1
|
Maryam B, Smith ME, Miller SJ, Natarajan H, Zimmerman KA. Macrophage Ontogeny, Phenotype, and Function in Ischemia Reperfusion-Induced Injury and Repair. KIDNEY360 2024; 5:459-470. [PMID: 38297436 PMCID: PMC11000738 DOI: 10.34067/kid.0000000000000376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 01/22/2024] [Indexed: 02/02/2024]
Abstract
AKI is characterized by a sudden, and usually reversible, decline in kidney function. In mice, ischemia-reperfusion injury (IRI) is commonly used to model the pathophysiologic features of clinical AKI. Macrophages are a unifying feature of IRI as they regulate both the initial injury response as well as the long-term outcome following resolution of injury. Initially, macrophages in the kidney take on a proinflammatory phenotype characterized by the production of inflammatory cytokines, such as CCL2 (monocyte chemoattractant protein 1), IL-6, IL-1 β , and TNF- α . Release of these proinflammatory cytokines leads to tissue damage. After resolution of the initial injury, macrophages take on a reparative role, aiding in tissue repair and restoration of kidney function. By contrast, failure to resolve the initial injury results in prolonged inflammatory macrophage accumulation and increased kidney damage, fibrosis, and the eventual development of CKD. Despite the extensive amount of literature that has ascribed these functions to M1/M2 macrophages, a recent paradigm shift in the macrophage field now defines macrophages on the basis of their ontological origin, namely monocyte-derived and tissue-resident macrophages. In this review, we focus on macrophage phenotype and function during IRI-induced injury, repair, and transition to CKD using both the classic (M1/M2) and novel (ontological origin) definition of kidney macrophages.
Collapse
Affiliation(s)
- Bibi Maryam
- Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Morgan E. Smith
- Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Sarah J. Miller
- Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Hariharasudan Natarajan
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kurt A. Zimmerman
- Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
2
|
Nemoto H, Sakai D, Watson D, Masuda K. Nuclear Factor-κB Decoy Oligodeoxynucleotide Attenuates Cartilage Resorption In Vitro. Bioengineering (Basel) 2024; 11:46. [PMID: 38247922 PMCID: PMC10813736 DOI: 10.3390/bioengineering11010046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Cartilage harvest and transplantation is a common surgery using costal, auricular, and septal cartilage for craniofacial reconstruction. However, absorption and warping of the cartilage grafts can occur due to inflammatory factors associated with wound healing. Transcription factor nuclear factor-κB (NF-κB) is activated by the various stimulation such as interleukin-1 (IL-1), and plays a central role in the transactivation of this inflammatory cytokine gene. Inhibition of NF-κB may have anti-inflammatory effects. The aim of this study was to explore the potential of an NF-κB decoy oligodeoxynucleotide (Decoy) as a chondroprotective agent. MATERIALS AND METHODS Safe and efficacious concentrations of Decoy were assessed using rabbit nasal septal chondrocytes (rNSChs) and assays for cytotoxicity, proteoglycan (PG) synthesis, and PG turnover were carried out. The efficacious concentration of Decoy determined from the rNSChs was then applied to human nasal septal cartilage (hNSC) in vitro and analyzed for PG turnover, the levels of inflammatory markers, and catabolic enzymes in explant-conditioned culture medium. RESULTS Over the range of Decoy conditions and concentrations, no inhibition of PG synthesis or cytotoxicity was observed. Decoy at 10 μM effectively inhibited PG degradation in the hNSC explant, prolonging PG half-life by 63% and decreasing matrix metalloprotease 3 (MMP-3) by 70.7% (p = 0.027). CONCLUSIONS Decoy may be considered a novel chondroprotective therapeutic agent in cartilage transplantation due to its ability to inhibit cartilage degradation due to inflammation cytokines.
Collapse
Affiliation(s)
- Hitoshi Nemoto
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of California, La Jolla, San Diego, CA 92093, USA
- Department of Plastic Surgery, School of Medicine, Tokai University, Isehara 259-1193, Kanagawa, Japan
| | - Daisuke Sakai
- Department of Orthopaedic Surgery, School of Medicine, University of California, La Jolla, San Diego, CA 92093, USA; (D.S.); (K.M.)
- Department of Orthopaedic Surgery, School of Medicine, Tokai University, Isehara 259-1193, Kanagawa, Japan
| | - Deborah Watson
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of California, La Jolla, San Diego, CA 92093, USA
| | - Koichi Masuda
- Department of Orthopaedic Surgery, School of Medicine, University of California, La Jolla, San Diego, CA 92093, USA; (D.S.); (K.M.)
| |
Collapse
|
3
|
Gwon MG, Leem J, An HJ, Gu H, Bae S, Kim JH, Park KK. The decoy oligodeoxynucleotide against HIF-1α and STAT5 ameliorates atopic dermatitis-like mouse model. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102036. [PMID: 37799329 PMCID: PMC10550406 DOI: 10.1016/j.omtn.2023.102036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023]
Abstract
Atopic dermatitis (AD) is a common inflammatory skin disease caused by an immune disorder. Mast cells are known to be activated and granulated to maintain an allergic reaction, including rhinitis, asthma, and AD. Although hypoxia-inducible factor-1 alpha (HIF-1α) and signal transducer and activator of transcription 5 (STAT5) play crucial roles in mast cell survival and granulation, their effects need to be clarified in allergic disorders. Thus, we designed decoy oligodeoxynucleotide (ODN) synthetic DNA, without open ends, containing complementary sequences for HIF-1α and STAT5 to suppress the transcriptional activities of HIF-1α and STAT5. In this study, we demonstrated the effects of HIF-1α/STAT5 ODN using AD-like in vivo and in vitro models. The HIF-1α/STAT5 decoy ODN significantly alleviated cutaneous symptoms similar to AD, including morphology changes, immune cell infiltration, skin barrier dysfunction, and inflammatory response. In the AD model, it also inhibited mast cell infiltration and degranulation in skin tissue. These results suggest that the HIF-1α/STAT5 decoy ODN ameliorates the AD-like disorder and immunoglobulin E (IgE)-induced mast cell activation by disrupting HIF-1α/STAT5 signaling pathways. Taken together, these findings suggest the possibility of HIF-1α/STAT5 as therapeutic targets and their decoy ODN as a potential therapeutic tool for AD.
Collapse
Affiliation(s)
- Mi-Gyeong Gwon
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Hyun-Jin An
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Hyemin Gu
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Seongjae Bae
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Jong Hyun Kim
- Department of Biochemistry, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Kwan-Kyu Park
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| |
Collapse
|
4
|
Yang Y, Jin S, Zhang J, Chen W, Lu Y, Chen J, Yan Z, Shen B, Ning Y, Shi Y, Chen J, Wang J, Xu S, Jia P, Teng J, Fang Y, Song N, Ding X. Acid-sensing ion channel 1a exacerbates renal ischemia-reperfusion injury through the NF-κB/NLRP3 inflammasome pathway. J Mol Med (Berl) 2023; 101:877-890. [PMID: 37246982 PMCID: PMC10300185 DOI: 10.1007/s00109-023-02330-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/30/2023]
Abstract
Ischemia-reperfusion injury (IRI) is the main cause of acute kidney injury (AKI), and there is no effective therapy. Microenvironmental acidification is generally observed in ischemic tissues. Acid-sensing ion channel 1a (ASIC1a) can be activated by a decrease in extracellular pH which mediates neuronal IRI. Our previous study demonstrated that, ASIC1a inhibition alleviates renal IRI. However, the underlying mechanisms have not been fully elucidated. In this study, we determined that renal tubule-specific deletion of ASIC1a in mice (ASIC1afl/fl/CDH16cre) attenuated renal IRI, and reduced the expression of NLRP3, ASC, cleaved-caspase-1, GSDMD-N, and IL-1β. Consistent with these in vivo results, inhibition of ASIC1a by the specific inhibitor PcTx-1 protected HK-2 cells from hypoxia/reoxygenation (H/R) injury, and suppressed H/R-induced NLRP3 inflammasome activation. Mechanistically, the activation of ASIC1a by either IRI or H/R induced the phosphorylation of NF-κB p65, which translocates to the nucleus and promotes the transcription of NLRP3 and pro-IL-1β. Blocking NF-κB by treatment with BAY 11-7082 validated the roles of H/R and acidosis in NLRP3 inflammasome activation. This further confirmed that ASIC1a promotes NLRP3 inflammasome activation, which requires the NF-κB pathway. In conclusion, our study suggests that ASIC1a contributes to renal IRI by affecting the NF-κB/NLRP3 inflammasome pathway. Therefore, ASIC1a may be a potential therapeutic target for AKI. KEY MESSAGES: Knockout of ASIC1a attenuated renal ischemia-reperfusion injury. ASIC1a promoted the NF-κB pathway and NLRP3 inflammasome activation. Inhibition of the NF-κB mitigated the NLRP3 inflammasome activation induced by ASIC1a.
Collapse
Affiliation(s)
- Yan Yang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Shi Jin
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Jian Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Weize Chen
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Yufei Lu
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Jun Chen
- Department of Pathology, Changzheng Hospital, Naval Military Medical University, Shanghai, China
| | - Zhixin Yan
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Bo Shen
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Yichun Ning
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Yiqin Shi
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Jing Chen
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Jialin Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Sujuan Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Ping Jia
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Jie Teng
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Yi Fang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Nana Song
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China.
- Fudan Zhangjiang Institute, Shanghai, China.
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China.
| |
Collapse
|
5
|
Song N, Xu Y, Paust HJ, Panzer U, de Las Noriega MM, Guo L, Renné T, Huang J, Meng X, Zhao M, Thaiss F. IKK1 aggravates ischemia-reperfusion kidney injury by promoting the differentiation of effector T cells. Cell Mol Life Sci 2023; 80:125. [PMID: 37074502 PMCID: PMC10115737 DOI: 10.1007/s00018-023-04763-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/10/2023] [Accepted: 03/20/2023] [Indexed: 04/20/2023]
Abstract
Ischemia-reperfusion injury (IRI) is one of the major causes of acute kidney injury (AKI), and experimental work has revealed detailed insight into the inflammatory response in the kidney. T cells and NFκB pathway play an important role in IRI. Therefore, we examined the regulatory role and mechanisms of IkappaB kinase 1 (IKK1) in CD4+T lymphocytes in an experimental model of IRI. IRI was induced in CD4cre and CD4IKK1Δ mice. Compared to control mice, conditional deficiency of IKK1 in CD4+T lymphocyte significantly decreased serum creatinine, blood urea nitrogen (BUN) level, and renal tubular injury score. Mechanistically, lack in IKK1 in CD4+T lymphocytes reduced the ability of CD4 lymphocytes to differentiate into Th1/Th17 cells. Similar to IKK1 gene ablation, pharmacological inhibition of IKK also protected mice from IRI. Together, lymphocyte IKK1 plays a pivotal role in IRI by promoting T cells differentiation into Th1/Th17 and targeting lymphocyte IKK1 may be a novel therapeutic strategy for IRI.
Collapse
Affiliation(s)
- Ning Song
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Youzheng St 23, Harbin, 150001, China
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany
| | - Yang Xu
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Hans-Joachim Paust
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany
| | - Ulf Panzer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany
| | | | - Linlin Guo
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany
| | - Thomas Renné
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, 55131, Germany
| | - Jiabin Huang
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Xianglin Meng
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Youzheng St 23, Harbin, 150001, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Mingyan Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Youzheng St 23, Harbin, 150001, China.
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| | - Friedrich Thaiss
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany.
| |
Collapse
|
6
|
Gu H, An HJ, Gwon MG, Bae S, Zouboulis CC, Park KK. The Effects of Synthetic SREBP-1 and PPAR-γ Decoy Oligodeoxynucleotide on Acne-like Disease In Vivo and In Vitro via Lipogenic Regulation. Biomolecules 2022; 12:1858. [PMID: 36551286 PMCID: PMC9775059 DOI: 10.3390/biom12121858] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/25/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Acne vulgaris has a pathogenesis that involves increased sebum production and perifollicular inflammation. Sterol regulatory element-binding protein-1 (SREBP-1) and peroxisome proliferator activated receptor-γ (PPAR-γ) are transcription factors that regulate numerous genes involved in lipid biosynthesis. To improve a new therapeutic approach, we designed the SREBP/PPAR decoy oligodeoxynucleotide (ODN), a synthetic short DNA containing complementary sequences for the SREBP and PPAR transcription factors. We aim to investigate the beneficial functions and the molecular mechanisms of the synthetic SREBP/PPAR decoy ODN in lipogenic models. C. acnes was intradermally injected with a 1.0 × 107 colony forming unit/20 μL. The synthetic SREBP/PPAR decoy ODN or scrambled decoy ODN (10 μg) was transferred via the mouse tail vein injection. SZ95 cells were transfected with 2 μg of synthetic ODNs. After transfection, the SZ95 cells were cultured in serum-free medium containing 20 ng/μL of insulin-like growth factor-1 (IGF)-1 for 24 h. To investigate the expression of gene and signaling pathways, we performed Western blotting. The distribution of the chimeric decoy ODN was confirmed by EMSA. Lipid levels were assessed by Nile red and Oil Red O staining. The cytokine levels were measured by ELISA kit. This study showed that C. acnes-injected mice and IGF-1-stimulated SZ95 cells exhibited increased expression of SREBP-1 and PPAR-γ compared to the normal controls. In contrast, the administration of the SREBP/PPAR chimeric decoy ODN significantly suppressed the upregulation of lipogenic genes. Furthermore, the SREBP/PPAR decoy ODN decreased the plasma cytokines and cytokine levels of total protein. These results suggested that the SREBP/PPAR decoy ODN exerts its anti-lipogenic effects by regulating lipid metabolism and by inhibiting lipogenesis through the inactivation of the SREBP and PPAR pathways. Therefore, the synthetic SREBP/PPAR ODN demonstrates substantial therapeutic feasibility for the treatment of acne vulgaris.
Collapse
Affiliation(s)
- Hyemin Gu
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Hyun-Jin An
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Mi-Gyeong Gwon
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Seongjae Bae
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Christos C. Zouboulis
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Medical Center, Brandenburg Medical School Theodor Fontane and Faculty of Health Sciences Brandenburg, Auenweg 38, 06847 Dessau, Germany
| | - Kwan-Kyu Park
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| |
Collapse
|
7
|
Khanahmad H, Mirbod SM, Karimi F, Kharazinejad E, Owjfard M, Najaflu M, Tavangar M. Pathological Mechanisms Induced by TRPM2 Ion Channels Activation in Renal Ischemia-Reperfusion Injury. Mol Biol Rep 2022; 49:11071-11079. [PMID: 36104583 DOI: 10.1007/s11033-022-07836-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/26/2022] [Accepted: 08/01/2022] [Indexed: 10/14/2022]
Abstract
Renal ischemia-reperfusion (IR) injury triggers a cascade of signaling reactions involving an increase in Ca2 + charge and reactive oxygen species (ROS) levels resulting in necrosis, inflammation, apoptosis, and subsequently acute kidney injury (AKI).Transient receptor potential (TRP) channels include an essential class of Ca2+ permeable cation channels, which are segregated into six main channels: the canonical channel (TRPC), the vanilloid-related channel (TRPV), the melastatin-related channel (TRPM), the ankyrin-related channel (TRPA), the mucolipin-related channel (TRPML) and polycystin-related channel (TRPP) or polycystic kidney disease protein (PKD2). TRP channels are involved in adjusting vascular tone, vascular permeability, cell volume, proliferation, secretion, angiogenesis and apoptosis.TRPM channels include eight isoforms (TRPM1-TRPM8) and TRPM2 is the second member of this subfamily that has been expressed in various tissues and organs such as the brain, heart, kidney and lung. Renal TRPM2 channels have an important role in renal IR damage. So that TRPM2 deficient mice are resistant to renal IR injury. TRPM2 channels are triggered by several chemicals including hydrogen peroxide, Ca2+, and cyclic adenosine diphosphate (ADP) ribose (cADPR) that are generated during AKI caused by IR injury, as well as being implicated in cell death caused by oxidative stress, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Hossein Khanahmad
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of medical science, Isfahan, Iran
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical sciences, Isfahan, Iran, Isfahan University of Medical sciences, Isfahan, Iran
| | - Seyedeh Mahnaz Mirbod
- Resident of Cardiology, Department of cardiology, Isfahan University of Medical Science, Isfahan, Iran
- Department of Cardiology, Isfahan University of Medical Sciences, Isfahan, Iran., Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farzaneh Karimi
- Behbahan Faculty of Medical Sciences, Behbahan, Iran.
- Behbahan Faculty of Medical Sciences, No.8, Shahid Zibaei Blvd. Behbahan city, Behbahan, Khozestan province, Iran.
- Department of Physiology, Behbahan Faculty of Medical Sciences, Behbahan, Iran., Behbahan Faculty of Medical Sciences, Behbahan, Iran.
| | - Ebrahim Kharazinejad
- Abadan University of Medical Sciences, Abadan, Iran
- Department of Anatomy, Abadan University of Medical Sciences, Abadan, Iran, Abadan University of Medical Sciences, Abadan , Iran
| | - Maryam Owjfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz University of Applied Science and Technology (UAST), Shiraz, Iran
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran. Shiraz University of Applied Science and Technology (UAST), Shiraz, Iran, Shiraz University of Applied Science and Technology (UAST), Shiraz, Iran
| | - Malihe Najaflu
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehrsa Tavangar
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
8
|
Lee SJ, Kim YA, Park KK. Anti-Fibrotic Effect of Synthetic Noncoding Decoy ODNs for TFEB in an Animal Model of Chronic Kidney Disease. Int J Mol Sci 2022; 23:8138. [PMID: 35897713 PMCID: PMC9330689 DOI: 10.3390/ijms23158138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 02/01/2023] Open
Abstract
Despite emerging evidence suggesting that autophagy occurs during renal interstitial fibrosis, the role of autophagy activation in fibrosis and the mechanism by which autophagy influences fibrosis remain controversial. Transcription factor EB (TFEB) is a master regulator of autophagy-related gene transcription, lysosomal biogenesis, and autophagosome formation. In this study, we examined the preventive effects of TFEB suppression on renal fibrosis. We injected synthesized TFEB decoy oligonucleotides (ODNs) into the tail veins of unilateral ureteral obstruction (UUO) mice to explore the regulation of autophagy in UUO-induced renal fibrosis. The expression of interleukin (IL)-1β, tumor necrosis factor-α (TNF-α), and collagen was decreased by TFEB decoy ODN. Additionally, TEFB ODN administration inhibited the expression of microtubule-associated protein light chain 3 (LC3), Beclin1, and hypoxia-inducible factor-1α (HIF-1α). We confirmed that TFEB decoy ODN inhibited fibrosis and autophagy in a UUO mouse model. The TFEB decoy ODNs also showed anti-inflammatory effects. Collectively, these results suggest that TFEB may be involved in the regulation of autophagy and fibrosis and that regulating TFEB activity may be a promising therapeutic strategy against kidney diseases.
Collapse
Affiliation(s)
| | | | - Kwan-Kyu Park
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Korea; (S.-J.L.); (Y.-A.K.)
| |
Collapse
|
9
|
Plants with Therapeutic Potential for Ischemic Acute Kidney Injury: A Systematic Review. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6807700. [PMID: 35656467 PMCID: PMC9152371 DOI: 10.1155/2022/6807700] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/30/2022] [Indexed: 11/18/2022]
Abstract
Acute kidney injury (AKI) is a complex condition which has an intricate pathology mostly involving hemodynamic, inflammatory, and direct toxic effects at the cellular level with high morbidity and mortality ratios. Renal ischemic reperfusion injury (RIRI) is the main factor responsible for AKI, most often observed in different types of shock, kidney transplantation, sepsis, and postoperative procedures. The RIRI-induced AKI is accompanied by increased reactive oxygen species generation together with the activation of various inflammatory pathways. In this context, plant-derived medicines have shown encouraging nephroprotective properties. Evidence provided in this systemic review leads to the conclusion that plant-derived extracts and compounds exhibit nephroprotective action against renal ischemic reperfusion induced-AKI by increasing endogenous antioxidants and decreasing anti-inflammatory cytokines. However, there is no defined biomarker or target which can be used for treating AKI completely. These plant-derived extracts and compounds are only tested in selected transgenic animal models. To develop the results obtained into a therapeutic entity, one should apply them in proper vertebrate multitransgenic animal models prior to further validation in humans.
Collapse
|
10
|
Deficiency of IKK α in Macrophages Mitigates Fibrosis Progression in the Kidney after Renal Ischemia-Reperfusion Injury. J Immunol Res 2021; 2021:5521051. [PMID: 34917688 PMCID: PMC8670970 DOI: 10.1155/2021/5521051] [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: 02/19/2021] [Revised: 09/03/2021] [Accepted: 11/06/2021] [Indexed: 11/17/2022] Open
Abstract
Aims. Acute kidney injury (AKI) can lead to chronic kidney disease (CKD), and macrophages play a key role in this process. The aim of this study was to discover the role of IκB kinase α (IKKα) in macrophages in the process of AKI-to-CKD transition. Main Methods. We crossed lyz2-Cre mice with IKKα-floxed mice to generate mice with IKKα ablation in macrophages (Mac IKKα-/-). A mouse renal ischemia/reperfusion injury (IRI) model was induced by clamping the renal artery for 45 minutes. Treated mice were evaluated for blood biochemistry, tissue histopathology, and fibrosis markers. Macrophages were isolated from the peritoneal cavity for coculturing with tubular epithelial cells (TECs) and flow cytometry analysis. Key Findings. We found that fibrosis and kidney function loss after IRI were significantly alleviated in Mac IKKα-/- mice compared with wild-type (WT) mice. The expression of fibrosis markers and the infiltration of M2 macrophages were decreased in the kidneys of Mac IKKα-/- mice after IRI. The in vitro experiment showed that the IRI TECs cocultured with IKKα-/- macrophages (KO MΦs) downregulated the fibrosis markers accompanied by a downregulation of Wnt/β-catenin signaling. Significance. These data support the hypothesis that IKKα is involved in mediating macrophage polarization and increasing the expression of fibrosis-promoting inflammatory factors in macrophages. Therefore, knockdown of IKKα in macrophages may be a potential method that can be used to alleviate the AKI-to-CKD transition after IRI.
Collapse
|
11
|
Jin Y, Zhang M, Li M, Zhang H, Zhang F, Zhang H, Yin Z, Zhou M, Wan X, Li R, Cao C. Generation of Urine-Derived Induced Pluripotent Stem Cell Line from Patients with Acute Kidney Injury. Cell Reprogram 2021; 23:290-303. [PMID: 34648385 DOI: 10.1089/cell.2021.0051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Acute kidney injury (AKI) is mainly characterized by rapid decline of renal function. Currently, the strategy of stem cells might be a therapy to treat AKI. The objective of this study was to obtain human urine-derived cells (HUCs) from patients with AKI, followed by establishing induced pluripotent stem (iPS) cell line. We isolated urine cells from patients with AKI and found that the cells could survive long term with epithelioid morphology and maintain a normal karyotype. The cell line had expression of renal-specific markers and renal development-related genes. After induction, the urine cells cotransfecting with TET-ON vectors were converted into iPS cells. The HUC-derived iPS (HUC-iPS) was positive for alkaline phosphatase staining, and had expression of pluripotency markers, consistent with human embryonic fibroblast-derived iPS cell. Notably, HUC-iPS could be induced to undergo directional kidney precursor cells (KPCs) differentiation under defined conditions, and transplantation of KPCs resulted in reducing kidney damage from ischemia-reperfusion injury in mice. Therefore, we successfully established HUC-iPS cell from patients with AKI and provided a novel stem cell resource for cell therapy in AKI.
Collapse
Affiliation(s)
- Yong Jin
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Manling Zhang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Meishuang Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Hao Zhang
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Feng Zhang
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hong Zhang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Zhibao Yin
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Meng Zhou
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Xin Wan
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Rongfeng Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Changchun Cao
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
12
|
Jin Y, Zhang M, Li M, Zhang H, Zhao L, Qian C, Li S, Zhang H, Gao M, Pan B, Li R, Wan X, Cao C. SIX1 Activation Is Involved in Cell Proliferation, Migration, and Anti-inflammation of Acute Ischemia/Reperfusion Injury in Mice. Front Mol Biosci 2021; 8:725319. [PMID: 34513929 PMCID: PMC8427868 DOI: 10.3389/fmolb.2021.725319] [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: 06/15/2021] [Accepted: 08/11/2021] [Indexed: 11/13/2022] Open
Abstract
Nephrogenic proteins are re-expressed after ischemia/reperfusion (I/R) injury; however, the role of these proteins is still unknown. We found that sine oculis homeobox 1 (SIX1), a developmentally regulated homeoprotein, is reactivated in tubular epithelial cells after I/R injury associated with cell proliferation/migration and anti-inflammation. We demonstrated that SIX1 promoted cell proliferation by upregulating cyclin and glycolytic genes, and might increase cell migration by upregulating the expression of matrix metalloproteinase 9 (MMP9) directly or indirectly in the cell model. Notably, SIX1 targeted the promoters of the amino-terminal enhancer of split (AES) and fused in sarcoma (FUS), which are cofactors of nuclear factor-κB (NF-κB) subunit RELA, and then inhibited the transactivation function of RELA. The expression of monocyte chemotactic protein-1 (MCP-1) was decreased by the SIX1-mediated NF-κB pathway. Our results showed that the expression of cyclin, glycolytic genes, and MMP9 were significantly increased, and the infiltration of monocytes/macrophages (Mophs) was suppressed in SIX1 overexpression kidney at 1, 2, and 3 days after reperfusion. The overexpression of SIX1 resulted in reducing kidney damage from I/R injury in mice by promoting cell proliferation and migration and by inhibiting inflammation. Our study provides evidence that SIX1 involved in cell proliferation, migration, and anti-inflammation in the I/R model, which might be a potential therapeutic target that could be used to ameliorate kidney damage.
Collapse
Affiliation(s)
- Yong Jin
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Manling Zhang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Meishuang Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Hong Zhang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Lihua Zhao
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Cheng Qian
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Shensen Li
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Hao Zhang
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Min Gao
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Binbin Pan
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Rongfeng Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Xin Wan
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Changchun Cao
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
13
|
Kim S, Lee SA, Yoon H, Kim MY, Yoo JK, Ahn SH, Park CH, Park J, Nam BY, Park JT, Han SH, Kang SW, Kim NH, Kim HS, Han D, Yook JI, Choi C, Yoo TH. Exosome-based delivery of super-repressor IκBα ameliorates kidney ischemia-reperfusion injury. Kidney Int 2021; 100:570-584. [PMID: 34051264 DOI: 10.1016/j.kint.2021.04.039] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 04/22/2021] [Accepted: 04/30/2021] [Indexed: 12/15/2022]
Abstract
Ischemia-reperfusion injury is a major cause of acute kidney injury. Recent studies on the pathophysiology of ischemia-reperfusion-induced acute kidney injury showed that immunologic responses significantly affect kidney ischemia-reperfusion injury and repair. Nuclear factor (NF)-ĸB signaling, which controls cytokine production and cell survival, is significantly involved in ischemia-reperfusion-induced acute kidney injury, and its inhibition can ameliorate ischemic acute kidney injury. Using EXPLOR, a novel, optogenetically engineered exosome technology, we successfully delivered the exosomal super-repressor inhibitor of NF-ĸB (Exo-srIĸB) into B6 wild type mice before/after kidney ischemia-reperfusion surgery, and compared outcomes with those of a control exosome (Exo-Naïve)-injected group. Exo-srIĸB treatment resulted in lower levels of serum blood urea nitrogen, creatinine, and neutrophil gelatinase-associated lipocalin in post-ischemic mice than in the Exo-Naïve treatment group. Systemic delivery of Exo-srIĸB decreased NF-ĸB activity in post-ischemic kidneys and reduced apoptosis. Post-ischemic kidneys showed decreased gene expression of pro-inflammatory cytokines and adhesion molecules with Exo-srIĸB treatment as compared with the control. Intravital imaging confirmed the uptake of exosomes in neutrophils and macrophages. Exo-srIĸB treatment also significantly affected post-ischemic kidney immune cell populations, lowering neutrophil, monocyte/macrophage, and T cell frequencies than those in the control. Thus, modulation of NF-ĸB signaling through exosomal delivery can be used as a novel therapeutic method for ischemia-reperfusion-induced acute kidney injury.
Collapse
Affiliation(s)
- Seonghun Kim
- Department of Oral Pathology, Oral Cancer Research Institute, College of Dentistry, Yonsei University, Seoul, South Korea
| | - Sul A Lee
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, South Korea; Department of Internal Medicine, MetroWest Medical Center, Framingham, Massachusetts, USA
| | - Heakyung Yoon
- ILIAS Innovation Center, ILIAS Biologics Inc., Daejeon, South Korea
| | - Myung Yoon Kim
- ILIAS Innovation Center, ILIAS Biologics Inc., Daejeon, South Korea
| | - Jae-Kwang Yoo
- ILIAS Innovation Center, ILIAS Biologics Inc., Daejeon, South Korea
| | - So-Hee Ahn
- ILIAS Innovation Center, ILIAS Biologics Inc., Daejeon, South Korea
| | | | - Jimin Park
- Severance Biomedical Science Institute, College of Medicine, Yonsei University, Seoul, South Korea
| | - Bo Young Nam
- Severance Biomedical Science Institute, College of Medicine, Yonsei University, Seoul, South Korea
| | - Jung Tak Park
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, South Korea
| | - Seung Hyeok Han
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, South Korea
| | - Shin-Wook Kang
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, South Korea
| | - Nam Hee Kim
- Department of Oral Pathology, Oral Cancer Research Institute, College of Dentistry, Yonsei University, Seoul, South Korea
| | - Hyun Sil Kim
- Department of Oral Pathology, Oral Cancer Research Institute, College of Dentistry, Yonsei University, Seoul, South Korea
| | - Dawool Han
- Department of Oral Pathology, Oral Cancer Research Institute, College of Dentistry, Yonsei University, Seoul, South Korea
| | - Jong In Yook
- Department of Oral Pathology, Oral Cancer Research Institute, College of Dentistry, Yonsei University, Seoul, South Korea.
| | - Chulhee Choi
- ILIAS Innovation Center, ILIAS Biologics Inc., Daejeon, South Korea; Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.
| | - Tae-Hyun Yoo
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, South Korea.
| |
Collapse
|
14
|
Wu G, Nie W, Wang Q, Hao Y, Gong S, Zheng Y, Lv H. Umbelliferone Ameliorates Complete Freund Adjuvant-Induced Arthritis via Reduction of NF-κB Signaling Pathway in Osteoclast Differentiation. Inflammation 2021; 44:1315-1329. [PMID: 33484396 DOI: 10.1007/s10753-021-01418-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/19/2020] [Accepted: 01/08/2021] [Indexed: 12/22/2022]
Abstract
Osteoclasts, bone-resorbing somatic cells, are directly responsible for bone destruction during rheumatoid arthritis. Complete Freund adjuvant (CFA) is a widely used animal model using rodents for studying rheumatoid arthritis (RA), which effectively manifests serious cartilage destruction and progressive bone erosion, affecting synovial joints and serious joint dysfunction. It was considered that joint injury in RA is induced through systemic inflammation pathway. Umbelliferone (UF), a coumarin derivative of Agele marmilosa, possesses anti-inflammatory activity. In the current study, we scrutinize the effect of umbelliferone on CFA-induced arthritis model and explore the possible mechanism on bone destruction. Intradermal administration of CFA (0.05 mL) was to induce RA manifestations in the experimental rats and the same oral administration of UF was received. The anti-arthritic activity of UF was determined by its inhibitory activity on various biochemical markers, viz., pro-inflammatory, inflammatory, antioxidant enzymes, and hematological parameters elevated during RA condition. We also estimated the mRNA expression of osteoclast parameters. Obtained result disclosed significant reduction in the paw edema and increment of the body weight after UF administration. UF reduce the inflammatory mediatory such as COX-2, PGE2, NF-kB, and VEGF; pro-inflammatory cytokines include TNF-α, IL-1β, IL-6, IL-10, and IL-17 significantly. Moreover, UF treatment significantly reduced the osteoclast number via modulating the RANKL/RANK/OPG ratio. Furthermore, administration of umbelliferone significantly (P < 0.001) suppressed the NF-κB and VEGF. Collectively, our results indicated the novel role of umbelliferone in osteoclastogenesis and proved that umbelliferone is a modern therapeutic tool as a natural agent for treating arthritis and other autoimmune disorders with bone degradation.
Collapse
Affiliation(s)
- Guofeng Wu
- Department of Articular Orthopaedics, The First People's Hospital of Changzhou, Changzhou, 213000, Jiangsu, China
| | - Wenbo Nie
- Department of Orthopaedics, Shanxian Central Hospital, Heze, 274300, Shandong, China
| | - Qiu Wang
- Department of Surgery, Hot Spring Sanatorium of Linyi, Shandong Coal (Linyi Hedong Central Hospital, Linyi, 276032, Shandong, China
| | - Youguo Hao
- Department of Rehabilitation, Shanghai Putuo People's Hospital, Shanghai, 200060, China
| | - Shaohua Gong
- Department of Spinal surgery, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, China
| | - Yuxin Zheng
- Department of Orthopedics, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200021, China
| | - Hao Lv
- Department of Orthopedics Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
| |
Collapse
|
15
|
Inhibition of BRD4 Reduces Neutrophil Activation and Adhesion to the Vascular Endothelium Following Ischemia Reperfusion Injury. Int J Mol Sci 2020; 21:ijms21249620. [PMID: 33348732 PMCID: PMC7767067 DOI: 10.3390/ijms21249620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023] Open
Abstract
Renal ischemia reperfusion injury (IRI) is associated with inflammation, including neutrophil infiltration that exacerbates the initial ischemic insult. The molecular pathways involved are poorly characterized and there is currently no treatment. We performed an in silico analysis demonstrating changes in NFκB-mediated gene expression in early renal IRI. We then evaluated NFκB-blockade with a BRD4 inhibitor on neutrophil adhesion to endothelial cells in vitro, and tested BRD4 inhibition in an in vivo IRI model. BRD4 inhibition attenuated neutrophil adhesion to activated endothelial cells. In vivo, IRI led to increased expression of cytokines and adhesion molecules at 6 h post-IRI with sustained up-regulated expression to 48 h post-IRI. These effects were attenuated, in part, with BRD4 inhibition. Absolute neutrophil counts increased significantly in the bone marrow, blood, and kidney 24 h post-IRI. Activated neutrophils increased in the blood and kidney at 6 h post-IRI and remained elevated in the kidney until 48 h post-IRI. BRD4 inhibition reduced both total and activated neutrophil counts in the kidney. IRI-induced tubular injury correlated with neutrophil accumulation and was reduced by BRD4 inhibition. In summary, BRD4 inhibition has important systemic and renal effects on neutrophils, and these effects are associated with reduced renal injury.
Collapse
|
16
|
Wang M, Deng J, Lai H, Lai Y, Meng G, Wang Z, Zhou Z, Chen H, Yu Z, Li S, Jiang H. Vagus Nerve Stimulation Ameliorates Renal Ischemia-Reperfusion Injury through Inhibiting NF- κB Activation and iNOS Protein Expression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7106525. [PMID: 32148655 PMCID: PMC7053466 DOI: 10.1155/2020/7106525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/17/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVE In renal ischemia/reperfusion injury (RIRI), nuclear factor κB (NF-κB (NF-κB (NF. METHODS Eighteen male Sprague-Dawley rats were randomly allocated into the sham group, the I/R group, and the VNS+I/R group, 6 rats per group. An RIRI model was induced by a right nephrectomy and blockade of the left renal pedicle vessels for 45 min. After 6 h of reperfusion, the blood samples and renal samples were collected. The VNS treatment was performed throughout the I/R process in the VNS+I/R group using specific parameters (20 Hz, 0.1 ms in duration, square waves) known to produce a small but reliable bradycardia. Blood was used for evaluation of renal function and inflammatory state. Renal injury was evaluated via TUNEL staining. Renal samples were harvested to evaluate renal oxidative stress, NF-κB (NF. RESULTS The VNS treatment reduces serum creatinine (Cr) and blood urea nitrogen (BUN) levels. Simultaneously, the levels of tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), and interleukin 1-beta (IL-1β) were significantly increased in the I/R group, but VNS treatment markedly ameliorated this inflammatory response. Furthermore, the VNS ameliorated oxidant stress and renal injury, indicated by a decrease in 3-nitrotyrosine (3-NT) formation and MDA and MPO levels and an increase in the SOD level compared to that in the I/R group. Finally, the VNS also significantly decreases NF-κB (NF. CONCLUSION Our findings indicate that NF-κB activation increased iNOS expression and promoted RIRI and that VNS treatment attenuated RIRI by inhibiting iNOS expression, oxidative stress, and inflammation via NF-κB inactivation.κB (NF-κB (NF.
Collapse
Affiliation(s)
- Meng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 Hubei, China
| | - Jielin Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 Hubei, China
| | - Huanzhu Lai
- Department of Cardiology, First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Yanqiu Lai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 Hubei, China
| | - Guannan Meng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 Hubei, China
| | - Zhenya Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 Hubei, China
| | - Zhen Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 Hubei, China
| | - Hu Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 Hubei, China
| | - Zhiyao Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 Hubei, China
| | - Shuyan Li
- Department of Cardiology, First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 Hubei, China
| |
Collapse
|
17
|
Shen GH, Song Y, Yao Y, Sun QF, Jing B, Wu J, Li SY, Liu SQ, Li HC, Yuan C, Liu GY, Li JB, Liu XY, Wang HY. Downregulation of DLGAP1-Antisense RNA 1 Alleviates Vascular Endothelial Cell Injury Via Activation of the Phosphoinositide 3-kinase/Akt Pathway Results from an Acute Limb Ischemia Rat Model. Eur J Vasc Endovasc Surg 2019; 59:98-107. [PMID: 31744785 DOI: 10.1016/j.ejvs.2019.06.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study aimed to investigate the effect of long non-coding RNA (lncRNA) DLGAP1 antisense RNA 1 (DLGAP1-AS1) on vascular endothelial cell (VEC) injury via the phosphoinositide 3-kinase (PI3K)/Akt pathway in rat models of acute lower limb ischaemia-reperfusion (I/R). METHODS Differentially expressed lncRNAs related to I/R were screened using the gene expression omnibus database. Acute lower limb I/R models were induced in male Wistar rats, in which the regulatory mechanisms of DLGAP1-AS1 silencing were analysed after the treatment of small interfering RNA (siRNA) against DLGAP1-AS1 or an inhibitor of the PI3K/Akt pathway. The relationship between DLGAP1-AS1 and the PI3K/Akt pathway was analysed. The levels of tumour necrosis factor (TNF)-α and vascular cell adhesion molecule-1 (VCAM-1), as well as malondialdehyde (MDA) concentration and creatine kinase (CK) activity, were measured. The number of circulating endothelial cells (CECs) and apoptosis of VECs were identified. RESULTS Microarray based analysis indicated that DLGAP1-AS1 was highly expressed in I/R, which was further confirmed by detection of expression in rat models of acute lower limb I/R. Notably, the treatment of siRNA against DLGAP1-AS1 led to the activation of the PI3K/Akt pathway. In response to siRNA against DLGAP1-AS1, the levels of TNF-α and VCAM-1 were decreased, and MDA concentration and CK activity was downregulated. Reduced CEC numbers and suppressed VEC apoptosis were also observed. CONCLUSION DLGAP1-AS1 silencing could further suppress the oxidative stress, exert an anti-apoptosis effect, and reduce inflammatory reaction, whereby VEC injury is alleviated by activation of the PI3K/Akt pathway in rats with acute lower limb I/R.
Collapse
Affiliation(s)
- Guang-Hui Shen
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Ye Song
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Ye Yao
- Department of Cardiac Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Qing-Feng Sun
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Bao Jing
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Jia Wu
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Shi-Yong Li
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Si-Qi Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Hao-Cheng Li
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Chao Yuan
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Gao-Yan Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Jing-Bo Li
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Xin-Yu Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Hai-Yang Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China.
| |
Collapse
|
18
|
Liang Y, Xue N, Wang X, Ding X, Fang Y. Superagonistic CD28 protects against renal ischemia injury induced fibrosis through a regulatory T-cell expansion dependent mechanism. BMC Nephrol 2019; 20:407. [PMID: 31706278 PMCID: PMC6842503 DOI: 10.1186/s12882-019-1581-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/03/2019] [Indexed: 11/10/2022] Open
Affiliation(s)
- Yiran Liang
- Department of Nephrology, Zhongshan Hospital, Fudan University, 111 Yixueyuan Road, Shanghai, 200032, China
| | - Ning Xue
- Department of Nephrology, Zhongshan Hospital, Fudan University, 111 Yixueyuan Road, Shanghai, 200032, China.,Shanghai Medical Center of Kidney, Shanghai, China
| | - Xiaoyan Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University, 111 Yixueyuan Road, Shanghai, 200032, China
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, 111 Yixueyuan Road, Shanghai, 200032, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China
| | - Yi Fang
- Department of Nephrology, Zhongshan Hospital, Fudan University, 111 Yixueyuan Road, Shanghai, 200032, China. .,Shanghai Medical Center of Kidney, Shanghai, China. .,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China. .,Shanghai Institute of Kidney and Dialysis, Shanghai, China.
| |
Collapse
|
19
|
Matrine alleviates Staphylococcus aureus lipoteichoic acid-induced endometritis via suppression of TLR2-mediated NF-κB activation. Int Immunopharmacol 2019; 70:201-207. [PMID: 30822611 DOI: 10.1016/j.intimp.2019.02.033] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/19/2019] [Accepted: 02/19/2019] [Indexed: 12/28/2022]
Abstract
Endometritis is one of the main diseases that causes great economic losses in the dairy industry. Recent studies have shown that matrine extracted from the traditional Chinese herb Sophora flavescens is an alkaloid with a broad range of bioactivities. Here, we aimed to investigate the protective effects of matrine on Staphylococcus aureus lipoteichoic acid (LTA)-induced endometritis in mice and elucidate the possible molecular mechanisms in vitro. Histopathological changes showed that matrine remarkably attenuated the uterus injury in a mouse model of LTA-induced endometritis. qPCR and ELISA results showed that matrine dose-dependently reduced the expression of pro-inflammatory cytokines (TNF-α and IL-1β). To further elucidate the underlying mechanisms of this protective effect of matrine, LTA-stimulated bovine endometrial epithelial cells (bEECs) were employed in this study. The results demonstrated that TLR2 expression and its downstream nuclear factor (NF)-κB activation were both suppressed by matrine treatment. Furthermore, a small interference RNA targeting TLR2 gene mimicked matrine in its inhibition on LTA-induced activation of TLR2 and NF-κB. In conclusion, these findings suggest the protective effect of matrine against LTA-induced endometritis through negative regulation of TLR2-mediated NF-κB pathway.
Collapse
|
20
|
Gu L, Tao Y, Chen C, Ye Y, Xiong X, Sun Y. Initiation of the inflammatory response after renal ischemia/reperfusion injury during renal transplantation. Int Urol Nephrol 2018; 50:2027-2035. [PMID: 29974405 DOI: 10.1007/s11255-018-1918-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 06/20/2018] [Indexed: 01/22/2023]
Abstract
Ischemia/reperfusion injury (IRI) occurs commonly during renal transplantation. It has been well demonstrated that the inflammatory response has an important role in the pathogenesis and pathological processes of IRI. However, the signaling events that trigger the activation of the inflammatory response are less clear. Accumulated evidence has identified the role of various injury factors released from or exposed in ischemic, damaged, or dying cells, which serve as initiators of the inflammatory response and exacerbate kidney injury after renal IRI. Signaling pathways triggered by these endogenous molecules that activate different pathogen recognition receptors have also been widely investigated. Here, we review the molecular signaling molecules that initiate the inflammatory response during renal IRI and that provide potential therapeutic options for the disease.
Collapse
Affiliation(s)
- Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Tao
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Cheng Chen
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yingze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaoxing Xiong
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yao Sun
- Key Laboratory of Pesticides and Chemical Biology, Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, Hubei International Scientific and Technology Cooperation Base of Pesticide and Green Synthesis, Chemical Biology Center, College of Chemistry, Central China Normal Universtiy, Wuhan, 430079, China.
| |
Collapse
|
21
|
Weng XF, Li ST, Song Q, Zhu Q, Song DD, Qin ZH, Xie Y. Protective Effect of Nicotinamide Adenine Dinucleotide Phosphate on Renal Ischemia-Reperfusion Injury. Kidney Blood Press Res 2018; 43:651-663. [PMID: 29734167 DOI: 10.1159/000489620] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 04/26/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Renal ischemia-reperfusion injury (IRI) is a common consequence of acute kidney injury. Nicotinamide adenine dinucleotide phosphate (NADPH), which is derived from the pentose phosphate pathway, is essential for the proper functioning of essential redox and antioxidant defense systems. Previous studies have indicated that NADPH is responsible for protecting the brain from ischemic injury. The goal of this study was to analyze the protective function of NADPH in renal IRI. METHODS The IRI animal model was generated through a midline laparotomy surgery that clamped both sides of the renal pedicles for 40 min to induce renal ischemia. The in vitro model was generated by removing oxygen and glucose from human kidney epithelial cells (HK-2 cells), followed by reoxygenation to imitate IRI. Renal function and histopathological changes were observed and evaluated. Additionally, malondialdehyde and glutathione levels were determined in renal tissue homogenate as indicators of oxidative stress. ROS production in cells was determined by DHE staining. Protein biomarker expression was evaluated by western blot, apoptosis was analyzed by TUNEL staining, and p65 nuclear translocation was visualized by immunofluorescence. RESULTS Our data indicated that NADPH safeguarded the kidneys from histological and functional damage, and significantly reduce cell injury along with preventing potential increases in blood urea nitrogen and creatinine levels. Furthermore, we observed that NADPH increased glutathione levels, while reducing levels of malondialdehyde and reactive oxygen species. Additionally, our results suggested that NADPH treatment may alleviate IRI-induced apoptosis and inflammation. CONCLUSION NADPH treatment may protect against renal IRI and should be further developed as a new treatment for acute kidney injury.
Collapse
Affiliation(s)
- Xiao-Fen Weng
- The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Municipal Hospital, Suzhou, China
| | - Song-Tao Li
- People's Hospital of Huangjing, Suzhou, China
| | - Qi Song
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Zhu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Medicine, Suzhou, China
| | - Dan-Dan Song
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Medicine, Suzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Medicine, Suzhou, China
| | - Yan Xie
- The First Affiliated Hospital of Soochow University, Suzhou, China,
| |
Collapse
|
22
|
Zhu X, Shi J, li H, Chen F. Retracted Article: PVT1 knockdown alleviates vancomycin-induced acute kidney injury by targeting miR-124 via inactivation of NF-κB signaling. RSC Adv 2018; 8:31725-31734. [PMID: 35548198 PMCID: PMC9086227 DOI: 10.1039/c8ra05724a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/27/2018] [Indexed: 01/31/2023] Open
Abstract
Acute kidney injury (AKI) is a serious threat for human health and life. High dose of vancomycin (VAN) can give rise to AKI. The roles and molecular basis of long noncoding RNA plasmacytoma variant translocation 1 (PVT1) in VAN-induced AKI have been poorly defined till now. Protein levels of p65, phosphorylated p65 (p-p65), NF-κB inhibitor alpha (IκBα), phosphorylated IκBα (p-IκBα), Bcl-2 and Bax were measured by western blot assay. RNA levels of PVT1 and microRNA-124 (miR-124) were determined by RT-qPCR assay. HK-2 cell apoptosis was detected by an Annexin V-FITC apoptosis detection assay. Kidney functions were assessed by blood urea nitrogen (BUN) level, serum creatinine (Scr) level, histopathologic analysis, and TUNEL assay. Bioinformatical analysis, luciferase reporter assay, RIP and RNA pull down assays were performed to explore whether PVT1 could interact with miR-124. PVT1 was highly expressed in VAN-induced AKI models. Functional analysis revealed that PVT1 knockdown ameliorated VAN-induced AKI in vivo. Further exploration manifested that PVT1 directly interacted with miR-124. Moreover, the silencing of PVT1 abated VAN-induced HK-2 cell apoptosis in vitro, while this effect was reversed by the miR-124 inhibitor. Also, VAN treatment resulted in the reduction of miR-124 expression and the activation of NF-κB signaling in HK-2 cells. The inhibition of NF-κB alleviated VAN-induced HK-2 cell apoptosis. PVT1 activated NF-κB signaling by targeting miR-124 in VAN-induced HK-2 cells. PVT1 knockdown lessened VAN-induced AKI by targeting miR-124 via inactivating the NF-κB signaling, elucidating the critical roles and molecular basis of PVT1 in VAN-induced AKI and highlighting the diagnostic and therapeutic values of PVT1 in AKI. PVT1 was highly expressed in the kidneys of VAN-induced AKI mice.![]()
Collapse
Affiliation(s)
- Xiaoguang Zhu
- Department of Nephrology
- Huaihe Hospital of Henan University
- Kaifeng
- China
| | - Jun Shi
- Department of Nephrology
- Huaihe Hospital of Henan University
- Kaifeng
- China
| | - Huicong li
- Department of Nephrology
- Huaihe Hospital of Henan University
- Kaifeng
- China
| | - Fang Chen
- Department of Nephrology
- Huaihe Hospital of Henan University
- Kaifeng
- China
| |
Collapse
|
23
|
Luteolin Treatment Protects against Renal Ischemia-Reperfusion Injury in Rats. Mediators Inflamm 2017; 2017:9783893. [PMID: 29358852 PMCID: PMC5735687 DOI: 10.1155/2017/9783893] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/28/2017] [Accepted: 10/11/2017] [Indexed: 01/07/2023] Open
Abstract
Renal ischemia-reperfusion (I/R) injury is a common but severe scientific problem. Luteolin has great anti-inflammatory and antioxidant effects. In this study, we studied the effect of luteolin on renal I/R injury in rats. Intragastric administration of luteolin or saline was performed in Sprague-Dawley rats before (40 mg/kg for three days) and after (one day) renal I/R modeling. Kidney and blood samples were harvested to detect the severity of renal injury 24 hours after operation. The results showed that luteolin-treated rats exhibited milder histomorphological changes with lower scores of renal histological lesions; lower blood urea nitrogen and creatinine levels; lower renal malondialdehyde (MDA), 8-oxo-deoxyguanosine (8-OHdG), and myeloperoxidase (MPO) levels; and higher superoxide dismutase (SOD) and catalase (CAT) activities in the kidney. Luteolin attenuated the increased levels of serum and renal tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6, renal high mobility group box-1 (HMGB1), and nuclear factor kappa β (NF-κB) expression levels in I/R rats. Furthermore, luteolin treatment significantly reduced renal cell apoptosis and endoplasmic reticulum (ER) stress caused by renal I/R injury. In conclusion, luteolin improved renal function in I/R rats by reducing oxidative stress, neutrophil infiltration, inflammation, renal cell apoptosis, and expression of HMGB1 and NF-κB, and ER stress.
Collapse
|
24
|
Yamashita M, Yoshida T, Hayashi M. Podocyte NF-κB is dispensable for the pathogenesis of renal ischemia-reperfusion injury. Physiol Rep 2017; 4:4/16/e12912. [PMID: 27565904 PMCID: PMC5002916 DOI: 10.14814/phy2.12912] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/01/2016] [Indexed: 11/24/2022] Open
Abstract
Podocytes play a central role in the formation of the glomerular filtration barrier in the kidney, and their dysfunction has been shown to result in multiple proteinuric kidney diseases. In this study, we sought to determine whether NF-κB, a proinflammatory signaling, within podocytes was involved in renal ischemia-reperfusion (I/R) injury. Podocyte-specific IκBΔN transgenic (Pod-IκBΔN) mice, in which NF-κB was inhibited specifically in podocytes, were generated by the Cre-loxP technology, and their phenotype was compared with control mice after bilateral renal ischemia. The effect of systemic administration of a NF-κB inhibitor, pyrrolidinedithiocarbamate (PDTC), on renal I/R injury was also examined. Pod-IκBΔN mice were phenotypically normal before surgery. Following renal I/R injury, serum concentrations of urea nitrogen and creatinine were elevated in both Pod-IκBΔN and control mice to a similar extent, whereas PDTC treatment attenuated the elevation of these parameters. Renal histological damage in I/R-injured Pod-IκBΔN mice was also similar to I/R-injured control mice, although it was improved by PDTC treatment. Moreover, I/R induced accumulation of inflammatory cells, such as neutrophils and macrophages, was reduced by PDTC treatment, but not by podocyte-specific NF-κB inhibition. These results provide evidence that the NF-κB activity in podocytes does not contribute to the pathogenesis of renal I/R injury.
Collapse
Affiliation(s)
- Maho Yamashita
- Apheresis and Dialysis Center, School of Medicine, Keio University, Tokyo, Japan
| | - Tadashi Yoshida
- Apheresis and Dialysis Center, School of Medicine, Keio University, Tokyo, Japan Department of General Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Matsuhiko Hayashi
- Apheresis and Dialysis Center, School of Medicine, Keio University, Tokyo, Japan Department of General Medicine, School of Medicine, Keio University, Tokyo, Japan
| |
Collapse
|
25
|
Kim JY, An HJ, Kim WH, Gwon MG, Gu H, Park YY, Park KK. Anti-fibrotic Effects of Synthetic Oligodeoxynucleotide for TGF-β1 and Smad in an Animal Model of Liver Cirrhosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:250-263. [PMID: 28918026 PMCID: PMC5511593 DOI: 10.1016/j.omtn.2017.06.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/29/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022]
Abstract
Liver fibrosis is characterized by changes in tissue architecture and extracellular matrix composition. Liver fibrosis affects not only hepatocytes but also the non-parenchymal cells such as hepatic stellate cells (HSCs), which are essential for maintaining an intact liver structure and function. Transforming growth factor β1 (TGF-β1) is a multifunctional cytokine that induces liver fibrosis through activation of Smad signaling pathways. To improve a new therapeutic approach, synthetic TGF-β1/Smad oligodeoxynucleotide (ODN) was used to suppress both TGF-β1 expression and Smad transcription factor using a combination of antisense ODN and decoy ODN. The aims of this study are to investigate the anti-fibrotic effects of TGF-β1/Smad ODN on simultaneous suppressions of both Smad transcription factor and TGF-β1 mRNA expression in the hepatic fibrosis model in vitro and in vivo. Synthetic TGF-β1/Smad ODN effectively inhibits Smad binding activity and TGF-β1 expression. TGF-β1/Smad ODN attenuated the epithelial mesenchymal transition (EMT) and activation of HSCs in TGF-β1-induced AML12 and HSC-T6 cells. TGF-β1/Smad ODN prevented the fibrogenesis and deposition of collagen in CCl4-treated mouse model. Synthetic TGF-β1/Smad ODN demonstrates anti-fibrotic effects that are mediated by the suppression of fibrogenic protein and inflammatory cytokines. Therefore, synthetic TGF-β1/Smad ODN has substantial therapeutic feasibility for the treatment of liver fibrotic diseases.
Collapse
Affiliation(s)
- Jung-Yeon Kim
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Hyun-Jin An
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Woon-Hae Kim
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Mi-Gyeong Gwon
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Hyemin Gu
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Yoon-Yub Park
- Department of Physiology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Kwan-Kyu Park
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea.
| |
Collapse
|
26
|
Chen DZ, Chen LQ, Lin MX, Gong YQ, Ying BY, Wei DZ. Esculentoside A inhibits LPS-induced acute kidney injury by activating PPAR-γ. Microb Pathog 2017; 110:208-213. [PMID: 28666844 DOI: 10.1016/j.micpath.2017.06.037] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/18/2017] [Accepted: 06/26/2017] [Indexed: 12/11/2022]
Abstract
Acute kidney injury (AKI) is a major clinical problem associated with high morbidity and mortality. Esculentoside A (EsA), a kind of saponin isolated from the root of the Chinese herb Phytolaca esculenta, has been reported to have anti-inflammatory effect. In this study, we aimed to investigate the protective effects of EsA on LPS-induced AKI in mice. The protective effects of EsA was evaluated by detecting kidney histological change, blood urea nitrogen (BUN) and creatinine levels, and inflammatory cytokines production. The results showed that EsA significantly attenuated LPS-induced kidney histological change, as well as BUN and creatinine levels. EsA also inhibited LPS-induced TNF-α, IL-1β, and IL-6 production. LPS-induced NF-κB activation was significantly suppressed by treatment of EsA. In addition, EsA up-regulated the expression of PPAR-γ in a dose-dependent manner. In conclusion, EsA protected mice effectively from LPS-induced AKI by PPAR-γ, which subsequently inhibited LPS-induced inflammatory response.
Collapse
Affiliation(s)
- De-Zhun Chen
- Department of Anesthesiology, Critical Care, and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Li-Qiong Chen
- Department of Anesthesiology, Critical Care, and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Meng-Xiang Lin
- Department of Anesthesiology, Critical Care, and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yu-Qiang Gong
- Department of Anesthesiology, Critical Care, and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Bin-Yu Ying
- Department of Anesthesiology, Critical Care, and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Da-Zhen Wei
- Department of Anesthesiology, Critical Care, and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China.
| |
Collapse
|
27
|
Wei L, Zhang X, Ye Q, Yang Y, Chen X. The transfection of A20 gene prevents kidney from ischemia reperfusion injury in rats. Mol Med Rep 2017; 16:1486-1492. [PMID: 29067462 DOI: 10.3892/mmr.2017.6725] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 03/24/2017] [Indexed: 11/06/2022] Open
Abstract
Ischemia/reperfusion may induce inflammation and cell death through the nuclear factor (NF)‑κB signaling pathway. As a negative regulator of NF‑κB, zinc finger A20 exhibits anti-apoptotic and anti‑inflammatory effects in vitro. The present study was designed to upregulate A20 expression using an A20 transfection approach to investigate the in vivo protective effects of the A20 gene on renal ischemia reperfusion injury. The A20 gene was cloned into a pcDNA3.1 vector to construct the expression plasmid pcDNA3.1‑A20. The plasmid was wrapped with a liposome and injected intravenously into rats 48 h prior to establishing the models of renal ischemia reperfusion injury. Saline and the empty plasmid pcDNA3.1 were used as controls. Following 24 h post‑operation, A20 expression was determined using reverse transcription‑quantitative polymerase chain reaction and western blotting. The renal function and structure were assessed by analyzing the concentrations of serum creatinine (Scr), blood urea nitrogen (BUN) and histological features. Renal tissues were additionally examined for renal tubular cell apoptosis and NF‑κB activity. The results demonstrated that in vivo transfection of pcDNA3.1‑A20 induced renal A20 expression in rats. A20 overexpression in vivo significantly reduced renal injury as demonstrated by the improved levels of Scr and BUN and the reduction in histological damage. These improvements were accompanied by a suppression of renal proximal tubular epithelial cell apoptosis and an inhibition of NF‑κB activity. These results indicated that transfection of the A20 gene upregulates the expression of A20 in vivo and protects the kidneys from ischemia reperfusion injury via inhibition of the NF‑κB signal transduction pathway.
Collapse
Affiliation(s)
- Lixin Wei
- Department of Nephropathy, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Xianghui Zhang
- Department of Nephropathy, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, Guangdong 519000, P.R. China
| | - Qiuping Ye
- Department of Nephropathy, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Yueer Yang
- Department of Nephropathy, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Xiaowen Chen
- Department of Nephropathy, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
28
|
Innate Immune Response in Kidney Ischemia/Reperfusion Injury: Potential Target for Therapy. J Immunol Res 2017; 2017:6305439. [PMID: 28676864 PMCID: PMC5476886 DOI: 10.1155/2017/6305439] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 05/17/2017] [Indexed: 01/06/2023] Open
Abstract
Acute kidney injury caused by ischemia and subsequent reperfusion is associated with a high rate of mortality and morbidity. Ischemia/reperfusion injury in kidney transplantation causes delayed graft function and is associated with more frequent episodes of acute rejection and progression to chronic allograft nephropathy. Alloantigen-independent inflammation is an important process, participating in pathogenesis of injurious response, caused by ischemia and reperfusion. This innate immune response is characterized by the activity of classical cells belonging to the immune system, such as neutrophils, macrophages, dendritic cells, lymphocytes, and also tubular epithelial cells and endothelial cells. These immune cells not only participate in inflammation after ischemia exerting detrimental influence but also play a protective role in the healing response from ischemia/reperfusion injury. Delineating of complex mechanisms of their actions could be fruitful in future prevention and treatment of ischemia/reperfusion injury. Among numerous so far conducted experiments, observed immunomodulatory role of adenosine and adenosine receptor agonists in complex interactions of dendritic cells, natural killer T cells, and T regulatory cells is emphasized as promising in the treatment of kidney ischemia/reperfusion injury. Potential pharmacological approaches which decrease NF-κB activity and antagonize mechanisms downstream of activated Toll-like receptors are discussed.
Collapse
|
29
|
Peng J, Ren X, Lan T, Chen Y, Shao Z, Yang C. Renoprotective effects of ursolic acid on ischemia/reperfusion‑induced acute kidney injury through oxidative stress, inflammation and the inhibition of STAT3 and NF‑κB activities. Mol Med Rep 2016; 14:3397-402. [PMID: 27573738 DOI: 10.3892/mmr.2016.5654] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 06/22/2016] [Indexed: 11/05/2022] Open
Abstract
Ursolic acid, a pentacyclic triterpene compound with low toxicity and easy availability, has a variety of biological activities, including antitumor, antioxidant, antihepatitis, anti‑inflammatory and antibacterial effects. The present study aimed to investigate the renoprotective effects of ursolic acid on ischemia/reperfusion‑induced acute kidney injury (I/R‑IAKI) in rats associated with its antioxidant and anti‑inflammatory effects, as well as interference with the signal transducer and activator of transcription (STAT)3/nuclear factor (NF)‑κB signaling pathway. The present study demonstrated that pre‑treatment with ursolic acid significantly increased renal functioning and attenuated increases of serum angiotensin II levels in rats subjected to I/R‑IAKI. In addition, I/R‑IAKI‑induced inflammation and oxidative stress were significantly reduced by pre‑treatment with ursolic acid. Furthermore, ursolic acid significantly suppressed the upregulation of STAT3, NF‑κB and caspase‑3 activities in rats following I/R‑IAKI. These results indicated that ursolic acid may be a potential drug for reducing I/R‑IAKI through suppression of inflammation and oxidative stress damage, as well as modulation of STAT3 and NF‑κB activities.
Collapse
Affiliation(s)
- Jun Peng
- Department of Nephrology, Wuhan General Hospital of Guangzhou Military Area, Wuhan, Hubei 430030, P.R. China
| | - Xingfeng Ren
- Department of Nephrology, Wuhan General Hospital of Guangzhou Military Area, Wuhan, Hubei 430030, P.R. China
| | - Tianbiao Lan
- Department of Nephrology, Wuhan General Hospital of Guangzhou Military Area, Wuhan, Hubei 430030, P.R. China
| | - Yan Chen
- Department of Nephrology, Wuhan General Hospital of Guangzhou Military Area, Wuhan, Hubei 430030, P.R. China
| | - Ziyun Shao
- Department of Nephrology, Wuhan General Hospital of Guangzhou Military Area, Wuhan, Hubei 430030, P.R. China
| | - Cheng Yang
- Department of Nephrology, Wuhan General Hospital of Guangzhou Military Area, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
30
|
Guo L, Lee HH, Noriega MDLM, Paust HJ, Zahner G, Thaiss F. Lymphocyte-specific deletion of IKK2 or NEMO mediates an increase in intrarenal Th17 cells and accelerates renal damage in an ischemia-reperfusion injury mouse model. Am J Physiol Renal Physiol 2016; 311:F1005-F1014. [PMID: 27582100 DOI: 10.1152/ajprenal.00242.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 08/29/2016] [Indexed: 12/22/2022] Open
Abstract
Acute kidney injury (AKI) is associated with poor patient outcome and a global burden for end-stage renal disease. Ischemia-reperfusion injury (IRI) is one of the major causes of AKI, and experimental work has revealed many details of the inflammatory response in the kidney, such as activation of the NF-κB pathway. Here, we investigated whether deletion of the NF-κB kinases IKK2 or NEMO in lymphocytes or systemic inhibition of IKK2 would cause different kidney inflammatory responses after IRI induction. Serum creatinine, blood urea nitrogen (BUN) level, and renal tubular injury score were significantly increased in CD4creIKK2f/f (CD4xIKK2Δ) and CD4creNEMOf/f (CD4xNEMOΔ) mice compared with CD4cre mice after IRI induction. The frequency of Th17 cells infiltrating the kidneys of CD4xIKK2Δ or CD4xNEMOΔ mice was also significantly increased at all time points. CCL20, an important chemokine in Th17 cell recruitment, was significantly increased at early time points after the induction of IRI. IL-1β, TNF-α, and CCL2 were also significantly increased in different patterns. A specific IKK2 inhibitor, KINK-1, reduced BUN and serum creatinine compared with nontreated mice after IRI induction, but the frequency of kidney Th17 cells was also significantly increased. In conclusion, although systemic IKK2 inhibition improved kidney function, lymphocyte-specific deletion of IKK2 or NEMO aggravated kidney injury after IRI, and, in both conditions, the percentage of Th17 cells was increased. Our findings demonstrate the critical role of the NF-κB pathway in Th17 activation, which advises caution when using systemic IKK2 inhibitors in patients with kidney injury, since they might impair the T cell response and aggravate renal disease.
Collapse
Affiliation(s)
- Linlin Guo
- III Medizinische Klinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Hannah Heejung Lee
- III Medizinische Klinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | | | - Hans J Paust
- III Medizinische Klinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Gunther Zahner
- III Medizinische Klinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Thaiss
- III Medizinische Klinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
31
|
Dai Y, Jia P, Fang Y, Liu H, Jiao X, He JC, Ding X. miR-146a is essential for lipopolysaccharide (LPS)-induced cross-tolerance against kidney ischemia/reperfusion injury in mice. Sci Rep 2016; 6:27091. [PMID: 27250735 PMCID: PMC4890025 DOI: 10.1038/srep27091] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 05/09/2016] [Indexed: 12/31/2022] Open
Abstract
MicroRNA-146a is one of most important microRNAs involved in development of endotoxin tolerance via (toll-like receptors) TLRs/ NF-κB pathway. In this study, we sought to identify the mechanistic role of miR-146a in mediating the protective effect of lipopolysaccharide (LPS) pretreatment on kidney ischemia/reperfusion injury. A locked nucleic acid–modified anti-miR-146a given before LPS treatment knocked down miR-146a expression and completely negated LPS-mediated protection against kidney ischemia/reperfusion injury. Knockdown of miR-146a resulted in significantly higher histopathological scores for tubular damage, expression of proinflammatory cytokines and chemokines, and neutrophil and macrophage infiltration. Furthermore, knockdown of miR-146a greatly up-regulated the protein levels of IL-1 receptor-associated kinase (IRAK-1) and tumor-necrosis factor (TNF) receptor-associated factor 6 (TRAF6), which are known target genes of miR-146a, leading to activation of NF-κB. Finally, elevation of nuclear translocation of NF-κB p65/p50 and caspase-3 expression, degradation of cytosolic IkBα and BcL-xL, and substantially exacerbation of tubular cell apoptosis were inversely correlated with miR-146a expression. Taken together, our results identify that miR146a exerts a kidney protective effect through negative regulation of acute inflammatory response by suppressing NF-κB activation and proinflammatory genes expression.
Collapse
Affiliation(s)
- Yan Dai
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China.,Kidney and Dialysis Institute of Shanghai, Shanghai, China.,Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China
| | - Ping Jia
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China.,Kidney and Dialysis Institute of Shanghai, Shanghai, China.,Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China
| | - Yi Fang
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hong Liu
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyan Jiao
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China.,Kidney and Dialysis Institute of Shanghai, Shanghai, China.,Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China
| | - John C He
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Kidney Section, James J Peter Veteran Administration Medical Center at Bronx, NY, United States
| | - Xiaoqiang Ding
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China.,Kidney and Dialysis Institute of Shanghai, Shanghai, China.,Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China
| |
Collapse
|
32
|
Mitochondria-Targeted Antioxidants: Future Perspectives in Kidney Ischemia Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:2950503. [PMID: 27313826 PMCID: PMC4894993 DOI: 10.1155/2016/2950503] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 04/28/2016] [Indexed: 02/03/2023]
Abstract
Kidney ischemia/reperfusion injury emerges in various clinical settings as a great problem complicating the course and outcome. Ischemia/reperfusion injury is still an unsolved puzzle with a great diversity of investigational approaches, putting the focus on oxidative stress and mitochondria. Mitochondria are both sources and targets of ROS. They participate in initiation and progression of kidney ischemia/reperfusion injury linking oxidative stress, inflammation, and cell death. The dependence of kidney proximal tubule cells on oxidative mitochondrial metabolism makes them particularly prone to harmful effects of mitochondrial damage. The administration of antioxidants has been used as a way to prevent and treat kidney ischemia/reperfusion injury for a long time. Recently a new method based on mitochondria-targeted antioxidants has become the focus of interest. Here we review the current status of results achieved in numerous studies investigating these novel compounds in ischemia/reperfusion injury which specifically target mitochondria such as MitoQ, Szeto-Schiller (SS) peptides (Bendavia), SkQ1 and SkQR1, and superoxide dismutase mimics. Based on the favorable results obtained in the studies that have examined myocardial ischemia/reperfusion injury, ongoing clinical trials investigate the efficacy of some novel therapeutics in preventing myocardial infarct. This also implies future strategies in preventing kidney ischemia/reperfusion injury.
Collapse
|
33
|
Yang Z, Zhong Z, Li M, Xiong Y, Wang Y, Peng G, Ye Q. Hypothermic machine perfusion increases A20 expression which protects renal cells against ischemia/reperfusion injury by suppressing inflammation, apoptosis and necroptosis. Int J Mol Med 2016; 38:161-71. [PMID: 27177159 PMCID: PMC4899006 DOI: 10.3892/ijmm.2016.2586] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 04/21/2016] [Indexed: 12/14/2022] Open
Abstract
There is an urgent need to improve the quality of donor organs obtained after cardiac death. In the present study, we examined the potential mechanisms through which A20 protects renal cells against ischemia/reperfusion injury (IRI) following either hypothermic machine perfusion (HMP) or static cold storage (CS) of the kidneys in a rabbit model. The expression of markers of apoptosis, necroptosis and inflammation in frozen kidney tissues were detected by western blot analysis, RT-qPCR and ELISA. Compared with the CS group, A20 expression was significantly higher in the tissue from the HMP group (P<0.01). By contrast, the expression of nuclear factor-κB (NF-κB) and tumor necrosis factor-α (TNF-α) was significantly lower in HMP group (P<0.01), whereas IκBα expression was significantly higher (P<0.01). The expression of apoptosis signal-regulating kinase 1 (ASK1), phosphorylated (p-)c-Jun N-terminal kinase (JNK) and activated caspase-3 in the HMP group was significantly downregulated compared with that in the CS group (all P<0.01). In addition, A20 inhibited receptor-interacting protein kinase 3 (RIPK3)-mediated necroptosis in the kidney. RIPK3 expression in the HMP group was significantly lower than that in the CS group (P<0.01), although the levels in both groups were higher than those in the sham group (P<0.01). Based on these findings, we propose a novel mechanism underlying the anti-apoptotic effect of A20 in renal cells in which A20 binds to ASK1 and promotes the degradation of ASK1 leading to the suppression of JNK activation and eventually, to the blockade of apoptosis. Thus, HMP reduces inflammation, apoptosis and necroptosis by upregulating the expression of A20; this mechanism may be responsible for protecting the kidney against IRI.
Collapse
Affiliation(s)
- Zixuan Yang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Zibiao Zhong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Mingxia Li
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Yan Xiong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Guizhu Peng
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
34
|
Yang Y, Song M, Liu Y, Liu H, Sun L, Peng Y, Liu F, Venkatachalam MA, Dong Z. Renoprotective approaches and strategies in acute kidney injury. Pharmacol Ther 2016; 163:58-73. [PMID: 27108948 DOI: 10.1016/j.pharmthera.2016.03.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 03/18/2016] [Indexed: 12/17/2022]
Abstract
Acute kidney injury (AKI) is a major renal disease associated with high mortality rate and increasing prevalence. Decades of research have suggested numerous chemical and biological agents with beneficial effects in AKI. In addition, cell therapy and molecular targeting have been explored for reducing kidney tissue damage and promoting kidney repair or recovery from AKI. Mechanistically, these approaches may mitigate oxidative stress, inflammation, cell death, and mitochondrial and other organellar damage, or activate cytoprotective mechanisms such as autophagy and pro-survival factors. However, none of these findings has been successfully translated into clinical treatment of AKI. In this review, we analyze these findings and propose experimental strategies for the identification of renoprotective agents or methods with clinical potential. Moreover, we propose the consideration of combination therapy by targeting multiple targets in AKI.
Collapse
Affiliation(s)
- Yuan Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Meifang Song
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Youming Peng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fuyou Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | | | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
35
|
Markó L, Vigolo E, Hinze C, Park JK, Roël G, Balogh A, Choi M, Wübken A, Cording J, Blasig IE, Luft FC, Scheidereit C, Schmidt-Ott KM, Schmidt-Ullrich R, Müller DN. Tubular Epithelial NF-κB Activity Regulates Ischemic AKI. J Am Soc Nephrol 2016; 27:2658-69. [PMID: 26823548 DOI: 10.1681/asn.2015070748] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 11/26/2015] [Indexed: 12/20/2022] Open
Abstract
NF-κB is a key regulator of innate and adaptive immunity and is implicated in the pathogenesis of AKI. The cell type-specific functions of NF-κB in the kidney are unknown; however, the pathway serves distinct functions in immune and tissue parenchymal cells. We analyzed tubular epithelial-specific NF-κB signaling in a mouse model of ischemia-reperfusion injury (IRI)-induced AKI. NF-κB reporter activity and nuclear localization of phosphorylated NF-κB subunit p65 analyses in mice revealed that IRI induced widespread NF-κB activation in renal tubular epithelia and in interstitial cells that peaked 2-3 days after injury. To genetically antagonize tubular epithelial NF-κB activity, we generated mice expressing the human NF-κB super-repressor IκBαΔN in renal proximal, distal, and collecting duct epithelial cells. Compared with control mice, these mice exhibited improved renal function, reduced tubular apoptosis, and attenuated neutrophil and macrophage infiltration after IRI-induced AKI. Furthermore, tubular NF-κB-dependent gene expression profiles revealed temporally distinct functional gene clusters for apoptosis, chemotaxis, and morphogenesis. Primary proximal tubular cells isolated from IκBαΔN-expressing mice and exposed to hypoxia-mimetic agent cobalt chloride exhibited less apoptosis and expressed lower levels of chemokines than cells from control mice did. Our results indicate that postischemic NF-κB activation in renal tubular epithelia aggravates tubular injury and exacerbates a maladaptive inflammatory response.
Collapse
Affiliation(s)
- Lajos Markó
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany; Max Delbrück Center for Molecular Medicine, Berlin, Germany;
| | - Emilia Vigolo
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | | | - Giulietta Roël
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - András Balogh
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany; Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Mira Choi
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Anne Wübken
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Jimmi Cording
- Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany; and
| | - Ingolf E Blasig
- Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany; and
| | - Friedrich C Luft
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany; Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | - Kai M Schmidt-Ott
- Max Delbrück Center for Molecular Medicine, Berlin, Germany; Department of Nephrology, Charité Medical Faculty, Berlin, Germany
| | | | - Dominik N Müller
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany; Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
36
|
O'Neill S, Humphries D, Tse G, Marson LP, Dhaliwal K, Hughes J, Ross JA, Wigmore SJ, Harrison EM. Heat shock protein 90 inhibition abrogates TLR4-mediated NF-κB activity and reduces renal ischemia-reperfusion injury. Sci Rep 2015; 5:12958. [PMID: 26248657 PMCID: PMC4528191 DOI: 10.1038/srep12958] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 07/06/2015] [Indexed: 12/17/2022] Open
Abstract
Renal ischemia-reperfusion injury (IRI) is a common cause of acute kidney injury. Toll-like receptor 4 (TLR4) mediates sterile inflammation following renal IRI. Heat shock protein 90 (Hsp90) inhibition is a potential strategy to reduce IRI, and AT13387 is a novel Hsp90 inhibitor with low toxicity. This study assessed if pre-treatment with AT13387 could reduce renal IRI and established if the mechanism of protection involved a reduction in inflammatory signalling. Mice were pre-treated with AT13387 prior to renal IRI. 24 h later, renal function was determined by serum creatinine, kidney damage by tubular necrosis score, renal TLR4 expression by PCR and inflammation by cytokine array. In vitro, human embryonic kidney cells were co-transfected to express TLR4 and a secreted alkaline phosphatase NF-κB reporter. Cells were pre-treated with AT13387 and exposed to endotoxin-free hyaluronan to stimulate sterile TLR4-specific NF-κB inflammatory activation. Following renal IRI, AT13387 significantly reduced serum creatinine, tubular necrosis, TLR4 expression and NF-κB-dependent chemokines. In vitro, AT13387-treatment resulted in breakdown of IκB kinase, which abolished TLR4-mediated NF-κB activation by hyaluronan. AT13387 is a new agent with translational potential that reduces renal IRI. The mechanism of protection may involve breakdown of IκB kinase and repression of TLR4-mediated NF-κB inflammatory activity.
Collapse
Affiliation(s)
- Stephen O'Neill
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, EH16 4SA
| | - Duncan Humphries
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, EH16 4SA
| | - George Tse
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, EH16 4SA
| | - Lorna P Marson
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, EH16 4SA
| | - Kevin Dhaliwal
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, EH16 4SA
| | - Jeremy Hughes
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, EH16 4SA
| | - James A Ross
- MRC Centre for Regenerative Medicine, University of Edinburgh, Royal Infirmary of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SA
| | - Stephen J Wigmore
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, EH16 4SA
| | - Ewen M Harrison
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, EH16 4SA
| |
Collapse
|
37
|
Wan X, Hou LJ, Zhang LY, Huang WJ, Liu L, Zhang Q, Hu B, Chen W, Chen X, Cao CC. IKKα is involved in kidney recovery and regeneration of acute ischemia/reperfusion injury in mice through IL10-producing regulatory T cells. Dis Model Mech 2015; 8:733-42. [PMID: 26035380 PMCID: PMC4486855 DOI: 10.1242/dmm.018200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 04/17/2015] [Indexed: 12/30/2022] Open
Abstract
The recovery phase after kidney ischemia/reperfusion (IR) injury is often associated with the suppression of inflammation and the proliferation of tubular epithelial cells (TECs). The duration of this phase is often determined by the suppression of inflammation and the proliferation of TECs. Several lines of evidence suggest that IκB kinase α (IKKα) not only promotes the production of anti-inflammatory factors and/or prevents the production of inflammatory factors, but also induces the accompanying cell differentiation and regeneration, and suppresses inflammation. We therefore hypothesized that IKKα could participate in the kidney repair after IR injury and have used a mouse model of acute kidney injury (AKI) to test this. We found that IKKα mediated the repair of the kidney via infiltrated regulatory T (Treg) cells, which can produce anti-inflammatory cytokine IL10, and that IKKα also increased the proliferation of the surviving TECs and suppressed of inflammation. In addition, the expression of indoleamine 2,3-dioxygenase (IDO) in TECs is consistent with the infiltration of IL10-producing Treg cells. We conclude that IKKα is involved in kidney recovery and regeneration through the Treg cells that can produce IL10, which might be a potential therapeutic target that can be used to promote kidney repair after IR injury.
Collapse
Affiliation(s)
- Xin Wan
- Division of Nephrology, Department of Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Li-Jun Hou
- Division of Neurosurgery, Department of Surgery and Shanghai Neurosurgical Institute, The Second Military Medical University, Changzheng Hospital, Shanghai 200003, China
| | - Li-Yuan Zhang
- Division of Nephrology, Department of Medicine, Affiliated Lianyungang Clinical Medical College of Nanjing Medical University, Lianyungang 222002, China
| | - Wen-Juan Huang
- Division of Nephrology, Department of Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Lin Liu
- Division of Nephrology, Department of Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Qian Zhang
- Division of Nephrology, Department of Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Bo Hu
- Division of Nephrology, Department of Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Wen Chen
- Division of Cardiovascular Surgery, Department of Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xin Chen
- Division of Cardiovascular Surgery, Department of Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Chang-Chun Cao
- Division of Nephrology, Department of Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| |
Collapse
|
38
|
Kumar D, Singla SK, Puri V, Puri S. The restrained expression of NF-kB in renal tissue ameliorates folic acid induced acute kidney injury in mice. PLoS One 2015; 10:e115947. [PMID: 25559736 PMCID: PMC4283964 DOI: 10.1371/journal.pone.0115947] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 11/27/2014] [Indexed: 01/13/2023] Open
Abstract
The Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) represent family of structurally-related eukaryotic transcription factors which regulate diverse array of cellular processes including immunological responses, inflammation, apoptosis, growth & development. Increased expression of NF-kB has often been seen in many diverse diseases, suggesting the importance of genomic deregulation to disease pathophysiology. In the present study we focused on acute kidney injury (AKI), which remains one of the major risk factor showing a high rate of mortality and morbidity. The pathology associated with it, however, remains incompletely known though inflammation has been reported to be one of the major risk factor in the disease pathophysiology. The role of NF-kB thus seemed pertinent. In the present study we show that high dose of folic acid (FA) induced acute kidney injury (AKI) characterized by elevation in levels of blood urea nitrogen & serum creatinine together with extensive tubular necrosis, loss of brush border and marked reduction in mitochondria. One of the salient observations of this study was a coupled increase in the expression of renal, relA, NF-kB2, and p53 genes and proteins during folic acid induced AKI (FA AKI). Treatment of mice with NF-kB inhibitor, pyrrolidine dithio-carbamate ammonium (PDTC) lowered the expression of these transcription factors and ameliorated the aberrant renal function by decreasing serum creatinine levels. In conclusion, our results suggested that NF-kB plays a pivotal role in maintaining renal function that also involved regulating p53 levels during FA AKI.
Collapse
Affiliation(s)
- Dev Kumar
- Department of Biochemistry, Panjab University, Chandigarh, India
| | | | - Veena Puri
- Centre for Systems Biology & Bioinformatics, Panjab University, Chandigarh, India
| | - Sanjeev Puri
- Biotechnology Branch, University Institute of Engineering & Technology, Panjab University, Chandigarh, India
- Centre for Stem Cell & Tissue Engineering, Panjab University, Chandigarh, India
- * E-mail:
| |
Collapse
|
39
|
Abstract
Acute kidney injury (AKI) prolongs hospital stay and increases mortality in various clinical settings. Ischaemia-reperfusion injury (IRI), nephrotoxic agents and infection leading to sepsis are among the major causes of AKI. Inflammatory responses substantially contribute to the overall renal damage in AKI. Both innate and adaptive immune systems are involved in the inflammatory process occurring in post-ischaemic AKI. Proinflammatory damage-associated molecular patterns, hypoxia-inducible factors, adhesion molecules, dysfunction of the renal vascular endothelium, chemokines, cytokines and Toll-like receptors are involved in the activation and recruitment of immune cells into injured kidneys. Immune cells of both the innate and adaptive immune systems, such as neutrophils, dendritic cells, macrophages and lymphocytes contribute to the pathogenesis of renal injury after IRI, and some of their subpopulations also participate in the repair process. These immune cells are also involved in the pathogenesis of nephrotoxic AKI. Experimental studies of immune cells in AKI have resulted in improved understanding of the immune mechanisms underlying AKI and will be the foundation for development of novel diagnostic and therapeutic targets. This Review describes what is currently known about the function of the immune system in the pathogenesis and repair of ischaemic and nephrotoxic AKI.
Collapse
Affiliation(s)
- Hye Ryoun Jang
- Nephrology Division, Department of Medicine, Samsung Medical Centre, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 81 Irwon-Ro Gangnam-gu, Seoul 135-710, South Korea
| | - Hamid Rabb
- Nephrology Division, Department of Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA
| |
Collapse
|
40
|
Shi Z, Lian A, Zhang F. Nuclear factor-κB activation inhibitor attenuates ischemia reperfusion injury and inhibits Hmgb1 expression. Inflamm Res 2014; 63:919-25. [PMID: 25209109 DOI: 10.1007/s00011-014-0765-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 06/08/2014] [Accepted: 07/26/2014] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE AND DESIGN To investigate the effects of nuclear factor-κB activation inhibitor dehydroxymethylepoxyquinomicin (DHMEQ) on cardiac ischemia reperfusion injury in a transplantation model. METHODS Hearts of C57BL/6 mice were flushed and stored in cold Bretschneider solution for 8 h and then transplanted into syngeneic recipient. Some mice were administrated intraperitoneally with DHMEQ (8 mg/kg) 1 h before reperfusion. For inhibition of Hmgb1, mice were treated with glycyrrhizin at 250 mg/kg prior to reperfusion. RESULTS DHMEQ decreased cardiomyocyte apoptosis and recruitment of neutrophils and macrophages. Troponin T (TnT) production on 24 h after myocardial IR injury was reduced by DHMEQ treatment. Cardiac output at 60 mmHg of afterload pressure was significantly increased in hearts with DHMEQ treatment (IR+DHMEQ: 58.6 ± 5.75 ml/min; IR: 25.9 ± 4.1 ml/min; P < 0.05). Furthermore, DHMEQ suppressed high mobility group protein (Hmgb1) expression. And the Caspase 3 activity, the number of TUNEL-positive cardiomyocytes and infiltrated neutrophil in cardiac allograft were markedly decreased with Hmgb1 inhibitor treatment. CONCLUSIONS Nuclear factor-κB activation inhibitor DHMEQ attenuates ischemia reperfusion injury in a cardiac transplantation model and it may be a suitable agent for the protection of the cardiac against ischemia reperfusion injury.
Collapse
Affiliation(s)
- Zhiyong Shi
- Department of Cardiothoracic Surgery, China Pingmei Shenma Medical Group General Hospital, Pingdingshan Kuanggong Road 1#, Henan, China,
| | | | | |
Collapse
|
41
|
Xu X, Kriegel AJ, Jiao X, Liu H, Bai X, Olson J, Liang M, Ding X. miR-21 in ischemia/reperfusion injury: a double-edged sword? Physiol Genomics 2014; 46:789-97. [PMID: 25159851 DOI: 10.1152/physiolgenomics.00020.2014] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
MicroRNAs (miRNAs or miRs) are endogenous, small RNA molecules that suppress expression of targeted mRNA. miR-21, one of the most extensively studied miRNAs, is importantly involved in divergent pathophysiological processes relating to ischemia/reperfusion (I/R) injury, such as inflammation and angiogenesis. The role of miR-21 in renal I/R is complex, with both protective and pathological pathways being regulated by miR-21. Preconditioning-induced upregulation of miR-21 contributes to the protection against subsequent renal I/R injury through the targeting of genes such as the proapoptotic gene programmed cell death 4 and interactions between miR-21 and hypoxia-inducible factor. Conversely, long-term elevation of miR-21 may be detrimental to the organ by promoting the development of renal interstitial fibrosis following I/R injury. miR-21 is importantly involved in several pathophysiological processes related to I/R injury including inflammation and angiogenesis as well as the biology of stem cells that could be used to treat I/R injury; however, the effect of miR-21 on these processes in renal I/R injury remains to be studied.
Collapse
Affiliation(s)
- Xialian Xu
- Division of Nephrology, Fudan University Zhongshan Hospital, Shanghai, Peoples Republic of China
| | - Alison J Kriegel
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Xiaoyan Jiao
- Division of Nephrology, Fudan University Zhongshan Hospital, Shanghai, Peoples Republic of China
| | - Hong Liu
- Division of Nephrology, Fudan University Zhongshan Hospital, Shanghai, Peoples Republic of China
| | - Xiaowen Bai
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jessica Olson
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mingyu Liang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Xiaoqiang Ding
- Division of Nephrology, Fudan University Zhongshan Hospital, Shanghai, Peoples Republic of China; Institutes of Biomedical Sciences of Shanghai Medical School, Fudan University, Shanghai, Peoples Republic of China; Kidney and Dialysis Institute of Shanghai, Shanghai, Peoples Republic of China; and Kidney and Blood Purification Laboratory of Shanghai, Shanghai, Peoples Republic of China
| |
Collapse
|
42
|
Kim KH, Lee WR, Kang YN, Chang YC, Park KK. Inhibitory effect of nuclear factor-κB decoy oligodeoxynucleotide on liver fibrosis through regulation of the epithelial-mesenchymal transition. Hum Gene Ther 2014; 25:721-729. [PMID: 24959740 PMCID: PMC4137351 DOI: 10.1089/hum.2013.106] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 06/20/2014] [Indexed: 01/11/2023] Open
Abstract
The epithelial-mesenchymal transition (EMT) has been recognized to occur during embryonic development, fibrosis, and tumor metastasis. Nuclear factor (NF)-κB plays a central role in mediating the inflammation and wound-healing responses during liver fibrogenesis. However, the involvement of NF-κB during EMT in liver cells remains unidentified. To develop a therapeutic approach to EMT during liver fibrosis, we examined the inhibition of transcription factor NF-κB, using a decoy oligodeoxynucleotide (ODN) strategy in liver fibrosis in vitro and in vivo. NF-κB decoy ODN contains consensus binding sequences of the NF-κB-binding site. NF-κB decoy ODN effectively suppresses transforming growth factor-β(1)-induced EMT in AML12 murine hepatocytes. Liver fibrosis induced by CCl(4) administration was suppressed by NF-κB decoy ODN. Furthermore, NF-κB decoy ODN was shown to inhibit the EMT process in fibrotic liver in vivo. This study demonstrates the feasibility of NF-κB decoy ODN treatment for preventing liver fibrosis via EMT processes.
Collapse
Affiliation(s)
- Kyung-Hyun Kim
- Department of Pathology, Catholic University of Daegu, College of Medicine, 3056-6 Daemyung 4-dong, Nam-gu, Daegu 705-718, Republic of Korea
| | - Woo-Ram Lee
- Department of Pathology, Catholic University of Daegu, College of Medicine, 3056-6 Daemyung 4-dong, Nam-gu, Daegu 705-718, Republic of Korea
| | - Yu-Na Kang
- Department of Pathology, Keimyung University, School of Medicine, Dalseong-ro 56, Jung-gu, Daegu 700-712, Republic of Korea
| | - Young-Chae Chang
- Department of Pathology, Catholic University of Daegu, College of Medicine, 3056-6 Daemyung 4-dong, Nam-gu, Daegu 705-718, Republic of Korea
| | - Kwan-Kyu Park
- Department of Pathology, Catholic University of Daegu, College of Medicine, 3056-6 Daemyung 4-dong, Nam-gu, Daegu 705-718, Republic of Korea
| |
Collapse
|
43
|
Abstract
China has a large population and bears a heavy burden of kidney diseases. Acute kidney injury (AKI) is increasingly prevalent in China and is associated with severe morbidity and mortality. The inadequacies of early diagnosis and management remain the major challenges to Chinese nephrologists. There is an urgent need for a nationwide, or even a global effort to increase an awareness of the devastating effects of AKI, to develop professional preventive strategies, as well as to achieve early diagnosis and early intervention. In this article, we review the increasing incidence of AKI and complexity behind prevention and management, focusing on differences in various clinical settings in China.
Collapse
Affiliation(s)
- Yi Fang
- Division of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | |
Collapse
|
44
|
Abstract
Many types of kidney injury induce inflammation as a protective response. However, unresolved inflammation promotes progressive renal fibrosis, which can culminate in end-stage renal disease. Kidney inflammation involves cells of the immune system as well as activation of intrinsic renal cells, with the consequent production and release of profibrotic cytokines and growth factors that drive the fibrotic process. In glomerular diseases, the development of glomerular inflammation precedes interstitial fibrosis; although the mechanisms linking these events are poorly understood, an important role for tubular epithelial cells in mediating this link is gaining support. Data have implicated macrophages in promoting both glomerular and interstitial fibrosis, whereas limited evidence suggests that CD4(+) T cells and mast cells are involved in interstitial fibrosis. However, macrophages can also promote renal repair when the cause of renal injury can be resolved, highlighting their plasticity. Understanding the mechanisms by which inflammation drives renal fibrosis is necessary to facilitate the development of therapeutics to halt the progression of chronic kidney disease.
Collapse
|
45
|
Xue C, Liu Y, Li C, Li Y, Yang T, Xie L, Zhou P. Powerful protection against renal ischemia reperfusion injury by T cell-specific NF-κB inhibition. Transplantation 2014; 97:391-6. [PMID: 24398854 DOI: 10.1097/01.tp.0000438622.89310.95] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND NF-κB plays a key role in ischemia reperfusion injury (IRI). Systemic inhibition of NF-κB by various methods has been proven to ameliorate IRI. However, NF-κB is also responsible for tissue protection against IRI. Systemic NF-κB inhibition may not be the optimal way for preventing IRI because of its complex roles. T cells are essential in mediating IRI. NF-κB is an important molecule during T cell activation. It is not clear the effect of T cell-specific NF-κB inhibition on IRI. We aimed to study the effect of T cell-specific NF-κB inhibition on renal IRI in IκBαΔN-Tg mice. We also compared the different effects between T cell-specific and systemic NF-κB inhibition on IRI. METHODS Renal IRI was induced by left renal pedicle clamping for 60 or 80 min in wild-type, ursolic acid-treated or IκBαΔN-Tg mice. Renal function, histologic examination and overall survival after lethal IRI was evaluated in each group. RESULTS Serum creatinine, BUN, and pathologic damage were all reduced in IκBαDN-Tg mice and ursolic acid-treated mice than those in the control group. All the above indexes were improved better in IκBαDN-Tg mice than those in ursolic acid-treated mice. The survival rate of IκBαDN-Tg mice was higher than that of ursolic acid-treated mice after lethal kidney ischemia reperfusion injury. Immunohistochemistry showed a significant reduced CD4+ T cells and neutrophil infiltration in IκBαDN-Tg mice. CONCLUSION T cell-specific NF-κB inhibition provides powerful protective effect against renal IRI.
Collapse
Affiliation(s)
- ChengBiao Xue
- 1 Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Ministry of Health, and Key Laboratory of Ministry of Education, China. 2 Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. 3 Address correspondence to: Ping Zhou, 1095 Jiefang Road, Tongji Hospital, Institute of Organ Transplantation, Wuhan, Hubei 430030, China
| | | | | | | | | | | | | |
Collapse
|
46
|
The effect of mTOR-inhibition on NF-κB activity in kidney ischemia-reperfusion injury in mice. Transplant Proc 2014; 45:1708-14. [PMID: 23769029 DOI: 10.1016/j.transproceed.2013.02.110] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 02/07/2013] [Accepted: 02/27/2013] [Indexed: 02/05/2023]
Abstract
Kidney ischemia-reperfusion injury (IRI) is associated with a robust inflammatory response, which is regulated by nuclear factor-kappaB (NF-κB), mainly its heterodimeric form p65/p50. Considering immunomodulatory properties of mammalian target of rapamycin (mTOR) inhibitors, the effect of everolimus on NF-κB activation in kidney IRI was determined in this study. IRI was induced in C57/BL6 mice by clamping both renal pedicles for 45 minutes. Application of everolimus (0.25 mg/kg bw subcutaneously daily) was started one day before IRI induction. Both everolimus-treated and nontreated mice were sacrificed at several times starting at 30 minutes and finishing on day 7 after IRI induction. The NF-κB activity, proinflammatory cytokines IL-1β, TNF-α, and anti-inflammatory cytokine IL-10 production were determined in kidneys. Compared with nontreated animals, everolimus-treated animals showed significantly increased TNF-α (2741.6 ± 201.72 pg/mg; 1925 ± 185.81 pg/mg, P < .05) and IL-1β (11.47 ± 1.2 pg/mg; 4.3 ± 0.13 pg/mg, P < .01) production on day 2 after IRI induction accompanied by significantly greater NF-κB/DNA binding activity and p65 nuclear expression (P < .01). Two hours after IRI induction, everolimus-treated animals showed significantly increased IL-1β mRNA expression (P < .05) followed by increased IL-1β protein concentrations when compared with nontreated animals measured 6 hours after IRI induction (11.71 ± 1.5 pg/mg; 7.5 ± 1.11 pg/mg, P < .01). Both experimental groups showed increased NF-κB/DNA binding activity at 7 days after IRI induction. Significantly increased nuclear p65 expression was measured in nontreated animals (P < .01), whereas everolimus-treated hosts showed significantly increased nuclear RelB expression (P < .01). These data suggested that everolimus potentiated innate immunity in the early phase of IRI, stimulating the production of NF-κB-driven proinflammatory cytokines such as TNF-α and IL-1β. The NF-κB activity was potentiated under m-TOR inhibition during kidney IRI, implicating a possible beneficial role of alternative NF-κB activation during the repair phase.
Collapse
|
47
|
Effect of a novel nuclear factor-κB activation inhibitor on renal ischemia-reperfusion injury. Transplantation 2014; 96:863-70. [PMID: 23958925 DOI: 10.1097/tp.0b013e3182a3df74] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND In kidney transplantation, the relationship between prolonged warm or cold ischemic storage of kidneys and a higher incidence of delayed graft function is previously known, and delayed graft function has been known to aggravate poor long-term graft survival. We investigated the effect of a novel nuclear factor-κB activation inhibitor, dehydroxymethylepoxyquinomicin (DHMEQ), on renal ischemia-reperfusion (I/R) injury. METHODS DHMEQ was administered to Lewis rats once just before renal artery clamping (DHMEQ pretreatment group), and the effect on I/R injury was investigated. RESULTS In the DHMEQ pretreatment group, the 24-hr urine volume on days 1 to 3 after I/R was significantly larger, and the protein concentration of the urine on days 2 to 7 was significantly smaller than in the untreated group. The serum creatinine level was significantly improved, and significantly lower levels of the inflammatory cells and inflammatory cytokines were present in the kidneys on day 1. The relative ratio of nuclear to cytoplasmic nuclear factor-κB and oxidative stress of kidney tissue on day 1 were significantly decreased. CONCLUSIONS Treatment with DHMEQ before renal artery clamping may therefore be useful for renal I/R injury and application to renal transplantation is expected.
Collapse
|
48
|
Sleeman P, Patel NN, Lin H, Walkden GJ, Ray P, Welsh GI, Satchell SC, Murphy GJ. High fat feeding promotes obesity and renal inflammation and protects against post cardiopulmonary bypass acute kidney injury in swine. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2013; 17:R262. [PMID: 24172587 PMCID: PMC4056797 DOI: 10.1186/cc13092] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Accepted: 10/10/2013] [Indexed: 01/07/2023]
Abstract
Introduction Obesity confers a survival advantage in the critically ill and in patients undergoing cardiac surgery. We explored whether an obesogenic high fat diet could confer protection against post cardiopulmonary bypass (CPB) acute kidney injury (AKI) in a swine model. Methods In this study, 28 anaesthetised adult female Landrace White swine (55 to 70 kg) were allocated into a 4 group design to either 2.5 hours of CPB or Sham operation with or without pre-procedural high fat (HF) feeding containing 15% lard, 1.5% cholesterol and 1% cholic acid for 12-weeks (Groups: Sham, CPB, CPB + HF and Sham + HF). Our primary endpoint was creatinine clearance measured at 1.5 and 24 hours post intervention. This is a validated index of the glomerular filtration rate (GFR) in swine and an endpoint used in our clinical studies. Secondary endpoints included measures of systemic and renal inflammation, endothelial homeostasis, tubular injury and dysfunction, and inflammatory cell signalling. Differences between groups were calculated using analysis of variance with adjustment for baseline differences for repeated measures. Results CPB in pigs fed a normal chow diet resulted in AKI. This was characterised by reductions in GFR sustained for up to 24 hours post injury relative to Sham operated pigs fed a normal diet; mean difference 50.2 ml/min (95% CI 5.9 to 94.4). Post CPB AKI was also characterised by renal inflammation, parallel activation of both pro-inflammatory (NF-kB, iNOS) and pro-survival pathways (pAkt, p70s6k, HIF-1α) and apoptosis. Pigs fed a 12-week high fat diet developed obesity and hyperlipidaemia. This was associated with increased redox sensitive pro-inflammatory and anti-apoptotic signalling, and tubular epithelial cell proliferation. High fat feeding also protected swine against post CPB AKI; mean difference in creatinine clearance CPB - CPB + HF −65.3 ml/min (95% CI −106.9 to −23.7), by preserving endothelial homeostasis and function, and preventing the reductions in GFR, loss of ATP and tubular apoptosis that characterise the extension phase of AKI in swine at 24 hours post injury. Reno-protection was not attributed to pAkt signaling. Conclusions A high fat diet promoted obesity and renal inflammation and prevented post CPB AKI in swine. This study provides insights into the obesity paradox and the failure of anti-inflammatory interventions to improve clinical outcomes in patients at risk of post cardiac surgery AKI.
Collapse
|
49
|
Hickey FB, Martin F. Diabetic kidney disease and immune modulation. Curr Opin Pharmacol 2013; 13:602-12. [DOI: 10.1016/j.coph.2013.05.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 04/23/2013] [Accepted: 05/03/2013] [Indexed: 12/11/2022]
|
50
|
Li Y, Raman I, Du Y, Yan M, Min S, Yang J, Fang X, Li W, Lu J, Zhou XJ, Mohan C, Li QZ. Kallikrein transduced mesenchymal stem cells protect against anti-GBM disease and lupus nephritis by ameliorating inflammation and oxidative stress. PLoS One 2013; 8:e67790. [PMID: 23935844 PMCID: PMC3720854 DOI: 10.1371/journal.pone.0067790] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 05/26/2013] [Indexed: 12/20/2022] Open
Abstract
Previously we have shown that kallikreins (klks) play a renoprotective role in nephrotoxic serum induced nephritis. In this study, we have used mesenchymal stem cells (MSCs) as vehicles to deliver klks into the injured kidneys and have measured their therapeutic effect on experimental antibody induced nephritis and lupus nephritis. Human KLK-1 (hKLK1) gene was transduced into murine MSCs using a retroviral vector to generate a stable cell line, hKLK1-MSC, expressing high levels of hKLK1. 129/svj mice subjected to anti-GBM induced nephritis were transplanted with 10(6) hKLK1-MSCs and hKLK1 expression was confirmed in the kidneys. Compared with vector-MSCs injected mice, the hKLK1-MSCs treated mice showed significantly reduced proteinuria, blood urea nitrogen (BUN) and ameliorated renal pathology. Using the same strategy, we treated lupus-prone B6.Sle1.Sle3 bicongenic mice with hKLK1-MSCs and demonstrated that hKLK1-MSCs delivery also attenuated lupus nephritis. Mechanistically, hKLK1-MSCs reduced macrophage and T-lymphocyte infiltration into the kidney by suppressing the expression of inflammation cytokines. Moreover, hKLK1 transduced MSCs were more resistant to oxidative stress-induced apoptosis. These findings advance genetically modified MSCs as potential gene delivery tools for targeting therapeutic agents to the kidneys in order to modulate inflammation and oxidative stress in lupus nephritis.
Collapse
Affiliation(s)
- Yajuan Li
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Indu Raman
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Yong Du
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Mei Yan
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Soyoun Min
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Jichen Yang
- Quantitative Biomedical Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Xiangdong Fang
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Wei Li
- Key Laboratory of Medical Genetics, Wenzhou Medical College School of Laboratory Medicine & Life Science, Wenzhou, China
| | - Jianxin Lu
- Key Laboratory of Medical Genetics, Wenzhou Medical College School of Laboratory Medicine & Life Science, Wenzhou, China
| | - Xin J. Zhou
- Renal Path Diagnostics, Pathologist BioMedical Laboratories, Lewisville, Texas, United States of America
| | - Chandra Mohan
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- BME Departments, University of Houston, Houston, Texas, United States of America
| | - Quan-Zhen Li
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Key Laboratory of Medical Genetics, Wenzhou Medical College School of Laboratory Medicine & Life Science, Wenzhou, China
| |
Collapse
|