fibroblasts were cocultured with Tnaive or Tr1 and the effect on extracellular matrix (ECM) regulation was analyzed. RESULTS The anti-inflammatory and antifibrotic effects of CD4+ cells on SCID wounds were lost with cells from IL-10-/- mice. Adoptive transfer of Tr1 into SCID mice resulted in accelerated wound closure at d7 with reduced fibrosis at d28, with Tr1 favoring hyaluronan production by fibroblasts, an ECM molecule implicated in IL-10-induced regenerative healing. CONCLUSIONS IL-10 producing T-lymphocytes, specifically Tr1, regulate inflammatory cell cytokine expression to promote HA-rich ECM deposition and attenuate fibrosis. Promoting IL-10 producing lymphocytes in wounds may be a therapeutic target to promote regenerative wound healing.
Collapse
Affiliation(s)
- Walker D Short
- Department of Surgery, Division of Pediatric Surgery, Laboratory for Regenerative Tissue Repair, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas
| | - Xinyi Wang
- Department of Surgery, Division of Pediatric Surgery, Laboratory for Regenerative Tissue Repair, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas
| | - Hui Li
- Department of Surgery, Division of Pediatric Surgery, Laboratory for Regenerative Tissue Repair, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas
| | - Ling Yu
- Department of Surgery, Division of Pediatric Surgery, Laboratory for Regenerative Tissue Repair, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas
| | - Aditya Kaul
- Department of Surgery, Division of Pediatric Surgery, Laboratory for Regenerative Tissue Repair, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas
| | - Gisele A Calderon
- Department of Surgery, Division of Pediatric Surgery, Laboratory for Regenerative Tissue Repair, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas
| | - Jamie Gilley
- Department of Surgery, Division of Pediatric Surgery, Laboratory for Regenerative Tissue Repair, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas
| | - Paul L Bollyky
- Department of Medicine, Division of Infectious Diseases, Stanford University School of Medicine, Stanford, California
| | - Swathi Balaji
- Department of Surgery, Division of Pediatric Surgery, Laboratory for Regenerative Tissue Repair, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas
| | - Sundeep G Keswani
- Department of Surgery, Division of Pediatric Surgery, Laboratory for Regenerative Tissue Repair, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas
| |
Collapse
|
12
|
Soranno DE, Kirkbride-Romeo L, Han D, Altmann C, Rodell CB. Measurement of glomerular filtration rate reveals that subcapsular injection of shear-thinning hyaluronic acid hydrogels does not impair kidney function in mice. J Biomed Mater Res A 2021; 110:652-658. [PMID: 34590787 PMCID: PMC9292789 DOI: 10.1002/jbm.a.37317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 11/07/2022]
Abstract
The continued development of minimally invasive therapeutic implants, such as injectable hydrogels, necessitates the concurrent advancement of methods to best assess their biocompatibility via functional outcomes in vivo. Biomaterial implants have been studied to treat kidney disease; however, assessment of biocompatibility has been limited to biomarker and histological assessments. Techniques now exist to measure kidney function serially in vivo in murine studies via transcutaneous measurements of glomerular filtration rate (tGFR). In this study, adult male and female wild-type BalbC mice underwent right unilateral nephrectomy. The remaining solitary left kidney was allowed 4 weeks to recover via compensatory hypertrophy, after which subcapsular injection of either saline or shear-thinning hyaluronic acid hydrogel was performed. Serial tGFR measurements before and after treatment were used to assess the effect of hydrogel injection on kidney filtration. Urine and serum biomarkers of kidney function, and kidney histology were also quantified. Hydrogel injection did not affect kidney function, as assessed by tGFR. Results were in agreement with standard metrics of serum and urine biomarkers of injury as well as histological assessment of inflammation. The model developed provides a direct functional assessment of implant compatibility for the treatment of kidney disease and impact on kidney function.
Collapse
Affiliation(s)
- Danielle E Soranno
- Department of Pediatrics, University of Colorado, Aurora, Colorado, USA.,Department of Bioengineering, University of Colorado, Aurora, Colorado, USA.,Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | | | - Daniel Han
- Department of Urology, Stanford University, CA, USA
| | | | - Christopher B Rodell
- School of Biomedical Engineering, Science and Health SystemsScience and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
13
|
Garantziotis S. Modulation of hyaluronan signaling as a therapeutic target in human disease. Pharmacol Ther 2021; 232:107993. [PMID: 34587477 DOI: 10.1016/j.pharmthera.2021.107993] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/14/2022]
Abstract
The extracellular matrix is an active participant, modulator and mediator of the cell, tissue, organ and organismal response to injury. Recent research has highlighted the role of hyaluronan, an abundant glycosaminoglycan constituent of the extracellular matrix, in many fundamental biological processes underpinning homeostasis and disease development. From this basis, emerging studies have demonstrated the therapeutic potential of strategies which target hyaluronan synthesis, biology and signaling, with significant promise as therapeutics for a variety of inflammatory and immune diseases. This review summarizes the state of the art in this field and discusses challenges and opportunities in what could emerge as a new class of therapeutic agents, that we term "matrix biologics".
Collapse
Affiliation(s)
- Stavros Garantziotis
- Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA.
| |
Collapse
|
14
|
Kaul A, Short WD, Wang X, Keswani SG. Hyaluronidases in Human Diseases. Int J Mol Sci 2021; 22:ijms22063204. [PMID: 33809827 PMCID: PMC8004219 DOI: 10.3390/ijms22063204] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/18/2021] [Accepted: 03/18/2021] [Indexed: 12/28/2022] Open
Abstract
With the burgeoning interest in hyaluronic acid (HA) in recent years, hyaluronidases (HYALs) have come to light for their role in regulating catabolism of HA and its molecular weight (MW) distribution in various tissues. Of the six hyaluronidase-like gene sequences in the human genome, HYALs 1 and 2 are of particular significance because they are the primary hyaluronidases active in human somatic tissue. Perhaps more importantly, for the sake of this review, they cleave anti-inflammatory and anti-fibrotic high-molecular-weight HA into pro-inflammatory and pro-fibrotic oligosaccharides. With this, HYALs regulate HA degradation and thus the development and progression of various diseases. Given the dearth of literature focusing specifically on HYALs in the past decade, this review seeks to expound their role in human diseases of the skin, heart, kidneys, and more. The review will delve into the molecular mechanisms and pathways of HYALs and discuss current and potential future therapeutic benefits of HYALs as a clinical treatment.
Collapse
Affiliation(s)
- Aditya Kaul
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, TX 77030, USA; (A.K.); (W.D.S.)
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Walker D. Short
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, TX 77030, USA; (A.K.); (W.D.S.)
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xinyi Wang
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, TX 77030, USA; (A.K.); (W.D.S.)
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence: (X.W.); (S.G.K.); Tel.: +832-824-0469 (X.W.); +832-822-3135 (S.G.K.); Fax: +832-825-3141 (X.W.); +832-825-3141 (S.G.K.)
| | - Sundeep G. Keswani
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, TX 77030, USA; (A.K.); (W.D.S.)
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence: (X.W.); (S.G.K.); Tel.: +832-824-0469 (X.W.); +832-822-3135 (S.G.K.); Fax: +832-825-3141 (X.W.); +832-825-3141 (S.G.K.)
| |
Collapse
|
15
|
Selman G, Martinez L, Lightle A, Aguilar A, Woltmann D, Xiao Y, Vazquez-Padron RI, Salman LH. A hyaluronan synthesis inhibitor delays the progression of diabetic kidney disease in a mouse experimental model. KIDNEY360 2021; 2:809-818. [PMID: 34350420 PMCID: PMC8330520 DOI: 10.34067/kid.0004642020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND The role of hyaluronan (HA) in the development and progression of diabetic kidney disease (DKD), as well as the precise mechanisms and consequences of HA involvement in this pathology are still to be clarified. METHODS In this study, we assayed the effects of the HA synthesis inhibitor 4-methylumbelliferone (4-MU) on the development of DKD. Diabetic type 2 model mice (eNOS-/- C57BLKS/Jdb) were fed artificial diets containing 5% 4-MU or not for 9 weeks. Plasma glucose, glomerular filtration rate (GFR), albumin to creatinine ratio (ACR), and biomarkers of kidney function and systemic inflammation were measured at baseline and after treatment. Diabetic nephropathy was further characterized in treated and control mice by histopathology. RESULTS Treated animals consumed a daily dose of approximately 6.2 g of 4-MU per kg of body weight. At the end of the experimental period, the 4-MU supplemented diet resulted in a significant decrease in non-fasting plasma glucose (516 [interquartile range 378-1170] vs. 1149 [875.8-1287] mg/dL, P=0.050) and a trend toward lower HA kidney content (5.6 ± 1.5 vs. 8.8 ± 3.1 ng/mg of kidney weight, P=0.070) compared to the control diet, respectively. Diabetic animals treated with 4-MU showed significantly higher GFR and lower urine ACR and plasma cystatin C levels than diabetic controls. Independent histological assessment of DKD also demonstrated a significant decrease in mesangial expansion score and glomerular injury index in 4-MU-treated mice compared to controls. Plasma glucose showed a strong correlation with kidney HA levels (r=0.66, P=0.0098). Both total hyaluronan (r=0.76, P=0.0071) and low-molecular-weight hyaluronan content (r=0.64, P=0.036) in the kidneys correlated with urine ACR in mice. CONCLUSION These results show that the hyaluronan synthesis inhibitor 4-MU effectively slowed the progression of DKD and constitutes a potential new therapeutic approach to treat DKD.
Collapse
Affiliation(s)
- Guillermo Selman
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida
| | - Andrea Lightle
- Department of Pathology, Albany Medical Center, Albany, New York
| | - Alejandra Aguilar
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Daniel Woltmann
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Yuxuan Xiao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Roberto I. Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida
| | - Loay H. Salman
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York,Division of Nephrology and Hypertension, Albany Medical Center, Albany, New York
| |
Collapse
|