1
|
Koyuncu S, Sipahioğlu H, Karakukcu C, Zararsız G, İçaçan G, Biçer NS, Kocyigit I. The relationship between changes in peritoneal permeability with CA-125 and HIF-1α. Ther Apher Dial 2024. [PMID: 39233434 DOI: 10.1111/1744-9987.14206] [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: 05/18/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Peritoneal fibrosis (PF) is a major, persistent complication of prolonged peritoneal dialysis that eventually leads to peritoneal ultrafiltration failure and termination of peritoneal dialysis. Prolonged exposure to high glucose concentrations, degradation products, uremic toxins, and episodes of peritonitis can cause some changes in the peritoneal membrane, resulting in intraperitoneal inflammation and PF, leading to failure of ultrafiltration and dialysis. CA-125 can be used as a biomarker of peritoneal mesothelial cell count in the peritoneal dialysate and for monitoring cell count in PD patients. Hypoxia-inducible factor 1-alpha (HIF-1α) has been reported to cause PF, but has not been reported to be associated with changes in peritoneal structure. We hypothesized that peritoneal adequacy can be followed using HIF-1α and CA-125 values. In the present study, therefore, we investigated the relationship between HIF-1α and CA-125 levels and parietal membrane permeability changes in PD patients. METHODS Forty-five patients were included in the study. Peritoneal permeability was constant in 20 of these, while peritoneal permeability increased in 11 and decreased in 14. The HIF-1α value from the blood samples of the patients and the CA-125 measurement from the peritoneal fluids were measured. The relationship between peritoneal variability and CA-125 and HIF levels after follow-up was investigated. RESULTS We compared serum HIF-1α and peritoneal fluid CA-125 levels in the three groups receiving peritoneal dialysis treatment. HIF-1α levels increased with peritoneal permeability changes, while CA-125 levels decreased. In patients with high to low permeability changes, HIF-1α levels were higher compared to those with stable or low to high changes, which was statistically significant. Conversely, CA-125 levels significantly decreased in patients whose peritoneal permeability changed from high to low, compared to the other two groups. CONCLUSION Changes in peritoneal structure can be followed with biomarkers. It has been shown that CA-125 and HIF-1α levels can guide the changes in the peritoneal membrane. This can be useful in the monitoring of peritoneal dialysis.
Collapse
Affiliation(s)
- Sumeyra Koyuncu
- Department of Nephrology, Kayseri Education and Research Hospital, Kayseri, Turkey
| | - Hilal Sipahioğlu
- Department of İntensive Care Unit, Kayseri Education and Research Hospital, Kayseri, Turkey
| | - Cigdem Karakukcu
- Department of Biochemistry, Erciyes Medical Faculty, Kayseri, Turkey
| | - Gökmen Zararsız
- Department of Biostatistics, Erciyes Medical Faculty, Kayseri, Turkey
| | - Gamze İçaçan
- Department of Nephrology, Izmir City Hospital, Izmir, Turkey
| | | | - Ismail Kocyigit
- Department of Nephrology, Erciyes Medical Faculty, Kayseri, Turkey
| |
Collapse
|
2
|
Kang Y, Liu Y, Fu P, Ma L. Peritoneal fibrosis: from pathophysiological mechanism to medicine. Front Physiol 2024; 15:1438952. [PMID: 39301425 PMCID: PMC11411570 DOI: 10.3389/fphys.2024.1438952] [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: 05/27/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024] Open
Abstract
Peritoneal dialysis (PD) is currently one of the effective methods for treating end-stage renal disease (ESRD). However, long-term exposure to high concentration glucose in peritoneal dialysis environment could lead to peritoneal fibrosis (PF), impaired peritoneal filtration function, decreased peritoneal dialysis efficiency, and even withdrawal from peritoneal dialysis in patients. Considerable evidence suggests that peritoneal fibrosis after peritoneal dialysis is related to crucial factors such as mesothelial-to-mesenchymal transition (MMT), inflammatory response, and angiogenesis, etc. In our review, we summarize the pathophysiological mechanisms and further illustrate the future strategies against PF.
Collapse
Affiliation(s)
- Yingxi Kang
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Yuan Liu
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ping Fu
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Liang Ma
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Lotfollahzadeh S, Vazirani A, Sellinger IE, Clovie J, Hoekstra I, Patel A, Malloum AB, Yin W, Paul H, Yadati P, Siracus J, Malikova M, Pernar LI, Francis J, Stern L, Chitalia VC. Aryl Hydrocarbon Receptor Pathway Augments Peritoneal Fibrosis in a Murine CKD Model Exposed to Peritoneal Dialysate. KIDNEY360 2024; 5:1238-1250. [PMID: 39235862 PMCID: PMC11441816 DOI: 10.34067/kid.0000000000000516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/11/2024] [Indexed: 09/07/2024]
Abstract
Key Points
CKD and high glucose–containing peritoneal dialysate alter peritoneal membrane contributing to peritoneal dialysis failure, with a poorly understood mechanism.CKD milieu activates the aryl hydrocarbon receptor pathway in the subperitoneal vasculature, increasing the peritoneal fibrosis and collagen deposition in humans and mice.An aryl hydrocarbon receptor inhibitor mitigates CKD and peritoneal dialysis–mediated peritoneal fibrosis, collagen deposition, and vasculogenesis in a mouse model.
Background
CKD is a proinflammatory and profibrotic condition and can independently alter the peritoneal membrane structure. Peritoneal dialysis (PD) results in profound alterations in the peritoneal membrane. The mechanisms contributing to the alterations of the peritoneal membrane structure in CKD milieu, along with PD, are poorly understood.
Methods
Here, we show that human CKD induces peritoneal membrane thickening, fibrosis, and collagen deposition and activates the aryl hydrocarbon receptor (AHR) pathway in the subperitoneal vasculature. Leveraging a novel model of PD in CKD mice, we confirm these CKD-induced changes in the peritoneal membrane, which are exacerbated on exposure to the peritoneal dialysate. Peritoneal dialysate further augmented the AHR activity in endothelial cells of peritoneal microvasculature in CKD mice.
Results
Treatment of CKD mice with an AHR inhibitor in peritoneal dialysate for 2 weeks resulted in a seven-fold reduction in AHR expression in the endothelial cells of subperitoneal capillaries, a five-fold decrease in subperitoneal space, and a nine-fold decrease in fibrosis and collagen deposition compared with vehicle-treated CKD mice. AHR inhibition reduced inflammation, subperitoneal neovascular areas, and its downstream target, tissue factor. The AHR inhibitor treatment normalized the peritoneal dialysate-induced proinflammatory and profibrotic cytokines, such as IL-6, monocyte chemoattractant protein-1, and macrophage inflammatory protein 1 levels, in CKD mice.
Conclusions
This study uncovers the activation of the AHR-cytokine axis in the endothelial cells of subperitoneal vessels in humans and mice with CKD, which is likely to prime the peritoneal membrane to peritoneal dialysate–mediated alterations. This study supports further exploration of AHR as a potential therapeutic target to preserve the structural and functional integrity of the peritoneal membrane in PD.
Collapse
Affiliation(s)
- Saran Lotfollahzadeh
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Aniket Vazirani
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Isaac E Sellinger
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Janelle Clovie
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Isaac Hoekstra
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Arjun Patel
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Abbas Brahim Malloum
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Wenqing Yin
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Herreet Paul
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Pranav Yadati
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Jeffrey Siracus
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Marina Malikova
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Luise I Pernar
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Jean Francis
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Lauren Stern
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Vipul C Chitalia
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
4
|
Shi SS, Zhang YQ, Zhang LQ, Li YF, Zhou XS, Li RS. Expression and significance of SIRT6 in human peritoneal dialysis effluents and peritoneal mesothelial cells. Int Urol Nephrol 2024; 56:2659-2670. [PMID: 38483736 PMCID: PMC11266209 DOI: 10.1007/s11255-024-03970-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/02/2024] [Indexed: 07/25/2024]
Abstract
Sirtuin 6 (SIRT6) can inhibit the fibrosis of many organs. However, the relationship between SIRT6 and peritoneal fibrosis (PF) in peritoneal dialysis (PD) remains unclear. We collected 110 PD patients with a duration of PD for more than 3 months and studied the influence of PD duration and history of peritonitis on SIRT6 levels in PD effluents (PDEs). We also analyzed the relationship between SIRT6 levels in PDEs and transforming growth factor beta 1 (TGF-β1), IL-6, PD duration, peritoneal function, PD ultrafiltration (UF), and glucose exposure. We extracted human peritoneal mesothelial cells (HPMCs) from PDEs and measured the protein and gene expression levels of SIRT6, E-cadherin, vimentin, and TGF-β1 in these cells. Based on the clinical results, we used human peritoneal mesothelial cells lines (HMrSV5) to observe the changes in SIRT6 levels and mesothelial-to-mesenchymal transition (MMT) after intervention with PD fluid. By overexpressing and knocking down SIRT6 expression, we investigated the effect of SIRT6 expression on E-cadherin, vimentin, and TGF-β1 expression to elucidate the role of SIRT6 in mesothelial-to-epithelial transition in PMCs. Results: (1) With the extension of PD duration, the influence of infection on SIRT6 levels in PDEs increased. Patients with the PD duration of more than 5 years and a history of peritonitis had the lowest SIRT6 levels. (2) SIRT6 levels in PDEs were negatively correlated with PD duration, total glucose exposure, TGF-β1, IL-6 levels, and the dialysate-to-plasma ratio of creatinine (Cr4hD/P), but positively correlated with UF. This indicates that SIRT6 has a protective effect on the peritoneum. (3) The short-term group (PD ≤ 1 year) had higher SIRT6 and E-cadherin gene and protein levels than the mid-term group (1 year < PD ≤ 5 years) and long-term group (PD > 5 years) in PMCs, while vimentin and TGF-β1 levels were lower in the mid-term group and long-term group. Patients with a history of peritonitis had lower SIRT6 and E-cadherin levels than those without such a history. (4) After 4.25% PD fluid intervention for HPMCs, longer intervention time resulted in lower SIRT6 levels. (5) Overexpressing SIRT6 can lead to increased E-cadherin expression and decreased vimentin and TGF-β1 expression in HPMCs. Knocking down SIRT6 expression resulted in decreased E-cadherin expression and increased vimentin and TGF-β1 expression in HPMCs. This indicates that SIRT6 expression can inhibit MMT in HPMCs, alleviate PF associated with PD, and have a protective effect on the peritoneum.
Collapse
Affiliation(s)
- Shuai-Shuai Shi
- Department of Nephrology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, Shanxi, China
- Department of Nephrology, Heji Hospital of Changzhi Medical College, Changzhi, 046011, Shanxi, China
| | - Yi-Qiang Zhang
- Department of Biochemistry, Changzhi Medical College, Changzhi, 046000, Shanxi, China
| | - Lu-Qi Zhang
- First Clinical Department of Changzhi Medical College, Changzhi, 046000, Shanxi, China
| | - Yun-Feng Li
- First Clinical Department of Changzhi Medical College, Changzhi, 046000, Shanxi, China
| | - Xiao-Shuang Zhou
- Department of Nephrology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, Shanxi, China
| | - Rong-Shan Li
- Department of Nephrology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, Shanxi, China.
| |
Collapse
|
5
|
Yang F, Zhang K, Dai X, Jiang W. Preliminary Exploration of Potential Active Ingredients and Molecular Mechanisms of Yanggan Yishui Granules for Treating Hypertensive Nephropathy Using UPLC-Q-TOF/MS Coupled with Network Pharmacology and Molecular Docking Strategy. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2024; 2024:7967999. [PMID: 38766523 PMCID: PMC11101260 DOI: 10.1155/2024/7967999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/07/2024] [Accepted: 04/26/2024] [Indexed: 05/22/2024]
Abstract
Hypertensive nephropathy (HN) is a prevalent complication of hypertension and stands as the second primary reason for end-stage renal disease. Research in clinical settings has revealed that Yanggan Yishui Granule (YGYSG) has significant therapeutic effects on HN. However, the material basis and action mechanisms of YGYSG against HN remain unclear. Consequently, this study utilized a comprehensive method integrating ultraperformance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS), network pharmacology, and molecular docking to delineate the active ingredients and potential therapeutic mechanisms of YGYSG for treating HN. Firstly, sixty distinct components were recognized in total as potential active ingredients in YGYSG by UPLC-Q-TOF/MS. Subsequently, the mechanisms of YGYSG against HN were revealed for the first time using network pharmacology. 23 ingredients played key roles in the complete network and were the key active ingredients, which could affect the renin-angiotensin system, fluid shear stress and atherosclerosis, HIF-1 signaling pathway, and AGE-RAGE signaling pathway in diabetic complications by regulating 29 key targets such as TNF, IL6, ALB, EGFR, ACE, and MMP2. YGYSG could treat HN through the suppression of inflammatory response and oxidative stress, attenuating the proliferation of renal vascular smooth muscle cells, lessening glomerular capillary systolic pressure, and ameliorating renal dysfunction and vascular damage through the aforementioned targets and pathways. Molecular docking results revealed that most key active ingredients exhibited a high affinity for binding to the key targets. This study pioneers in clarifying the bioactive compounds and molecular mechanisms of YGYSG against HN and offers scientific reference into the clinical application.
Collapse
Affiliation(s)
- Fan Yang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
- Department of Cardiology, The First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei, Anhui 230000, China
| | - Kailun Zhang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230000, China
| | - Xiaohua Dai
- Department of Cardiology, The First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei, Anhui 230000, China
| | - Weimin Jiang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| |
Collapse
|
6
|
Song Q, Li H, Yan H, Yu Z, Li Z, Yuan J, Jiang N, Ni Z, Gu L, Fang W. Inhibition of STAT3 by S3I-201 suppress peritoneal fibroblast phenotype conversion and alleviate peritoneal fibrosis. J Cell Mol Med 2024; 28:e18381. [PMID: 38780509 PMCID: PMC11114217 DOI: 10.1111/jcmm.18381] [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: 03/14/2024] [Revised: 04/22/2024] [Accepted: 04/27/2024] [Indexed: 05/25/2024] Open
Abstract
Peritoneal fibrosis is a common pathological response to long-term peritoneal dialysis (PD) and a major cause for PD discontinuation. Understanding the cellular and molecular mechanisms underlying the induction and progression of peritoneal fibrosis is of great interest. In our study, in vitro study revealed that signal transducer and activator of transcription 3 (STAT3) is a key factor in fibroblast activation and extracellular matrix (ECM) synthesis. Furthermore, STAT3 induced by IL-6 trans-signalling pathway mediate the fibroblasts of the peritoneal stroma contributed to peritoneal fibrosis. Inhibition of STAT3 exerts an antifibrotic effect by attenuating fibroblast activation and ECM production with an in vitro co-culture model. Moreover, STAT3 plays an important role in the peritoneal fibrosis in an animal model of peritoneal fibrosis developed in mice. Blocking STAT3 can reduce the peritoneal morphological changes induced by chlorhexidine gluconate. In conclusion, our findings suggested STAT3 signalling played an important role in peritoneal fibrosis. Therefore, blocking STAT3 might become a potential treatment strategy in peritoneal fibrosis.
Collapse
Affiliation(s)
- Qianhui Song
- Department of Nephrology, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- Shanghai Center for Peritoneal Dialysis ResearchShanghaiPeople's Republic of China
| | - Han Li
- Department of Nephrology, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- Shanghai Center for Peritoneal Dialysis ResearchShanghaiPeople's Republic of China
| | - Hao Yan
- Department of Nephrology, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- Shanghai Center for Peritoneal Dialysis ResearchShanghaiPeople's Republic of China
| | - Zanzhe Yu
- Department of Nephrology, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- Shanghai Center for Peritoneal Dialysis ResearchShanghaiPeople's Republic of China
| | - Zhenyuan Li
- Department of Nephrology, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- Shanghai Center for Peritoneal Dialysis ResearchShanghaiPeople's Republic of China
| | - Jiangzi Yuan
- Department of Nephrology, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- Shanghai Center for Peritoneal Dialysis ResearchShanghaiPeople's Republic of China
| | - Na Jiang
- Department of Nephrology, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- Shanghai Center for Peritoneal Dialysis ResearchShanghaiPeople's Republic of China
| | - Zhaohui Ni
- Department of Nephrology, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- Shanghai Center for Peritoneal Dialysis ResearchShanghaiPeople's Republic of China
| | - Leyi Gu
- Department of Nephrology, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- Shanghai Center for Peritoneal Dialysis ResearchShanghaiPeople's Republic of China
| | - Wei Fang
- Department of Nephrology, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- Shanghai Center for Peritoneal Dialysis ResearchShanghaiPeople's Republic of China
| |
Collapse
|
7
|
Sheng L, Shan Y, Dai H, Yu M, Sun J, Huang L, Wang F, Sheng M. Intercellular communication in peritoneal dialysis. Front Physiol 2024; 15:1331976. [PMID: 38390449 PMCID: PMC10882094 DOI: 10.3389/fphys.2024.1331976] [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: 11/02/2023] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Long-term peritoneal dialysis (PD) causes structural and functional alterations of the peritoneal membrane. Peritoneal deterioration and fibrosis are multicellular and multimolecular processes. Under stimulation by deleterious factors such as non-biocompatibility of PD solution, various cells in the abdominal cavity show differing characteristics, such as the secretion of different cytokines, varying protein expression levels, and transdifferentiation into other cells. In this review, we discuss the role of various cells in the abdominal cavity and their interactions in the pathogenesis of PD. An in-depth understanding of intercellular communication and inter-organ communication in PD will lead to a better understanding of the pathogenesis of this disease, enabling the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Li Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Shan
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Huibo Dai
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Manshu Yu
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinyi Sun
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liyan Huang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Funing Wang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meixiao Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
8
|
Si Z, Su W, Zhou Z, Li J, Su C, Zhang Y, Hu Z, Huang Z, Zhou H, Cong A, Zhou Z, Cao W. Hyperglycolysis in endothelial cells drives endothelial injury and microvascular alterations in peritoneal dialysis. Clin Transl Med 2023; 13:e1498. [PMID: 38037461 PMCID: PMC10689974 DOI: 10.1002/ctm2.1498] [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: 05/29/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Endothelial cell (EC) dysfunction leading to microvascular alterations is a hallmark of technique failure in peritoneal dialysis (PD). However, the mechanisms underlying EC dysfunction in PD are poorly defined. METHODS We combined RNA sequencing with metabolite set analysis to characterize the metabolic profile of peritoneal ECs from a mouse model of PD. This was combined with EC-selective blockade of glycolysis by genetic or pharmacological inhibition of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) in vivo and in vitro. We also investigated the association between peritoneal EC glycolysis and microvascular alterations in human peritoneal samples from patients with end-stage kidney disease (ESKD). RESULTS In a mouse model of PD, peritoneal ECs had a hyperglycolytic metabolism that shunts intermediates into nucleotide synthesis. Hyperglycolytic mouse peritoneal ECs displayed a unique active phenotype with increased proliferation, permeability and inflammation. The active phenotype of mouse peritoneal ECs can be recapitulated in human umbilical venous ECs and primary human peritoneal ECs by vascular endothelial growth factor that was released from high glucose-treated mesothelial cells. Importantly, reduction of peritoneal EC glycolysis, via endothelial deficiency of the glycolytic activator PFKFB3, inhibited PD fluid-induced increases in peritoneal capillary density, vascular permeability and monocyte extravasation, thereby protecting the peritoneum from the development of structural and functional damages. Mechanistically, endothelial PFKFB3 deficiency induced the protective effects in part by inhibiting cell proliferation, VE-cadherin endocytosis and monocyte-adhesion molecule expression. Pharmacological PFKFB3 blockade induced a similar therapeutic benefit in this PD model. Human peritoneal tissue from patients with ESKD also demonstrated evidence of increased EC PFKFB3 expression associated with microvascular alterations and peritoneal dysfunction. CONCLUSIONS These findings reveal a critical role of glycolysis in ECs in mediating the deterioration of peritoneal function and suggest that strategies targeting glycolysis in peritoneal ECs may be of therapeutic benefit for patients undergoing PD.
Collapse
Affiliation(s)
- Zekun Si
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Wenyan Su
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Zhuoyu Zhou
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Jinjin Li
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Cailing Su
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Ying Zhang
- Division of NephrologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhouP. R. China
| | - Zuoyu Hu
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Zhijie Huang
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Hong Zhou
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Ansheng Cong
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Zhanmei Zhou
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Wei Cao
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| |
Collapse
|
9
|
Mukherjee S, Ghosh S, Bawali S, Chatterjee R, Saha A, Sengupta A, Keswani T, Sarkar S, Ghosh P, Chakraborty S, Khamaru P, Bhattacharyya A. Administration of soluble gp130Fc disrupts M-1 macrophage polarization, dendritic cell activation, MDSC expansion and Th-17 induction during experimental cerebral malaria. Int Immunopharmacol 2023; 123:110671. [PMID: 37494839 DOI: 10.1016/j.intimp.2023.110671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 07/28/2023]
Abstract
Regulatory effect of IL-6 on various immune cells plays a crucial role during experimental cerebral malaria pathogenesis. IL-6 neutralization can restore distorted ratios of myeloid dendritic cells and plasmacytoid dendritic cells as well as the balance between Th-17 and T-regulatory cells. IL-6 can also influence immune cells through classical and trans IL-6 signalling pathways. As trans IL-6 signalling is reportedly involved during malaria pathogenesis, we focused on studying the effects of trans IL-6 signalling blockade on various immune cell populations and how they regulate ECM progression. Results show that administration of sgp130Fc recombinant chimera protein lowers the parasitemia, increases the survivability of Plasmodium berghei ANKA infected mice, and restores the distorted ratios of M1/M2 macrophage, mDC/pDC, and Th-17/Treg. IL-6 trans signalling blockade has been found to affect both expansion of myeloid derived suppressor cells (MDSCs) and expression of inflammatory markers on them during Plasmodium berghei ANKA infection indicating that trans IL-6 signalling might regulate various immune cells and their function during ECM. In this work for the first time, we delineate the effect of sgp130Fc administration on influencing the immunological changes within the host secondary lymphoid organ during ECM induced by Plasmodium berghei ANKA infection.
Collapse
Affiliation(s)
- Saikat Mukherjee
- Immunology Laboratory, Department of Zoology, University of Calcutta. 35, Ballygunge Circular Road, Kolkata-700019, West Bengal, India
| | - Soubhik Ghosh
- Immunology Laboratory, Department of Zoology, University of Calcutta. 35, Ballygunge Circular Road, Kolkata-700019, West Bengal, India
| | - Sriparna Bawali
- Immunology Laboratory, Department of Zoology, University of Calcutta. 35, Ballygunge Circular Road, Kolkata-700019, West Bengal, India
| | - Rimbik Chatterjee
- Immunology Laboratory, Department of Zoology, University of Calcutta. 35, Ballygunge Circular Road, Kolkata-700019, West Bengal, India
| | - Atreyee Saha
- Immunology Laboratory, Department of Zoology, University of Calcutta. 35, Ballygunge Circular Road, Kolkata-700019, West Bengal, India
| | - Anirban Sengupta
- Department of Biomedical and Clinical Sciences, Linköping University, 58185 Linköping, Sweden
| | - Tarun Keswani
- Center for Immunological and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA 149 13th Street Charlestown, MA 02129, USA
| | - Samrat Sarkar
- Immunology Laboratory, Department of Zoology, University of Calcutta. 35, Ballygunge Circular Road, Kolkata-700019, West Bengal, India
| | - Pronabesh Ghosh
- Immunology Laboratory, Department of Zoology, University of Calcutta. 35, Ballygunge Circular Road, Kolkata-700019, West Bengal, India
| | - Sayan Chakraborty
- Immunology Laboratory, Department of Zoology, University of Calcutta. 35, Ballygunge Circular Road, Kolkata-700019, West Bengal, India
| | - Poulomi Khamaru
- Immunology Laboratory, Department of Zoology, University of Calcutta. 35, Ballygunge Circular Road, Kolkata-700019, West Bengal, India
| | - Arindam Bhattacharyya
- Immunology Laboratory, Department of Zoology, University of Calcutta. 35, Ballygunge Circular Road, Kolkata-700019, West Bengal, India.
| |
Collapse
|
10
|
Li J, Liu Y, Liu J. A review of research progress on mechanisms of peritoneal fibrosis related to peritoneal dialysis. Front Physiol 2023; 14:1220450. [PMID: 37817984 PMCID: PMC10560738 DOI: 10.3389/fphys.2023.1220450] [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: 05/10/2023] [Accepted: 09/13/2023] [Indexed: 10/12/2023] Open
Abstract
Peritoneal dialysis (PD) is an effective alternative treatment for patients with end-stage renal disease (ESRD) and is increasingly being adopted and promoted worldwide. However, as the duration of peritoneal dialysis extends, it can expose problems with dialysis inadequacy and ultrafiltration failure. The exact mechanism and aetiology of ultrafiltration failure have been of great concern, with triggers such as biological incompatibility of peritoneal dialysis solutions, uraemia toxins, and recurrent intraperitoneal inflammation initiating multiple pathways that regulate the release of various cytokines, promote the transcription of fibrosis-related genes, and deposit extracellular matrix. As a result, peritoneal fibrosis occurs. Exploring the pathogenic factors and molecular mechanisms can help us prevent peritoneal fibrosis and prolong the duration of Peritoneal dialysis.
Collapse
Affiliation(s)
- Jin’e Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yinghong Liu
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Jianping Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
11
|
Zhao T, Sun Z, Lai X, Lu H, Liu L, Li S, Yuan JH, Guo Z. Tamoxifen exerts anti-peritoneal fibrosis effects by inhibiting H19-activated VEGFA transcription. J Transl Med 2023; 21:614. [PMID: 37697303 PMCID: PMC10494369 DOI: 10.1186/s12967-023-04470-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/25/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Peritoneal dialysis (PD) remains limited due to dialysis failure caused by peritoneal fibrosis. Tamoxifen (TAM), an inhibitor of estrogen receptor 1 (ESR1), has been reported to treat fibrosis, but the underlying mechanism remains unknown. In this study, we sought to explore whether tamoxifen played an anti-fibrotic role by affecting transcription factor ESR1. METHODS ESR1 expression was detected in the human peritoneum. Mice were daily intraperitoneally injected with 4.25% glucose PD dialysate containing 40 mM methylglyoxal for 2 weeks to establish PD-induced peritoneal fibrosis. Tamoxifen was administrated by daily gavage, at the dose of 10 mg/kg. Chromatin immunoprecipitation (ChIP) and dual-luciferase reporter assay were performed to validate ESR1 bound H19 promoter. Gain-of-function and loss-of-function experiments were performed to investigate the biological roles of H19 on the mesothelial-mesenchymal transition (MMT) of human peritoneal mesothelial cells (HPMCs). Intraperitoneal injection of nanomaterial-wrapped 2'-O-Me-modified small interfering RNA was applied to suppress H19 in the mouse peritoneum. RNA immunoprecipitation and RNA pull-down assays demonstrated binding between H19 and p300. Exfoliated peritoneal cells were obtained from peritoneal dialysis effluent to analyze the correlations between ESR1 (or H19) and peritoneal solute transfer rate (PSTR). RESULTS ESR1 was increased significantly in the peritoneum after long-term exposure to PD dialysate. Tamoxifen treatment ameliorated high glucose-induced MMT of HPMCs, improved ultrafiltration rate, and decreased PSTR of mouse peritoneum. Tamoxifen reduced the H19 level by decreasing the ESR1 transcription of H19. Depletion of H19 reversed the pro-fibrotic effect of high glucose while ectopic expression of H19 exacerbated fibrotic pathological changes. Intraperitoneal injection of nanomaterial-wrapped 2'-O-Me-modified siRNAs targeting H19 mitigated PD-related fibrosis in mice. RNA immunoprecipitation (RIP) and RNA pull-down results delineated that H19 activated VEGFA expression by binding p300 to the VEGFA promoter and inducing histone acetylation of the VEGFA promoter. ESR1 and H19 were promising targets to predict peritoneal function. CONCLUSIONS High glucose-induced MMT of peritoneal mesothelial cells in peritoneal dialysis via activating ESR1. In peritoneal mesothelial cells, ESR1 transcribed the H19 and H19 binds to transcription cofactor p300 to activate the VEGFA. Targeting ESR1/H19/VEGFA pathway provided new hope for patients undergoing peritoneal dialysis.
Collapse
Affiliation(s)
- Tingting Zhao
- Department of Nephrology, First Affiliated Hospital of Naval Medical University, Shanghai Changhai Hospital, Shanghai, 200433, China
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200433, China
| | - Zhengyu Sun
- Department of Nephrology, First Affiliated Hospital of Naval Medical University, Shanghai Changhai Hospital, Shanghai, 200433, China
| | - Xueli Lai
- Department of Nephrology, First Affiliated Hospital of Naval Medical University, Shanghai Changhai Hospital, Shanghai, 200433, China
| | - Hongtao Lu
- Department of Nutrition, Naval Medical University, Shanghai, 200433, China
| | - Lulu Liu
- Department of Nephrology, First Affiliated Hospital of Naval Medical University, Shanghai Changhai Hospital, Shanghai, 200433, China
| | - Shuangxi Li
- Department of Nephrology, First Affiliated Hospital of Naval Medical University, Shanghai Changhai Hospital, Shanghai, 200433, China
| | - Ji-Hang Yuan
- Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China.
| | - Zhiyong Guo
- Department of Nephrology, First Affiliated Hospital of Naval Medical University, Shanghai Changhai Hospital, Shanghai, 200433, China.
| |
Collapse
|
12
|
Zhao H, Zhang HL, Jia L. High glucose dialysate-induced peritoneal fibrosis: Pathophysiology, underlying mechanisms and potential therapeutic strategies. Biomed Pharmacother 2023; 165:115246. [PMID: 37523983 DOI: 10.1016/j.biopha.2023.115246] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/02/2023] Open
Abstract
Peritoneal dialysis is an efficient renal replacement therapy for patients with end-stage kidney disease. However, continuous exposure of the peritoneal membrane to dialysate frequently leads to peritoneal fibrosis, which alters the function of the peritoneal membrane and results in withdrawal from peritoneal dialysis in patients. Among others, high glucose dialysate is considered as a predisposing factor for peritoneal fibrosis in patients on peritoneal dialysis. Glucose-induced inflammation, metabolism disturbance, activation of the renin-angiotensin-aldosterone system, angiogenesis and noninflammation-induced reactive oxygen species are implicated in the pathogenesis of high glucose dialysate-induced peritoneal fibrosis. Specifically, high glucose causes chronic inflammation and recurrent peritonitis, which could cause migration and polarization of inflammatory cells, as well as release of cytokines and fibrosis. High glucose also interferes with lipid metabolism and glycolysis by activating the sterol-regulatory element-binding protein-2/cleavage-activating protein pathway and increasing hypoxia inducible factor-1α expression, leading to angiogenesis and peritoneal fibrosis. Activation of the renin-angiotensin-aldosterone system and Ras-mitogen activated protein kinase signaling pathway is another contributing factor in high glucose dialysate-induced fibrosis. Ultimately, activation of the transforming growth factor-β1/Smad pathway is involved in mesothelial-mesenchymal transition or epithelial-mesenchymal transition, which leads to the development of fibrosis. Although possible intervention strategies for peritoneal dialysate-induced fibrosis by targeting the transforming growth factor-β1/Smad pathway have occasionally been proposed, lack of laboratory evidence renders clinical decision-making difficult. We therefore aim to revisit the upstream pathways of transforming growth factor-beta1/Smad and propose potential therapeutic targets for high glucose-induced peritoneal fibrosis.
Collapse
Affiliation(s)
- Hanxue Zhao
- First Clinical Medical College, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Beijing 100053, China
| | - Hong-Liang Zhang
- Department of Life Sciences, National Natural Science Foundation of China, No. 83 Shuangqing Road, Beijing 100085, China.
| | - Linpei Jia
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Beijing 100053, China.
| |
Collapse
|
13
|
Su HY, Yang JJ, Zou R, An N, Chen XC, Yang C, Yang HJ, Yao CW, Liu HF. Autophagy in peritoneal fibrosis. Front Physiol 2023; 14:1187207. [PMID: 37256065 PMCID: PMC10226653 DOI: 10.3389/fphys.2023.1187207] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/03/2023] [Indexed: 06/01/2023] Open
Abstract
Peritoneal dialysis (PD) is a widely accepted renal replacement therapy for patients with end-stage renal disease (ESRD). Morphological and functional changes occur in the peritoneal membranes (PMs) of patients undergoing long-term PD. Peritoneal fibrosis (PF) is a common PD-related complication that ultimately leads to PM injury and peritoneal ultrafiltration failure. Autophagy is a cellular process of "self-eating" wherein damaged organelles, protein aggregates, and pathogenic microbes are degraded to maintain intracellular environment homeostasis and cell survival. Growing evidence shows that autophagy is involved in fibrosis progression, including renal fibrosis and hepatic fibrosis, in various organs. Multiple risk factors, including high-glucose peritoneal dialysis solution (HGPDS), stimulate the activation of autophagy, which participates in PF progression, in human peritoneal mesothelial cells (HPMCs). Nevertheless, the underlying roles and mechanisms of autophagy in PF progression remain unclear. In this review, we discuss the key roles and potential mechanisms of autophagy in PF to offer novel perspectives on future therapy strategies for PF and their limitations.
Collapse
|
14
|
Lee Y, Lee J, Park M, Seo A, Kim KH, Kim S, Kang M, Kang E, Yoo KD, Lee S, Kim DK, Oh KH, Kim YS, Joo KW, Yang SH. Inflammatory chemokine (C-C motif) ligand 8 inhibition ameliorates peritoneal fibrosis. FASEB J 2023; 37:e22632. [PMID: 36468785 DOI: 10.1096/fj.202200784r] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 10/09/2022] [Accepted: 10/18/2022] [Indexed: 12/12/2022]
Abstract
Peritoneal fibrosis (PF) is an irreversible complication of peritoneal dialysis (PD) that leads to loss of peritoneal membrane function. We investigated PD effluent and serum levels and the tissue expression of chemokine (C-C motif) ligand 8 (CCL8) in patients with PD. Additionally, we investigated their association with PF in a mouse model. Eighty-two end-stage renal disease (ESRD) patients with PD were examined. CCL8 levels were measured via enzyme-linked immunosorbent assays in PD effluents and serum and analyzed with peritoneal transport parameters. Human peritoneal mesothelial cells (hPMCs) were obtained from the PD effluents of 20 patients. Primary cultured hPMCs were treated with recombinant (r) transforming growth factor (TGF)-β, and CCL8 expression was assessed via western blotting. As the duration of PD increased, the concentration of CCL8 in PD effluents significantly increased. Correlations between peritoneal transport parameters and dialysate CCL8 levels were observed. Western blotting analysis showed that CCL8 was upregulated via rTGF-β treatment, accompanied by increases in markers of inflammation, fibrosis, senescence, and apoptosis in hPMCs after induction of fibrosis with rTGF-β. Anti-CCL8 monoclonal antibody (mAb) treatment suppressed the rTGF-β-induced increase in all analyzed markers. Immunohistochemical analysis revealed that CCL8 along with fibrosis- and inflammation-related markers were significantly increased in the PF mouse model. Functional blockade of CCL8 using a CCR8 inhibitor (R243) abrogated peritoneal inflammation and fibrosis in vivo. In conclusion, high CCL8 levels in PD effluents may be associated with an increased risk of PD failure, and the CCL8 pathway is associated with PF. CCL8 blockade can ameliorate peritoneal inflammation and fibrosis.
Collapse
Affiliation(s)
- Yeonhee Lee
- Department of Internal Medicine, Uijeongbu Eulji Medical Center, Eulji University, Gyeonggi-do, Republic of Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jangwook Lee
- Department of Internal Medicine, Dongguk University Ilsan Hospital, Gyeonggi-do, Republic of Korea
| | - Minkyoung Park
- Kidney Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Areum Seo
- Kidney Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Kyu Hyeon Kim
- Kidney Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Seonmi Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Minjung Kang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eunjeong Kang
- Department of Internal Medicine, Ewha Womans University Seoul Hospital, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Kyung Don Yoo
- Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Sunhwa Lee
- Department of Internal Medicine, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Kidney Research Institute, Seoul National University, Seoul, Republic of Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Kidney Research Institute, Seoul National University, Seoul, Republic of Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Kidney Research Institute, Seoul National University, Seoul, Republic of Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kwon Wook Joo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Kidney Research Institute, Seoul National University, Seoul, Republic of Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung Hee Yang
- Kidney Research Institute, Seoul National University, Seoul, Republic of Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
15
|
Blockade of the protease ADAM17 ameliorates experimental pancreatitis. Proc Natl Acad Sci U S A 2022; 119:e2213744119. [PMID: 36215509 PMCID: PMC9586293 DOI: 10.1073/pnas.2213744119] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Acute and chronic pancreatitis, the latter associated with fibrosis, are multifactorial inflammatory disorders and leading causes of gastrointestinal disease-related hospitalization. Despite the global health burden of pancreatitis, currently, there are no effective therapeutic agents. In this regard, the protease A Disintegrin And Metalloproteinase 17 (ADAM17) mediates inflammatory responses through shedding of bioactive inflammatory cytokines and mediators, including tumor necrosis factor α (TNFα) and the soluble interleukin (IL)-6 receptor (sIL-6R), the latter of which drives proinflammatory IL-6 trans-signaling. However, the role of ADAM17 in pancreatitis is unclear. To address this, Adam17ex/ex mice-which are homozygous for the hypomorphic Adam17ex allele resulting in marked reduction in ADAM17 expression-and their wild-type (WT) littermates were exposed to the cerulein-induced acute pancreatitis model, and acute (1-wk) and chronic (20-wk) pancreatitis models induced by the cigarette smoke carcinogen nicotine-derived nitrosamine ketone (NNK). Our data reveal that ADAM17 expression was up-regulated in pancreatic tissues of animal models of pancreatitis. Moreover, the genetic (Adam17ex/ex mice) and therapeutic (ADAM17 prodomain inhibitor [A17pro]) targeting of ADAM17 ameliorated experimental pancreatitis, which was associated with a reduction in the IL-6 trans-signaling/STAT3 axis. This led to reduced inflammatory cell infiltration, including T cells and neutrophils, as well as necrosis and fibrosis in the pancreas. Furthermore, up-regulation of the ADAM17/IL-6 trans-signaling/STAT3 axis was a feature of pancreatitis patients. Collectively, our findings indicate that the ADAM17 protease plays a pivotal role in the pathogenesis of pancreatitis, which could pave the way for devising novel therapeutic options to be deployed against this disease.
Collapse
|
16
|
Wang R, Guo T, Li J. Mechanisms of Peritoneal Mesothelial Cells in Peritoneal Adhesion. Biomolecules 2022; 12:biom12101498. [PMID: 36291710 PMCID: PMC9599397 DOI: 10.3390/biom12101498] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/08/2022] [Accepted: 10/14/2022] [Indexed: 11/24/2022] Open
Abstract
A peritoneal adhesion (PA) is a fibrotic tissue connecting the abdominal or visceral organs to the peritoneum. The formation of PAs can induce a variety of clinical diseases. However, there is currently no effective strategy for the prevention and treatment of PAs. Damage to peritoneal mesothelial cells (PMCs) is believed to cause PAs by promoting inflammation, fibrin deposition, and fibrosis formation. In the early stages of PA formation, PMCs undergo mesothelial–mesenchymal transition and have the ability to produce an extracellular matrix. The PMCs may transdifferentiate into myofibroblasts and accelerate the formation of PAs. Therefore, the aim of this review was to understand the mechanism of action of PMCs in PAs, and to offer a theoretical foundation for the treatment and prevention of PAs.
Collapse
Affiliation(s)
- Ruipeng Wang
- The First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou 730030, China
| | - Tiankang Guo
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730030, China
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730030, China
| | - Junliang Li
- The First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou 730030, China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730030, China
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730030, China
- Correspondence:
| |
Collapse
|
17
|
Song Q, Yang X, Shi Y, Yan H, Yu Z, Li Z, Yuan J, Ni Z, Gu L, Fang W. High intraperitoneal interleukin-6 levels predict ultrafiltration (UF) insufficiency in peritoneal dialysis patients: A prospective cohort study. Front Med (Lausanne) 2022; 9:836861. [PMID: 36035388 PMCID: PMC9400905 DOI: 10.3389/fmed.2022.836861] [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: 12/16/2021] [Accepted: 07/18/2022] [Indexed: 11/20/2022] Open
Abstract
Introduction UF insufficiency is a major limitation in PD efficiency and sustainability. Our study object to investigate the efficacy of intraperitoneal inflammation marker, IL-6 level as a predictor of UF insufficiency in continuous ambulatory peritoneal dialysis (CAPD) patients. Methods Stable prevalent CAPD patients were enrolled in this prospective study. IL-6 concentration in the overnight effluent was determined and expressed as the IL-6 appearance rate (IL-6 AR). Patients were divided into two groups according to the median of IL-6 AR and prospectively followed up until death, transfer to permanent HD, recovery of renal function, kidney transplantation, transfer to other centers, lost to follow-up or to the end of study (January 31, 2021). Factors associated with UF capacity as well as dialysate IL-6 AR were assessed by multivariable linear regression. Cox proportional hazards model was used to examine the association between dialysate IL-6 AR and UF insufficiency. Results A total of 291 PD patients were enrolled, including 148 males (51%) with a mean age of 56.6 ± 14.1 years and a median PD duration of 33.4 (12.7-57.5) months. No correlation was found between dialysate IL-6 AR and UF capacity at baseline. PD duration was found positively correlated with baseline dialysate IL-6 AR, while 24h urine volume was negatively correlated with baseline dialysate IL-6 AR (P < 0.05). By the end of study, UF insufficiency was observed in 56 (19.2%) patients. Patients in the high IL-6 AR group showed a significantly inferior UF insufficiency-free survival when compared with their counterparts in the low IL-6 AR group (P = 0.001). In the multivariate Cox regression analysis, after adjusting for DM, previous peritonitis episode and 24h urine volume, higher baseline dialysate IL-6 AR (HR 3.639, 95% CI 1.776-7.456, P = 0.002) were associated with an increased risk of UF insufficiency. The area under the ROC curve (AUC) for baseline IL-6 AR to predict UF insufficiency was 0.663 (95% CI, 0.580-0.746; P < 0.001). Conclusion Our study suggested that the dialysate IL-6 AR could be a potential predictor of UF insufficiency in patients undergoing PD.
Collapse
Affiliation(s)
- Qianhui Song
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Xiaoxiao Yang
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Yuanyuan Shi
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Hao Yan
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Zanzhe Yu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Zhenyuan Li
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Jiangzi Yuan
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Zhaohui Ni
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Leyi Gu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Wei Fang
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| |
Collapse
|
18
|
Sacnun JM, Herzog R, Kratochwill K. Proteomic study of mesothelial and endothelial cross-talk: key lessons. Expert Rev Proteomics 2022; 19:289-296. [PMID: 36714918 DOI: 10.1080/14789450.2023.2174851] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION The peritoneum, pleura, and pericardium are yet understudied multicellular systems where mesothelial cells (MCs) and endothelial cells (ECs) are in close proximity. Crosstalk between these cell types likely plays role in molecular transport, immunological reactions, and metabolic processes in health, disease, and therapeutic intervention. AREAS COVERED In this review, we discuss recent proteomic efforts to characterize the crosstalk between MC and EC. We describe the proteomic methods necessary for investigation of crosstalk between MC and EC, as well as the in-vitro models that can be employed. Potential experimental approaches range from conditioned medium, via co-culture on semi-permeable membranes, to 3D cell culture based organoid models. While the biological and clinical relevance of the models may increase with their ability to mimic close cell communication, the practicality of these complex experiments corresponds vice versa, making standardization more difficult and expensive. EXPERT OPINION Currently, data and reports on mesothelial-to-endothelial crosstalk are still very scarce. In our opinion, the in-vitro model using semi-permeable cell culture inserts will allow to establish a basic understanding of cellular crosstalk that may occur between those cell types. Later-on, more sophisticated 3D cell cultures may be better able to simulate the transport dynamics within the peritoneal membrane.
Collapse
Affiliation(s)
- Juan Manuel Sacnun
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Rebecca Herzog
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Klaus Kratochwill
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
19
|
Wang J, Sun Q, Wang G, Wang H, Liu H. The effects of blunt snout bream (Megalobrama amblycephala) IL-6 trans-signaling on immunity and iron metabolism via JAK/STAT3 pathway. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 131:104372. [PMID: 35217123 DOI: 10.1016/j.dci.2022.104372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 06/14/2023]
Abstract
Interleukin-6 (IL-6) is a pleiotropic inflammatory cytokine, which plays a dual role in mammalian inflammation through both classical signaling (IL-6 binds to IL-6 receptor/IL-6R) and trans-signaling (IL-6 binds to soluble IL-6R). However, the function of IL-6, especially the regulatory mechanism of IL-6 trans-signaling in immunity and iron metabolism remains largely unclear in teleost. Here, L8824 cells (Ctenopharyngodon idella hepatic cells) were stimulated with blunt snout bream (Megalobrama amblycephala) IL-6 combination with sIL-6R protein (rmaIL-6+rmasIL-6R/maIL-6 trans-signaling) or STAT3 inhibitor (c188-9), and RNA-sequencing, global transcriptional analyses. The enrichment analysis of GO and KEGG showed that maIL-6 trans-signaling is mainly involved in stress and inflammation response, and the activation of STAT3 is mainly related to cell proliferation, apoptosis and immune regulation. Furthermore, after treated L8824 cells with JAK2 inhibitors, it was found that the induction of IL-6 trans-signaling on the selected immune-related genes could be inhibited. These results implied that in early stage after rmaIL-6+rmasIL-6R treatment, the maIL-6 trans-signaling played an important role in the immune regulation through the JAK2/STAT3 pathway. By extending the rmaIL-6+rmasIL-6R treatment time, it was found that maIL-6 trans-signaling could promote the expression of iron metabolism related genes (ft, tf, tfr1, hamp and fpn1) in L8824 cells, indicating that maIL-6 trans-signaling may be involved in iron metabolism in the non-acute immune phase. Finally, after treated L8824 cells with JAK2 and STAT3 inhibitors, it was found that only tf and fpn1 were regulated by maIL-6 trans-signaling through the JAK2/STAT3 pathway. These findings provided novel insights into IL-6 trans-signaling regulatory mechanism in teleost, enriching our knowledge of fish immunity and iron metabolism.
Collapse
Affiliation(s)
- Jixiu Wang
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affair / Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Qianhui Sun
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affair / Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Guowen Wang
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affair / Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Huanling Wang
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affair / Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Hong Liu
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affair / Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China.
| |
Collapse
|
20
|
Fibrosis of Peritoneal Membrane as Target of New Therapies in Peritoneal Dialysis. Int J Mol Sci 2022; 23:ijms23094831. [PMID: 35563220 PMCID: PMC9102299 DOI: 10.3390/ijms23094831] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 01/27/2023] Open
Abstract
Peritoneal dialysis (PD) is an efficient renal replacement therapy for patients with end-stage renal disease. Even if it ensures an outcome equivalent to hemodialysis and a better quality of life, in the long-term, PD is associated with the development of peritoneal fibrosis and the consequents patient morbidity and PD technique failure. This unfavorable effect is mostly due to the bio-incompatibility of PD solution (mainly based on high glucose concentration). In the present review, we described the mechanisms and the signaling pathway that governs peritoneal fibrosis, epithelial to mesenchymal transition of mesothelial cells, and angiogenesis. Lastly, we summarize the present and future strategies for developing more biocompatible PD solutions.
Collapse
|
21
|
PM2.5 Induces Early Epithelial Mesenchymal Transition in Human Proximal Tubular Epithelial Cells through Activation of IL-6/STAT3 Pathway. Int J Mol Sci 2021; 22:ijms222312734. [PMID: 34884542 PMCID: PMC8657854 DOI: 10.3390/ijms222312734] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 11/23/2021] [Indexed: 12/21/2022] Open
Abstract
Particulate matter exposure has been known as a potential risk for the global burden of disease, such as respiratory and cardiovascular diseases. Accumulating evidence suggests that PM2.5 (particulate matter with a diameter less than 2.5 μm) is associated with increased risk of kidney disease, but the mechanisms underlying the renal injury caused by PM2.5 remain to be elucidated. This study investigated the effects of PM2.5 on human proximal tubular epithelial (HK-2) cells by monolayer and 3D spheroid cultures and explored the potential mechanisms. The typical morphology of HK-2 cells showed epithelial–mesenchymal transition (EMT), resulting in reduced adhesion and enhanced migration after PM2.5 exposure, and was accompanied by decreased E-cadherin expression and increased vimentin and α-SMA expressions. Exposure to PM2.5 in the HK-2 cells could lead to an increase in interleukin-6 (IL-6) levels and cause the activation of signal transducer and activator of transcription 3 (STAT3), which is involved in EMT features of HK-2 cells. Furthermore, blocking IL-6/STAT3 signaling by an IL-6 neutralizing antibody or STAT3 inhibitor was sufficient to reverse PM2.5-induced EMT characteristics of the HK-2 cells. Our study suggests that PM2.5 could induce early renal tubule cell injury, contributing to EMT change, and the induction of IL-6/STAT3 pathway may play an important role in this process.
Collapse
|
22
|
Interleukin-17 activates JAK2/STAT3, PI3K/Akt and nuclear factor-κB signaling pathway to promote the tumorigenesis of cervical cancer. Exp Ther Med 2021; 22:1291. [PMID: 34630646 PMCID: PMC8461522 DOI: 10.3892/etm.2021.10726] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 04/27/2021] [Indexed: 12/17/2022] Open
Abstract
Interleukin (IL)-17 has been regarded as a significant factor in inflammation. In addition, IL-17 is known to be involved in the progression of cancers; however, the function of IL-17 in cervical cancer remains unclear. In the present study, cell viability was detected by Cell Counting Kit-8 assay. Quantitative PCR and western blotting were performed to detect gene and protein expression levels, respectively, in cancer cells or tissues. Ki-67 staining was used to evaluate cell proliferation. Wound-healing assay was used to detect cell migration. Moreover, Transwell assay was performed to investigate the invasion of cervical cancer cells. The results revealed that IL-17 significantly promoted the proliferation of cervical cancer cells. Additionally, IL-17 notably enhanced the migration and invasion of cervical cancer cells in vitro. IL-17 promoted the progression of cervical cancer via the activation of JAK2/STAT3 and PI3K/Akt/NF-κB signaling. In conclusion, IL-17 was a key regulator during the progression of cervical cancer through the JAK2/STAT3 and PI3K/Akt/nuclear factor-κB signaling pathway, which may serve as a novel target for the treatment of cervical cancer.
Collapse
|
23
|
Liang Y, Yan B, Meng Z, Xie M, Liang Z, Zhu Z, Meng Y, Ma J, Ma B, Yao X, Luo J. Comparison of Inflammatory and Angiogenic Factors in the Aqueous Humor of Vitrectomized and Non-Vitrectomized Eyes in Diabetic Macular Edema Patients. Front Med (Lausanne) 2021; 8:699254. [PMID: 34568366 PMCID: PMC8455809 DOI: 10.3389/fmed.2021.699254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/09/2021] [Indexed: 12/03/2022] Open
Abstract
Objectives: To compare the aqueous concentrations of inflammatory and angiogenetic factors in vitrectomized vs. non-vitrectomized eyes with diabetic macular edema (DME). Methods: Aqueous samples were obtained from 107 eyes with DME before intravitreal injection of anti-VEGF, 36 eyes with previous pars plana vitrectomy (PPV) combined with pan-retinal endolaser photocoagulation (PRP), and 71 treatment-naïve. Interleukin (IL)-6, IL-8, interferon-induced protein (IP)-10, monocyte chemoattractant protein (MCP)-1, and vascular endothelial growth factor (VEGF) were measured by cytometric bead array (CBA). Optical coherence tomography (OCT) was used for measuring central retinal thickness (CRT). Results: IL-6, IL-8, IP-10, and MCP-1 in aqueous humor of DME vitrectomized eyes were significantly higher than in non-vitrectomized DME eyes, while VEGF was lower than in non-vitrectomized DME eyes. VEGF in aqueous humor significantly correlated with CRT for DME in non-vitrectomized DME eyes. IL-6, IL-8, IP-10, and MCP-1 in aqueous humor were not significantly associated with VEGF for DME in vitrectomized eyes. Conclusions: Inflammation might play an important role in the pathogenesis of DME in vitrectomized eyes. Moreover, inflammation might play a central role in the development of DME via the VEGF-independent pathway. Thus, anti-inflammatory therapy might be a strategy for DME in vitrectomized eyes.
Collapse
Affiliation(s)
- Youling Liang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bin Yan
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhishang Meng
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Manyun Xie
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhou Liang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ziyi Zhu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yongan Meng
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiayue Ma
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bosheng Ma
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxi Yao
- Shenzhen College of International Education, Shenzhen, China
| | - Jing Luo
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
24
|
Yang X, Bao M, Fang Y, Yu X, Ji J, Ding X. STAT3/HIF-1α signaling activation mediates peritoneal fibrosis induced by high glucose. J Transl Med 2021; 19:283. [PMID: 34193173 PMCID: PMC8246671 DOI: 10.1186/s12967-021-02946-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/17/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) of mesothelial cells is a key step in the peritoneal fibrosis (PF). Recent evidence indicates that signal transducer and activator of transcription 3 (STAT3) might mediate the process of renal fibrosis, which could induce the expression of hypoxia-inducible factor-1α (HIF-1α). Here, we investigated the effect of STAT3 activation on HIF-1α expression and the EMT of mesothelial cells, furthermore the role of pharmacological blockade of STAT3 in the process of PF during peritoneal dialysis (PD) treatment. METHODS Firstly, we investigated the STAT3 signaling in human peritoneal mesothelial cells (HPMCs) from drained PD effluent. Secondly, we explored the effect of STAT3 signaling activation on the EMT and the expression of HIF-1α in human mesothelial cells (Met-5A) induced by high glucose. Finally, peritoneal fibrosis was induced by daily intraperitoneal injection with peritoneal dialysis fluid (PDF) so as to explore the role of pharmacological blockade of STAT3 in this process. RESULTS Compared with the new PD patient, the level of phosphorylated STAT3 was up-regulated in peritoneal mesothelial cells from long-term PD patients. High glucose (60 mmol/L) induced over-expression of Collagen I, Fibronectin, α-SMA and reduced the expression of E-cadherin in Met-5A cells, which could be abrogated by STAT3 inhibitor S3I-201 pretreatment as well as by siRNA for STAT3. Furthermore, high glucose-mediated STAT3 activation in mesothelial cells induced the expression of HIF-1α and the profibrotic effect of STAT3 signaling was alleviated by siRNA for HIF-1α. Daily intraperitoneal injection of high-glucose based dialysis fluid (HG-PDF) induced peritoneal fibrosis in the mice, accompanied by the phosphorylation of STAT3. Immunostaining showed that phosphorylated STAT3 was expressed mostly in α-SMA positive cells in the peritoneal membrane induced by HG-PDF. Administration of S3I-201 prevented the progression of peritoneal fibrosis, angiogenesis, macrophage infiltration as well as the expression of HIF-1α in the peritoneal membrane induced by high glucose. CONCLUSIONS Taken together, these findings identified a novel mechanism linking STAT3/HIF-1α signaling to peritoneal fibrosis during long-term PD treatment. It provided the first evidence that pharmacological inhibition of STAT3 signaling attenuated high glucose-mediated mesothelial cells EMT as well as peritoneal fibrosis.
Collapse
Affiliation(s)
- Xiaoxiao Yang
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, People's Republic of China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Manchen Bao
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, People's Republic of China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Yi Fang
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, People's Republic of China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Xiaofang Yu
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, People's Republic of China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Jun Ji
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, People's Republic of China. .,Shanghai Medical Center of Kidney, Shanghai, China. .,Shanghai Institute of Kidney and Dialysis, Shanghai, China. .,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China. .,Hemodialysis Quality Control Center of Shanghai, Shanghai, China.
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, People's Republic of China. .,Shanghai Medical Center of Kidney, Shanghai, China. .,Shanghai Institute of Kidney and Dialysis, Shanghai, China. .,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China. .,Hemodialysis Quality Control Center of Shanghai, Shanghai, China.
| |
Collapse
|
25
|
Masola V, Bonomini M, Onisto M, Ferraro PM, Arduini A, Gambaro G. Biological Effects of XyloCore, a Glucose Sparing PD Solution, on Mesothelial Cells: Focus on Mesothelial-Mesenchymal Transition, Inflammation and Angiogenesis. Nutrients 2021; 13:2282. [PMID: 34209455 PMCID: PMC8308380 DOI: 10.3390/nu13072282] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022] Open
Abstract
Glucose-based solutions remain the most used osmotic agents in peritoneal dialysis (PD), but unavoidably they contribute to the loss of peritoneal filtration capacity. Here, we evaluated at a molecular level the effects of XyloCore, a new PD solution with a low glucose content, in mesothelial and endothelial cells. Cell viability, integrity of mesothelial and endothelial cell membrane, activation of mesothelial and endothelial to mesenchymal transition programs, inflammation, and angiogenesis were evaluated by several techniques. Results showed that XyloCore preserves mesothelial and endothelial cell viability and membrane integrity. Moreover XyloCore, unlike glucose-based solutions, does not exert pro-fibrotic, -inflammatory, and -angiogenic effects. Overall, the in vitro evidence suggests that XyloCore could represent a potential biocompatible solution promising better outcomes in clinical practice.
Collapse
Affiliation(s)
- Valentina Masola
- Division of Nephrology and Dialysis, Department of Medicine, Piazzale A. Stefani 1, 37126 Verona, Italy;
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121 Padova, Italy;
| | - Mario Bonomini
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS.Annunziata Hospital, Via dei Vestini, 66013 Chieti, Italy;
| | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121 Padova, Italy;
| | - Pietro Manuel Ferraro
- U.O.S. Terapia Conservativa della Malattia Renale Cronica, U.O.C. Nefrologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00178 Rome, Italy;
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00178 Rome, Italy
| | - Arduino Arduini
- R&D Department, Iperboreal Pharma Srl, 65122 Pescara, Italy;
| | - Giovanni Gambaro
- Division of Nephrology and Dialysis, Department of Medicine, Piazzale A. Stefani 1, 37126 Verona, Italy;
| |
Collapse
|
26
|
Shi Y, Ni J, Tao M, Ma X, Wang Y, Zang X, Hu Y, Qiu A, Zhuang S, Liu N. Elevated expression of HDAC6 in clinical peritoneal dialysis patients and its pathogenic role on peritoneal angiogenesis. Ren Fail 2021; 42:890-901. [PMID: 32862739 PMCID: PMC7472510 DOI: 10.1080/0886022x.2020.1811119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Peritoneal dialysis (PD) is an important renal replacement therapy for end-stage renal disease (ESRD) patients. However, its complications, such as peritoneal fibrosis (PF) and angiogenesis can cause ultrafiltration failure and PD termination. Histone deacetylase 6 (HDAC6) has been demonstrated to be involved in PF. However, its underlying role in peritoneal angiogenesis is still unknown and clinical value needs to be explored. In this study, we analyzed the expression of HDAC6 in the peritoneum from patients with non-PD and PD-related peritonitis and dialysis effluent from stable PD patients. Our study revealed that HDAC6 expressed highly in the peritoneum with peritonitis and co-stained with α-smooth muscle actin (α-SMA), a biomarker of the myofibroblast. And the level of HDAC6 in the dialysate increased with time and positively correlated with transforming growth factor-β1 (TGF-β1), interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF), and negatively with cancer antigen 125 (CA125). In vitro, blockading HDAC6 with a selective inhibitor tubastatin A (TA) or silencing HDAC6 with a small interfering RNA (siRNA) prominently decreased IL-6-stimulated VEGF expression in cultured human peritoneal mesothelial cells (HPMCs), and inhibited proliferation and vasoformation of human umbilical vein endothelial cells (HUVECs). TA or HDAC6 siRNA also suppressed the expression of Wnt1, β-catenin, and the phosphorylation of STAT3 in IL-6-treated HPMCs. In summary, HDAC6 inhibition protects against PD-induced angiogenesis through suppression of IL-6/STAT3 and Wnt1/β-catenin signaling pathway, subsequently reducing the VEGF production and angiogenesis. It could become a new therapeutic target or forecast biomarker for PF, inflammation, and angiogenesis in the future.
Collapse
Affiliation(s)
- Yingfeng Shi
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun Ni
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Tao
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoyan Ma
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Nephrology, Baoshan Branch of Shanghai First People's Hospital, Shanghai, China
| | - Yi Wang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiujuan Zang
- Department of Nephrology, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Yan Hu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Andong Qiu
- School of Life Science and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai, China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, USA
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
27
|
Shi Y, Hu Y, Cui B, Zhuang S, Liu N. Vascular endothelial growth factor-mediated peritoneal neoangiogenesis in peritoneal dialysis. Perit Dial Int 2021; 42:25-38. [PMID: 33823711 DOI: 10.1177/08968608211004683] [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] [Indexed: 12/16/2022] Open
Abstract
Peritoneal dialysis (PD) is an important renal replacement therapy for patients with end-stage renal diseases, which is limited by peritoneal neoangiogenesis leading to ultrafiltration failure (UFF). Vascular endothelial growth factor (VEGF) and its receptors are key angiogenic factors involved in almost every step of peritoneal neoangiogenesis. Impaired mesothelial cells are the major sources of VEGF in the peritoneum. The expression of VEGF will be up-regulated in specific pathological conditions in PD patients, such as with non-biocompatible peritoneal dialysate, uremia and inflammation, and so on. Other working cells (i.e. vascular endothelial cells, macrophages and adipocytes) can also stimulate the secretion of VEGF. Meanwhile, hypoxia and activation of complement system further aggravate peritoneal injury and contribute to neoangiogenesis. There are several signalling pathways participating in VEGF-mediated peritoneal neoangiogenesis including tumour growth factor-β, Wnt/β-catenin, Notch and interleukin-6/signal transducer and activator of transcription 3. Moreover, VEGF is highly expressed in dialysate effluent of long-term PD patients and is associated with peritoneal transport function, which supports its role in the alteration of peritoneal structure and function. In this review, we systematically summarize the angiogenic effect of VEGF and evaluate it as a potential target for the prevention of peritoneal neoangiogenesis and UFF. Preservation of the peritoneal membrane using targeted therapy of VEGF-mediated peritoneal neoangiogenesis may increase the longevity of the PD modality for those who require life-long dialysis.
Collapse
Affiliation(s)
- Yingfeng Shi
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yan Hu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Binbin Cui
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, USA
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Zhou L, Xing C, Zhou D, Yang R, Cai M. Downregulation of lncRNA FGF12-AS2 suppresses the tumorigenesis of NSCLC via sponging miR-188-3p. Open Med (Wars) 2020; 15:986-996. [PMID: 33344773 PMCID: PMC7724005 DOI: 10.1515/med-2020-0219] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Background Non-small-cell lung carcinoma (NSCLC) seriously threatens the health of human beings. Aberrant expression of lncRNAs has been confirmed to be related with the progression of multiple malignant tumors, including NSCLC. LncRNA FGF12-AS2 has been considered to be upregulated in NSCLC. However, the mechanism by which FGF12-AS2 promotes the tumorigenesis of NSCLC remains elusive. Methods Gene and protein expressions in NSCLC cells were measured by q-PCR and western blot, respectively. CCK-8 and immunofluorescence staining were performed to detect the cell proliferation. Cell apoptosis was tested by flow cytometry. Transwell assay was used to detect the cell migration and invasion. Finally, the dual luciferase report assay was used to verify the relation among FGF12-AS2, miR-188-3p, and NCAPG2. Results Downregulation of FGF12-AS2 significantly inhibited the proliferation of NSCLC cells via inducing apoptosis. In addition, FGF12-AS2 silencing notably suppressed the migration and invasion of A549 cells. Meanwhile, FGF12-AS2 modulated the progression of NSCLC via regulation of miR-188-3p/NCAPG2 axis. Finally, knockdown of FGF12-AS2 inhibited the tumorigenesis of NSCLC via suppressing the EMT process of NSCLC. Conclusion Downregulation of lncRNA FGF12-AS2 suppressed the tumorigenesis of NSCLC via sponging miR-188-3p. Thus, FGF12-AS2 may serve as a potential target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Lili Zhou
- Department of Oncology, Yancheng Second People’s Hospital, No. 135 Kaifang Avenue, Yancheng 224003, Jiangsu, China
| | - Chen Xing
- Department of Oncology, Yancheng Second People’s Hospital, No. 135 Kaifang Avenue, Yancheng 224003, Jiangsu, China
| | - Dongxia Zhou
- Department of Oncology, Yancheng Second People’s Hospital, No. 135 Kaifang Avenue, Yancheng 224003, Jiangsu, China
| | - Rong Yang
- Department of Oncology, Yancheng Second People’s Hospital, No. 135 Kaifang Avenue, Yancheng 224003, Jiangsu, China
| | - Maohuai Cai
- Department of Oncology, Yancheng Second People’s Hospital, No. 135 Kaifang Avenue, Yancheng 224003, Jiangsu, China
| |
Collapse
|
29
|
Angiopoietin-2 is released during anaphylactic hypotension in anesthetized and unanesthetized rats. PLoS One 2020; 15:e0242026. [PMID: 33201925 PMCID: PMC7671552 DOI: 10.1371/journal.pone.0242026] [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: 04/14/2020] [Accepted: 10/26/2020] [Indexed: 11/19/2022] Open
Abstract
Angiopoietin (Angpt)-2, a permeability-increasing growth factor, is involved in vascular leakage of sepsis and acute lung injury, and could be released from endothelium in response to anaphylaxis-related secretagogues such as histamine and leukotrienes, or cytokines. However, roles of Angpt-2 in the hyperpermeability during systemic anaphylaxis are not known. Thus, we determined plasma levels of Angpt-2 and cytokines and vascular permeability during anaphylactic hypotension in unanesthetized rats. Anaphylaxis was induced by an intravenous injection of ovalbumin antigen. Mean arterial blood pressure (MBP) was measured, and hematocrit (Hct) and plasma levels of Angpt-2 and cytokines were assessed for 24 h after antigen injection. Separately, vascular permeability was measured in various organs using the Evans blue dye method, and Angpt-2 mRNA expression in liver was measured. After antigen injection, MBP decreased to the nadir at 6 min, and returned to baseline at 45 min, and Hct peaked at 20 min and thereafter progressively declined, suggesting that vascular leak and hypotension occurred within 20 min. Plasma Angpt-2 levels began to increase significantly at 1 h after antigen, reaching the peak 2.7-fold baseline at 6 h with a return to baseline at 24 h. Detected cytokines of IL-1α, IL-1β, IL-6, IL-10, and TNF-α peaked 1 or 2 h after antigen. Angpt-2 mRNA increased at 2 h and showed an increasing tendency at 6 h. Vascular permeability in bronchus, trachea, intestines, mesentery and skeletal muscle was increased at 10 min but not at 6 h after antigen. In addition, we confirmed using anesthetized rat anaphylaxis models that plasma Angpt-2 levels increased at 1 h after antigen. In conclusion, plasma Angpt-2 is elevated presumably due to increased cytokines and enhanced gene transcription during anaphylaxis in anesthetized and unanesthetized rats.
Collapse
|
30
|
Balzer MS. Molecular pathways in peritoneal fibrosis. Cell Signal 2020; 75:109778. [PMID: 32926960 DOI: 10.1016/j.cellsig.2020.109778] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 01/02/2023]
Abstract
Peritoneal dialysis (PD) is a renal replacement therapy for patients with end-stage renal disease that is equivalent to hemodialysis with respect to adequacy, mortality, and other outcome parameters, yet providing superior quality-of-life measures and cost savings. However, long-term usage of the patient's peritoneal membrane as a dialyzer filter is unphysiological and leads to peritoneal fibrosis, which is a major factor of patient morbidity and PD technique failure, resulting in a transfer to hemodialysis or death. Peritoneal fibrosis pathophysiology involves chronic inflammation and the fibrotic process itself. Frequently, inflammation precedes membrane fibrosis development, although a bidirectional relationship of one inducing the other exists. This review aims at highlighting the histopathological definition of peritoneal fibrosis, outlining the interplay of fibrosis, angiogenesis and epithelial-to-mesenchymal transition (EMT), delineating important fibrogenic pathways involving Smad-dependent and Smad-independent transforming growth factor-β (TGF-β) as well as connective tissue growth factor (CTGF) signaling, and summarizing historic and recent studies of inflammatory pathways involving NOD-like receptor protein 3 (NLRP3)/interleukin (IL)-1β, IL-6, IL-17, and other cytokines.
Collapse
Affiliation(s)
- Michael S Balzer
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany.
| |
Collapse
|