1
|
Qian Z, Zhang X, Huang J, Niu X, Zhu C, Tai Z, Zhu Q, Chen Z, Zhu T, Wu G. ROS-responsive MSC-derived Exosome Mimetics Carrying MHY1485 Alleviate Renal Ischemia Reperfusion Injury through Multiple Mechanisms. ACS OMEGA 2024; 9:24853-24863. [PMID: 38882096 PMCID: PMC11170644 DOI: 10.1021/acsomega.4c01624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 06/18/2024]
Abstract
Renal ischemia reperfusion (IR) injury is a prevalent inflammatory nephropathy in surgeries such as renal transplantation or partial nephrectomy, damaging renal function through inducing inflammation and cell death in renal tubules. Mesenchymal stromal/stem cell (MSC)-based therapies, common treatments to attenuate inflammation in IR diseases, fail to exhibit satisfying effects on cell death in renal IR. In this study, we prepared MSC-derived exosome mimetics (EMs) carrying the mammalian target of the rapamycin (mTOR) agonist to protect kidneys in proinflammatory environments under IR conditions. The thioketal-modified EMs carried the mTOR agonist and bioactive molecules in MSCs and responsively released them in kidney IR areas. MSC-derived EMs and mTOR agonists protected kidneys synergistically from IR through alleviating inflammation, apoptosis, and ferroptosis. The current study indicates that MSC-TK-MHY1485 EMs (MTM-EM) are promising therapeutic biomaterials for renal IR injury.
Collapse
Affiliation(s)
- Zhiyu Qian
- Department of Urology, Zhongshan Hospital Fudan University, 170 Fenglin Road, Shanghai 200030, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200030, China
| | - Xinyue Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China
| | - Jiahua Huang
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai 201500, China
| | - Xinhao Niu
- Department of Urology, Zhongshan Hospital Fudan University, 170 Fenglin Road, Shanghai 200030, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200030, China
| | - Cuisong Zhu
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai 201500, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital Fudan University, 170 Fenglin Road, Shanghai 200030, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200030, China
| | - Guoyi Wu
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai 201500, China
| |
Collapse
|
2
|
Saiz ML, Lozano-Chamizo L, Florez AB, Marciello M, Diaz-Bulnes P, Corte-Iglesias V, Bernet CR, Rodrigues-Diez RR, Martin-Martin C, Rodriguez-Santamaria M, Fernandez-Vega I, Rodriguez RM, Diaz-Corte C, Suarez-Alvarez B, Filice M, Lopez-Larrea C. BET inhibitor nanotherapy halts kidney damage and reduces chronic kidney disease progression after ischemia-reperfusion injury. Biomed Pharmacother 2024; 174:116492. [PMID: 38537579 DOI: 10.1016/j.biopha.2024.116492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 05/01/2024] Open
Abstract
Targeting epigenetic mechanisms has emerged as a potential therapeutic approach for the treatment of kidney diseases. Specifically, inhibiting the bromodomain and extra-terminal (BET) domain proteins using the small molecule inhibitor JQ1 has shown promise in preclinical models of acute kidney injury (AKI) and chronic kidney disease (CKD). However, its clinical translation faces challenges due to issues with poor pharmacokinetics and side effects. Here, we developed engineered liposomes loaded with JQ1 with the aim of enhancing kidney drug delivery and reducing the required minimum effective dose by leveraging cargo protection. These liposomes efficiently encapsulated JQ1 in both the membrane and core, demonstrating superior therapeutic efficacy compared to freely delivered JQ1 in a mouse model of kidney ischemia-reperfusion injury. JQ1-loaded liposomes (JQ1-NPs) effectively targeted the kidneys and only one administration, one-hour after injury, was enough to decrease the immune cell (neutrophils and monocytes) infiltration to the kidney-an early and pivotal step to prevent damage progression. By inhibiting BRD4, JQ1-NPs suppress the transcription of pro-inflammatory genes, such as cytokines (il-6) and chemokines (ccl2, ccl5). This success not only improved early the kidney function, as evidenced by decreased serum levels of BUN and creatinine in JQ1-NPs-treated mice, along with reduced tissue expression of the damage marker, NGAL, but also halted the production of extracellular matrix proteins (Fsp-1, Fn-1, α-SMA and Col1a1) and the fibrosis development. In summary, this work presents a promising nanotherapeutic strategy for AKI treatment and its progression and provides new insights into renal drug delivery.
Collapse
Affiliation(s)
- Maria Laura Saiz
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain
| | - Laura Lozano-Chamizo
- Nanobiotechnology for Life Sciences Laboratory, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, Madrid E-28040, Spain; Microscopy and Dynamic Imaging Unit, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Calle Melchor Fernández Almagro 3, Madrid E-28029, Spain; Atrys Health, Madrid E-28001, Spain
| | - Aida Bernardo Florez
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain
| | - Marzia Marciello
- Nanobiotechnology for Life Sciences Laboratory, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, Madrid E-28040, Spain; Microscopy and Dynamic Imaging Unit, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Calle Melchor Fernández Almagro 3, Madrid E-28029, Spain
| | - Paula Diaz-Bulnes
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain
| | - Viviana Corte-Iglesias
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain; Department of Immunology, Hospital Universitario Central de Asturias, Oviedo 33011, Spain
| | - Cristian Ruiz Bernet
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain
| | - Raul R Rodrigues-Diez
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain
| | - Cristina Martin-Martin
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain
| | - Mar Rodriguez-Santamaria
- Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain
| | - Ivan Fernandez-Vega
- Department of Pathology, Hospital Universitario Central de Asturias, Oviedo 33001, Spain; Biobank of Principality of Asturias, Oviedo 33011, Spain
| | - Ramon M Rodriguez
- Lipids in Human Pathology, Institut d'Investigació Sanitària Illes Balears (IdISBa), Ctra. Valldemossa 79, Palma, Balearic Islands E-07120, Spain; Research Unit, University Hospital Son Espases, Ctra. Valldemossa79, Palma, Balearic Islands E-07120, Spain
| | - Carmen Diaz-Corte
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; Department of Nephrology, Hospital Universitario Central de Asturias, Oviedo 33001, Spain
| | - Beatriz Suarez-Alvarez
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain.
| | - Marco Filice
- Nanobiotechnology for Life Sciences Laboratory, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, Madrid E-28040, Spain; Microscopy and Dynamic Imaging Unit, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Calle Melchor Fernández Almagro 3, Madrid E-28029, Spain.
| | - Carlos Lopez-Larrea
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain; Department of Immunology, Hospital Universitario Central de Asturias, Oviedo 33011, Spain
| |
Collapse
|
3
|
El-Aziz Fathy EA, Abdel-Gaber SAW, Gaber Ibrahim MF, Thabet K, Waz S. Downregulation of IL-1β/p38 mitogen activated protein kinase pathway by diacerein protects against kidney ischemia/reperfusion injury in rats. Cytokine 2024; 176:156511. [PMID: 38290257 DOI: 10.1016/j.cyto.2024.156511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/08/2023] [Accepted: 01/16/2024] [Indexed: 02/01/2024]
Abstract
Renal ischemia-reperfusion (I/R) can be precipitated by multiple clinical situations that lead to impaired renal function and associated mortality. The resulting tubular cell damage is the outcome of complex disorders including, an inflammatory process with an overproduction of cytokines. Here, diacerein (DIA), an inhibitor of proinflammatory cytokine interleukin-1 beta (IL-1β), was investigated against renal I/R in rats. DIA was orally administrated (50 mg/kg/day) for ten days before bilateral ischemia for 45 min with subsequent 2 hr. reperfusion. Interestingly, DIA alleviated the renal dysfunction and histopathological damage in the renal tissues. Pretreatment with DIA corrected the oxidative imbalance by prevented reduction in antioxidant levels of GSH and SOD, while it decreased the elevation of the oxidative marker, MDA. In addition, DIA downregulated IL-1β and TNF-α expression in the renal tissues. Consequent to inhibition of the oxidative stress and inflammatory cascades, DIA inhibited the phosphorylation of p38 mitogen-activated protein kinase (p38 MAPK). Therefore, downstream targets for p38 MAPK were also inhibited via DIA which prevented further increases of inflammatory cytokines and the apoptotic marker, caspase-3. Collectively, this study revealed the renoprotective role of DIA for renal I/R and highlighted the role of p38 MAPK encountered in its therapeutic application in renal disease.
Collapse
Affiliation(s)
- Eman Abd El-Aziz Fathy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, El-Minia 61511, Egypt.
| | | | - Manar Fouli Gaber Ibrahim
- Department of Histology and Cell Biology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt.
| | - Khaled Thabet
- Department of Biochemistry, Faculty of Pharmacy, Minia University, El-Minia 61511, Egypt.
| | - Shaimaa Waz
- Department of Biochemistry, Faculty of Pharmacy, Minia University, El-Minia 61511, Egypt.
| |
Collapse
|
4
|
Liu X, Hu J, Liao G, Liu D, Zhou S, Zhang J, Liao J, Guo Z, Li Y, Yang S, Li S, Chen H, Guo Y, Li M, Fan L, Li L, Zhao M, Liu Y. The role of regulatory T cells in the pathogenesis of acute kidney injury. J Cell Mol Med 2023; 27:3202-3212. [PMID: 37667551 PMCID: PMC10568672 DOI: 10.1111/jcmm.17771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 09/06/2023] Open
Abstract
The incidence of acute kidney injury (AKI) is on the rise and is associated with high mortality; however, there are currently few effective treatments. Moreover, the relationship between Tregs and other components of the immune microenvironment (IME) in the pathogenesis of AKI remains unclear. We downloaded four publicly accessible AKI datasets, GSE61739, GSE67401, GSE19130, GSE81741, GSE19288 and GSE106993 from the gene expression omnibus (GEO) database. Additionally, we gathered two kidney single-cell sequencing (scRNA-seq) samples from the Department of Organ Transplantation at Zhujiang Hospital of Southern Medical University to investigate chronic kidney transplant rejection (CKTR). Moreover, we also collected three samples of normal kidney tissue from GSE131685. By analysing the differences in immune cells between the AKI and Non-AKI groups, we discovered that the Non-AKI group contained a significantly greater number of Tregs than the AKI group. Additionally, the activation of signalling pathways, such as inflammatory molecules secretion, immune response, glycolytic metabolism, NOTCH, FGF, NF-κB and TLR4, was significantly greater in the AKI group than in the Non-AKI group. Additionally, analysis of single-cell sequencing data revealed that Tregs in patients with chronic kidney rejection and in normal kidney tissue have distinct biology, including immune activation, cytokine production, and activation fractions of signalling pathways such as NOTCH and TLR4. In this study, we found significant differences in the IME between AKI and Non-AKI, including differences in Tregs cells and activation levels of biologically significant signalling pathways. Tregs were associated with lower activity of signalling pathways such as inflammatory response, inflammatory molecule secretion, immune activation, glycolysis.
Collapse
Affiliation(s)
- Xiaoyou Liu
- Department of Organ transplantationThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Jianmin Hu
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| | - Guorong Liao
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| | - Ding Liu
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| | - Song Zhou
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| | - Jie Zhang
- Department of Organ transplantationThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Jun Liao
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| | - Zefeng Guo
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| | - Yuzhu Li
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| | - Siqiang Yang
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| | - Shichao Li
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| | - Hua Chen
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| | - Ying Guo
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| | - Min Li
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| | - Lipei Fan
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| | - Liuyang Li
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| | - Ming Zhao
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| | - Yongguang Liu
- Department of Organ transplantationZhujiang Hospital of the Southern Medical UniversityGuangzhouChina
| |
Collapse
|
5
|
Zhao X, Li Y, Wu S, Wang Y, Liu B, Zhou H, Li F. Role of extracellular vesicles in pathogenesis and therapy of renal ischemia-reperfusion injury. Biomed Pharmacother 2023; 165:115229. [PMID: 37506581 DOI: 10.1016/j.biopha.2023.115229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023] Open
Abstract
Renal ischemia-reperfusion injury (RIRI) is a complex disorder characterized by both intrinsic damage to renal tubular epithelial cells and extrinsic inflammation mediated by cytokines and immune cells. Unfortunately, there is no cure for this devastating condition. Extracellular vesicles (EVs) are nanosized membrane-bound vesicles secreted by various cell types that can transfer bioactive molecules to target cells and modulate their function. EVs have emerged as promising candidates for cell-free therapy of RIRI, owing to their ability to cross biological barriers and deliver protective signals to injured renal cells. In this review, we provide an overview of EVs, focusing on their functional role in RIRI and the signaling messengers responsible for EV-mediated crosstalk between various cell types in renal tissue. We also discuss the renoprotective role of EVs and their use as therapeutic agents for RIRI, highlighting the advantages and challenges encountered in the therapeutic application of EVs in renal disease.
Collapse
Affiliation(s)
- Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Shouwang Wu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yuxiong Wang
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
6
|
Zhang T, Li Y, Wise AF, Kulkarni K, Aguilar MI, Samuel CS, Del Borgo M, Widdop RE, Ricardo SD. The protective effects of a novel AT 2 receptor agonist, β-Pro 7Ang III in ischemia-reperfusion kidney injury. Biomed Pharmacother 2023; 161:114556. [PMID: 36948137 DOI: 10.1016/j.biopha.2023.114556] [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: 01/29/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/24/2023] Open
Abstract
BACKGROUND AND PURPOSE This study investigated the reno-protective effects of a highly selective AT2R agonist peptide, β-Pro7Ang III in a mouse model of acute kidney injury (AKI). METHODS C57BL/6 J mice underwent either sham surgery or unilateral kidney ischemia-reperfusion injury (IRI) for 40 min. IRI mice were treated with either β-Pro7Ang III or perindopril and at 7 days post-surgery the kidneys analysed for histopathology and the development of fibrosis and matrix metalloproteinase (MMP)-2 and -9 activity. The association of the therapeutic effects of β-Pro7Ang III with macrophage number and phenotype was determined in vivo and in vitro. KEY RESULTS Decreased kidney tubular injury, interstitial matrix expansion and reduced interstitial immune cell infiltration in IRI mice receiving β-Pro7Ang III treatment was observed at day 7, compared to IRI mice without treatment. This correlated to reduced collagen accumulation and MMP-2 activity in IRI mice following β-Pro7Ang III treatment. FACS analysis showed a reduced number and proportion of CD45+CD11b+F4/80+ macrophages in IRI kidneys in response to β-Pro7Ang III, correlating with a significant increase in M2 macrophage markers and decreased M1 markers at day 3 and 7 post-IR injury, respectively. In vitro analysis of cultured THP-1 cells showed that β-Pro7Ang III attenuated lipopolysaccharide (LPS)-induced tumour necrosis factor-α (TNF-α) and interleukin (IL)- 6 production but increased IL-10 secretion, compared to LPS alone. CONCLUSION Administration of β-Pro7Ang III via mini-pump improved kidney structure and reduced interstitial collagen accumulation, in parallel with an alteration of macrophage phenotype and anti-inflammatory cytokine release, therefore mitigating the downstream progression of ischemic AKI.
Collapse
Affiliation(s)
- Tingfang Zhang
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Yifang Li
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Andrea F Wise
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Ketav Kulkarni
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Chrishan S Samuel
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Mark Del Borgo
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Robert E Widdop
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Sharon D Ricardo
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
7
|
Haga S, Kanno A, Morita N, Jin S, Matoba K, Ozawa T, Ozaki M. Poly(ADP-ribose) Polymerase (PARP) is Critically Involved in Liver Ischemia/reperfusion-injury. J Surg Res 2021; 270:124-138. [PMID: 34656890 DOI: 10.1016/j.jss.2021.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 08/27/2021] [Accepted: 09/14/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Poly(ADP-ribose) polymerase (PARP) is a DNA-repairing enzyme activated by extreme genomic stress, and therefore is potently activated in the remnant liver suffering from ischemia after surgical resection. However, the impact of PARP on post-ischemic liver injury has not been elucidated yet. MATERIALS AND METHODS We investigated the impact of PARP on murine hepatocyte/liver injury induced by hypoxia/ischemia, respectively. RESULTS PJ34, a specific inhibitor of PARP, markedly protected against hypoxia/reoxygenation (H/R)-induced cell death, though z-VAD-fmk, a pan-caspase inhibitor similarly showed the protective effect. PJ34 did not affect H/R-induced caspase activity or caspase-mediated cell death. z-VAD-fmk also did not affect the production of PAR (i.e., PARP activity). Therefore, PARP- and caspase-mediated cell death occurred in a mechanism independent of each other in H/R. H/R immediately induced activation of PARP and cell death afterwards, both of which were suppressed by PJ34 or Trolox, an antioxidant. This suggests that H/R-induced cell death occurred redox-dependently through PARP activation. H/R and OS induced nuclear translocation of apoptosis inducing factor (AIF, a marker of parthanatos) and RIP1-RIP3 interaction (a marker of necroptosis), both of which were suppressed by PJ34. H/R induced PARP-mediated parthanatos and necroptosis redox-dependently. In mouse experiments, PJ34 significantly reduced serum levels of AST, ALT & LDH and areas of hepatic necrosis after liver ischemia/reperfusion, similar to z-VAD-fmk or Trolox. CONCLUSION PARP, activated by ischemic damage and/or oxidative stress, may play a critical role in post-ischemic liver injury by inducing programmed necrosis (parthanatos and necroptosis). PARP inhibition may be one of the promising strategies against post-ischemic liver injury.
Collapse
Affiliation(s)
- Sanae Haga
- Department of Biological Response and Regulation, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Akira Kanno
- Department of Environmental Applied Chemistry, University of Toyama, Toyama, Toyama, Japan
| | - Naoki Morita
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Sapporo, Hokkaido, Japan
| | - Shigeki Jin
- Department of Forensic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kotaro Matoba
- Department of Forensic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo, Japan
| | - Michitaka Ozaki
- Department of Biological Response and Regulation, Hokkaido University, Sapporo, Hokkaido, Japan; Laboratory of Molecular and Functional Bio-Imaging, Hokkaido University, Sapporo, Hokkaido, Japan.
| |
Collapse
|
8
|
Lack of gamma delta T cells ameliorates inflammatory response after acute intestinal ischemia reperfusion in mice. Sci Rep 2021; 11:18628. [PMID: 34545104 PMCID: PMC8452610 DOI: 10.1038/s41598-021-96525-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/29/2021] [Indexed: 01/07/2023] Open
Abstract
T-cells have been demonstrated to modulate ischemia–reperfusion injury (IRI) in the kidney, lung, liver, and intestine. Whereas most T-cell subpopulations contribute primarily to the antigen-specific effector and memory phases of immunity, γδ-T-cells combine adaptive features with rapid, innate-like responses that can place them in the initiation phase of immune reactions. Therefore, we aimed to clarify the role of γδ-T-cells in intestinal IRI. Adult wild-type (WT) and γδ-T-cell-deficient mice were subjected to acute intestinal IRI. Gene expression of pro-inflammatory cytokines and influx of leukocyte subpopulations in the gut were assessed by qPCR and flow cytometry. Serum transaminases were measured as an indicator of distant organ IRI. Intestinal IRI led to increased influx of neutrophils, pro-inflammatory cytokine expression and LDH/ALT/AST elevation. Selective deficiency of γδ-T-cells significantly decreased pro-inflammatory cytokine levels and neutrophil infiltration in the gut following IRI compared to controls. Furthermore, γδ-T-cell deficiency resulted in decreased LDH and transaminases levels in sera, indicating amelioration of distant organ injury. Increasing evidence demonstrates a key role of T-cell subpopulations in IRI. We demonstrate that γδ-T-cell deficiency ameliorated pro-inflammatory cytokine production, neutrophil recruitment and distant organ injury. Thus, γδ-T-cells may be considered as mediators contributing to the inflammatory response in the acute phase of intestinal IRI.
Collapse
|
9
|
Battistone MA, Mendelsohn AC, Spallanzani RG, Allegretti AS, Liberman RN, Sesma J, Kalim S, Wall SM, Bonventre JV, Lazarowski ER, Brown D, Breton S. Proinflammatory P2Y14 receptor inhibition protects against ischemic acute kidney injury in mice. J Clin Invest 2020; 130:3734-3749. [PMID: 32287042 PMCID: PMC7324186 DOI: 10.1172/jci134791] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 04/08/2020] [Indexed: 12/24/2022] Open
Abstract
Ischemic acute kidney injury (AKI), a complication that frequently occurs in hospital settings, is often associated with hemodynamic compromise, sepsis, cardiac surgery, or exposure to nephrotoxins. Here, using a murine renal ischemia/reperfusion injury (IRI) model, we show that intercalated cells (ICs) rapidly adopted a proinflammatory phenotype after IRI. Wwe demonstrate that during the early phase of AKI either blockade of the proinflammatory P2Y14 receptor located on the apical membrane of ICs or ablation of the gene encoding the P2Y14 receptor in ICs (a) inhibited IRI-induced increase of chemokine expression in ICs, (b) reduced neutrophil and monocyte renal infiltration, (c) reduced the extent of kidney dysfunction, and (d) attenuated proximal tubule damage. These observations indicate that the P2Y14 receptor participates in the very first inflammatory steps associated with ischemic AKI. In addition, we show that the concentration of the P2Y14 receptor ligand UDP-glucose (UDP-Glc) was higher in urine samples from intensive care unit patients who developed AKI compared with patients without AKI. In particular, we observed a strong correlation between UDP-Glc concentration and the development of AKI in cardiac surgery patients. Our study identifies the UDP-Glc/P2Y14 receptor axis as a potential target for the prevention and/or attenuation of ischemic AKI.
Collapse
Affiliation(s)
- Maria Agustina Battistone
- Program in Membrane Biology, Division of Nephrology, Department of Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alexandra C. Mendelsohn
- Program in Membrane Biology, Division of Nephrology, Department of Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Raul German Spallanzani
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Andrew S. Allegretti
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Rachel N. Liberman
- Program in Membrane Biology, Division of Nephrology, Department of Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Juliana Sesma
- Marsico Lung Institute, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Sahir Kalim
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Susan M. Wall
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Eduardo R. Lazarowski
- Marsico Lung Institute, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Dennis Brown
- Program in Membrane Biology, Division of Nephrology, Department of Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sylvie Breton
- Program in Membrane Biology, Division of Nephrology, Department of Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
10
|
do Valle Duraes F, Lafont A, Beibel M, Martin K, Darribat K, Cuttat R, Waldt A, Naumann U, Wieczorek G, Gaulis S, Pfister S, Mertz KD, Li J, Roma G, Warncke M. Immune cell landscaping reveals a protective role for regulatory T cells during kidney injury and fibrosis. JCI Insight 2020; 5:130651. [PMID: 32051345 DOI: 10.1172/jci.insight.130651] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 01/15/2020] [Indexed: 12/30/2022] Open
Abstract
Acute kidney injury (AKI) and chronic kidney diseases are associated with high mortality and morbidity. Although the underlying mechanisms determining the transition from acute to chronic injury are not completely understood, immune-mediated processes are critical in renal injury. We have performed a comparison of 2 mouse models leading to either kidney regeneration or fibrosis. Using global gene expression profiling we could identify immune-related pathways accounting for the majority of the observed transcriptional changes during fibrosis. Unbiased examination of the immune cell composition, using single-cell RNA sequencing, revealed major changes in tissue-resident macrophages and T cells. Following injury, there was a marked increase in tissue-resident IL-33R+ and IL-2Ra+ regulatory T cells (Tregs). Expansion of this population before injury protected the kidney from injury and fibrosis. Transcriptional profiling of Tregs showed a differential upregulation of regenerative and proangiogenic pathways during regeneration, whereas in the fibrotic environment they expressed markers of hyperactivation and fibrosis. Our data point to a hitherto underappreciated plasticity in Treg function within the same tissue, dictated by environmental cues. Overall, we provide a detailed cellular and molecular characterization of the immunological changes during kidney injury, regeneration, and fibrosis.
Collapse
Affiliation(s)
| | - Armelle Lafont
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Martin Beibel
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Kea Martin
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Katy Darribat
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Rachel Cuttat
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Annick Waldt
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Ulrike Naumann
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | - Swann Gaulis
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Sabina Pfister
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Kirsten D Mertz
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Jianping Li
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Guglielmo Roma
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Max Warncke
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| |
Collapse
|
11
|
Ghayur A, Padwal MK, Liu L, Zhang J, Margetts PJ. SMAD3-dependent and -independent pathways in glomerular injury associated with experimental glomerulonephritis. Am J Physiol Renal Physiol 2019; 317:F152-F162. [PMID: 31141397 DOI: 10.1152/ajprenal.00406.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Glomerulonephritis (GN) is a common cause of end-stage kidney disease and is characterized by glomerular inflammation, hematuria, proteinuria, and progressive renal dysfunction. Transforming growth factor (TGF)-β is involved in glomerulosclerosis and interstitial fibrosis. TGF-β activates multiple signaling pathways, including the canonical SMAD pathway. We evaluated the role of SMAD signaling in renal injury and proteinuria in a murine model of GN. SMAD3+/+ or SMAD3-/- mice received anti-glomerular basement membrane antibodies to induce GN. We confirmed previous reports that demonstrated that SMAD3 is an important mediator of glomerulosclerosis and renal interstitial fibrosis. Proteinuria was highly SMAD3 dependent. We found differential effects of SMAD3 deletion on podocytes and glomerular endothelial cells. GN led to podocyte injury, including foot process effacement and loss of podocyte-specific markers. Interestingly, these changes were not SMAD3 dependent. Furthermore, there were significant changes to glomerular endothelial cells, including loss of fenestrations, swelling, and basement membrane reduplication, which were SMAD3 dependent. Despite ongoing markers of podocyte injury in SMAD3-/- mice, proteinuria was transient. Renal injury in the setting of GN involves TGF-β and SMAD3 signaling. Cell populations within the glomerulus respond differently to SMAD3 deletion. Proteinuria correlated more with endothelial cell changes as opposed to podocyte injury in this model.
Collapse
Affiliation(s)
- Ayesha Ghayur
- Department of Medicine, McMaster University , Hamilton, Ontario , Canada
| | | | - Limin Liu
- Department of Medicine, McMaster University , Hamilton, Ontario , Canada
| | - Jing Zhang
- Department of Medicine, McMaster University , Hamilton, Ontario , Canada
| | - Peter J Margetts
- Department of Medicine, McMaster University , Hamilton, Ontario , Canada
| |
Collapse
|
12
|
Yu Y, Feng X, Vieten G, Dippel S, Imvised T, Gueler F, Ure BM, Kuebler JF, Klemann C. Conventional alpha beta (αβ) T cells do not contribute to acute intestinal ischemia-reperfusion injury in mice. PLoS One 2017; 12:e0181326. [PMID: 28704542 PMCID: PMC5509314 DOI: 10.1371/journal.pone.0181326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 06/29/2017] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Ischemia-reperfusion injury (IRI) is associated with significant patient mortality and morbidity. The complex cascade of IRI is incompletely understood, but inflammation is known to be a key mediator. In addition to the predominant innate immune responses, previous research has also indicated that αβ T cells contribute to IRI in various organ models. The aim of this study was to clarify the role αβ T cells play in IRI to the gut. METHODS Adult wild-type (WT) and αβ T cell-deficient mice were subjected to acute intestinal IRI with 30min ischemia followed by 4h reperfusion. The gene expression of pro-inflammatory cytokines was measured by qPCR, and the influx of leukocyte subpopulations in the gut was assessed via flow cytometry and histology. Pro-inflammatory cytokines in the serum were measured, and transaminases were assessed as an indicator of distant organ IRI. RESULTS Intestinal IRI led to an increased expression of pro-inflammatory cytokines in the gut tissue and an influx of leukocytes that predominantly consisted of neutrophils and macrophages. Furthermore, intestinal IRI increased serum IL-6, TNF-α, and ALT/AST levels. The αβ T cell-deficient mice did not exhibit a more significant increase in pro-inflammatory cytokines in the gut or serum following IR than the WT mice. There was also no difference between WT- and αβ T cell-deficient mice in terms of neutrophil infiltration or macrophage activation. Furthermore, the increase in transaminases was equal in both groups indicating that the level of distant organ injury was comparable. CONCLUSION An increasing body of evidence demonstrates that αβ T cells play a key role in IRI. In the gut, however, αβ T cells are not pivotal in the first hours following acute IRI as deficiency does not impact cytokine production, neutrophil recruitment, macrophage activation, or distant organ injury. Thus, αβ T cells may be considered innocent bystanders during the acute phase of intestinal IRI.
Collapse
Affiliation(s)
- Yi Yu
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
| | - Xiaoyan Feng
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
| | - Gertrud Vieten
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
| | - Stephanie Dippel
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
| | - Tawan Imvised
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
| | - Faikah Gueler
- Department of Nephrology, Hannover Medical School, Hanover, Germany
| | - Benno M. Ure
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
| | - Jochen F. Kuebler
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
| | - Christian Klemann
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hanover, Germany
| |
Collapse
|