1
|
Yao C, Li Z, Sun K, Zhang Y, Shou S, Jin H. Mitochondrial dysfunction in acute kidney injury. Ren Fail 2024; 46:2393262. [PMID: 39192578 PMCID: PMC11360640 DOI: 10.1080/0886022x.2024.2393262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
Acute kidney injury (AKI) is a systemic clinical syndrome increasing morbidity and mortality worldwide in recent years. Renal tubular epithelial cells (TECs) death caused by mitochondrial dysfunction is one of the pathogeneses. The imbalance of mitochondrial quality control is the main cause of mitochondrial dysfunction. Mitochondrial quality control plays a crucial role in AKI. Mitochondrial quality control mechanisms are involved in regulating mitochondrial integrity and function, including antioxidant defense, mitochondrial quality control, mitochondrial DNA (mtDNA) repair, mitochondrial dynamics, mitophagy, and mitochondrial biogenesis. Currently, many studies have used mitochondrial dysfunction as a targeted therapeutic strategy for AKI. Therefore, this review aims to present the latest research advancements on mitochondrial dysfunction in AKI, providing a valuable reference and theoretical foundation for clinical prevention and treatment of this condition, ultimately enhancing patient prognosis.
Collapse
Affiliation(s)
- Congcong Yao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Ziwei Li
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Keke Sun
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Zhang
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Songtao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Heng Jin
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
2
|
Jamadar A, Ward CJ, Remadevi V, Varghese MM, Pabla NS, Gumz ML, Rao R. Circadian Clock Disruption and Growth of Kidney Cysts in Autosomal Dominant Polycystic Kidney Disease. J Am Soc Nephrol 2024:00001751-990000000-00444. [PMID: 39401086 DOI: 10.1681/asn.0000000528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024] Open
Abstract
Key Points
Lack of Bmal1, a circadian clock protein in renal collecting ducts disrupted the clock and increased cyst growth and fibrosis in an autosomal dominant polycystic kidney disease mouse model.
Bmal1 gene deletion increased cell proliferation by increasing lipogenesis in kidney cells.Thus, circadian clock disruption could be a risk factor for accelerated disease progression in patients with autosomal dominant polycystic kidney disease.
Background
Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in the PKD1 and PKD2 genes and often progresses to kidney failure. ADPKD progression is not uniform among patients, suggesting that factors secondary to the PKD1/2 gene mutation could regulate the rate of disease progression. Here, we tested the effect of circadian clock disruption on ADPKD progression. Circadian rhythms are regulated by cell-autonomous circadian clocks composed of clock proteins. BMAL1 is a core constituent of the circadian clock.
Methods
To disrupt the circadian clock, we deleted Bmal1 gene in the renal collecting ducts of the Pkd1
RC/RC (RC/RC) mouse model of ADPKD (RC/RC;Bmal1
f/f;Pkhd1
cre, called double knockout [DKO] mice) and in Pkd1 knockout mouse inner medullary collecting duct cells (Pkd1Bmal1KO mouse renal inner medullary collecting duct cells). Only male mice were used.
Results
Human nephrectomy ADPKD kidneys showed altered clock gene expression when compared with normal control human kidneys. When compared with RC/RC kidneys, DKO kidneys showed significantly altered clock gene expression, increased cyst growth, cell proliferation, apoptosis, and fibrosis. DKO kidneys also showed increased lipogenesis and cholesterol synthesis–related gene expression and increased tissue triglyceride levels compared with RC/RC kidneys. Similarly, in vitro, Pkd1Bmal1KO cells showed altered clock genes, increased lipogenesis and cholesterol synthesis–related genes, and reduced fatty acid oxidation–related gene expression compared with Pkd1KO cells. The Pkd1Bmal1KO cells showed increased cell proliferation compared with Pkd1KO cells, which was rescued by pharmacological inhibition of lipogenesis.
Conclusions
Renal collecting duct–specific Bmal1 gene deletion disrupted the circadian clock and triggered accelerated ADPKD progression by altering lipid metabolism–related gene expression.
Collapse
Affiliation(s)
- Abeda Jamadar
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
- Department of Medicine, Division of Nephrology, University of Kansas Medical Center, Kansas City, Kansas
| | - Christopher J Ward
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
- Department of Medicine, Division of Nephrology, University of Kansas Medical Center, Kansas City, Kansas
| | - Viji Remadevi
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
- Department of Medicine, Division of Nephrology, University of Kansas Medical Center, Kansas City, Kansas
| | - Meekha M Varghese
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
- Department of Medicine, Division of Nephrology, University of Kansas Medical Center, Kansas City, Kansas
| | - Navjot S Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Michelle L Gumz
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Physiology and Aging, Department of Medicine, University of Florida, Gainesville, Florida
| | - Reena Rao
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
- Department of Medicine, Division of Nephrology, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
3
|
Li J, Zhang Y, Tang R, Liu H, Li X, Lei W, Chen J, Jin Z, Tang J, Wang Z, Yang Y, Wu X. Glycogen synthase kinase-3β: A multifaceted player in ischemia-reperfusion injury and its therapeutic prospects. J Cell Physiol 2024; 239:e31335. [PMID: 38962880 DOI: 10.1002/jcp.31335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/05/2024] [Accepted: 05/22/2024] [Indexed: 07/05/2024]
Abstract
Ischemia-reperfusion injury (IRI) results in irreversible metabolic dysfunction and structural damage to tissues or organs, posing a formidable challenge in the field of organ implantation, cardiothoracic surgery, and general surgery. Glycogen synthase kinase-3β (GSK-3β) a multifunctional serine/threonine kinase, is involved in a variety of biological processes, including cell proliferation, apoptosis, and immune response. Phosphorylation of its tyrosine 216 and serine 9 sites positively and negatively regulates the activation and inactivation of the enzyme. Significantly, inhibition or inactivation of GSK-3β provides protection against IRI, making it a viable target for drug development. Though numerous GSK-3β inhibitors have been identified to date, the development of therapeutic treatments remains a considerable distance away. In light of this, this review summarizes the complicated network of GSK-3β roles in IRI. First, we provide an overview of GSK-3β's basic background. Subsequently, we briefly review the pathological mechanisms of GSK-3β in accelerating IRI, and highlight the latest progress of GSK-3β in multiorgan IRI, encompassing heart, brain, kidney, liver, and intestine. Finally, we discuss the current development of GSK-3β inhibitors in various organ IRI, offering a thorough and insightful reference for GSK-3β as a potential target for future IRI therapy.
Collapse
Affiliation(s)
- Jiayan Li
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yan Zhang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Ran Tang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Hui Liu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Xiayun Li
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Wangrui Lei
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Junmin Chen
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Zhenxiao Jin
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiayou Tang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Yang Yang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Xiaopeng Wu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| |
Collapse
|
4
|
Jamadar A, Ward CJ, Remadevi V, Varghese MM, Pabla NS, Gumz ML, Rao R. Circadian clock disruption and growth of kidney cysts in autosomal dominant polycystic kidney disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606676. [PMID: 39211074 PMCID: PMC11361200 DOI: 10.1101/2024.08.05.606676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in the PKD1 and PKD2 genes, and often progresses to kidney failure. ADPKD progression is not uniform among patients, suggesting that factors secondary to the PKD1/2 gene mutation could regulate the rate of disease progression. Here we tested the effect of circadian clock disruption on ADPKD progression. Circadian rhythms are regulated by cell-autonomous circadian clocks composed of clock proteins. BMAL1 is a core constituent of the circadian clock. Methods To disrupt the circadian clock, we deleted Bmal1 gene in the renal collecting ducts of the Pkd1 RC/RC (RC/RC) mouse model of ADPKD (RC/RC; Bmal1 f/f ; Pkhd1 cre , called DKO mice), and in Pkd1 knockout mouse inner medullary collecting duct cells ( Pkd1Bmal1 KO mIMCD3 cells). Only male mice were used. Results Human nephrectomy ADPKD kidneys and Pkd1 KO mIMCD3 cells showed reduced Bmal1 gene expression compared to normal controls. When compared to RC/RC kidneys, DKO kidneys showed significantly altered clock gene expression, increased cyst growth, cell proliferation, apoptosis and fibrosis. DKO kidneys also showed increased lipogenesis and cholesterol synthesis-related gene expression, and increased tissue triglyceride levels compared to RC/RC kidneys. Similarly, in vitro, Pkd1Bmal1 KO cells showed altered clock genes, increased lipogenesis and cholesterol synthesis-related genes, and reduced fatty-acid oxidation-related gene expression compared to Pkd1KO cells. The Pkd1Bmal1 KO cells showed increased cell proliferation compared to Pkd1KO cells, which was rescued by pharmacological inhibition of lipogenesis. Conclusion Renal collecting duct specific Bmal1 gene deletion disrupts the circadian clock and triggers accelerated ADPKD progression by altering lipid metabolism-related gene expression. Key points Lack of BMAL1, a circadian clock protein in renal collecting ducts disrupted the clock and increased cyst growth and fibrosis in an ADPKD mouse model.BMAL1 gene deletion increased cell proliferation by increasing lipogenesis in kidney cells.Thus, circadian clock disruption could be a risk factor for accelerated disease progression in patients with ADPKD.
Collapse
|
5
|
Salminen A. AMPK signaling inhibits the differentiation of myofibroblasts: impact on age-related tissue fibrosis and degeneration. Biogerontology 2024; 25:83-106. [PMID: 37917219 PMCID: PMC10794430 DOI: 10.1007/s10522-023-10072-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 09/26/2023] [Indexed: 11/04/2023]
Abstract
Disruption of the extracellular matrix (ECM) and an accumulation of fibrotic lesions within tissues are two of the distinctive hallmarks of the aging process. Tissue fibroblasts are mesenchymal cells which display an impressive plasticity in the regulation of ECM integrity and thus on tissue homeostasis. Single-cell transcriptome studies have revealed that tissue fibroblasts exhibit a remarkable heterogeneity with aging and in age-related diseases. Excessive stress and inflammatory insults induce the differentiation of fibroblasts into myofibroblasts which are fusiform contractile cells and abundantly secrete the components of the ECM and proteolytic enzymes as well as many inflammatory mediators. Detrimental stresses can also induce the transdifferentiation of certain mesenchymal and myeloid cells into myofibroblasts. Interestingly, many age-related stresses, such as oxidative and endoplasmic reticulum stresses, ECM stiffness, inflammatory mediators, telomere shortening, and several alarmins from damaged cells are potent inducers of myofibroblast differentiation. Intriguingly, there is convincing evidence that the signaling pathways stimulated by the AMP-activated protein kinase (AMPK) are potent inhibitors of myofibroblast differentiation and accordingly AMPK signaling reduces fibrotic lesions within tissues, e.g., in age-related cardiac and pulmonary fibrosis. AMPK signaling is not only an important regulator of energy metabolism but it is also able to control cell fate determination and many functions of the immune system. It is known that AMPK signaling can delay the aging process via an integrated signaling network. AMPK signaling inhibits myofibroblast differentiation, e.g., by suppressing signaling through the TGF-β, NF-κB, STAT3, and YAP/TAZ pathways. It seems that AMPK signaling can alleviate age-related tissue fibrosis and degeneration by inhibiting the differentiation of myofibroblasts.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
6
|
Khan H, Bangar A, Grewal AK, Singh TG. Mechanistic Implications of GSK and CREB Crosstalk in Ischemia Injury. Neurotox Res 2023; 42:1. [PMID: 38091155 DOI: 10.1007/s12640-023-00680-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 11/03/2023] [Accepted: 11/11/2023] [Indexed: 12/18/2023]
Abstract
Ischemia-reperfusion (IR) injury is a damage to an organ when the blood supply is less than the demand required for normal functioning, leading to exacerbation of cellular dysfunction and death. IR injury occurs in different organs like the kidney, liver, heart, brain, etc., and may not only involve the ischemic organ but also cause systemic damage to distant organs. Oxygen-glucose deprivation in cells causes oxidative stress, calcium overloading, inflammation, and apoptosis. CREB is an essential integrator of the body's various physiological systems, and it is widely accepted that dysfunction of CREB signaling is involved in many diseases, including ischemia-reperfusion injury. The activation of CREB can provide life to a cell and increase the cell's survival after ischemia. Hence, GSK/CREB signaling pathway can provide significant protection to cells of different organs after ischemia and emerges as a futuristic strategy for managing ischemia-reperfusion injury. Different signaling pathways such as MAPK/ERK, TLR4/MyD88, RISK, Nrf2, and NF-κB, get altered during IR injury by the modulation of GSK-3 and CREB (cyclic AMP response element (CRE)-binding protein). GSK-3 (protein kinase B) and CREB are the downstream targets for fulfilling the roles of various signaling pathways. Calcium overloading during ischemia increases the expression of calcium-calmodulin-dependent protein kinase (CaMK), which subsequently activates CREB-mediated transcription, thus promoting the survival of cells. Furthermore, this review highlights the crosstalk between GSK-3 and CREB, promoting survival and rendering the cells resistant to subsequent severe ischemia.
Collapse
Affiliation(s)
- Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Annu Bangar
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | | | | |
Collapse
|
7
|
Yakupova EI, Abramicheva PA, Bocharnikov AD, Andrianova NV, Plotnikov EY. Biomarkers of the End-Stage Renal Disease Progression: Beyond the GFR. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1622-1644. [PMID: 38105029 DOI: 10.1134/s0006297923100164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 07/20/2023] [Accepted: 08/20/2023] [Indexed: 12/19/2023]
Abstract
Chronic kidney disease can progress to the end-stage renal disease (ESRD) characterized by a high risk of morbidity and mortality. ESRD requires immediate therapy or even dialysis or kidney transplantation, therefore, its timely diagnostics is critical for many patients. ESRD is associated with pathological changes, such as inflammation, fibrosis, endocrine disorders, and epigenetic changes in various cells, which could serve as ESRD markers. The review summarizes information on conventional and new ESRD biomarkers that can be assessed in kidney tissue, blood, and urine. Some biomarkers are specific to a particular pathology, while others are more universal. Here, we suggest several universal inflammatory, fibrotic, hormonal, and epigenetic markers indicative of severe deterioration of renal function and ESRD progression for improvement of ESRD diagnostics.
Collapse
Affiliation(s)
- Elmira I Yakupova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
| | - Polina A Abramicheva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Alexey D Bocharnikov
- International School of Medicine of the Future, Sechenov First Moscow State Medical University, Moscow, 119992, Russia
| | - Nadezda V Andrianova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Egor Y Plotnikov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
- Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, 117997, Russia
| |
Collapse
|
8
|
Wu Y, Bai Y, Feng Y, Zhang Q, Diao Z, Liu W. Renalase Prevents Renal Fibrosis by Inhibiting Endoplasmic Reticulum Stress and Down-Regulating GSK-3β/Snail Signaling. Int J Med Sci 2023; 20:669-681. [PMID: 37082730 PMCID: PMC10110476 DOI: 10.7150/ijms.82192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/24/2023] [Indexed: 04/22/2023] Open
Abstract
Background: Treating renal fibrosis is crucial to delaying chronic kidney disease. The glycogen synthase kinase-3β (GSK-3β)/Snail pathway regulates renal fibrosis and Renalase can ameliorate renal interstitial fibrosis. However, it is not clear whether GSK-3β/Snail signaling affects Renalase action. Here, we explored the role and mechanism of GSK-3β/Snail in the anti-fibrosis action of Renalase. Materials and methods: We used mice with complete unilateral ureteral obstruction (UUO) and human proximal renal tubular epithelial (HK-2) cells with transforming growth factor-β1 (TGF-β1)-induced fibrosis to explore the role and regulatory mechanism of the GSK-3β/Snail pathway in the amelioration of renal fibrosis by Renalase. Results: In UUO mice and TGF-β1-induced fibrotic HK-2 cells, the expression of p-GSK-3β-Tyr216/p-GSK-3β-Ser9, GSK-3β and Snail was significantly increased, and endoplasmic reticulum (ER) stress was activated. After Renalase supplementation, fibrosis was alleviated, ER stress was inhibited and p-GSK-3β-Tyr216/p-GSK-3β-Ser9, GSK-3β and Snail were significantly down-regulated. The amelioration of renal fibrosis by Renalase and its inhibitory effect on GSK-3β/Snail were reversed by an ER stress agonist. Furthermore, when an adeno-associated virus or plasmid was used to overexpress GSK-3β, the effect of Renalase on delaying renal fibrosis was counteracted, although ER stress markers did not change. Conclusion: Renalase prevents renal fibrosis by down-regulating GSK-3β/Snail signaling through inhibition of ER stress. Exogenous Renalase may be an effective method of slowing or stopping chronic kidney disease progression.
Collapse
Affiliation(s)
- Yiru Wu
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing 100050, P. R. China
| | - Yu Bai
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing 100050, P. R. China
| | - Yiduo Feng
- Department of Nephrology, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring Road West, Fengtai District, Beijing,100070, P. R. China
| | - Qidong Zhang
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing 100050, P. R. China
| | - Zongli Diao
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing 100050, P. R. China
| | - Wenhu Liu
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing 100050, P. R. China
- ✉ Corresponding author: Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P. R. China. Tel.: +86-10-63138579; Fax: +86-10-63139144; E-mail:
| |
Collapse
|
9
|
Jamadar A, Dwivedi N, Mathew S, Calvet JP, Thomas SM, Rao R. Vasopressin Receptor Type-2 Mediated Signaling in Renal Cell Carcinoma Stimulates Stromal Fibroblast Activation. Int J Mol Sci 2022; 23:7601. [PMID: 35886951 PMCID: PMC9325308 DOI: 10.3390/ijms23147601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/01/2022] [Accepted: 07/07/2022] [Indexed: 01/27/2023] Open
Abstract
Vasopressin type-2 receptor (V2R) is ectopically expressed and plays a pathogenic role in clear cell renal cell carcinoma (ccRCC) tumor cells. Here we examined how V2R signaling within human ccRCC tumor cells (Caki1 cells) stimulates stromal cancer-associated fibroblasts (CAFs). We found that cell culture conditioned media from Caki1 cells increased activation, migration, and proliferation of fibroblasts in vitro, which was inhibited by V2R gene silencing in Caki1 cells. Analysis of the conditioned media and mRNA of the V2R gene silenced and control Caki1 cells showed that V2R regulates the production of CAF-activating factors. Some of these factors were also found to be regulated by YAP in these Caki1 cells. YAP expression colocalized and correlated with V2R expression in ccRCC tumor tissue. V2R gene silencing or V2R antagonist significantly reduced YAP in Caki1 cells. Moreover, the V2R antagonist reduced YAP expression and myofibroblasts in mouse xenograft tumors. These results suggest that V2R plays an important role in secreting pro-fibrotic factors that stimulate fibroblast activation by a YAP-dependent mechanism in ccRCC tumors. Our results demonstrate a novel role for the V2R-YAP axis in the regulation of myofibroblasts in ccRCC and a potential therapeutic target.
Collapse
Affiliation(s)
- Abeda Jamadar
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA; (A.J.); (N.D.); (J.P.C.)
| | - Nidhi Dwivedi
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA; (A.J.); (N.D.); (J.P.C.)
| | - Sijo Mathew
- Department of Pharmaceutical Sciences, School of Pharmacy, North Dakota State University, Fargo, ND 58105, USA;
| | - James P. Calvet
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA; (A.J.); (N.D.); (J.P.C.)
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sufi M. Thomas
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Reena Rao
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA; (A.J.); (N.D.); (J.P.C.)
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Medicine, 5040 WHE, The Jared Grantham Kidney Institute, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| |
Collapse
|
10
|
Recent Advances in Diabetic Kidney Diseases: From Kidney Injury to Kidney Fibrosis. Int J Mol Sci 2021; 22:ijms222111857. [PMID: 34769288 PMCID: PMC8584225 DOI: 10.3390/ijms222111857] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/08/2021] [Accepted: 10/30/2021] [Indexed: 12/14/2022] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of chronic kidney disease and end-stage renal disease. The natural history of DKD includes glomerular hyperfiltration, progressive albuminuria, declining estimated glomerular filtration rate, and, ultimately, kidney failure. It is known that DKD is associated with metabolic changes caused by hyperglycemia, resulting in glomerular hypertrophy, glomerulosclerosis, and tubulointerstitial inflammation and fibrosis. Hyperglycemia is also known to cause programmed epigenetic modification. However, the detailed mechanisms involved in the onset and progression of DKD remain elusive. In this review, we discuss recent advances regarding the pathogenic mechanisms involved in DKD.
Collapse
|
11
|
Zhang X, Agborbesong E, Li X. The Role of Mitochondria in Acute Kidney Injury and Chronic Kidney Disease and Its Therapeutic Potential. Int J Mol Sci 2021; 22:ijms222011253. [PMID: 34681922 PMCID: PMC8537003 DOI: 10.3390/ijms222011253] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/10/2021] [Accepted: 10/13/2021] [Indexed: 12/19/2022] Open
Abstract
Mitochondria are heterogeneous and highly dynamic organelles, playing critical roles in adenosine triphosphate (ATP) synthesis, metabolic modulation, reactive oxygen species (ROS) generation, and cell differentiation and death. Mitochondrial dysfunction has been recognized as a contributor in many diseases. The kidney is an organ enriched in mitochondria and with high energy demand in the human body. Recent studies have been focusing on how mitochondrial dysfunction contributes to the pathogenesis of different forms of kidney diseases, including acute kidney injury (AKI) and chronic kidney disease (CKD). AKI has been linked to an increased risk of developing CKD. AKI and CKD have a broad clinical syndrome and a substantial impact on morbidity and mortality, encompassing various etiologies and representing important challenges for global public health. Renal mitochondrial disorders are a common feature of diverse forms of AKI and CKD, which result from defects in mitochondrial structure, dynamics, and biogenesis as well as crosstalk of mitochondria with other organelles. Persistent dysregulation of mitochondrial homeostasis in AKI and CKD affects diverse cellular pathways, leading to an increase in renal microvascular loss, oxidative stress, apoptosis, and eventually renal failure. It is important to understand the cellular and molecular events that govern mitochondria functions and pathophysiology in AKI and CKD, which should facilitate the development of novel therapeutic strategies. This review provides an overview of the molecular insights of the mitochondria and the specific pathogenic mechanisms of mitochondrial dysfunction in the progression of AKI, CKD, and AKI to CKD transition. We also discuss the possible beneficial effects of mitochondrial-targeted therapeutic agents for the treatment of mitochondrial dysfunction-mediated AKI and CKD, which may translate into therapeutic options to ameliorate renal injury and delay the progression of these kidney diseases.
Collapse
Affiliation(s)
- Xiaoqin Zhang
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA; (X.Z.); (E.A.)
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ewud Agborbesong
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA; (X.Z.); (E.A.)
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA; (X.Z.); (E.A.)
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Correspondence: ; Tel.: +507-266-0110
| |
Collapse
|
12
|
Jamadar A, Suma SM, Mathew S, Fields TA, Wallace DP, Calvet JP, Rao R. The tyrosine-kinase inhibitor Nintedanib ameliorates autosomal-dominant polycystic kidney disease. Cell Death Dis 2021; 12:947. [PMID: 34650051 PMCID: PMC8517027 DOI: 10.1038/s41419-021-04248-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/13/2021] [Accepted: 09/28/2021] [Indexed: 12/27/2022]
Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease and is characterized by progressive growth of fluid-filled cysts. Growth factors binding to receptor tyrosine kinases (RTKs) stimulate cell proliferation and cyst growth in PKD. Nintedanib, a triple RTK inhibitor, targets the vascular endothelial growth-factor receptor (VEGFR), platelet-derived growth-factor receptor (PDGFR), and fibroblast growth-factor receptor (FGFR), and is an approved drug for the treatment of non-small-cell lung carcinoma and idiopathic lung fibrosis. To determine if RTK inhibition using nintedanib can slow ADPKD progression, we tested its effect on human ADPKD renal cyst epithelial cells and myofibroblasts in vitro, and on Pkd1f/fPkhd1Cre and Pkd1RC/RC, orthologous mouse models of ADPKD. Nintedanib significantly inhibited cell proliferation and in vitro cyst growth of human ADPKD renal cyst epithelial cells, and cell viability and migration of human ADPKD renal myofibroblasts. Consistently, nintedanib treatment significantly reduced kidney-to-body-weight ratio, renal cystic index, cystic epithelial cell proliferation, and blood-urea nitrogen levels in both the Pkd1f/fPkhd1Cre and Pkd1RC/RC mice. There was a corresponding reduction in ERK, AKT, STAT3, and mTOR activity and expression of proproliferative factors, including Yes-associated protein (YAP), c-Myc, and Cyclin D1. Nintedanib treatment significantly reduced fibrosis in Pkd1RC/RC mice, but did not affect renal fibrosis in Pkd1f/fPkhd1Cre mice. Overall, these results suggest that nintedanib may be repurposed to effectively slow cyst growth in ADPKD.
Collapse
Affiliation(s)
- Abeda Jamadar
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sreenath M Suma
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sijo Mathew
- Department of Pharmaceutical Sciences, School of Pharmacy, North Dakota State University, Fargo, ND, USA
| | - Timothy A Fields
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Darren P Wallace
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - James P Calvet
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Reena Rao
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA.
- Department of Medicine, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
13
|
Liu S, Lv X, Wei X, Liu C, Li Q, Min J, Hua F, Zhang X, Li K, Li P, Xiao Y, Hu Z, Cui B. TRIB3‒GSK-3 β interaction promotes lung fibrosis and serves as a potential therapeutic target. Acta Pharm Sin B 2021; 11:3105-3119. [PMID: 34729304 PMCID: PMC8546892 DOI: 10.1016/j.apsb.2021.06.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 11/15/2022] Open
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive, fatal interstitial lung disease with limited available therapeutic strategies. We recently reported that the protein kinase glycogen synthase kinase-3β (GSK-3β) interacts with and inactivates the ubiquitin-editing enzyme A20 to suppress the degradation of the transcription factor CCAAT/enhancer-binding protein beta (C/EBPβ) in alveolar macrophages (AMs), resulting in a profibrotic phenotype of AMs and promoting the development of PF. Here, we showed that chronic lung injury upregulated the stress response protein tribbles homolog 3 (TRIB3), which interacted with GSK-3β and stabilized GSK-3β from ubiquitination and degradation. Elevated GSK-3β expression phosphorylated A20 to inhibit its ubiquitin-editing activity, causing the accumulation of C/EBPβ and the production of several profibrotic factors in AMs and promoting PF development. Activated C/EBPβ, in turn, increased the transcription of TRIB3 and GSK-3β, thereby establishing a positive feedback loop in AMs. The knockdown of TRIB3 expression or the pharmacologic disruption of the TRIB3‒GSK-3β interaction was an effective PF treatment. Our study reveals an intact profibrotic axis of TRIB3‒GSK-3β‒A20‒C/EBPβ in AMs, which represents a target that may provide a promising treatment strategy for PF.
Collapse
|
14
|
Liu Q, Kong Y, Guo X, Liang B, Xie H, Hu S, Han M, Zhao X, Feng P, Lyu Q, Dong W, Liang X, Wang W, Li C. GSK-3β inhibitor TDZD-8 prevents reduction of aquaporin-1 expression via activating autophagy under renal ischemia reperfusion injury. FASEB J 2021; 35:e21809. [PMID: 34314052 DOI: 10.1096/fj.202100549r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/12/2021] [Accepted: 07/06/2021] [Indexed: 11/11/2022]
Abstract
Renal ischemia/reperfusion (I/R) injury is a main cause of acute kidney injury (AKI). Aquaporin (AQP)-1 water channel in the kidney is critical for the maintenance of water homeostasis and the urinary concentrating ability. Increasing evidence supports an important role of autophagy in the pathogenesis of AKI induced by renal I/R. The purpose of the present study is to investigate whether activation of autophagy prevents downregulation of AQP1 protein induced by renal I/R and potential molecular mechanisms. Renal I/R induced consistently reduced protein expression of AQP1, 2, and 3, as well as sodium cotransporters Na+ -K+ -2Cl- cotransporter and α-Na,K-ATPase, which was associated with increased urine output and decreased creatinine clearance in rats. Renal I/R also suppressed autophagy and increased inflammatory responses in the kidney. 4-Benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8), the glycogen synthase kinase-3β inhibitor, ameliorated renal injury under I/R, activated autophagy and markedly increased expression of AQPs and sodium transporters in the kidney, which was associated with improved urine output and creatinine clearance in rats. Hypoxia/reoxygenation (H/R) induced suppression of autophagy and downregulation of AQP1 in murine inner medullary collecting duct 3 (IMCD3) cells, which was fully prevented by TDZD-8 treatment. Inhibition of autophagy by 3-methyladenine or Atg5 gene knockdown attenuated recovery of AQP1 protein expression induced by TDZD-8 in IMCD3 cells with H/R. Interleukin-1 beta (IL-1β) decreased the abundance of AQP1 protein in IMCD3 cells. H/R induced increases in protein expression of nod-like receptor pyrin domain-containing 3 and IL-1β, which was reversed by TDZD-8. In conclusion, TDZD-8 treatment prevented downregulation of AQP1 expression under renal I/R injury, likely via activating autophagy and decreasing IL-1β production.
Collapse
Affiliation(s)
- Qiaojuan Liu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yonglun Kong
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiangdong Guo
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Baien Liang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Haixia Xie
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shan Hu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Mengke Han
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoduo Zhao
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Pinning Feng
- Department of Clinical Laboratory, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qianqian Lyu
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Wei Dong
- Division of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xinling Liang
- Division of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Weidong Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Nephrology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Chunling Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
15
|
Deng W, Wei X, Dong Z, Zhang J, Huang Z, Na N. Identification of fibroblast activation-related genes in two acute kidney injury models. PeerJ 2021; 9:e10926. [PMID: 33777519 PMCID: PMC7982076 DOI: 10.7717/peerj.10926] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/20/2021] [Indexed: 12/27/2022] Open
Abstract
Background Ischemia-reperfusion injury and drug-induced nephrotoxicity are the two most common reasons for acute kidney injury (AKI). However, little attention has been paid to early activation of fibroblasts in the progression of AKI to chronic kidney disease (CKD). The present study aimed to identify related genes and pathways on fibroblast activation in two mouse models of AKI: ischemia-reperfusion injury (IRI) model and folic acid (FA)-induced injury model. Methods The microarray expression profiles of GSE62732 and GSE121190 were downloaded from the GEO database, and the differentially expressed genes (DEGs) was analyzed using the Limma package of R software. Principal component analysis (PCA) was also performed using R. The functional information of gene products was annotated by Gene Ontology (GO) and DAVID online database, and the pathway analysis was carried out by using the Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) database. Protein-protein interactions (PPI) network was constructed by STRING and Cytoscape. Furthermore, in the Hypoxia/Reoxygenation (H/R) model, the morphological changes of cells were observed under microscope and the expression of the hub genes in NRK-49F cells were validated by qRT-PCR assays. Results A total of 457 DEGs were identified. Among these, 215 DEGs were upregulated and 242 DEGs were downregulated in the acute injured samples compared with uninjured samples. The GO enrichment analysis indicated that these DEGs were mainly involved in transport, the oxidation-reduction process, the metabolic process, metal ion binding, hydrolase activity, and oxidoreductase activity. The KEGG analysis revealed that these DEGs were significantly enriched in the PI3K-Akt signaling pathway, protein digestion and absorption pathway, and focal adhesion pathway. The hub genes including Hnf4α, Pck1 and Timp1 were validated by the qRT-PCR assay in NRK-49F cells in the H/R model. Conclusions Hnf4α, Pck1 and Timp-1 may play a pivotal role in the early activation of fibroblasts, providing novel therapeutic strategies for early prediction and treatment of renal fibrosis.
Collapse
Affiliation(s)
- Weiming Deng
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiangling Wei
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhanwen Dong
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jinhua Zhang
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhengyu Huang
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ning Na
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
16
|
Hao H, Ma S, Zheng C, Wang Q, Lin H, Chen Z, Xie J, Chen L, Chen K, Wang Y, Huang X, Cao S, Liao W, Bin J, Liao Y. Excessive fibroblast growth factor 23 promotes renal fibrosis in mice with type 2 cardiorenal syndrome. Aging (Albany NY) 2021; 13:2982-3009. [PMID: 33460402 PMCID: PMC7880350 DOI: 10.18632/aging.202448] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023]
Abstract
Cardiorenal syndrome (CRS) has a high mortality, but its pathogenesis remains elusive. Fibroblast growth factor 23 (FGF23) is increased in both renal dysfunction and cardiac dysfunction, and FGF receptor 4 (FGFR4) has been identified as a receptor for FGF23. Deficiency of FGF23 causes growth retardation and shortens the lifespan, but it is unclear whether excess FGF23 is detrimental in CRS. This study sought to investigate whether FGF23 plays an important role in CRS-induced renal fibrosis. A mouse model of CRS was created by surgical myocardial infarction for 12 weeks. CRS mice showed a significant increase of circulatory and renal FGF23 protein levels, as well as an upregulation of p-GSK, active-β-catenin, TGF-β, collagen I and vimentin, a downregulation of renal Klotho expression and induction of cardiorenal dysfunction and cardiorenal fibrosis. These changes were enhanced by cardiac overexpression of FGF23 and attenuated by FGF receptor blocker PD173074 or β-catenin blocker IGC001. In fibroblasts (NRK-49F), expression of FGFR4 rather than Klotho was detected. Recombinant FGF23 upregulated the expression of p-GSK, active-β-catenin, TGF-β, collagen I and vimentin proteins. These changes were attenuated by FGFR4 blockade with BLU9931 or β-catenin blockade with IGC001. We concluded that FGF23 promotes CRS-induced renal fibrosis mediated by partly activating FGFR4/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Huixin Hao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Siyuan Ma
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Cankun Zheng
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qiancheng Wang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hairuo Lin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhenhuan Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiahe Xie
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lin Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kaitong Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuegang Wang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaobo Huang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Shiping Cao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jianping Bin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yulin Liao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
17
|
Huang G, Zhang J, Qing G, Liu D, Wang X, Chen Y, Li Y, Guo S. S100A2 Silencing Relieves Epithelial-Mesenchymal Transition in Pulmonary Fibrosis by Inhibiting the Wnt/β-Catenin Signaling Pathway. DNA Cell Biol 2020; 40:18-25. [PMID: 33306933 DOI: 10.1089/dna.2020.6030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Pulmonary fibrosis (PF) is a progressive and lethal disease with poor prognosis. S100A2 plays an important role in the progression of cancer. However, the role of S100A2 in PF has not yet been reported. In this study, we explored the potential role of S100A2 in PF and its potential molecular mechanisms. Increased expression of S100A2 was first observed in lung tissues of PF patients. We found that downregulation of S100A2 inhibited the transforming growth factor-β1 (TGF-β1)-induced epithelial-mesenchymal transition (EMT) in A549 cells. Mechanically, TGF-β1 upregulated β-catenin and the phosphorylation of glycogen synthase kinase-3β, which was blocked by silencing S100A2 in vitro. Furthermore, lithium chloride (activator of the Wnt/β-catenin signaling pathway) effectively rescued S100A2 knockdown-mediated inhibition of EMT in PF. In conclusion, these findings demonstrate that downregulation of S100A2 alleviated PF through inhibiting EMT. S100A2 is a promising potential target for further understanding the mechanism and developing a strategy for the treatment of PF and other EMT-associated diseases.
Collapse
Affiliation(s)
- Guichuan Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Zhang
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Gang Qing
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | - Xin Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yishi Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuliang Guo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
18
|
Zheng H, Yang Z, Xin Z, Yang Y, Yu Y, Cui J, Liu H, Chen F. Glycogen synthase kinase-3β: a promising candidate in the fight against fibrosis. Theranostics 2020; 10:11737-11753. [PMID: 33052244 PMCID: PMC7545984 DOI: 10.7150/thno.47717] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 09/12/2020] [Indexed: 02/07/2023] Open
Abstract
Fibrosis exists in almost all organs/tissues of the human body, plays an important role in the occurrence and development of diseases and is also a hallmark of the aging process. However, there is no effective prevention or therapeutic method for fibrogenesis. As a serine/threonine (Ser/Thr)-protein kinase, glycogen synthase kinase-3β (GSK-3β) is a vital signaling mediator that participates in a variety of biological events and can inhibit extracellular matrix (ECM) accumulation and the epithelial-mesenchymal transition (EMT) process, thereby exerting its protective role against the fibrosis of various organs/tissues, including the heart, lung, liver, and kidney. Moreover, we further present the upstream regulators and downstream effectors of the GSK-3β pathway during fibrosis and comprehensively summarize the roles of GSK-3β in the regulation of fibrosis and provide several potential targets for research. Collectively, the information reviewed here highlights recent advances vital for experimental research and clinical development, illuminating the possibility of GSK-3β as a novel therapeutic target for the management of tissue fibrosis in the future.
Collapse
Affiliation(s)
- Hanxue Zheng
- Lab of Tissue Engineering, Faculty of Life Sciences, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Zhi Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Zhenlong Xin
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yuan Yu
- Lab of Tissue Engineering, Faculty of Life Sciences, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Jihong Cui
- Lab of Tissue Engineering, Faculty of Life Sciences, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Hongbo Liu
- Lab of Tissue Engineering, Faculty of Life Sciences, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Fulin Chen
- Lab of Tissue Engineering, Faculty of Life Sciences, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| |
Collapse
|
19
|
Sinha S, Dwivedi N, Woodgett J, Tao S, Howard C, Fields TA, Jamadar A, Rao R. Glycogen synthase kinase-3β inhibits tubular regeneration in acute kidney injury by a FoxM1-dependent mechanism. FASEB J 2020; 34:13597-13608. [PMID: 32813289 DOI: 10.1096/fj.202000526rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/10/2020] [Accepted: 07/27/2020] [Indexed: 12/29/2022]
Abstract
Acute kidney injury (AKI) is characterized by injury to the tubular epithelium that leads to the sudden loss of renal function. Proper tubular regeneration is essential to prevent progression to chronic kidney disease. In this study, we examined the role of FoxM1, a forkhead box family member transcription factor in tubular repair after AKI. Renal FoxM1 expression increased after renal ischemia/reperfusion (I/R)-induced AKI in mouse kidneys. Treatment with thiostrepton, a FoxM1 inhibitor, reduced FoxM1 regulated pro-proliferative factors and cell proliferation in vitro, and tubular regeneration in mouse kidneys after AKI. Glycogen synthase kinase-3 (GSK3) was found to be an upstream regulator of FoxM1 because GSK3 inhibition or renal tubular GSK3β gene deletion significantly increased FoxM1 expression, and improved tubular repair and renal function. GSK3 inactivation increased β-catenin, Cyclin D1, and c-Myc, and reduced cell cycle inhibitors p21 and p27. Importantly, thiostrepton treatment abolished the improved tubular repair in GSK3β knockout mice following AKI. These results demonstrate that FoxM1 is important for renal tubular regeneration following AKI and that GSK3β suppresses tubular repair by inhibiting FoxM1.
Collapse
Affiliation(s)
- Sonali Sinha
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Nidhi Dwivedi
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - James Woodgett
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital and Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Shixin Tao
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Christianna Howard
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Timothy A Fields
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Pathology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Abeda Jamadar
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Reena Rao
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
20
|
Jamadar A, Rao R. Glycogen Synthase Kinase-3 Signaling in Acute Kidney Injury. Nephron Clin Pract 2020; 144:609-612. [PMID: 32726778 DOI: 10.1159/000509354] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/10/2020] [Indexed: 12/16/2022] Open
Abstract
Acute kidney injury (AKI) is a common clinical syndrome that involves renal tubular epithelial cell death and leads to acute decline in renal function. Improper tubular regeneration following AKI often leads to CKD. We discuss the role of a serine/threonine protein kinase called glycogen synthase kinase-3 (GSK3) in renal tubular injury and renal fibrosis. We also highlight the importance of GSK3 as a potential drug target in AKI patients and molecular mechanisms promoting tissue regeneration.
Collapse
Affiliation(s)
- Abeda Jamadar
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Reena Rao
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA, .,Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA,
| |
Collapse
|
21
|
Zhang T, Zhou J, Yue H, Du C, Xiao Z, Zhao W, Li N, Wang X, Liu X, Li Y, Geng X, Zhang Y, Li L, Tian J. Glycogen synthase kinase-3β promotes radiation-induced lung fibrosis by regulating β-catenin/lin28 signaling network to determine type II alveolar stem cell transdifferentiation state. FASEB J 2020; 34:12466-12480. [PMID: 32706136 DOI: 10.1096/fj.202001518] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 07/06/2020] [Indexed: 01/17/2023]
Abstract
The role of type II alveolar epithelial stem cells (AEC II) for alveolar repair in radiation-induced lung fibrosis (RILF) remains largely unknown, mainly because of AEC II phenotype's spontaneous change in vitro. Cell differentiation status is determined by Lin28 and let-7 miRNAs in see-saw-pattern. Lin28, a repressor of let-7 and a stem cell marker, is activated by β-catenin. The expression of β-catenin is regulated by GSK-3β/TGF-β1 signaling. To understand the true role of AEC II in RILF, we freshly isolated primary AEC II directly from thoracically irradiated lungs. We then explored the expressions of cell phenotype markers and differentiation regulators in these isolated AEC II to analyze the correlation between GSK-3β/TGF-β1/β-catenin signaling pathway, lin28/let-7 balance, and AEC II phenotypes at different injury phases following irradiation. Results showed that isolated single primary cells displayed AEC II ultrastructural features and proSP-C positive. The gene expressions of prosp-c (an AEC II biomarker) and hopx (an AEC I marker) significantly increased in isolated AEC II during injury repair phase (P < .001 and P < .05) but decreased at end-stage of injury, while mesenchymal markers increased in both isolated AEC II and irradiated lungs. mRNA levels of gsk-3β, tgf-β1, and β-catenin increased in all irradiated AEC II, but more pronounced in the second half of injury phase (P < .05-P < .001). Similarly, the expression of lin28 was also significantly elevated in isolated AEC II at the late phase (P < .05-P < .001). Four let-7 miRNAs were significantly upregulated in all irradiated AEC II groups (P < .05-P < .001). The time-dependent and highly consistent uptrends for four lin28/let-7 ratios in sorted AEC II contrasted to downtrends in irradiated lungs. In conclusion, RILF occurred when GSK-3β/TGF-β1 signaling increased β-catenin levels, which led to the augmentation of AEC II population by elevated lin28/let-7 ratio and the transcription of profibrotic cytokines and factors, thereby inducing AEC II to undergo transdifferentiation into mesenchymal cells.
Collapse
Affiliation(s)
- Tingting Zhang
- Center for Clinical Single-Cell Biomedicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Jianwei Zhou
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Haodi Yue
- Center for Clinical Single-Cell Biomedicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Chunyan Du
- Laboratory Animal Center, School of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ziting Xiao
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Wendi Zhao
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Na Li
- Henan Provincial Key Laboratory for Kidney Disease and Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Xiangdong Wang
- Center for Clinical Single-Cell Biomedicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Xiaozhuan Liu
- Center for Clinical Single-Cell Biomedicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Yanjun Li
- Center for Clinical Single-Cell Biomedicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Xiwen Geng
- Center for Clinical Single-Cell Biomedicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Yuwei Zhang
- Henan Provincial Key Laboratory for Kidney Disease and Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Li Li
- Department of Scientific Research and Discipline Construction, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Jian Tian
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| |
Collapse
|
22
|
Liu J, Zhu YM, Guo Y, Lin L, Wang ZX, Gu F, Dong XY, Zhou M, Wang YF, Zhang HL. Inhibition of GSK3β and RIP1K Attenuates Glial Scar Formation Induced by Ischemic Stroke via Reduction of Inflammatory Cytokine Production. Front Pharmacol 2020; 11:812. [PMID: 32595496 PMCID: PMC7303311 DOI: 10.3389/fphar.2020.00812] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 05/18/2020] [Indexed: 01/05/2023] Open
Abstract
In the chronic phase following ischemic stroke, glial scars can prevent axonal regeneration and the intensification of inflammation. The protective effect of inhibition of glycogen synthase kinase-3β (GSK3β) or receptor-interacting protein 1 kinase (RIP1K) on ischemic stroke has been previously reported. The current study examined the effects of RIP1K and GSK3β on ischemic stroke-induced glial scar formation. To investigate this, we used an in vivo model of ischemic stroke based on middle cerebral artery occlusion for 90 min followed by reperfusion for 7 d, and an in vitro model in primary cultured astrocytes involving oxygen and glucose deprivation for 6 h followed by reoxygenation for 24 h. Both in vivo and in vitro, we found that SB216763, a GSK3β inhibitor, and necrostatin-1 (Nec-1), a RIP1K inhibitor, decreased levels of glial scar markers, including glial fibrillary acidic protein (GFAP), neurocan, and phosphacan. SB216763 and Nec-1 also decreased levels of inflammatory related cytokines, including interleukin-6 (IL-6), interleukin-1 β (IL-1β), and tumor necrosis factor-α (TNF-α). However, only Nec-1 increased the level of interleukin-1 receptor antagonist. Concurrent neutralization of TNF-α, IL-1β, and IL-6 with their antibodies provided better reduction in oxygen and glucose deprivation-induced increases in scar markers than obtained with separate use of each antibody. Further investigations showed that SB216763 reduced the levels of necroptosis-related proteins, including RIP1K, p-RIP1K, RIP3K, p-RIP3K, mixed lineage kinase domain-like protein (MLKL), and p-MLKL, while Nec-1 decreased the expression of p-GSK3β. Compared with Nec-1 (10 μM) and SB216763 (1 μM) alone, Nec-1 and SB216763 in combination reduced levels of GFAP, neurocan, and inflammatory-related cytokines. In conclusion, inhibition of GSK3β or RIP1K reduced glial scar formation induced by ischemic stroke. The underlying mechanisms might be at least, partially related to reducing levels of inflammatory-related cytokines and to blocking an interaction between GSK3β- and RIP1K-mediated pathways.
Collapse
Affiliation(s)
- Jin Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Yong-Ming Zhu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Yi Guo
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Liang Lin
- Department of Anesthesiology, Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Zhan-Xiang Wang
- Department of Anesthesiology, Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Feng Gu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Xin-Yi Dong
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Ming Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Yi-Fan Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Hui-Ling Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| |
Collapse
|
23
|
Nobakht N, Hanna RM, Al-Baghdadi M, Ameen KM, Arman F, Nobahkt E, Kamgar M, Rastogi A. Advances in Autosomal Dominant Polycystic Kidney Disease: A Clinical Review. Kidney Med 2020; 2:196-208. [PMID: 32734239 PMCID: PMC7380379 DOI: 10.1016/j.xkme.2019.11.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Polycystic kidney disease (PKD) is a multiorgan disorder resulting in fluid-filled cyst formation in the kidneys and other systems. The replacement of kidney parenchyma with an ever-increasing volume of cysts eventually leads to kidney failure. Recently, increased understanding of the pathophysiology of PKD and genetic advances have led to new approaches of treatment targeting physiologic pathways, which has been proven to slow the progression of certain types of the disease. We review the pathophysiologic patterns and recent advances in the clinical pharmacotherapy of autosomal dominant PKD. A multipronged approach with pharmacologic and nonpharmacologic treatments can be successfully used to slow down the rate of progression of autosomal dominant PKD to kidney failure.
Collapse
Affiliation(s)
- Niloofar Nobakht
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Ramy M. Hanna
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Division of Nephrology, Department of Medicine, University of California Irvine, Orange, CA
| | - Maha Al-Baghdadi
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Department of Medicine, University of Alabama Birmingham Huntsville Regional Campus, Huntsville, AL
| | - Khalid Mohammed Ameen
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Farid Arman
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Department of Medicine, University of Pennsylvania Medical Center, Philadelphia, PA
| | - Ehsan Nobahkt
- Division of Renal Diseases and Hypertension, Department of Medicine, George Washington University, Washington, DC
| | - Mohammad Kamgar
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Anjay Rastogi
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
24
|
Lu M, Wang P, Qiao Y, Jiang C, Ge Y, Flickinger B, Malhotra DK, Dworkin LD, Liu Z, Gong R. GSK3β-mediated Keap1-independent regulation of Nrf2 antioxidant response: A molecular rheostat of acute kidney injury to chronic kidney disease transition. Redox Biol 2019; 26:101275. [PMID: 31349118 PMCID: PMC6669347 DOI: 10.1016/j.redox.2019.101275] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/05/2019] [Accepted: 07/16/2019] [Indexed: 01/07/2023] Open
Abstract
Transition of acute kidney injury (AKI) to chronic kidney disease (CKD) represents an important cause of kidney failure. However, how AKI is transformed into CKD remains elusive. Following folic acid injury, mice developed AKI with ensuing CKD transition, featured by variable degrees of interstitial fibrosis and tubular cell atrophy and growth arrest. This lingering injury of renal tubules was associated with sustained oxidative stress that was concomitant with an impaired Nrf2 antioxidant defense, marked by mitigated Nrf2 nuclear accumulation and blunted induction of its target antioxidant enzymes, like heme oxygenase (HO)-1. Activation of the canonical Keap1/Nrf2 signaling, nevertheless, seems intact during CKD transition because Nrf2 in injured tubules remained activated and elevated in cytoplasm. Moreover, oxidative thiol modification and activation of Keap1, the cytoplasmic repressor of Nrf2, was barely associated with CKD transition. In contrast, glycogen synthase kinase (GSK)3β, a key modulator of the Keap1-independent Nrf2 regulation, was persistently overexpressed and hyperactive in injured tubules. Likewise, in patients who developed CKD following AKI due to diverse etiologies, like volume depletion and exposure to radiocontrast agents or aristolochic acid, sustained GSK3β overexpression was evident in renal tubules and coincided with oxidative damages, impaired Nrf2 nuclear accumulation and mitigated induction of antioxidant gene expression. Mechanistically, the Nrf2 response against oxidative insult was sabotaged in renal tubular cells expressing a constitutively active mutant of GSK3β, but reinforced by ectopic expression of dominant negative GSK3β in a Keap1-independent manner. In vivo in folic acid-injured mice, targeting GSK3β in renal tubules via conditional knockout or by weekly microdose lithium treatment reinstated Nrf2 antioxidant response in the kidney and hindered AKI to CKD transition. Ergo, our findings suggest that GSK3β-mediated Keap1-independent regulation of Nrf2 may serve as an actionable therapeutic target for modifying the long-term sequelae of AKI. AKI to CKD transition involves sustained GSK3β overactivation and impaired Nrf2 response in injured renal tubules. Microdose lithium rectifies GSK3β overactivity in the kidney, reinstates Nrf2 response and hinders AKI to CKD transition. GSK3β-mediated Keap1-independent regulation of Nrf2 is a novel therapeutic target for modifying long-term sequelae of AKI.
Collapse
Affiliation(s)
- Minglei Lu
- Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Division of Kidney Disease and Hypertension, Brown University School of Medicine, Providence, RI, 02903, United States; Division of Nephrology, University of Toledo College of Medicine, Toledo, OH, 43614, United States
| | - Pei Wang
- Division of Kidney Disease and Hypertension, Brown University School of Medicine, Providence, RI, 02903, United States
| | - Yingjin Qiao
- Division of Kidney Disease and Hypertension, Brown University School of Medicine, Providence, RI, 02903, United States
| | - Chunming Jiang
- Division of Kidney Disease and Hypertension, Brown University School of Medicine, Providence, RI, 02903, United States
| | - Yan Ge
- Division of Kidney Disease and Hypertension, Brown University School of Medicine, Providence, RI, 02903, United States; Division of Nephrology, University of Toledo College of Medicine, Toledo, OH, 43614, United States
| | | | - Deepak K Malhotra
- Division of Nephrology, University of Toledo College of Medicine, Toledo, OH, 43614, United States
| | - Lance D Dworkin
- Division of Kidney Disease and Hypertension, Brown University School of Medicine, Providence, RI, 02903, United States; Division of Nephrology, University of Toledo College of Medicine, Toledo, OH, 43614, United States; Department of Medicine, University of Toledo College of Medicine, Toledo, OH, 43614, United States
| | - Zhangsuo Liu
- Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Rujun Gong
- Division of Kidney Disease and Hypertension, Brown University School of Medicine, Providence, RI, 02903, United States; Division of Nephrology, University of Toledo College of Medicine, Toledo, OH, 43614, United States; Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, 43614, United States.
| |
Collapse
|
25
|
Yao X, Cheng F, Yu W, Rao T, Li W, Zhao S, Zhou X, Ning J. Cathepsin S regulates renal fibrosis in mouse models of mild and severe hydronephrosis. Mol Med Rep 2019; 20:141-150. [PMID: 31115520 PMCID: PMC6580002 DOI: 10.3892/mmr.2019.10230] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 05/02/2019] [Indexed: 12/11/2022] Open
Abstract
As a member of the cysteine protease family, cathepsin S (CTSS) serves an important role in diseases such as cancer, arthritis and atherosclerosis. Nevertheless, its role in renal fibrosis is unknown. In the present study, the effects of CTSS on renal fibrosis in mild (group M) and severe (group S) hydronephrosis were studied by reverse transcription‑-quantitative PCR (RT‑qPCR), western blot analysis (WB), Masson's trichrome staining and immunohistochemical staining in mouse models. The effects of CTSS on extracellular matrix (ECM) deposition and epithelial‑mesenchymal transition (EMT) and the potential mechanisms were further studied by RT‑qPCR and WB in transforming growth factor (TGF‑β1)‑stimulated TCMK‑1 cells. Compared with group N (no hydronephrosis), the expression levels of CTSS in the M and S groups were significantly higher, and a significant increase in ECM deposition was observed in the S group. In addition, compared with group N, the expression levels of TGF‑β1, α‑smooth muscle actin (α‑SMA), SMAD2, SMAD3, phosphorylated (p)SMAD2 and pSMAD3 in groups M and S were significantly higher, whereas the expression of E‑cadherin was significantly lower. Inhibition of CTSS expression increased the expression levels of TGF‑β1, α‑SMA, fibronectin, collagen‑I, SMAD2, SMAD3, pSMAD2 and pSMAD3, whereas E‑cadherin expression decreased. A significant increase in CTSS was observed in the TGF‑β1‑stimulated TCMK‑1 cell line. ECM deposition and EMT were also intensified. The opposite outcomes occurred after intervention with small interfering RNA targeting CTSS. In conclusion, CTSS affected EMT and the deposition of ECM. CTSS may mediate the regulation of fibrosis by the TGF‑β/SMAD signaling pathway. CTSS may serve an important role in the treatment of renal fibrosis.
Collapse
Affiliation(s)
- Xiaobing Yao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Weiming Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Ting Rao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Sheng Zhao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jinzhuo Ning
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
26
|
Guo Q, Wang J, Ge Y, Malhotra DK, Dworkin LD, Wang P, Gong R. Brain natriuretic peptide mitigates TIMP2 induction and reinstates extracellular matrix catabolic activity via GSK3β inhibition in glomerular podocytes exposed to a profibrogenic milieu. Am J Transl Res 2019; 11:964-973. [PMID: 30899395 PMCID: PMC6413260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 12/03/2018] [Indexed: 06/09/2023]
Abstract
Brain natriuretic peptide (BNP) has a demonstrable anti-fibrotic effect on diverse organ systems, including the kidney. To understand the molecular mechanism underlying this renoprotective effect, the efficacy of BNP was examined in an in vitro model of glomerular sclerosis by exposing glomerular podocytes to transforming growth factor (TGF)β1-containing media that recapitulates the profibrogenic milieu in chronic glomerular disease. BNP mitigates extracellular matrix (ECM) accumulation in TGFβ1-treated podocytes, as evidenced by Sirius red assay and staining, concomitant with a restoration of the ECM catabolizing activity, as assessed by pulse chase analysis. This effect was in parallel with a mitigating effect on TGFβ1-elicited overexpression of tissue inhibitor of metalloproteinases (TIMP)2, a key inhibitor of a multitude of ECM-degrading metalloproteinases. Mechanistically, glycogen synthase kinase (GSK)3β, a key player in pathogenesis of podocyte injury and glomerulopathies, seems to be involved. BNP treatment considerably induced GSK3β inhibition, marked by inhibitory phosphorylation at the serine 9 residue, and this significantly correlated with the abrogated TIMP2 induction in TGFβ1-injured podocytes. Moreover, genetic knockout of GSK3β in podocytes is sufficient to attenuate the TGFβ1 induced TIMP2 expression and ECM deposition, reminiscent of the effect of BNP. Conversely, ectopic expression of a nonphosphorylatable GSK3β mutant abolished the inhibitory effect of BNP on TGFβ1-elicited TIMP2 overexpression and ECM accumulation, signifying an essential role of GSK3β inhibition in mediating the effect of BNP. Collectively, BNP possesses an anti-fibrotic activity in glomerular epithelial cells. This finding, if validated in vivo, may open a new avenue to the treatment of glomerulosclerosis.
Collapse
Affiliation(s)
- Qiongqiong Guo
- Department of Hemopurification Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and TechnologyLuoyang, China
| | - Junxia Wang
- Department of Hemopurification Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and TechnologyLuoyang, China
| | - Yan Ge
- Institute of Nephrology, Blood Purification Center, The First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
- Division of Nephrology, Department of Medicine, University of Toledo College of MedicineToledo, Ohio, USA
| | - Deepak K Malhotra
- Division of Nephrology, Department of Medicine, University of Toledo College of MedicineToledo, Ohio, USA
| | - Lance D Dworkin
- Division of Nephrology, Department of Medicine, University of Toledo College of MedicineToledo, Ohio, USA
| | - Pei Wang
- Institute of Nephrology, Blood Purification Center, The First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Rujun Gong
- Division of Nephrology, Department of Medicine, University of Toledo College of MedicineToledo, Ohio, USA
| |
Collapse
|
27
|
Niclosamide alleviates pulmonary fibrosis in vitro and in vivo by attenuation of epithelial-to-mesenchymal transition, matrix proteins & Wnt/β-catenin signaling: A drug repurposing study. Life Sci 2019; 220:8-20. [PMID: 30611787 DOI: 10.1016/j.lfs.2018.12.061] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/31/2018] [Accepted: 12/31/2018] [Indexed: 01/09/2023]
Abstract
Drug repurposing off late has been emerging as an inspiring alternative approach to conventional, exhaustive and arduous process of drug discovery. It is a process of identifying new therapeutic values for a drug already established for the treatment of a certain condition. Our current study is aimed at repurposing the old anti-helimenthic drug Niclosamide as an anti-fibrotic drug against pulmonary fibrosis (PF). PF is most common lethal interstitial lung disease hallmarked by deposition of extracelluar matrix and scarring of lung. Heterogenous nature, untimely diagnosis and lack of appropriate treatment options make PF an inexorable lung disorder. Prevailing void in PF treatment and drug repositioning strategy of drugs kindled our interest to demonstrate the anti-fibrotic activity of Niclosamide. Our study is aimed at investigating the anti-fibrotic potential of Niclosamide in TGF-β1 induced in vitro model of PF and 21-day model of Bleomycin induced PF in vivo respectively. Our study results showed that Niclosamide holds the potential to exert anti-fibrotic effect by hampering fibroblast migration, attenuating EMT, inhibiting fibrotic signaling and by regulating WNT/β-catenin signaling as evident from protein expression studies. Our study findings can give new directions to development of Niclosamide as an anti-fibrotic agent for treatment of pulmonary fibrosis.
Collapse
|
28
|
Kidney fibrosis induced by various irrigation pressures in mouse models of mild and severe hydronephrosis. Int Urol Nephrol 2018; 51:215-222. [PMID: 30483999 DOI: 10.1007/s11255-018-2040-5] [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: 09/30/2018] [Accepted: 11/21/2018] [Indexed: 10/27/2022]
Abstract
OBJECTIVE We want to study whether the degree of fibrosis in the mild and severe hydronephrosis is different, and whether the irrigation pressure will affect the fibrosis of the hydronephrosis. METHODS Animal models of mild and severe hydronephrosis in the left kidney were established: 72 healthy C57BL/6 mice were randomly divided into nine groups (eight in each group). The N group was used as a control group, and 0 mmHg pressure perfusion was given. The M and S groups were used as mild and severe hydronephrosis groups, respectively. The mild and severe hydronephrosis groups were subdivided into eight subgroups, M0-M3 and S0-S3. Among them, groups 0, 1, 2, and 3 were perfused with 0 mmHg, 20 mmHg, 60 mmHg, and 100 mmHg, respectively. We investigated the effects of irrigation pressures on renal fibrosis in mild (group M) and heavy (group S) hydronephrosis by quantitative real-time polymerase chain reaction, Western blot analysis, Masson staining and immunohistochemistry staining in mouse models. RESULTS Compared with group N, EMT and ECM deposits were significantly aggravated in both the mild and severe hydronephrosis groups, TGF-β signaling pathway-related molecules significantly changed too. In terms of ECM deposition, S2 and S3 are significantly increased compared to S0.The EMT of M2 and M3 changed significantly compared with M0; the EMT of S1, S2 and S3 changed significantly compared with S0.The molecules related to TGF-β signaling pathway also changed: M0 and S0 changed significantly compared with N; M1, M2 and M3 changed significantly compared with M0; compared with S0, S1, S2 and S3 changed significantly. CONCLUSION Compared with mild hydronephrosis, renal fibrosis in severe hydronephrosis is more severe and its tolerance to perfusion pressure is lower. These changes may be related to the TGF-β signalling pathway.
Collapse
|
29
|
Singh SP, Huck O, Abraham NG, Amar S. Kavain Reduces Porphyromonas gingivalis-Induced Adipocyte Inflammation: Role of PGC-1α Signaling. THE JOURNAL OF IMMUNOLOGY 2018; 201:1491-1499. [PMID: 30037847 DOI: 10.4049/jimmunol.1800321] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022]
Abstract
A link between obesity and periodontitis has been suggested because of compromised immune response and chronic inflammation in obese patients. In this study, we evaluated the anti-inflammatory properties of Kavain, an extract from Piper methysticum, on Porphyromonas gingivalis-induced inflammation in adipocytes with special focus on peroxisome proliferation-activated receptor γ coactivator α (PGC-1α) and related pathways. The 3T3-L1 mouse preadipocytes and primary adipocytes harvested from mouse adipose tissue were infected with P. gingivalis, and inflammation (TNF-α; adiponectin/adipokines), oxidative stress, and adipogenic marker (FAS, CEBPα, and PPAR-γ) expression were measured. Furthermore, effect of PGC-1α knockdown on Kavain action was evaluated. Results showed that P. gingivalis worsens adipocyte dysfunction through increase of TNF-α, IL-6, and iNOS and decrease of PGC-1α and adiponectin. Interestingly, although Kavain obliterated P. gingivalis-induced proinflammatory effects in wild-type cells, Kavain did not affect PGC-1α-deficient cells, strongly advocating for Kavain effects being mediated by PGC-1α. In vivo adipocytes challenged with i.p. injection of P. gingivalis alone or P. gingivalis and Kavain displayed the same phenotype as in vitro adipocytes. Altogether, our findings established anti-inflammatory and antioxidant effects of Kavain on adipocytes and emphasized protective action against P. gingivalis-induced adipogenesis. The use of compounds such as Kavain offer a portal to potential therapeutic approaches to counter chronic inflammation in obesity-related diseases.
Collapse
Affiliation(s)
- Shailendra P Singh
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595
| | - Olivier Huck
- INSERM, UMR 1260, Regenerative Nanomedicine (Fédération de Médicine Translationalle de Strasbourg), 67000 Strasbourg, France; and.,Periodontology, Dental Faculty, University of Strasbourg, 67000 Strasbourg, France
| | - Nader G Abraham
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595
| | - Salomon Amar
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595;
| |
Collapse
|
30
|
Dexmedetomidine attenuates renal fibrosis via α2-adrenergic receptor-dependent inhibition of cellular senescence after renal ischemia/reperfusion. Life Sci 2018; 207:1-8. [PMID: 29729264 DOI: 10.1016/j.lfs.2018.05.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/23/2018] [Accepted: 05/02/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Renal ischemia/reperfusion (IR) can induce acute kidney injury (AKI), which often progresses to chronic kidney disease (CKD). Dexmedetomidine (Dex), a highly selective α2 adrenergic receptor (α2-AR) agonist, protects against acute renal IR-induced injury. However, the effects of Dex on the transition of AKI to CKD remain unclear. Therefore, we investigated the mechanisms of Dex on renal fibrosis. METHODS Adult male C57BL/6 mice were pretreated with Dex, a specific α2A-adrenergic receptor (AR) blocker (BRL-44408), or a cell senescence inhibitor (rapamycin) in a surgical bilateral renal IR model. The diagnoses of AKI and chronic renal fibrosis were performed by histopathological staining and western blotting. Histopathological changes, cell senescence, tubular fibrotic markers, and the expression of inflammatory factors were studied. RESULTS Pretreatment with Dex alleviated renal IR-induced AKI and chronic tubulointerstitial fibrosis in later stages. Similar to the effects of rapamycin, pretreatment with Dex also decreased the number of senescent tubular cells and weakened the protein expression of senescence-associated markers such as p53, p21, and p16. Furthermore, the expression of inflammatory markers was also decreased in Dex-treated IR mice; and these protective effects of Dex could be abolished by treatment with the specific α2A-AR blocker, BRL-44408. CONCLUSIONS The administration of a single dose of Dex protects against AKI and CKD. Dex inhibits tubular cell senescence and inflammation as well as improves renal fibrosis to moderate the AKI-to-CKD transition. The renal protective potential of Dex may provide a novel treatment strategy for high-risk renal injury patients.
Collapse
|
31
|
Potz BA, Scrimgeour LA, Sabe SA, Clements RT, Sodha NR, Sellke FW. Glycogen synthase kinase 3β inhibition reduces mitochondrial oxidative stress in chronic myocardial ischemia. J Thorac Cardiovasc Surg 2018. [PMID: 29523407 DOI: 10.1016/j.jtcvs.2017.12.127] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVES Glycogen synthase kinase 3β (GSK-3β) inhibition has been reported to increase microvascular density and improve myocardial blood flow in a porcine model of chronic myocardial ischemia and metabolic syndrome. Inhibition of GSK-3β can also be cardioprotective by modulating fibrosis signaling and mitochondrial-induced apoptosis. We hypothesized GSK-3β inhibition would have a beneficial effect on myocardial fibrosis and oxidative stress in a porcine model of chronic myocardial ischemia and metabolic syndrome. METHODS Pigs were fed a high fat diet for 4 weeks followed by placement of an ameroid constrictor to the left circumflex coronary artery. Three weeks later animals received either no drug or a GSK-3β inhibitor. The diets and placebo/GSK-3β inhibition were continued for an additional 5 weeks, the pigs were then euthanized, and the myocardial tissue was harvested. Collagen expression was analyzed via Picrosirius staining. Oxidative stress was analyzed via Oxyblot analysis. Protein expression was analyzed via Western blot. RESULTS GSK-3β inhibition was associated with decreased collagen expression and oxidative stress in the ischemic and nonischemic myocardial tissue compared with control. There was a decrease in profibrotic proteins transforming growth factor-β, p-SMAD2/3, and matrix metalloproteinase-9, and in proapoptotic and oxidative stress proteins, apoptosis inducing factor, the cleaved caspase 3/caspase 3 protein ratio and phosphorylated myeloid cell leukemia sequence-1 in the GSK-3β inhibited group compared with the control. CONCLUSIONS In the setting of metabolic syndrome and chronic myocardial ischemia, inhibition of GSK-3β decreases collagen formation and oxidative stress in myocardial tissue. GSK-3β inhibition might be having this beneficial effect by downregulating transforming growth factor-β/SMAD2/3 signaling and decreasing mitochondrial induced cellular stress.
Collapse
Affiliation(s)
- Brittany A Potz
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Laura A Scrimgeour
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Sharif A Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Richard T Clements
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Neel R Sodha
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI.
| |
Collapse
|
32
|
Amini-Nik S, Yousuf Y, Jeschke MG. Scar management in burn injuries using drug delivery and molecular signaling: Current treatments and future directions. Adv Drug Deliv Rev 2018; 123:135-154. [PMID: 28757325 PMCID: PMC5742037 DOI: 10.1016/j.addr.2017.07.017] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/14/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022]
Abstract
In recent decades, there have been tremendous improvements in burn care that have allowed patients to survive severe burn injuries that were once fatal. However, a major limitation of burn care currently is the development of hypertrophic scars in approximately 70% of patients. This significantly decreases the quality of life for patients due to the physical and psychosocial symptoms associated with scarring. Current approaches to manage scarring include surgical techniques and non-surgical methods such as laser therapy, steroid injections, and compression therapy. These treatments are limited in their effectiveness and regularly fail to manage symptoms. As a result, the development of novel treatments that aim to improve outcomes and quality of life is imperative. Drug delivery that targets the molecular cascades of wound healing to attenuate or prevent hypertrophic scarring is a promising approach that has therapeutic potential. In this review, we discuss current treatments for scar management after burn injury, and how drug delivery targeting molecular signaling can lead to new therapeutic strategies.
Collapse
Affiliation(s)
- Saeid Amini-Nik
- Sunnybrook Research Institute, Toronto, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada.
| | - Yusef Yousuf
- Institute of Medical Science, University of Toronto, Toronto, Canada; Sunnybrook Research Institute, Toronto, Canada
| | - Marc G Jeschke
- Institute of Medical Science, University of Toronto, Toronto, Canada; Sunnybrook Research Institute, Toronto, Canada; Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada; Department of Immunology, University of Toronto, Toronto, Canada; Ross-Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Canada.
| |
Collapse
|
33
|
Inhibition of Glycogen Synthase Kinase 3β Blocks Mesomesenchymal Transition and Attenuates Streptococcus pneumonia-Mediated Pleural Injury in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2461-2472. [PMID: 29073967 DOI: 10.1016/j.ajpath.2017.07.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 07/07/2017] [Accepted: 07/17/2017] [Indexed: 01/13/2023]
Abstract
Pleural loculation affects about 30,000 patients annually in the United States and in severe cases can resolve with restrictive lung disease and pleural fibrosis. Pleural mesothelial cells contribute to pleural rind formation by undergoing mesothelial mesenchymal transition (MesoMT), whereby they acquire a profibrotic phenotype characterized by increased expression of α-smooth muscle actin and collagen 1. Components of the fibrinolytic pathway (urokinase plasminogen activator and plasmin) are elaborated in pleural injury and strongly induce MesoMT in vitro. These same stimuli enhance glycogen synthase kinase (GSK)-3β activity through increased phosphorylation of Tyr-216 in pleural mesothelial cells and GSK-3β mobilization from the cytoplasm to the nucleus. GSK-3β down-regulation blocked induction of MesoMT. Likewise, GSK-3β inhibitor 9ING41 blocked induction of MesoMT and reversed established MesoMT. Similar results were demonstrated in a mouse model of Streptococcus pneumoniae-induced empyema. Intraperitoneal administration of 9ING41, after the induction of pleural injury, attenuated injury progression and improved lung function (lung volume and compliance; P < 0.05 compared with untreated and vehicle controls). MesoMT marker α-smooth muscle actin was reduced in 9ING41-treated mice. Pleural thickening was also notably reduced in 9ING41-treated mice (P < 0.05). Collectively, these studies identify GSK-3β as a newly identified target for amelioration of empyema-related pleural fibrosis and provide a strong rationale for further investigation of GSK-3β signaling in the control of MesoMT and pleural injury.
Collapse
|
34
|
Basile DP, Mehrotra P. Surprising Enhancement of Fibrosis by Tubule-Specific Deletion of the TGF- β Receptor: A New Twist on an Old Paradigm. J Am Soc Nephrol 2017; 28:3427-3429. [PMID: 29074738 DOI: 10.1681/asn.2017080947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- David P Basile
- Department of Cellular and Integrative Physiology, Indiana University, Indianapolis, Indiana
| | - Purvi Mehrotra
- Department of Cellular and Integrative Physiology, Indiana University, Indianapolis, Indiana
| |
Collapse
|
35
|
Wang L, Zhu Y, Wang L, Hou J, Gao Y, Shen L, Zhang J. Effects of chronic alcohol exposure on ischemia-reperfusion-induced acute kidney injury in mice: the role of β-arrestin 2 and glycogen synthase kinase 3. Exp Mol Med 2017. [PMID: 28642577 PMCID: PMC5519017 DOI: 10.1038/emm.2017.76] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Little is known about the effects of chronic alcohol intake on the outcome of acute kidney injury (AKI). Hence, we examined the effects of chronic alcohol intake on the development of renal fibrosis following AKI in an animal model of bilateral renal ischemia–reperfusion (IR) injury. We first found that chronic alcohol exposure exacerbated bilateral IR-induced renal fibrosis and renal function impairment. This phenomenon was associated with increased bilateral IR-induced extracellular matrix deposition and an increased myofibroblast population as well as increased bilateral IR-induced expression of fibrosis-related genes in the kidneys. To explore the mechanisms underlying this phenomenon, we showed that chronic alcohol exposure enhanced β-arrestin 2 (Arrb2) expression and Akt and glycogen synthase kinase-3 (GSK3)β activation in the kidneys. Importantly, pharmacological GSK3 inhibition alleviated bilateral IR-induced renal fibrosis and renal function impairment. Furthermore, we demonstrated that Arrb2−/− mice exhibited resistance to IR-induced renal fibrosis and renal function impairment following chronic alcohol exposure, and these effects were associated with attenuated GSK3β activation in the kidneys. Taken together, our results suggest that chronic alcohol exposure may potentiate AKI via β-arrestin 2/Akt/GSK3β-mediated signaling in the kidney.
Collapse
Affiliation(s)
- Lihua Wang
- Division of Blood Purification, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yifei Zhu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lili Wang
- Division of Blood Purification, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jingjing Hou
- Division of Blood Purification, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yongning Gao
- Division of Blood Purification, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lei Shen
- Division of Blood Purification, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jingyu Zhang
- Department of Hematology, The Second Hospital of Hebei Medical University, Key Laboratory of Hematology of Hebei Province, Shijiazhuang, Hebei, China
| |
Collapse
|
36
|
Di Gregorio J, Sferra R, Speca S, Vetuschi A, Dubuquoy C, Desreumaux P, Pompili S, Cristiano L, Gaudio E, Flati V, Latella G. Role of glycogen synthase kinase-3β and PPAR-γ on epithelial-to-mesenchymal transition in DSS-induced colorectal fibrosis. PLoS One 2017; 12:e0171093. [PMID: 28207769 PMCID: PMC5313173 DOI: 10.1371/journal.pone.0171093] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 01/15/2017] [Indexed: 02/07/2023] Open
Abstract
Background Intestinal fibrosis is characterized by abnormal production and deposition of extracellular matrix (ECM) proteins by activated myofibroblasts. The main progenitor cells of activated myofibroblasts are the fibroblasts and the epithelial cells, the latter through the epithelial-mesenchymal transition (EMT). Aim To evaluate the action of the new PPAR-γ modulator, GED-0507-34 Levo (GED) on the expression of EMT associated and regulatory proteins such as TGF-β, Smad3, E-cadherin, Snail, ZEB1, β-catenin, and GSK-3β, in a mouse model of DSS-induced intestinal fibrosis. Methods Chronic colitis and fibrosis were induced by oral administration of 2.5% DSS (w/v) for 6 weeks. GW9662 (GW), a selective PPAR-γ inhibitor, was also administered by intraperitoneal injection at the dose of 1 mg/kg/day combined with GED treatment. All drugs were administered at the beginning of the second cycle of DSS (day 12). 65 mice were randomly divided into five groups (H2O as controls n = 10, H2O+GED n = 10, DSS n = 15, DSS+GED n = 15, DSS+GED+GW n = 15). The colon was excised for macroscopic examination and histological and morphometric analyses. The level of expression of molecules involved in EMT and fibrosis, like TGF-β, Smad3, E-cadherin, Snail, ZEB1, β-catenin, GSK-3β and PPAR-γ, was assessed by immunohistochemistry, immunofluorescence, western blot and Real Time PCR. Results GED improved the DSS-induced chronic colitis and fibrosis. GED was able to reduce the expression of the main fibrosis markers (α-SMA, collagen I-III and fibronectin) as well as the pivotal pro-fibrotic molecules IL-13, TGF-β and Smad3, while it increased the anti-fibrotic PPAR-γ. All these GED effects were nullified by co-administration of GW with GED. Furthermore, GED was able to normalize the expression levels of E-cadherin and β-catenin and upregulated GSK-3β, that are all known to be involved both in EMT and fibrosis. Conclusions The DSS-induced intestinal fibrosis was improved by the new PPAR-γ modulator GED-0507-34 Levo through the modulation of EMT mediators and pro-fibrotic molecules and through GSK-3β induction.
Collapse
Affiliation(s)
- Jacopo Di Gregorio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Roberta Sferra
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Silvia Speca
- University of Lille, U995, Lille Inflammation Research International Center (LIRIC), F-59000 Lille, France
- IBD, Lille, France
| | - Antonella Vetuschi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
- * E-mail:
| | | | - Pierre Desreumaux
- University of Lille, U995, Lille Inflammation Research International Center (LIRIC), F-59000 Lille, France
- IBD, Lille, France
- CHR Lille, Service des Maladies de l’Appareil Digestif et de la Nutrition, Hôpital Claude Huriez, Lille, France
| | - Simona Pompili
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Loredana Cristiano
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L’Aquila, Italy
| | - Eugenio Gaudio
- Department of Human Anatomy, University of Rome La Sapienza, Rome, Italy
| | - Vincenzo Flati
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Giovanni Latella
- Department of Life, Health and Environmental Sciences, Gastroenterology Unit, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
37
|
Wu X, Wang Y, Wang S, Xu R, Lv X. Purinergic P2X7 receptor mediates acetaldehyde-induced hepatic stellate cells activation via PKC-dependent GSK3β pathway. Int Immunopharmacol 2017; 43:164-171. [PMID: 28061416 DOI: 10.1016/j.intimp.2016.12.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 11/22/2016] [Accepted: 12/12/2016] [Indexed: 12/31/2022]
Abstract
The activation of hepatic stellate cells (HSCs) is an essential part in the development of alcoholic liver fibrosis (ALF). In this study, stimulated HSCs with 200μM acetaldehyde for 48h was used to imitate alcoholic liver fibrosis in vitro. The western blot and qRT-PCR results showed that P2X7R expression was significantly increased in the activation of HSCs after acetaldehyde treatment. Interestingly, activation of P2X7R by stimulating with P2X7R agonist BzATP significantly promoted acetaldehyde-induced CyclinD1 expression, cell proportion in S phase, inflammatory response, and the protein and mRNA levels of α-SMA, collagen I. In contrast, blockage of P2X7R by stimulating with the inhibitor A438079 or transfecting with specific siRNA dramatically suppressed acetaldehyde-induced HSCs activation. Furthermore, PKC activation treated with PMA could obviously up-regulate the expression of α-SMA and collagen I and the phosphorylation of GSK3β, while inhibition of PKC significantly reduced GSK3β activation. Moreover, GSK3β inhibition harvested a dramatic decrease of the mRNA and protein levels of α-SMA and collagen I by suppressing GSK3β phosphorylation. Taken together, these results suggested that purinergic P2X7R mediated acetaldehyde-induced activation of HSCs via PKC-dependent GSK3β pathway, which maybe a novel target for limiting HSCs activation.
Collapse
Affiliation(s)
- Xiaojuan Wu
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Yuhui Wang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Sheng Wang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Rixiang Xu
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Xiongwen Lv
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
38
|
Hsieh PF, Liu SF, Hung TJ, Hung CY, Liu GZ, Chuang LY, Chen MF, Wang JL, Shi MD, Hsu CH, Shiue YL, Yang YL. Elucidation of the therapeutic role of mitochondrial biogenesis transducers NRF-1 in the regulation of renal fibrosis. Exp Cell Res 2016; 349:23-31. [PMID: 27634749 DOI: 10.1016/j.yexcr.2016.09.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 09/07/2016] [Accepted: 09/11/2016] [Indexed: 11/25/2022]
Abstract
BACKGROUND Mitochondrial dysfunction is a newly established risk factor for the development of renal fibrosis. Cell survival and injury repair is facilitated by mitochondrial biogenesis. Nuclear respiratory factor 1 (NRF-1) is a transcriptional regulation factor that plays a central role in the regulation of mitochondrial biogenesis. However, the transcription factor of this process in renal fibrosis is unknown. Thus, we hereby discussed the correlations of NRF-1 and renal interstitial fibrosis. MATERIALS AND METHODS In vitro fibrosis model was established by treatment with transforming growth factor-β1 (TGF-β1) in NRK-49F (Normal Rat kidney fibroblast). We investigated the ROS production, mitochondrial biogenesis and fibrogenic marker (e.q. fibronectin) during the progression of renal fibrosis by kit and Western blotting assay. Here, we used that two distinct mechanisms regulate NRF-1 activation and degradation of NRF-1. NRF-1 was transfect by pcDNA-NRF-1 overexpression gene to evaluate the NRF-1 activity of the therapeutic effect in renal fibrosis. In addition, NRF-1 was silenced by shRNA-NRF-1 to evaluate the significance of NRF-1. ELISA was used to evaluate the secreted fibronectin. Immunofluorescence staining was used to assay the in situ expression of proteins (e.g. fibronectin, NRF-1). RESULTS Under renal fibrosis conditions, TGF-β1 (5ng/ml) increased ROS. Simultaneously, TGF-β1-induced extracellular fibronectin by ELISA assay. In addition, TGF-β1 decreased expression of mitochondrial biogenesis. This is the first time to demonstrate that expression of NRF-1 is significantly decreased in renal fibrosis. However, NRK49F was a transfection with pcDNA-NRF-1 (2μg/ml) expression vector dramatically reverse TGF-β1-induced cellular fibrosis concomitantly with the suppression of fibronectin (both intracellular and extracellular fibronectin). More importantly, transfection with shRNA-NRF-1 (2μg/ml) significantly increased the expression of fibronectin of both intercellular and extracellular origins in NRK-49F cells. DISCUSSION These finding suggest that NRF-1 plays a pivotal role on renal cellular fibrosis. Moreover, NRF-1 might act as a novel renal fibrosis antagonist by down-regulating fibrosis signaling in renal fibroblast cells.
Collapse
Affiliation(s)
- Pei-Fang Hsieh
- Graduate Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, Taiwan; Graduate Institute of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Shu-Fen Liu
- Department of Internal Medicine, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung, Taiwan
| | - Tsung-Jen Hung
- Graduate Institute of Biomedical Science, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Chien-Ya Hung
- Department of Food Nutrition, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Guo-Zheng Liu
- Graduate Institute of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Lea-Yea Chuang
- Department of Biochemistry, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Fen Chen
- Department of Acupressure Technology, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Jue-Long Wang
- Department of Physical Medicine and Rehabilitation, Kaohsiung Veterans General Hospital, Taiwan
| | - Ming-Der Shi
- Graduate Institute of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Tainan, Taiwan; Department of Medical Technology, Kaohsiung Veterans General Hospital Tainan Branch, Tainan, Taiwan
| | - Chen Hung Hsu
- Department of Biological Science and Technology, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Yow-Ling Shiue
- Graduate Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, Taiwan.
| | - Yu-Lin Yang
- Graduate Institute of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Tainan, Taiwan; Graduate Institute of Biomedical Science, Chung Hwa University of Medical Technology, Tainan, Taiwan.
| |
Collapse
|
39
|
Ruan XZ, Guan Y, Liu ZH, Eckardt KU, Unwin R. Summary of ISN Forefronts Symposium 2015: ‘Immunomodulation of Cardio-Renal Function’. Kidney Int Rep 2016. [PMCID: PMC5678622 DOI: 10.1016/j.ekir.2016.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The International Society of Nephrology Forefronts Symposium Immunomodulation of Cardio-Renal Function took place October 22 to 25, 2015, in Shenzhen, China. The program covered basic and clinical aspects of cardio-renal pathophysiology and immunity. Leading scientists from different and related disciplines of clinical and basic research described and reviewed recent discoveries, and discussed emerging topics under the headings “Immunity and Renal Pathophysiology”; “Autoimmunity and the Inflammasome”; “Immunity and the Gut Microbiome”; “Immuno-Metabolism”; “Immunogenetics, Transcriptomics and Epigenetics; “Immunity and Hypertension”; and “Immunity, Fibrosis, and Kidney Disease.”
Collapse
|
40
|
Hsieh PF, Liu SF, Hung TJ, Hung CY, Liu GZ, Chuang LY, Chen MF, Wang JL, Shi MD, Hsu CH, Shiue YL, Yang YL. Treatment with cytokine thymic stromal lymphopoietin short hairpin RNA substantially reduces TGF-β1-induced interstitial cellular fibrosis. Exp Cell Res 2016; 347:153-160. [PMID: 27492484 DOI: 10.1016/j.yexcr.2016.07.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 07/28/2016] [Accepted: 07/30/2016] [Indexed: 11/26/2022]
Abstract
Thymic stromal lymphopoietin (TSLP) has previously been linked to allergic inflammatory diseases, and tissue fibrosis and organ dysfunction may also arise from such inflammation. It remains unclear, however, whether TSLP plays any role in the occurrence of renal fibrosis, so this study investigated that possibility. An in vitro fibrosis model was established by treating normal rat kidney fibroblast (NRK-49F) cells with transforming growth factor-β1 (TGF-β1), after which the levels of various fibrogenic markers (e.g., fibronectin) and downstream fibrogenic signal proteins (e.g., smad 7) were investigated. Also, TSLP shRNA was used to silence the effects of TSLP, while an ELISA was conducted to evaluate the fibronectin secretions. The level of fibronectin in the NRK-49F cells was dose- and time-dependently increased by the administration of exogenous TSLP (P<0.05). TSLP also significantly increased the level of fibrosis signaling, in addition to inducing a marked decrease in the down-regulation of Smad7. Interestingly, the application of TSLP shRNA caused a stark reversal of the TGF-β1-induced cellular fibrosis while simultaneously leading to the suppression of fibronectin and fibrogenic signal proteins. Taken together, these observations provide insights into how extracellular matrices develop and could thus lead to potential therapeutic interventions for the suppression of renal fibrosis.
Collapse
Affiliation(s)
- Pei-Fang Hsieh
- Graduate Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan; Graduate Institute of Medical Laboratory Science and Biotechnology, Chung Hwa Medical University, Tainan, Taiwan
| | - Shu-Fen Liu
- Department of Internal Medicine, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung, Taiwan
| | - Tsung-Jen Hung
- Graduate Institute of Biomedical Science, Chung Hwa Medical University, Tainan, Taiwan
| | - Chien-Ya Hung
- Department of Food nutrition, Chung Hwa Medical University, Tainan, Taiwan
| | - Guo-Zheng Liu
- Graduate Institute of Medical Laboratory Science and Biotechnology, Chung Hwa Medical University, Tainan, Taiwan
| | - Lea-Yea Chuang
- Department of Biochemistry, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Fen Chen
- Department of Acupressure Technology, Chung Hwa Medical University, Tainan, Taiwan
| | - Jue-Long Wang
- Department of Physical Medicine and Rehabilitation, Kaohsiung Veterans General Hospital, Taiwan
| | - Ming-Der Shi
- Graduate Institute of Medical Laboratory Science and Biotechnology, Chung Hwa Medical University, Tainan, Taiwan; Department of Medical Technology, Kaohsiung Veterans General Hospital Tainan Branch, Tainan, Taiwan
| | - Chen Hung Hsu
- Department of Biological Science and Technology, Chung Hwa University of Medical Technology, Tainan, Taiwan; Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yow-Ling Shiue
- Graduate Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan.
| | - Yu-Lin Yang
- Graduate Institute of Medical Laboratory Science and Biotechnology, Chung Hwa Medical University, Tainan, Taiwan.
| |
Collapse
|
41
|
Kakade VR, Tao S, Rajagopal M, Zhou X, Li X, Yu ASL, Calvet JP, Pandey P, Rao R. A cAMP and CREB-mediated feed-forward mechanism regulates GSK3β in polycystic kidney disease. J Mol Cell Biol 2016; 8:464-476. [PMID: 27190311 DOI: 10.1093/jmcb/mjw022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/05/2016] [Accepted: 02/28/2016] [Indexed: 12/12/2022] Open
Abstract
Glycogen synthase kinase 3β (GSK3β), a serine/threonine protein kinase, is commonly known to be regulated at the level of its activity. However, in some diseases including polycystic kidney disease (PKD), GSK3β expression is increased and plays a pathophysiological role. The current studies aimed to determine the mechanism for the increased GSK3β expression in PKD and its significance to disease progression. In mouse models of PKD, increases in renal GSK3β corresponded with increases in renal cAMP levels and disease progression. In vivo and in vitro studies revealed that GSK3β is a cAMP-responsive gene, and elevated cAMP levels, as seen in PKD, can increase GSK3β expression. In normal mice, vasopressin signaling induced by water deprivation increased GSK3β expression, which decreased following rehydration. Examination of the GSK3β promoter revealed five potential binding sites for the transcription factor, cAMP response element binding protein (CREB). CREB was found to bind to GSK3β promoter and essential for cAMP-mediated regulation of GSK3β. Importantly, this regulation was demonstrated to be part of a feed-forward loop in which cAMP through CREB regulates GSK3β expression, and GSK3β in turn positively regulates cAMP generation. GSK3β or CREB inhibition reduced transepithelial fluid secretion and cyst expansion in vitro Thus, disruption at any point of this destructive cycle may be therapeutically useful to reduce cyst expansion and preserve renal function in PKD.
Collapse
Affiliation(s)
- Vijayakumar R Kakade
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Shixin Tao
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Madhumitha Rajagopal
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xia Zhou
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xiaogang Li
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Alan S L Yu
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - James P Calvet
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Pankaj Pandey
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Reena Rao
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
42
|
Yu AS, Zhao L. Effects of the GSK-3β inhibitor (2Z,3E)-6-bromoindirubin-3'-oxime upon ovarian cancer cells. Tumour Biol 2015; 37:4857-64. [PMID: 26526575 DOI: 10.1007/s13277-015-4344-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 10/28/2015] [Indexed: 02/08/2023] Open
Abstract
Ovarian cancer (OC) is a deadly disease, and despite improvements in treatment, overall 5-year survival is low. Glycogen synthase kinase (GSK)-3β is a multifunctional serine/threonine kinase. We wished to ascertain if the GSK-3β inhibitor (2Z,3E)-6-bromoindirubin-3'-oxime, known as "BIO," can suppress OC development. The OC cell lines A2780 and OVCAR3 were exposed to BIO. At different time points, cell proliferation, apoptosis, cell cycle, and cell invasion/cell migration assays were carried out. Phalloidin staining was undertaken to observe lamellipodia formation. Real-time reverse transcription-polymerase chain reaction and western blotting were used to assess expression of messenger RNA (mRNA) and protein of GSK-3β, cyclin D1, matrix metalloproteinase (MMP)-9, and p21. BIO suppressed the proliferation, invasion, and migration of OC cells; reduced lamellipodia formation; and induced G1 arrest of the cell cycle. BIO exposure led to a significant downregulation of mRNA and protein expression of cyclin D1 and MMP9 in comparison with untreated control cells. In contrast, BIO exposure upregulated mRNA and protein expression of p21 in comparison with untreated control cells. Besides, GSK-3β small interfering RNA (siRNA) transfection in ovarian cancer cells also downregulated GSK-3β, cyclin D1, and MMP9 protein expression while upregulated p21 expression. These data suggest that BIO, as an inhibitor of GSK-3β, can suppress OC development. Therefore, BIO could be a candidate drug for the treatment of OC.
Collapse
Affiliation(s)
- Ai-Song Yu
- Department of Preventive Medicine, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Lin Zhao
- Department of Breast Surgery, Liaoning Cancer Hospital & Institute, Shenyang, 110042, People's Republic of China. .,Clinical Oncology College, Dalian Medical University, Shenyang, 110042, People's Republic of China.
| |
Collapse
|