1
|
Zhang C, Guo J. Cell cycle disorders in podocytes: an emerging and increasingly recognized phenomenon. Cell Death Discov 2025; 11:182. [PMID: 40246828 PMCID: PMC12006314 DOI: 10.1038/s41420-025-02486-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/01/2025] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
Proteinuria is observed in various kidney diseases and is frequently associated with a compromised glomerular filtration barrier. Podocytes, as a crucial component of this barrier, play an essential role in preserving the kidney's normal filtration function. Podocytes are terminally differentiated cells that typically do not proliferate. However, certain harmful stimuli can trigger podocytes to re-enter the cell cycle. Due to its unique cytoskeletal structure, podocytes are unable to maintain the structure of the foot process and complete cell division at the same time, eventually form binucleated or multinucleated podocytes. Studies have found that podocytes re-entering the cell cycle are more susceptible to injury, and are prone to detachment from the basement membrane or apoptosis, which are accompanied by the widening of foot processes. This eventually leads to podocyte mitotic catastrophe and the development of proteinuria. Podocyte cell cycle disorders have previously been found mainly in focal segmental glomerulosclerosis and IgA nephropathy. In recent years, this phenomenon has been frequently identified in diabetic kidney disease and lupus nephritis. An expanding body of research has begun to investigate the mechanisms underlying podocyte cell cycle disorders, including cell cycle re-entry, cell cycle arrest, and mitotic catastrophe. This review consolidates the existing literature on podocyte cell cycle disorders in renal diseases and summarizes the molecules that trigger podocyte re-entry into the cell cycle, thereby providing new drug targets for mitigating podocyte damage. This is essential for alleviating podocyte injury, reducing proteinuria, and delaying the progression of kidney diseases.
Collapse
Affiliation(s)
- Chaojie Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jia Guo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, Henan, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
2
|
Yousefi T, Mohammadi Jobani B, Taebi R, Qujeq D. Innovating Cancer Treatment Through Cell Cycle, Telomerase, Angiogenesis, and Metastasis. DNA Cell Biol 2024; 43:438-451. [PMID: 39018567 DOI: 10.1089/dna.2024.0109] [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] [Indexed: 07/19/2024] Open
Abstract
Cancer remains a formidable challenge in the field of medicine, necessitating innovative therapeutic strategies to combat its relentless progression. The cell cycle, a tightly regulated process governing cell growth and division, plays a pivotal role in cancer development. Dysregulation of the cell cycle allows cancer cells to proliferate uncontrollably. Therapeutic interventions designed to disrupt the cell cycle offer promise in restraining tumor growth and progression. Telomerase, an enzyme responsible for maintaining telomere length, is often overactive in cancer cells, conferring them with immortality. Targeting telomerase presents an opportunity to limit the replicative potential of cancer cells and hinder tumor growth. Angiogenesis, the formation of new blood vessels, is essential for tumor growth and metastasis. Strategies aimed at inhibiting angiogenesis seek to deprive tumors of their vital blood supply, thereby impeding their progression. Metastasis, the spread of cancer cells from the primary tumor to distant sites, is a major challenge in cancer therapy. Research efforts are focused on understanding the underlying mechanisms of metastasis and developing interventions to disrupt this deadly process. This review provides a glimpse into the multifaceted approach to cancer therapy, addressing critical aspects of cancer biology-cell cycle regulation, telomerase activity, angiogenesis, and metastasis. Through ongoing research and innovative strategies, the field of oncology continues to advance, offering new hope for improved treatment outcomes and enhanced quality of life for cancer patients.
Collapse
Affiliation(s)
- Tooba Yousefi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahareh Mohammadi Jobani
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reyhaneh Taebi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Durdi Qujeq
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
3
|
Liebisch M, Wolf G. Role of Epigenetic Changes in the Pathophysiology of Diabetic Kidney Disease. GLOMERULAR DISEASES 2024; 4:211-226. [PMID: 39649441 PMCID: PMC11623970 DOI: 10.1159/000541923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/08/2024] [Indexed: 12/10/2024]
Abstract
Background Diabetic kidney disease (DKD) is a global health issue. Epigenetic changes play an important role in the pathogenesis of this disease. Summary DKD is currently the leading cause of kidney failure worldwide. Although much is known about the pathophysiology of DKD, the research field of epigenetics is relatively new. Several recent studies have demonstrated that diabetes-induced dysregulation of epigenetic mechanisms alters the expression of pathological genes in kidney cells. If these changes persist for a long time, the so-called "metabolic memory" could be established. In this review, we highlight diabetes-induced epigenetic modifications associated with DKD. While there is a substantial amount of literature on epigenetic changes, only a few studies describe the underlying molecular mechanisms. Detailed analyses have shown that epigenetic changes play an important role in known pathological features of DKD, such as podocyte injury, fibrosis, accumulation of extracellular matrix, or oxidative injury, all of which contribute to the pathophysiology of disease. The transforming growth factor-β plays a key role as it is involved in all-mentioned epigenetic types of regulation. Key Messages Epigenetic is crucial for the development and progression of DKD, but the detailed molecular mechanisms have to be further analyzed more in detail.
Collapse
Affiliation(s)
- Marita Liebisch
- Department of Internal Medicine III, University Hospital Jena, Jena, Germany
| | - Gunter Wolf
- Department of Internal Medicine III, University Hospital Jena, Jena, Germany
| |
Collapse
|
4
|
Chevalier RL. Why is chronic kidney disease progressive? Evolutionary adaptations and maladaptations. Am J Physiol Renal Physiol 2023; 325:F595-F617. [PMID: 37675460 DOI: 10.1152/ajprenal.00134.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/08/2023] [Accepted: 08/27/2023] [Indexed: 09/08/2023] Open
Abstract
Despite significant advances in renal physiology, the global prevalence of chronic kidney disease (CKD) continues to increase. The emergence of multicellular organisms gave rise to increasing complexity of life resulting in trade-offs reflecting ancestral adaptations to changing environments. Three evolutionary traits shape CKD over the lifespan: 1) variation in nephron number at birth, 2) progressive nephron loss with aging, and 3) adaptive kidney growth in response to decreased nephron number. Although providing plasticity in adaptation to changing environments, the cell cycle must function within constraints dictated by available energy. Prioritized allocation of energy available through the placenta can restrict fetal nephrogenesis, a risk factor for CKD. Moreover, nephron loss with aging is a consequence of cell senescence, a pathway accelerated by adaptive nephron hypertrophy that maintains metabolic homeostasis at the expense of increased vulnerability to stressors. Driven by reproductive fitness, natural selection operates in early life but diminishes thereafter, leading to an exponential increase in CKD with aging, a product of antagonistic pleiotropy. A deeper understanding of the evolutionary constraints on the cell cycle may lead to manipulation of the balance between progenitor cell renewal and differentiation, regulation of cell senescence, and modulation of the balance between cell proliferation and hypertrophy. Application of an evolutionary perspective may enhance understanding of adaptation and maladaptation by nephrons in the progression of CKD, leading to new therapeutic advances.
Collapse
Affiliation(s)
- Robert L Chevalier
- Department of Pediatrics, The University of Virginia, Charlottesville, Virginia, United States
| |
Collapse
|
5
|
Suvakov S, Kattah AG, Gojkovic T, Enninga EAL, Pruett J, Jayachandran M, Sousa C, Santos J, Abou Hassan C, Gonzales-Suarez M, Garovic VD. Impact of Aging and Cellular Senescence in the Pathophysiology of Preeclampsia. Compr Physiol 2023; 13:5077-5114. [PMID: 37770190 DOI: 10.1002/cphy.c230003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
The incidence of hypertensive disorders of pregnancy is increasing, which may be due to several factors, including an increased age at pregnancy and more comorbid health conditions during reproductive years. Preeclampsia, the most severe hypertensive disorder of pregnancy, has been associated with an increased risk of future disease, including cardiovascular and kidney diseases. Cellular senescence, the process of cell cycle arrest in response to many physiologic and maladaptive stimuli, may play an important role in the pathogenesis of preeclampsia and provide a mechanistic link to future disease. In this article, we will discuss the pathophysiology of preeclampsia, the many mechanisms of cellular senescence, evidence for the involvement of senescence in the development of preeclampsia, as well as evidence that cellular senescence may link preeclampsia to the risk of future disease. Lastly, we will explore how a better understanding of the role of cellular senescence in preeclampsia may lead to therapeutic trials. © 2023 American Physiological Society. Compr Physiol 13:5077-5114, 2023.
Collapse
Affiliation(s)
- Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea G Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamara Gojkovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Elizabeth A L Enninga
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Jacob Pruett
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Ciria Sousa
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Janelle Santos
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
6
|
Ida-Naitoh M, Tokuyama H, Futatsugi K, Yasuda M, Adachi K, Kanda T, Tanabe Y, Wakino S, Itoh H. Proximal-tubule molecular relay from early Protein diaphanous homolog 1 to late Rho-associated protein kinase 1 regulates kidney function in obesity-induced kidney damage. Kidney Int 2022; 102:798-814. [PMID: 35716954 DOI: 10.1016/j.kint.2022.05.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 04/28/2022] [Accepted: 05/09/2022] [Indexed: 01/11/2023]
Abstract
The small GTPase protein RhoA has two effectors, ROCK (Rho-associated protein kinase 1) and mDIA1 (Protein diaphanous homolog 1), which cooperate reciprocally. However, temporal regulation of RhoA and its effectors in obesity-induced kidney damage remains unclear. Here, we investigated the role of RhoA activation in the proximal tubules at the early and late stages of obesity-induced kidney damage. In mice, a three week high-fat diet induced proximal tubule hypertrophy and damage without increased albuminuria, and RhoA/mDIA1 activation without ROCK activation. Conversely, a 12- week high-fat diet induced proximal tubule hypertrophy, proximal tubule damage, increased albuminuria, and RhoA/ROCK activation without mDIA1 elevation. Proximal tubule hypertrophy resulting from cell cycle arrest accompanied by downregulation of the multifunctional cyclin-dependent kinase inhibitor p27Kip1 was elicited by RhoA activation. Mice overexpressing proximal tubule-specific and dominant-negative RHOA display amelioration of high-fat diet-induced kidney hypertrophy, cell cycle abnormalities, inflammation, and renal impairment. In human proximal tubules cells, mechanical stretch mimicking hypertrophy activated ROCK, which triggered inflammation. In human kidney samples from normal individuals with a body mass index of about 25, proximal tubule cell size correlated with body mass index, proximal tubule cell damages, and mDIA1 expression. Thus, RhoA activation in proximal tubules is critical for the initiation and progression of obesity-induced kidney damage. Hence, the switch in the downstream RhoA effector in proximal tubule represents a transition from normal to pathogenic kidney adaptation and to body weight gain, leading to obesity-induced kidney damage.
Collapse
Affiliation(s)
- Makiko Ida-Naitoh
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Hirobumi Tokuyama
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Koji Futatsugi
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Marie Yasuda
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Keika Adachi
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Takeshi Kanda
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Yoshiyuki Tanabe
- Department of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan
| | - Shu Wakino
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan.
| | - Hiroshi Itoh
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| |
Collapse
|
7
|
Effect of 5'-fluoro-2'-deoxycytidine, 5-azacytidine, and 5-aza-2'–deoxycytidine on DNA Methyltransferase 1, CIP/KIP Family, and INK4a/ARF in Colon Cancer HCT-116 Cell Line. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2022. [DOI: 10.5812/ijcm.110419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Cyclin-dependent kinase inhibitors (CKIs) are the negative regulator of cell cycle progression, which inhibits cyclin-cdk complexes, resulting in cell cycle arrest. Recently, we evaluated the effect of 5-Aza-CdR on DNMT1 gene expression in the WCH-17 hepatocellular carcinoma (HCC) cell line. Objectives: The current study was designed to analyze the effects of 5-aza-2'–deoxycytidine (5-Aza-CdR, decitabine), 5-azacytidine (5-AzaC, vidaza), and 5'-fluoro-2'-deoxycytidine (FdCyd) on INK4a/ARF, CIP/KIP, and DNA methyltransferase 1 gene expression, apoptosis induction, and cell growth inhibition in colon cancer HCT-116 cell line. Methods: The colon cancer HCT-116 cell line was treated with 5-azaC, 5-Aza-CdR, and FdCyd at 24 and 48h. To determine colon cancer HCT-116 cell viability, cell apoptosis, and the relative expression level of the INK4a/ARF, CIP/KIP, and DNA methyltransferase 1 genes, MTT assay, flow cytometry, and qRT-PCR were done, respectively. Results: 5-azaC, 5-Aza-CdR, and FdCyd significantly inhibited colon cancer HCT-116 cell growth and induced apoptosis. Besides, they significantly increased CIP/KIP (p21CIP1, p27KIP1, and p57KIP2) and INK4 (p14ARF, p15INK4b, and p16INK4a) and decreased DNMT1 gene expression. Besides, minimal and maximal apoptosis were seen in the groups treated with FdCyd and 5-Aza-CdR, respectively. The IC50 for CAF for FdCyd was 1.72 ± 0.23 and 1.63 ± 0.21μM at 24 and 48h, respectively. The IC50 for CAF for 5-AzaC was 2.18 ± 0.33 and 1.98 ± 0.29 μM at 24 and 48h, respectively. The IC50 for CAF for 5-Aza-CdR was 4.08 ± 0.61 and 3.18 ± 0.50 μM at 24 and 48h, respectively. Conclusions: The 5-azac, 5-Aza-CdR, and FdCyd can reactivate the INK4a/ARF and CIP/KIP families through inhibition of DNMT1 activity.
Collapse
|
8
|
Yin L, Yu L, He JC, Chen A. Controversies in Podocyte Loss: Death or Detachment? Front Cell Dev Biol 2021; 9:771931. [PMID: 34881244 PMCID: PMC8645964 DOI: 10.3389/fcell.2021.771931] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/03/2021] [Indexed: 12/21/2022] Open
Abstract
Glomerular podocytes are characterized by terminally differentiated epithelial cells with limited proliferating ability; thus, podocyte loss could not be fully compensated by podocyte regeneration. A large body of clinical studies collectively demonstrated that podocyte loss correlated with glomerular diseases progression. Both podocyte death and podocyte detachment lead to podocyte loss; however, which one is the main cause remains controversial. Up to date, multiple mechanisms are involved in podocyte death, including programmed apoptotic cell death (apoptosis and anoikis), programmed nonapoptotic cell death (autophagy, entosis, and podoptosis), immune-related cell death (pyroptosis), and other types of cell death (necroptosis and mitotic catastrophe-related cell death). Apoptosis is considered a common mechanism of podocyte loss; however, most of the data were generated in vitro and the evidence of in vivo podocyte apoptosis is limited. The isolation of podocytes in the urine and subsequent culture of urinary podocytes in vitro suggest that detachment of viable podocytes could be another important mechanism for podocyte loss. In this review, we summarize recent advances that address this controversial topic on the specific circumstances of podocyte loss.
Collapse
Affiliation(s)
- Lijun Yin
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Lu Yu
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China.,Department of Health Sciences, Boston University College of Health and Rehabilitation Sciences: Sargent College, Boston University, Boston, MA, United States
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Renal Program, James J. Peters Veterans Affairs Medical Center at Bronx, New York, NY, United States
| | - Anqun Chen
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| |
Collapse
|
9
|
Zhou J, Yang J, Wang YM, Ding H, Li TS, Liu ZH, Chen L, Jiao RQ, Zhang DM, Kong LD. IL-6/STAT3 signaling activation exacerbates high fructose-induced podocyte hypertrophy by ketohexokinase-A-mediated tristetraprolin down-regulation. Cell Signal 2021; 86:110082. [PMID: 34252535 DOI: 10.1016/j.cellsig.2021.110082] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/25/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023]
Abstract
Glomerular hypertrophy is a crucial factor of severe podocyte damage and proteinuria. Our previous study showed that high fructose induced podocyte injury. The current study aimed to explore a novel molecular mechanism underlying podocyte hypertrophy induced by high fructose. Here we demonstrated for the first time that high fructose significantly initiated the hypertrophy in rat glomeruli and differentiated human podocytes (HPCs). Consistently, it induced inflammatory response with the down-regulation of anti-inflammatory factor zinc-finger protein tristetraprolin (TTP) and the activation of interleukin-6 (IL-6)/signal transducer and activator of transcription 3 (STAT3) signaling in these animal and cell models. Subsequently, high-expression of microRNA-92a-3p (miR-92a-3p) and its target protein cyclin-dependent kinase inhibitor p57 (P57) down-regulation, representing abnormal proliferation and apoptosis, were observed in vivo and in vitro. Moreover, high fructose increased ketohexokinase-A (KHK-A) expression in rat glomeruli and differentiated HPCs. Exogenous IL-6 stimulation up-regulated IL-6/STAT3 signaling and miR-92a-3p, reduced P57 expression and promoted podocyte proliferation, apoptosis and hypertrophy in vitro. The data from anti-inflammatory agent maslinic acid treatment or TTP siRNA transfection showed that high fructose may decrease TTP to activate IL-6/STAT3 signaling in podocyte overproliferation and apoptosis, causing podocyte hypertrophy. Whereas, KHK-A siRNA transfection remarkably restored high fructose-induced TTP down-regulation, IL-6/STAT3 signaling activation, podocyte overproliferation, apoptosis and hypertrophy in differentiated HPCs. Taken together, these results suggested that high fructose possibly increased KHK-A expression to down-regulate TTP, subsequently activated IL-6/STAT3 signaling to interfere with podocyte proliferation and apoptosis by up-regulating miR-92a-3p to suppress P57 expression, causing podocyte hypertrophy. Therefore, the inactivation of IL-6/STAT3 to relieve podocyte hypertrophy mediated by inhibiting KHK-A to increase TTP may be a novel strategy for high fructose diet-associated podocyte injury and proteinuria.
Collapse
Affiliation(s)
- Jie Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, PR China
| | - Jie Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, PR China
| | - Yu-Meng Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, PR China
| | - Hong Ding
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, PR China
| | - Tu-Shuai Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, PR China
| | - Zhi-Hong Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, PR China
| | - Li Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, PR China
| | - Rui-Qing Jiao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, PR China
| | - Dong-Mei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, PR China
| | - Ling-Dong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, PR China.
| |
Collapse
|
10
|
Cheval L, Viollet B, Klein C, Rafael C, Figueres L, Devevre E, Zadigue G, Azroyan A, Crambert G, Vogt B, Doucet A. Acidosis-induced activation of distal nephron principal cells triggers Gdf15 secretion and adaptive proliferation of intercalated cells. Acta Physiol (Oxf) 2021; 232:e13661. [PMID: 33840159 DOI: 10.1111/apha.13661] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 04/03/2021] [Accepted: 04/05/2021] [Indexed: 12/11/2022]
Abstract
AIM Type A intercalated cells of the renal collecting duct participate in the maintenance of the acid/base balance through their capacity to adapt proton secretion to homeostatic requirements. We previously showed that increased proton secretion stems in part from the enlargement of the population of proton secreting cells in the outer medullary collecting duct through division of fully differentiated cells, and that this response is triggered by growth/differentiation factor 15. This study aimed at deciphering the mechanism of acid load-induced secretion of Gdf15 and its mechanism of action. METHODS We developed an original method to evaluate the proliferation of intercalated cells and applied it to genetically modified or pharmacologically treated mice under basal and acid-loaded conditions. RESULTS Gdf15 is secreted by principal cells of the collecting duct in response to the stimulation of vasopressin receptors. Vasopressin-induced production of cAMP triggers activation of AMP-stimulated kinases and of Na,K-ATPase, and induction of p53 and Gdf15. Gdf15 action on intercalated cells is mediated by ErbB2 receptors, the activation of which triggers the expression of cyclin d1, of p53 and anti-proliferative genes, and of Egr1. CONCLUSION Acidosis-induced proliferation of intercalated cells results from a cross talk with principal cells which secrete Gdf15 in response to their stimulation by vasopressin. Thus, vasopressin is a major determinant of the collecting duct cellular homeostasis as it promotes proliferation of intercalated cells under acidosis conditions and of principal cells under normal acid-base status.
Collapse
Affiliation(s)
- Lydie Cheval
- Centre de Recherche des Cordeliers INSERMSorbonne UniversitéUniversité de Paris Paris France
- CNRS ERL 8228 ‐ Laboratoire de Physiologie Rénale et Tubulopathies Paris France
| | - Benoit Viollet
- Université de ParisInstitut CochinINSERMCNRS Paris France
| | - Christophe Klein
- Centre de Recherche des Cordeliers INSERMSorbonne UniversitéUniversité de Paris Paris France
| | - Chloé Rafael
- Centre de Recherche des Cordeliers INSERMSorbonne UniversitéUniversité de Paris Paris France
- CNRS ERL 8228 ‐ Laboratoire de Physiologie Rénale et Tubulopathies Paris France
| | - Lucile Figueres
- Centre de Recherche des Cordeliers INSERMSorbonne UniversitéUniversité de Paris Paris France
- CNRS ERL 8228 ‐ Laboratoire de Physiologie Rénale et Tubulopathies Paris France
| | - Estelle Devevre
- Centre de Recherche des Cordeliers INSERMSorbonne UniversitéUniversité de Paris Paris France
| | - Georges Zadigue
- Centre de Recherche des Cordeliers INSERMSorbonne UniversitéUniversité de Paris Paris France
| | - Anie Azroyan
- Program in Membrane Biology Nephrology Division Center for Systems Biology Massachusetts General Hospital Harvard Medical School Boston MA USA
| | - Gilles Crambert
- Centre de Recherche des Cordeliers INSERMSorbonne UniversitéUniversité de Paris Paris France
- CNRS ERL 8228 ‐ Laboratoire de Physiologie Rénale et Tubulopathies Paris France
| | - Bruno Vogt
- Department of Nephrology and Hypertension, Inselspital Bern University Hospital Bern Switzerland
| | - Alain Doucet
- Centre de Recherche des Cordeliers INSERMSorbonne UniversitéUniversité de Paris Paris France
- CNRS ERL 8228 ‐ Laboratoire de Physiologie Rénale et Tubulopathies Paris France
| |
Collapse
|
11
|
Lee K, Gusella GL, He JC. Epithelial proliferation and cell cycle dysregulation in kidney injury and disease. Kidney Int 2021; 100:67-78. [PMID: 33831367 PMCID: PMC8855879 DOI: 10.1016/j.kint.2021.03.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 03/05/2021] [Accepted: 03/11/2021] [Indexed: 02/08/2023]
Abstract
Various cellular insults and injury to renal epithelial cells stimulate repair mechanisms to adapt and restore the organ homeostasis. Renal tubular epithelial cells are endowed with regenerative capacity, which allows for a restoration of nephron function after acute kidney injury. However, recent evidence indicates that the repair is often incomplete, leading to maladaptive responses that promote the progression to chronic kidney disease. The dysregulated cell cycle and proliferation is also a key feature of renal tubular epithelial cells in polycystic kidney disease and HIV-associated nephropathy. Therefore, in this review, we provide an overview of cell cycle regulation and the consequences of dysregulated cell proliferation in acute kidney injury, polycystic kidney disease, and HIV-associated nephropathy. An increased understanding of these processes may help define better targets for kidney repair and combat chronic kidney disease progression.
Collapse
Affiliation(s)
- Kyung Lee
- Department of Medicine, Nephrology Division, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| | - G Luca Gusella
- Department of Medicine, Nephrology Division, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - John Cijiang He
- Department of Medicine, Nephrology Division, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Renal Program, James J. Peters Veterans Affairs Medical Center, Bronx, New York, USA.
| |
Collapse
|
12
|
Kawaguchi T, Hasegawa K, Yasuda I, Muraoka H, Umino H, Tokuyama H, Hashiguchi A, Wakino S, Itoh H. Diabetic condition induces hypertrophy and vacuolization in glomerular parietal epithelial cells. Sci Rep 2021; 11:1515. [PMID: 33452384 PMCID: PMC7810998 DOI: 10.1038/s41598-021-81027-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 12/09/2020] [Indexed: 01/29/2023] Open
Abstract
Diabetic nephropathy (DN) is accompanied by characteristic changes in the glomerulus, but little is known about the effect of diabetes on parietal epithelial cells (PECs). In this study, a descriptive analysis of PECs was undertaken in diabetic db/db mice and in diabetic patients. PEC hypertrophy was significantly more prominent in diabetic mice than in nondiabetic mice, and this was evident even at the early stage. Additionally, the number of vacuoles in PECs was markedly increased in diabetic mice, suggesting the presence of cellular injury in PECs in DN. Although rare, binuclear cells were observed in mice with early diabetes. In cultured PECs, a high glucose condition, compared with normal glucose condition, induced cellular hypertrophy and apoptosis. Flow cytometry showed that some PECs in the G0 phase reentered the cell cycle but got arrested in the S phase. Finally, in human diabetic subjects, hypertrophy and vacuolization were observed in the PECs. Our data showed that PECs undergo substantial changes in DN and may participate in rearrangement for differentiation into podocytes.
Collapse
Affiliation(s)
- Takahisa Kawaguchi
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Kazuhiro Hasegawa
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Itaru Yasuda
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Hirokazu Muraoka
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Hiroyuki Umino
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Hirobumi Tokuyama
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Akinori Hashiguchi
- grid.26091.3c0000 0004 1936 9959Department of Pathology, School of Medicine, Keio University, Tokyo, 160-8582 Japan
| | - Shu Wakino
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Hiroshi Itoh
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| |
Collapse
|
13
|
Zhou X, Lu Y, Guo P, Zhou C. Upregulation of microRNA‑140‑3p mediates dachshund family transcription factor 1 expression in immunoglobulin A nephropathy through cell cycle‑dependent mechanisms. Mol Med Rep 2020; 23:134. [PMID: 33313942 PMCID: PMC7751451 DOI: 10.3892/mmr.2020.11773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/11/2020] [Indexed: 12/03/2022] Open
Abstract
Immunoglobulin A nephropathy (IgAN) is a kidney disease and one of the commonest forms of glomerulonephritis worldwide. The present study investigated the role of dachshund family transcription factor 1 (DACH1) in IgAN and identified one of its binding microRNAs (miRNAs). The expression of DACH1 in human mesangial cells (HMCs) incubated with polymeric IgA (pIgA) isolated and purified from the serum of patients with IgAN or healthy individuals was evaluated by reverse transcription-quantitative (RT-q) PCR and western blotting. Cell proliferation and cell cycle assays were performed in DACH1-overexpressing HMCs to identify the role of DACH1 in IgAN and enzyme-linked immunosorbent assay was carried out to verify the release of inflammatory factors from HMCs. The target miRNAs of DACH1 were predicted using bioinformatics software and miR-140-3p was identified as a target of DACH1 by luciferase report assay, RT-qPCR and western blotting. The results demonstrated that DACH1 was downregulated in HMCs cultured with pIgA-IgAN at both mRNA and protein levels. Overexpression of DACH1 suppressed HMC growth and inhibited inflammatory cytokine release from HMCs cultured with pIgA-IgAN. The expression of DACH1 was negatively regulated by miR-140-3p in IgAN and miR-140-3p inhibition suppressed HMC growth and inhibited inflammatory cytokine release from HMCs cultured with pIgA-IgAN. The findings of the present study demonstrated that DACH1 decreased HMC growth and the release of inflammatory cytokines from HMCs may be targeted by miR-140-3p. The results suggested that DACH1 could be associated with the progression of IgAN and provide a potential target for further studies related to the mechanism of IgAN.
Collapse
Affiliation(s)
- Xiaobin Zhou
- Department of Clinical Laboratory, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Yao Lu
- Department of Teaching Research of Medical Technology, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Pengfei Guo
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, Shanghai 200072, P.R. China
| | - Chenglin Zhou
- Department of Clinical Laboratory, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| |
Collapse
|
14
|
Targeting AMP-activated protein kinase (AMPK) for treatment of autosomal dominant polycystic kidney disease. Cell Signal 2020; 73:109704. [DOI: 10.1016/j.cellsig.2020.109704] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 02/06/2023]
|
15
|
Inhibitor of growth 2 regulates the high glucose-induced cell cycle arrest and epithelial-to-mesenchymal transition in renal proximal tubular cells. J Physiol Biochem 2020; 76:373-382. [PMID: 32424454 DOI: 10.1007/s13105-020-00743-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/23/2020] [Indexed: 02/06/2023]
Abstract
The epithelial-to-mesenchymal transition (EMT)-based tubulointerstitial fibrosis is the major pathological feature of diabetic kidney disease (DKD). While several studies have linked cell cycle dysregulation to various kidney injuries in recent years, its involvement in fibrosis of DKD is far from being clarified. ING2 (inhibitor of growth 2) is the second member of the inhibitor of growth family and participates in the regulation of many cellular processes. So far the role of ING2 in DKD remains largely unknown. In the present study, ING2 expression was detected by western blotting and immunofluorescent staining both in vitro high glucose-stimulated human proximal tubular epithelial cells (HK-2) and in vivo streptozotocin-induced diabetic mice. Cell proliferation was analyzed by CCK-8 and EdU assay, and cell cycle arrest was measured by flow cytometry. Quantitative polymerase chain reaction (qPCR) and western blotting were used to detect the EMT markers, and the p53 signaling activation was evaluated by chromatin immunoprecipitation (ChIP), qPCR, and western blotting. We found that the proliferation of the cells was reduced upon high glucose stimulation, which was accompanied by cell cycle arrest. The expression of ING2 was increased in hyperglycemia conditions both in vivo and in vitro. ING2 suppression ameliorated the reduced proliferation and cell cycle arrest induced by high glucose in HK-2 cells. Moreover, ING2 knockdown suppressed p21 expression by reducing p53 acetylation and finally alleviated the EMT progress in the high glucose-stimulated HK-2 cells. Our study demonstrated that cell cycle regulation is bound up with the kidney fibrosis in DKD, suggesting a novel function of ING2 as a potential therapeutic strategy targeting cell cycle arrest for DKD.
Collapse
|
16
|
Matsushita K, Toyoda T, Yamada T, Morikawa T, Ogawa K. Comprehensive expression analysis of mRNA and microRNA for the investigation of compensatory mechanisms in the rat kidney after unilateral nephrectomy. J Appl Toxicol 2020; 40:1373-1383. [PMID: 32369870 DOI: 10.1002/jat.3990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 01/02/2023]
Abstract
Compensation is a physiological response that occurs during chemical exposure to maintain homeostasis. Because compensatory responses are not usually considered adverse effects, it is important to understand compensatory mechanisms for chemical risk assessment. Although the kidney is a major target organ for toxicity, there is controversy over whether hyperplasia or hypertrophy contributes to the compensatory mechanism, and there is limited information to apply for chemical risk assessment. In the present study, compensatory mechanisms of the kidney were investigated in a unilateral nephrectomy (UNx) model using adult male and female F344 rats. In residual kidneys of male and female rats after UNx, 5-bromo-2'-deoxyuridine-labeling indices and mRNA expression of cell cycle-related genes were increased, although there were no fluctuations in mRNA expression of transforming growth factor-β1, which contributes to hypertrophy in renal tubules. Pathway analysis using mRNA expression data from a complementary DNA (cDNA) microarray revealed that canonical pathways related to cell proliferation were mainly activated and that forkhead box M1 (FOXM1) was an upstream regulator of compensatory cell proliferation in residual kidneys of male and female rats. cDNA microarray for microRNAs (miRNAs) demonstrated that nine miRNAs were downregulated in residual kidneys, and mRNA/miRNA integrated analysis indicated that miRNAs were associated with the expression of factors downstream of FOXM1. Overall, these results suggested that FOXM1-mediated hyperplasia rather than hypertrophy contributed to compensatory mechanisms in the kidney and that miRNAs regulated downstream FOXM1 signaling. These results will be beneficial for evaluating nephrotoxicity in chemical risk assessment and for developing new biomarkers to predict nephrotoxicity.
Collapse
Affiliation(s)
- Kohei Matsushita
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Takeshi Toyoda
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Takanori Yamada
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan.,Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Tomomi Morikawa
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Kumiko Ogawa
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| |
Collapse
|
17
|
Lysophosphatidic acid increases mesangial cell proliferation in models of diabetic nephropathy via Rac1/MAPK/KLF5 signaling. Exp Mol Med 2019; 51:1-10. [PMID: 30770784 PMCID: PMC6377648 DOI: 10.1038/s12276-019-0217-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 11/20/2018] [Accepted: 11/23/2018] [Indexed: 12/11/2022] Open
Abstract
Mesangial cell proliferation has been identified as a major factor contributing to glomerulosclerosis, which is a typical symptom of diabetic nephropathy (DN). Lysophosphatidic acid (LPA) levels are increased in the glomerulus of the kidney in diabetic mice. LPA is a critical regulator that induces mesangial cell proliferation; however, its effect and molecular mechanisms remain unknown. The proportion of α-SMA+/PCNA+ cells was increased in the kidney cortex of db/db mice compared with control mice. Treatment with LPA concomitantly increased the proliferation of mouse mesangial cells (SV40 MES13) and the expression of cyclin D1 and CDK4. On the other hand, the expression of p27Kip1 was decreased. The expression of Krüppel-like factor 5 (KLF5) was upregulated in the kidney cortex of db/db mice and LPA-treated SV40 MES13 cells. RNAi-mediated silencing of KLF5 reversed these effects and inhibited the proliferation of LPA-treated cells. Mitogen-activated protein kinases (MAPKs) were activated, and the expression of early growth response 1 (Egr1) was subsequently increased in LPA-treated SV40 MES13 cells and the kidney cortex of db/db mice. Moreover, LPA significantly increased the activity of the Ras-related C3 botulinum toxin substrate (Rac1) GTPase in SV40 MES13 cells, and the dominant-negative form of Rac1 partially inhibited the phosphorylation of p38 and upregulation of Egr1 and KLF5 induced by LPA. LPA-induced hyperproliferation was attenuated by the inhibition of Rac1 activity. Based on these results, the Rac1/MAPK/KLF5 signaling pathway was one of the mechanisms by which LPA induced mesangial cell proliferation in DN models.
Collapse
|
18
|
Khamchun S, Thongboonkerd V. Cell cycle shift from G0/G1 to S and G2/M phases is responsible for increased adhesion of calcium oxalate crystals on repairing renal tubular cells at injured site. Cell Death Discov 2018; 4:106. [PMID: 30774989 PMCID: PMC6374384 DOI: 10.1038/s41420-018-0123-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/24/2018] [Accepted: 11/05/2018] [Indexed: 12/19/2022] Open
Abstract
Renal tubular cell injury can enhance calcium oxalate monohydrate (COM) crystal adhesion at the injured site and thus may increase the stone risk. Nevertheless, underlying mechanism of such enhancement remained unclear. In the present study, confluent MDCK renal tubular cell monolayers were scratched to allow cells to proliferate and repair the injured site. At 12-h post-scratch, the repairing cells had significant increases in crystal adhesion capacity and cell proliferation as compared to the control. Cell cycle analysis using flow cytometry demonstrated that the repairing cells underwent cell cycle shift from G0/G1 to S and G2/M phases. Cyclosporin A (CsA) and hydroxyurea (HU) at sub-toxic doses caused cell cycle shift mimicking that observed in the repairing cells. Crystal-cell adhesion assay confirmed the increased crystal adhesion capacity of the CsA-treated and HU-treated cells similar to that of the repairing cells. These findings provide evidence indicating that cell cycle shift from G0/G1 to S and G2/M phases is responsible, at least in part, for the increased adhesion of COM crystals on repairing renal tubular cells at the injured site.
Collapse
Affiliation(s)
- Supaporn Khamchun
- 1Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,2Department of Immunology and Immunology Graduate Program, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Visith Thongboonkerd
- 1Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,3Center for Research in Complex Systems Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
19
|
Kizhedathu A, Bagul AV, Guha A. Negative regulation of G2-M by ATR (mei-41)/Chk1(Grapes) facilitates tracheoblast growth and tracheal hypertrophy in Drosophila. eLife 2018; 7:29988. [PMID: 29658881 PMCID: PMC5953539 DOI: 10.7554/elife.29988] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 04/12/2018] [Indexed: 11/21/2022] Open
Abstract
Imaginal progenitors in Drosophila are known to arrest in G2 during larval stages and proliferate thereafter. Here we investigate the mechanism and implications of G2 arrest in progenitors of the adult thoracic tracheal epithelium (tracheoblasts). We report that tracheoblasts pause in G2 for ~48–56 h and grow in size over this period. Surprisingly, tracheoblasts arrested in G2 express drivers of G2-M like Cdc25/String (Stg). We find that mechanisms that prevent G2-M are also in place in this interval. Tracheoblasts activate Checkpoint Kinase 1/Grapes (Chk1/Grp) in an ATR/mei-41-dependent manner. Loss of ATR/Chk1 led to precocious mitotic entry ~24–32 h earlier. These divisions were apparently normal as there was no evidence of increased DNA damage or cell death. However, induction of precocious mitoses impaired growth of tracheoblasts and the tracheae they comprise. We propose that ATR/Chk1 negatively regulate G2-M in developing tracheoblasts and that G2 arrest facilitates cellular and hypertrophic organ growth. Every organism begins as a single cell. That cell, and all the other cells it generates over time, need to divide at the right time and in the right place to develop into an adult. As they do so, they pass through the stages of the cell cycle. As cells prepare to divide they enter into the first growth phase, G1, ramping up their metabolic activity. They then enter S phase, duplicate their DNA, and subsequently a second growth phase G2. Finally, during the mitotic phase, the chromosome separate and cells undergo cytokinesis to form new cells. Dividing cells can pause at certain stages of the cell cycle to assess whether the conditions are suitable to proceed. The length of the pause depends on the stage of development and the cell type. Signals around the cell provide the cues that it needs to make the decision. The fruit fly Drosophila melanogaster, for example, undergoes metamorphosis during development, meaning it transforms from a larva into an adult. The larva contains small patches of ‘progenitor’ cells that form the adult tissue. These remain paused for various intervals during larval life and restart their cell cycle as the animal develops. A key challenge in biology is to understand how these progenitors pause and what makes them start dividing again. Here, Kizhedathu, Bagul and Guha uncover a new mechanism that pauses the cell cycle in developing animal cells. Progenitors of the respiratory system in the adult fruit fly pause at the G2 stage of the cell cycle during larval life. Some of these progenitors, from a part of the larva called the dorsal trunk, go on to form the structures of the adult respiratory system. By counting the cells and tracking their dynamics with fluorescent labels, Kizhedathu et al. revealed that the progenitor cells pause for between 48 and to 56 hours. Previous research suggested that this pause happens because the cells lack a protein essential for mitosis called Cdc25/String. However, these progenitors were producing Cdc25/String. They stopped dividing because they also made another protein, known as Checkpoint Kinase 1/Grapes (Chk1/Grp). Chk1 is known to add a chemical modification to Cdc25, which dampens its activity and stops the cell cycle from progressing. This is likely what allow the flies to co-ordinate their development and give the cells more time to grow. When Chk1 was experimentally removed, it reactivated the paused cells sooner, resulting in smaller cells and a smaller respiratory organ. This work extends our understanding of stem cell dynamics and growth during development. Previous work has shown that cells that give rise to muscles and the neural tube (the precursor of the central nervous system) also pause their cell cycle in G2. Understanding more about how this happens could open new avenues for research into developmental disease.
Collapse
Affiliation(s)
- Amrutha Kizhedathu
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India.,SASTRA University, Thanjavur, India
| | - Archit V Bagul
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
| | - Arjun Guha
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
| |
Collapse
|
20
|
Yu YW, Li MX, Zhang ZY, Yu H. The deficiency of CX3CL1/CX3CR1 system ameliorates high fructose diet-induced kidney injury by regulating NF-κB pathways in CX3CR1-knock out mice. Int J Mol Med 2018; 41:3577-3585. [PMID: 29568873 PMCID: PMC5881704 DOI: 10.3892/ijmm.2018.3573] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 01/12/2018] [Indexed: 01/14/2023] Open
Abstract
Fructose, the most important functional food additive from the last century, has been widely used in industry, agriculture, light industry, food and medicine. With the improvement of people's living standard and economic level, excess intake of fructose results in metabolic symptoms, including hyperleptinemia, insulin resistance and neuroinflammation is causing high risk of chronic kidney disease development in humans and animals. However, the underlying molecular mechanism of renal injury is still not fully understood, and the development of effective drugs and treatments are delayed. Hence, we investigated the role of crosstalk of CX3CL1-CX3CR1 axis and nuclear factor-κB (NF-κB) signaling pathway in the development of renal injury. CX3CL1-knock-out C57BL/6 mice were constructed and used to analyze the influence of CX3CL1-related signaling pathways on kidney injury of wild-type (WT) mice and CXECR1 deficiency mice, which were administrated with 30% fructose water. Western blotting, quantitative RT-PCR (qRT-PCR), immunohistochemistry, ELISA, flow cytometry and biochemical indicator analysis were used to determine the levels of renal injury and key signaling pathway associated with renal damage. The results indicated that administration of high fructose intake can cause typical renal inflammatory responses in serum and tissues. Fructose enhances the CX3CL1-CX3CR1 axis and NF-κB activation, and promotes crosstalk of CX3CL1-CX3CR1 and NF-κB pathways. The phosphorylated AKT could be significantly activated in fructose-induced renal injury via CX3CL1-CX3CR1 axis. CX3CR1 expression between WT and CX3CR1−/− mice were evaluated to establish their relationship with injury. Our results indicated that CX3CR1 may be the central and major indicator in the process of renal injury, which mediate AKT pathway and further enhance the NF-κB activation. These findings demonstrated that crosstalk of CX3CL1-CX3CR1 axis and NF-κB signaling pathway play a direct role in fructose-induced kidney injury. Inhibition of CX3CL1-CX3CR1 pathway may suppress renal-related diseases. It may be a potential treatment choice for the clinical diagnoses and treatment in the future.
Collapse
Affiliation(s)
- Yong-Wu Yu
- Department of Nephrology, Navy General Hospital of Chinese PLA, Beijing 100048, P.R. China
| | - Ming-Xu Li
- Department of Nephrology, Navy General Hospital of Chinese PLA, Beijing 100048, P.R. China
| | - Zhi-Yong Zhang
- Department of Nephrology, Navy General Hospital of Chinese PLA, Beijing 100048, P.R. China
| | - Hai Yu
- Department of Nephrology, Navy General Hospital of Chinese PLA, Beijing 100048, P.R. China
| |
Collapse
|
21
|
Henique C, Bollée G, Loyer X, Grahammer F, Dhaun N, Camus M, Vernerey J, Guyonnet L, Gaillard F, Lazareth H, Meyer C, Bensaada I, Legrès L, Satoh T, Akira S, Bruneval P, Dimmeler S, Tedgui A, Karras A, Thervet E, Nochy D, Huber TB, Mesnard L, Lenoir O, Tharaux PL. Genetic and pharmacological inhibition of microRNA-92a maintains podocyte cell cycle quiescence and limits crescentic glomerulonephritis. Nat Commun 2017; 8:1829. [PMID: 29184126 PMCID: PMC5705755 DOI: 10.1038/s41467-017-01885-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 10/23/2017] [Indexed: 01/07/2023] Open
Abstract
Crescentic rapidly progressive glomerulonephritis (RPGN) represents the most aggressive form of acquired glomerular disease. While most therapeutic approaches involve potentially toxic immunosuppressive strategies, the pathophysiology remains incompletely understood. Podocytes are glomerular epithelial cells that are normally growth-arrested because of the expression of cyclin-dependent kinase (CDK) inhibitors. An exception is in RPGN where podocytes undergo a deregulation of their differentiated phenotype and proliferate. Here we demonstrate that microRNA-92a (miR-92a) is enriched in podocytes of patients and mice with RPGN. The CDK inhibitor p57Kip2 is a major target of miR-92a that constitutively safeguards podocyte cell cycle quiescence. Podocyte-specific deletion of miR-92a in mice de-repressed the expression of p57Kip2 and prevented glomerular injury in RPGN. Administration of an anti-miR-92a after disease initiation prevented albuminuria and kidney failure, indicating miR-92a inhibition as a potential therapeutic strategy for RPGN. We demonstrate that miRNA induction in epithelial cells can break glomerular tolerance to immune injury. Crescentic rapidly progressive glomerulonephritis is a severe form of glomerula disease characterized by podocyte proliferation and migration. Here Henique et al. demonstrate that inhibition of miRNA-92a prevents kidney failure by promoting the expression of CDK inhibitor p57Kip2 that regulates podocyte cell cycle.
Collapse
Affiliation(s)
- Carole Henique
- Paris Cardiovascular Research Centre-PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75015, France. .,Paris Descartes University, Sorbonne Paris Cité, Paris, 75006, France. .,Institut Mondor de Recherche Biomédicale, team 21, Unité Mixte de Recherche (UMR) 955, Institut National de la Santé et de la Recherche Médicale (INSERM), Créteil, 94000, France. .,Université Paris-Est Créteil, Créteil, 94000, France.
| | - Guillaume Bollée
- Paris Cardiovascular Research Centre-PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75015, France.,Paris Descartes University, Sorbonne Paris Cité, Paris, 75006, France.,Centre de Recherche, Centre Hospitalier de l'Université de Montréal, Montréal, H2X 0A9, QC, Canada
| | - Xavier Loyer
- Paris Cardiovascular Research Centre-PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75015, France.,Paris Descartes University, Sorbonne Paris Cité, Paris, 75006, France
| | - Florian Grahammer
- III. Medizinische Klinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, 20246, Germany.,Department of Medicine IV, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, P.O. Box 79085, Germany.,BIOSS Centre for Biological Signalling Studies and Center for Biological Systems Analysis (ZBSA), Albert-Ludwigs-University, Freiburg, 79104, Germany
| | - Neeraj Dhaun
- Paris Cardiovascular Research Centre-PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75015, France.,British Heart Foundation Centre of Research Excellence (BHF CoRE), Edinburgh, EH16 4TJ, UK
| | - Marine Camus
- Paris Cardiovascular Research Centre-PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75015, France
| | - Julien Vernerey
- Paris Cardiovascular Research Centre-PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75015, France
| | - Léa Guyonnet
- Paris Cardiovascular Research Centre-PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75015, France.,Paris Descartes University, Sorbonne Paris Cité, Paris, 75006, France
| | - François Gaillard
- Paris Cardiovascular Research Centre-PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75015, France.,Paris Descartes University, Sorbonne Paris Cité, Paris, 75006, France
| | - Hélène Lazareth
- Paris Cardiovascular Research Centre-PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75015, France.,Paris Descartes University, Sorbonne Paris Cité, Paris, 75006, France
| | - Charlotte Meyer
- BIOSS Centre for Biological Signalling Studies and Center for Biological Systems Analysis (ZBSA), Albert-Ludwigs-University, Freiburg, 79104, Germany
| | - Imane Bensaada
- Paris Cardiovascular Research Centre-PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75015, France.,Paris Descartes University, Sorbonne Paris Cité, Paris, 75006, France
| | - Luc Legrès
- Unité Mixte de Recherche (UMR_S) 1165, Institut National de la Santé et de la Recherche Médicale (INSERM), Plateforme MicroLaser Biotech, Paris, 75010, France
| | - Takashi Satoh
- Laboratory of Host Defense, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka, 565-0871, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka, 565-0871, Japan
| | - Patrick Bruneval
- Paris Descartes University, Sorbonne Paris Cité, Paris, 75006, France.,Department of Pathology, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, 75015, France.,Département Hospitalo-Universitaire, Paris Descartes University-Hôpitaux Universitaires Paris Ouest, Paris, 75015, France
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, 60590, Germany
| | - Alain Tedgui
- Paris Cardiovascular Research Centre-PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75015, France.,Paris Descartes University, Sorbonne Paris Cité, Paris, 75006, France
| | - Alexandre Karras
- Paris Cardiovascular Research Centre-PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75015, France.,Paris Descartes University, Sorbonne Paris Cité, Paris, 75006, France.,Département Hospitalo-Universitaire, Paris Descartes University-Hôpitaux Universitaires Paris Ouest, Paris, 75015, France.,Renal Division, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, 75015, France
| | - Eric Thervet
- Paris Cardiovascular Research Centre-PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75015, France.,Paris Descartes University, Sorbonne Paris Cité, Paris, 75006, France.,Département Hospitalo-Universitaire, Paris Descartes University-Hôpitaux Universitaires Paris Ouest, Paris, 75015, France.,Renal Division, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, 75015, France
| | - Dominique Nochy
- Paris Descartes University, Sorbonne Paris Cité, Paris, 75006, France.,Department of Pathology, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, 75015, France.,Département Hospitalo-Universitaire, Paris Descartes University-Hôpitaux Universitaires Paris Ouest, Paris, 75015, France
| | - Tobias B Huber
- III. Medizinische Klinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, 20246, Germany.,Department of Medicine IV, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, P.O. Box 79085, Germany.,BIOSS Centre for Biological Signalling Studies and Center for Biological Systems Analysis (ZBSA), Albert-Ludwigs-University, Freiburg, 79104, Germany
| | - Laurent Mesnard
- Unité Mixte de Recherche (UMR) 702, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75020, France.,Faculty of Medicine, University Pierre and Marie Curie, Paris, 75020, France
| | - Olivia Lenoir
- Paris Cardiovascular Research Centre-PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75015, France.,Paris Descartes University, Sorbonne Paris Cité, Paris, 75006, France
| | - Pierre-Louis Tharaux
- Paris Cardiovascular Research Centre-PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, 75015, France. .,Paris Descartes University, Sorbonne Paris Cité, Paris, 75006, France. .,Renal Division, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, 75015, France.
| |
Collapse
|
22
|
Bai J, Li Y, Zhang G. Cell cycle regulation and anticancer drug discovery. Cancer Biol Med 2017; 14:348-362. [PMID: 29372101 PMCID: PMC5785171 DOI: 10.20892/j.issn.2095-3941.2017.0033] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 10/13/2017] [Indexed: 02/05/2023] Open
Abstract
Cellular growth, development, and differentiation are tightly controlled by a conserved biological mechanism: the cell cycle. This cycle is primarily regulated by cyclin-dependent kinase (CDK)-cyclin complexes, checkpoint kinases, and CDK inhibitors. Deregulation of the cell cycle is a hallmark of the transformation of normal cells into tumor cells. Given its importance in tumorigenesis, several cell cycle inhibitors have emerged as potential therapeutic drugs for the treatment of cancers-both as single-agent therapy and in combination with traditional cytotoxic or molecular targeting agents. In this review, we discuss the mechanisms underlying cell cycle regulation and present small-molecule anticancer drugs that are under development, including both pan-CDK inhibitors and CDK4/6-selective inhibitors. In addition, we provide an outline of some promising CDK inhibitors currently in preclinical and clinical trials that target cell cycle abnormalities in various cancers.
Collapse
Affiliation(s)
- Jingwen Bai
- Department of Oncology, Xiang’an Hospital of Xiamen University, Xiamen 361005, China
- Changjiang Scholar’s Laboratory of Shantou University Medical College (SUMC), Shantou 515041, China
| | - Yaochen Li
- Department of Oncology, Xiang’an Hospital of Xiamen University, Xiamen 361005, China
| | - Guojun Zhang
- Department of Oncology, Xiang’an Hospital of Xiamen University, Xiamen 361005, China
- Changjiang Scholar’s Laboratory of Shantou University Medical College (SUMC), Shantou 515041, China
| |
Collapse
|
23
|
Sheu ML, Shen CC, Jheng JR, Chiang CK. Activation of PI3K in response to high glucose leads to regulation of SOCS-3 and STAT1/3 signals and induction of glomerular mesangial extracellular matrix formation. Oncotarget 2017; 8:16925-16938. [PMID: 28129651 PMCID: PMC5370011 DOI: 10.18632/oncotarget.14808] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 12/13/2016] [Indexed: 01/27/2023] Open
Abstract
Excessive deposition of extracellular matrix (ECM) in the glomerulus contributed by mesangial cells is the hallmark of diabetic nephropathy, eventually leading to glomerulosclerosis. In this study, we examined the regulatory signals involved in the high glucose (HG)-induced overproduction of ECM in rat mesangial cells (RMCs). We disclosed excessive fibronectin and collagen IV production, tyrosine phosphorylation of signal transducer and activator of transcription 1 and 3 (STAT1/3), and up-regulation of suppressor of cytokine signaling-3 (SOCS-3) expression in HG-treated RMCs. STAT1/STAT3 binding element was essential for SOCS-3 promoter activity stimulated by HG. HG was capable of promoting the specific DNA binding activities to an oligonucleotide probe containing the SOCS-3 sequence. The selective phosphoinositide 3-kinase (PI3K) inhibitor LY294002 and dominant negative p85 vector (DNΔp85) transfection effectively abolished these HG-induced responses. Moreover, HG markedly increased the cyclin kinase inhibitor p27Kip1 protein expression, which could be inhibited by LY294002 or transfection of DNΔp85. Taken together, these results suggest that HG-induced SOCS-3 upregulation depends upon the presence of STAT-binding element in the SOCS-3 promoter, which is specifically activated by STAT1/3. The PI3K/STAT1/3 signaling pathway mediated HG-triggered ECM accumulation and SOCS-3 upregulation in RMCs.
Collapse
Affiliation(s)
- Meei-Ling Sheu
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.,Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.,Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chin-Chang Shen
- Chemical Engineering Division, Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Jia-Rong Jheng
- Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chih-Kang Chiang
- Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Integrated Diagnostics & Therapeutics, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
24
|
Thomasova D, Ebrahim M, Fleckinger K, Li M, Molnar J, Popper B, Liapis H, Kotb AM, Siegerist F, Endlich N, Anders HJ. MDM2 prevents spontaneous tubular epithelial cell death and acute kidney injury. Cell Death Dis 2016; 7:e2482. [PMID: 27882940 PMCID: PMC5260907 DOI: 10.1038/cddis.2016.390] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/21/2016] [Accepted: 10/25/2016] [Indexed: 12/24/2022]
Abstract
Murine double minute-2 (MDM2) is an E3-ubiquitin ligase and the main negative regulator of tumor suppressor gene p53. MDM2 has also a non-redundant function as a modulator of NF-kB signaling. As such it promotes proliferation and inflammation. MDM2 is highly expressed in the unchallenged tubular epithelial cells and we hypothesized that MDM2 is necessary for their survival and homeostasis. MDM2 knockdown by siRNA or by genetic depletion resulted in demise of tubular cells in vitro. This phenotype was completely rescued by concomitant knockdown of p53, thus suggesting p53 dependency. In vivo experiments in the zebrafish model demonstrated that the tubulus cells of the larvae undergo cell death after the knockdown of mdm2. Doxycycline-induced deletion of MDM2 in tubular cell-specific MDM2-knockout mice Pax8rtTa-cre; MDM2f/f caused acute kidney injury with increased plasma creatinine and blood urea nitrogen and sharp decline of glomerular filtration rate. Histological analysis showed massive swelling of renal tubular cells and later their loss and extensive tubular dilation, markedly in proximal tubules. Ultrastructural changes of tubular epithelial cells included swelling of the cytoplasm and mitochondria with the loss of cristae and their transformation in the vacuoles. The pathological phenotype of the tubular cell-specific MDM2-knockout mouse model was completely rescued by co-deletion of p53. Tubular epithelium compensates only partially for the cell loss caused by MDM2 depletion by proliferation of surviving tubular cells, with incomplete MDM2 deletion, but rather mesenchymal healing occurs. We conclude that MDM2 is a non-redundant survival factor for proximal tubular cells by protecting them from spontaneous p53 overexpression-related cell death.
Collapse
Affiliation(s)
- Dana Thomasova
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der LMU München, Munich, Germany
| | - Martrez Ebrahim
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der LMU München, Munich, Germany
| | - Kristina Fleckinger
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der LMU München, Munich, Germany
| | - Moying Li
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der LMU München, Munich, Germany
| | - Jakob Molnar
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der LMU München, Munich, Germany
| | - Bastian Popper
- Department of Anatomy and Cell Biology, Ludwig-Maximilians Universität, Munich, Germany
| | - Helen Liapis
- Pathology & Immunology & Internal Medicine (Renal), Washington University, School of Medicine, St Louis, MO, USA
| | - Ahmed M Kotb
- Department of Anatomy and Cell Biology, Universitätsmedizin Greifswald, Greifswald, Germany
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Florian Siegerist
- Department of Anatomy and Cell Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Hans-Joachim Anders
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der LMU München, Munich, Germany
| |
Collapse
|
25
|
Histone Lysine Methylation in TGF-β1 Mediated p21 Gene Expression in Rat Mesangial Cells. BIOMED RESEARCH INTERNATIONAL 2016; 2016:6927234. [PMID: 27247942 PMCID: PMC4876202 DOI: 10.1155/2016/6927234] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 04/11/2016] [Indexed: 01/11/2023]
Abstract
Transforming growth factor beta1- (TGF-β1-) induced p21-dependent mesangial cell (MC) hypertrophy plays a key role in the pathogenesis of chronic renal diseases including diabetic nephropathy (DN). Increasing evidence demonstrated the role of posttranscriptional modifications (PTMs) in the event; however, the precise regulatory mechanism of histone lysine methylation remains largely unknown. Here, we examined the roles of both histone H3 lysine 4 and lysine 9 methylations (H3K4me/H3K9me) in TGF-β1 induced p21 gene expression in rat mesangial cells (RMCs). Our results indicated that TGF-β1 upregulated the expression of p21 gene in RMCs, which was positively correlated with the increased chromatin marks associated with active genes (H3K4me1/H3K4me2/H3K4me3) and negatively correlated with the decreased levels of repressive marks (H3K9me2/H3K9me3) at p21 gene promoter. TGF-β1 also elevated the recruitment of the H3K4 methyltransferase (HMT) SET7/9 to the p21 gene promoter. SET7/9 gene silencing with small interfering RNAs (siRNAs) significantly abolished the TGF-β1 induced p21 gene expression. Taken together, these results reveal the key role of histone H3Kme in TGF-β1 mediated p21 gene expression in RMC, partly through HMT SET7/9 occupancy, suggesting H3Kme and SET7/9 may be potential renoprotective agents in managing chronic renal diseases.
Collapse
|
26
|
Lu Y, Wen J, Chen D, Wu L, Li Q, Xie Y, Wu D, Liu X, Chen X. Modulation of cyclins and p53 in mesangial cell proliferation and apoptosis during Habu nephritis. Clin Exp Nephrol 2016; 20:178-86. [PMID: 26359229 PMCID: PMC4819602 DOI: 10.1007/s10157-015-1163-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 08/30/2015] [Indexed: 11/08/2022]
Abstract
BACKGROUND Mesangial cell (MC) proliferation and apoptosis are the main pathological changes observed in mesangial proliferative nephritis. In this study, we explored the role of cyclins and p53 in modulating MC proliferation and apoptosis in a mouse model of Habu nephritis. METHODS The Habu nephritis group was prepared by injection of Habu toxin. Mesangiolysis and mesangial expansion were determined by periodic acid-Schiff (PAS) reagent staining. Immunohistochemical analysis of PCNA and KI67, and TUNEL staining were used to detect cell proliferation and apoptosis, respectively. Expression levels of cyclins and p53 were examined by Western blotting. RESULTS PAS staining showed that mesangial dissolution appeared on days 1 and 3, and mesangial proliferation with extracellular matrix accumulation was apparent by days 7 and 14. Both PCNA and KI67 immunohistochemical analysis showed that MC proliferation began on day 3, peaked on day 3 and 7, and recovered by day 14. TUNEL staining results showed that MC apoptosis began to increase on day 1, continued to rise on day 7, and peaked on day 14. Western blot analysis showed that cyclin D1 was upregulated on day 1, cyclins A2 and E were upregulated on days 3 and 7, and p53 was upregulated on days 3, 7 and 14. There was no change in the expression levels of Bax or p21. CONCLUSION We explored the tendency for MC proliferation and apoptosis during the process of Habu nephritis and found that cyclins and p53 may modulate the disease pathology. This will help us determine the molecular pathogenesis of MC proliferation and provide new targets for disease intervention.
Collapse
Affiliation(s)
- Yang Lu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing, 100853, People's Republic of China
| | - Jun Wen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing, 100853, People's Republic of China
| | - DaPeng Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing, 100853, People's Republic of China
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - LingLing Wu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing, 100853, People's Republic of China
| | - QingGang Li
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing, 100853, People's Republic of China
| | - Yuansheng Xie
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing, 100853, People's Republic of China
| | - Di Wu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing, 100853, People's Republic of China
| | - Xiaoluan Liu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing, 100853, People's Republic of China
| | - XiangMei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing, 100853, People's Republic of China.
| |
Collapse
|
27
|
Li X, Li C, Li X, Cui P, Li Q, Guo Q, Han H, Liu S, Sun G. Involvement of Histone Lysine Methylation in p21 Gene Expression in Rat Kidney In Vivo and Rat Mesangial Cells In Vitro under Diabetic Conditions. J Diabetes Res 2016; 2016:3853242. [PMID: 27652271 PMCID: PMC5019898 DOI: 10.1155/2016/3853242] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 07/27/2016] [Indexed: 01/21/2023] Open
Abstract
Diabetic nephropathy (DN), a common complication associated with type 1 and type 2 diabetes mellitus (DM), characterized by glomerular mesangial expansion, inflammation, accumulation of extracellular matrix (ECM) protein, and hypertrophy, is the major cause of end-stage renal disease (ESRD). Increasing evidence suggested that p21-dependent glomerular and mesangial cell (MC) hypertrophy play key roles in the pathogenesis of DN. Recently, posttranscriptional modifications (PTMs) have uncovered novel molecular mechanisms involved in DN. However, precise regulatory mechanism of histone lysine methylation (HKme) mediating p21 related hypertrophy associated with DN is not clear. We evaluated the roles of HKme and histone methyltransferase (HMT) SET7/9 in p21 gene expression in glomeruli of diabetic rats and in high glucose- (HG-) treated rat mesangial cells (RMCs). p21 gene expression was upregulated in diabetic rats glomeruli; chromatin immunoprecipitation (ChIP) assays showed decreased histone H3-lysine9-dimethylation (H3K9me2) accompanied with enhanced histone H3-lysine4-methylation (H3K4me1/3) and SET7/9 occupancies at the p21 promoter. HG-treated RMCs exhibited increased p21 mRNA, H3K4me level, SET7/9 recruitment, and inverse H3K9me, which were reversed by TGF-β1 antibody. These data uncovered key roles of H3Kme and SET7/9 responsible for p21 gene expression in vivo and in vitro under diabetic conditions and confirmed preventive effect of TGF-β1 antibody on DN.
Collapse
Affiliation(s)
- Xiangjun Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Science, Jilin University, Changchun, Jilin 130021, China
| | - Chaoyuan Li
- Department of Nephrology, 2nd Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Xiaoxia Li
- Department of Nephrology, 2nd Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Peihe Cui
- Department of Nephrology, 2nd Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Qifeng Li
- Department of Nephrology, 2nd Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Qiaoyan Guo
- Department of Nephrology, 2nd Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Hongbo Han
- Department of Endocrinology, 208th Hospital of Chinese PLA, Changchun, Jilin 130062, China
| | - Shujun Liu
- Department of Nephrology, 2nd Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Guangdong Sun
- Department of Nephrology, 2nd Hospital of Jilin University, Changchun, Jilin 130041, China
- *Guangdong Sun:
| |
Collapse
|
28
|
Yang H, Yin P, Shi Z, Ma Y, Zhao C, Zheng J, Chen T. Sinomenine, a COX-2 inhibitor, induces cell cycle arrest and inhibits growth of human colon carcinoma cells in vitro and in vivo. Oncol Lett 2015; 11:411-418. [PMID: 26870226 DOI: 10.3892/ol.2015.3838] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 09/22/2015] [Indexed: 11/06/2022] Open
Abstract
Certain non-steroidal anti-inflammatory drugs may possess anti-tumorigenic effects in certain cancer cell types. Sinomenine (SIN) is an alkaloid from Sinomenium acutum, a Chinese medicinal plant that inhibits inflammatory reactions and that has been used in the treatment of neuralgia and rheumatic diseases. In this study, we investigated the anticancer effects of SIN against colorectal cancer in vitro and in vivo, as well as the underlying mechanisms. The effects of SIN on proliferation, cell cycle progression and cyclooxygenase (COX)-2 expression were examined in human colorectal cancer-derived SW1116 cells. The in vivo effects of SIN were examined in a model of SW1116 tumor xenograft growth in athymic nude mice. Changes in COX-2 expression induced by the biological effects of SIN were analyzed by western blot analysis. The effects of SIN treatment on G1 phase cell cycle regulators in xenografts were analyzed by immunohistochemistry. Our findings demonstrate that SIN inhibits the proliferation of SW1116 cells by promoting their accumulation in the G1 phase, with concomitant suppression of COX-2 expression. Time- and dose-dependent inhibition of tumor growth and reduced toxicity were observed in nude mice administered daily intraperitoneal injections of SIN at doses of 25, 50 and 100 mg/kg. SIN-treated tumors also exhibited reduced COX-2 expression, a marked increase in Cip1/p21 protein levels and a decrease in the levels of cyclin D1 and cyclin E. SIN may be an effective chemopreventive agent against colorectal cancer. The growth inhibitory properties of SIN against colorectal cancer may be mediated via a COX-2 inhibitory effect and cell cycle arrest in the G1 phase.
Collapse
Affiliation(s)
- Haibo Yang
- Department of Colorectal Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Peihao Yin
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Zhan Shi
- Department of Colorectal Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Yanchun Ma
- Experimental Center, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Chenggen Zhao
- Department of Oncology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Jampu Zheng
- Experimental Center, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Teng Chen
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| |
Collapse
|
29
|
Thomasova D, Anders HJ. Cell cycle control in the kidney. Nephrol Dial Transplant 2014; 30:1622-30. [PMID: 25538161 DOI: 10.1093/ndt/gfu395] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 11/27/2014] [Indexed: 01/01/2023] Open
Abstract
Proper control of the cell cycle is mandatory during homeostasis and disease. The balance of p53 and MDM2 integrates numerous signalling pathways to regulate the cell cycle, which is executed by multiple proteins including the cyclins, cyclin kinases and cyclin kinase inhibitors. Mutations or environmental factors that affect cell cycle control can lead to inappropriate hyperplasia or cancer as well as to cell loss and tissue atrophy. Normal kidney function is maintained largely by post-mitotic quiescent cells in the G0 phase with a low turnover. Early cell cycle activation during kidney injury contributes to cell death via mitotic catastrophe, i.e. death via mitosis, e.g. of cell with significant DNA damage. At later stages, cell cycle entry supports tissue regeneration and functional reconstitution via cell hypertrophy and/or cell proliferation. It is of note that so-called proliferation markers such as Ki67, PCNA or BrdU identify only cell cycle entry without telling whether this results in cell hypertrophy, cell division or mitotic catastrophe. With this in mind, some established concepts on kidney injury and regeneration are to be re-evaluated. Here, we discuss the components and functional roles of p53/MDM2-mediated cell cycle regulation in kidney homeostasis and disease.
Collapse
Affiliation(s)
- Dana Thomasova
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Munich, Germany
| | - Hans-Joachim Anders
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Munich, Germany
| |
Collapse
|
30
|
Tian J, Wang Y, Liu X, Zhou X, Li R. Rapamycin ameliorates IgA nephropathy via cell cycle-dependent mechanisms. Exp Biol Med (Maywood) 2014; 240:936-45. [PMID: 25349217 DOI: 10.1177/1535370214555666] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 09/07/2014] [Indexed: 12/14/2022] Open
Abstract
IgA nephropathy is the most frequent type of glomerulonephritis worldwide. The role of cell cycle regulation in the pathogenesis of IgA nephropathy has been studied. The present study was designed to explore whether rapamycin ameliorates IgA nephropathy via cell cycle-dependent mechanisms. After establishing an IgA nephropathy model, rats were randomly divided into four groups. Coomassie Brilliant Blue was used to measure the 24-h urinary protein levels. Renal function was determined using an autoanalyzer. Proliferation was assayed via Proliferating Cell Nuclear Antigen (PCNA) immunohistochemistry. Rat mesangial cells were cultured and divided into the six groups. Methylthiazolyldiphenyl-tetrazolium bromide (MTT) and flow cytometry were used to detect cell proliferation and the cell cycle phase. Western blotting was performed to determine cyclin E, cyclin-dependent kinase 2, p27(Kip1), p70S6K/p-p70S6K, and extracellular signal-regulated kinase 1/2/p- extracellular signal-regulated kinase 1/2 protein expression. A low dose of the mammalian target of rapamycin (mTOR) inhibitor rapamycin prevented an additional increase in proteinuria, protected kidney function, and reduced IgA deposition in a model of IgA nephropathy. Rapamycin inhibited mesangial cell proliferation and arrested the cell cycle in the G1 phase. Rapamycin did not affect the expression of cyclin E and cyclin-dependent kinase 2. However, rapamycin upregulated p27(Kip1) at least in part via AKT (also known as protein kinase B)/mTOR. In conclusion, rapamycin can affect cell cycle regulation to inhibit mesangial cell proliferation, thereby reduce IgA deposition, and slow the progression of IgAN.
Collapse
Affiliation(s)
- Jihua Tian
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, 030012, China Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Yanhong Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xinyan Liu
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, 030012, China
| | - Xiaoshuang Zhou
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, 030012, China
| | - Rongshan Li
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, 030012, China
| |
Collapse
|
31
|
Dvela-Levitt M, Cohen-Ben Ami H, Rosen H, Ornoy A, Hochner-Celnikier D, Granat M, Lichtstein D. Reduction in maternal circulating ouabain impairs offspring growth and kidney development. J Am Soc Nephrol 2014; 26:1103-14. [PMID: 25294233 DOI: 10.1681/asn.2014020130] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 08/13/2014] [Indexed: 12/20/2022] Open
Abstract
Ouabain, a steroid present in the circulation and in various tissues, was shown to affect the growth and viability of various cells in culture. To test for the possible influence of this steroid on growth and viability in vivo, we investigated the involvement of maternal circulating ouabain in the regulation of fetal growth and organ development. We show that intraperitoneal administration of anti-ouabain antibodies to pregnant mice resulted in a >80% decline in the circulating ouabain level. This reduction caused a significant decrease in offspring body weight, accompanied by enlargement of the offspring heart and inhibition of kidney and liver growth. Kidney growth inhibition was manifested by a decrease in the size and number of nephrons. After the reduction in maternal circulating ouabain, kidney expression of cyclin D1 was reduced and the expression of the α1 isoform of the Na(+), K(+)-ATPase was increased. In addition, the elevation of proliferation signals including ERK1/2, p-90RSK, Akt, PCNA, and Ki-67, and a reduction in apoptotic factors such as Bax, caspase-3, and TUNEL were detected. During human pregnancy, the circulating maternal ouabain level increased and the highest concentration of the steroid was found in the placenta. Furthermore, circulating ouabain levels in women with small-for-gestational age neonates were significantly lower than the levels in women with normal-for-gestational age newborns. These results support the notion that ouabain is a growth factor and suggest that a reduction in the concentration of this hormone during pregnancy may increase the risk of impaired growth and kidney development.
Collapse
Affiliation(s)
- Moran Dvela-Levitt
- Departments of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Hagit Cohen-Ben Ami
- Departments of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Haim Rosen
- Microbiology and Molecular Genetics, and Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Asher Ornoy
- Departments of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | | | | - David Lichtstein
- Departments of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
32
|
Mello CP, Morais IC, Menezes RR, Pereira GJ, Torres AF, Lima DB, Pereira TP, Toyama MH, Monteiro HS, Smaili SS, Martins AM. Bothropoides insularis venom cytotoxicity in renal tubular epithelia cells. Toxicon 2014; 88:107-14. [DOI: 10.1016/j.toxicon.2014.05.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 04/02/2014] [Accepted: 05/06/2014] [Indexed: 02/02/2023]
|
33
|
PARK SUNGSOO, KIM WUNJAE, MOON SUNGKWON. Gleditsia sinensis thorn extract inhibits the proliferation and migration of PDGF-induced vascular smooth muscle cells. Mol Med Rep 2014; 10:2031-8. [DOI: 10.3892/mmr.2014.2422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 03/19/2014] [Indexed: 11/06/2022] Open
|
34
|
Wang L, Zhang Y, Yuan L, Liu C, Fu L, Mei C. Cyclin-dependent kinase inhibitor p18INK4c is involved in protective roles of heme oxygenase-1 in cisplatin-induced acute kidney injury. Int J Mol Med 2014; 34:911-7. [PMID: 24993528 DOI: 10.3892/ijmm.2014.1828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 06/26/2014] [Indexed: 11/05/2022] Open
Abstract
Experimental studies have demonstrated the protective effect of heme oxygenase (HO)-1 and cyclin‑dependent kinase inhibitors (CDKIs) in acute kidney injury (AKI), and it has been documented that some of the protective effect of HO-1 is mediated by CDKIs. However, the role of p18INK4c (p18), an inhibitor of CDK4 (INK4), which is a family member of CDKIs, has not been well characterized in kidney diseases. The aim of the present study was to demonstrate p18 protection from the relationship between p18 and HO-1 in cisplatin-induced AKI. Upregulation of p18 and HO-1 was demonstrated by quantitative polymerase chain reaction (qPCR) and western blotting in cisplatin-induced AKI in vitro and in vivo. The effect of HO-1 on p18 was determined by western blotting using the inducer and inhibitor of HO-1 in vitro. The potential effect of p18 on HO-1 in cisplatin‑induced AKI was examined by p18 gene knockout mice in vivo. The results showed that p18 and HO-1 were upregulated in cisplatin‑induced AKI in vitro and in vivo. Deletion of the p18 gene did not affect the basal and inducible expression of HO-1 in the AKI animals, while hemin (10 µM) and znpp (10 µM), the inducer and inhibitor, respectively, of HO-1, regulated p18 expression when incubated with the cells. The results indicated that p18 may play protective roles and may be associated with or partially account for the cytoprotective effects of HO-1 in cisplatin-induced AKI.
Collapse
Affiliation(s)
- Liang Wang
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116021, P.R. China
| | - Yi Zhang
- Department of Nephrology, Changzheng Hospital, Shanghai 200003, P.R. China
| | - Li Yuan
- Department of Nephrology, Changzheng Hospital, Shanghai 200003, P.R. China
| | - Chunyan Liu
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116021, P.R. China
| | - Lili Fu
- Department of Nephrology, Changzheng Hospital, Shanghai 200003, P.R. China
| | - Changlin Mei
- Department of Nephrology, Changzheng Hospital, Shanghai 200003, P.R. China
| |
Collapse
|
35
|
The progression of the tubulointerstitial fibrosis driven by stress-induced “proliferation–death” vicious circle. Med Hypotheses 2014; 82:643-7. [DOI: 10.1016/j.mehy.2014.01.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 01/05/2014] [Accepted: 01/13/2014] [Indexed: 12/26/2022]
|
36
|
Wang L, Chen W, Zhang Y, Liu C, Yuan L, Fu L, Mei C. Deletion of p18(INK4c) aggravates cisplatin-induced acute kidney injury. Int J Mol Med 2014; 33:1621-6. [PMID: 24714825 DOI: 10.3892/ijmm.2014.1725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 03/27/2014] [Indexed: 11/05/2022] Open
Abstract
Protection of cyclin-dependent kinase inhibitors (CDKIs) has been demonstrated in acute kidney injury (AKI). However, previous studies on CDKIs have mainly focused on CIP/KIP family members, with INK4 family members rarely being investigated. This study investigated the behaviors of p18(INK4c) (p18) in cisplatin-induced AKI using p18 gene knockout mice (p18-/-). AKI was induced in p18-/- and wild-type (p18+/+) mice after a single cisplatin (12.5 mg/kg) intraperitoneal injection. Protection by p18 was identified by a comparison of survival, renal function and morphological injuries between p18-/- and p18+/+ mice. Further investigation of endoplasmic reticulum stress (ERS) was performed by western blot analysis in p18-/- and p18+/+ kidneys at day 3 after cisplatin injection. The results revealed that after cisplatin injection, the survival of p18-/- mice was significantly shorter than that of p18+/+ mice, accompanied by aggravated renal function and more severe morphological injuries. Deletion of p18 also significantly aggravated ERS in cisplatin-induced AKI. In conclusion, p18 exerts protective effects on cisplatin‑induced AKI, which may be associated with the effect of p18 on cell death pathways, such as the ERS pathway.
Collapse
Affiliation(s)
- Liang Wang
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116021, P.R. China
| | - Wei Chen
- Department of Nephrology, Changzheng Hospital, Shanghai 200003, P.R. China
| | - Yi Zhang
- Department of Nephrology, Changzheng Hospital, Shanghai 200003, P.R. China
| | - Chunyan Liu
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116021, P.R. China
| | - Li Yuan
- Department of Nephrology, Changzheng Hospital, Shanghai 200003, P.R. China
| | - Lili Fu
- Department of Nephrology, Changzheng Hospital, Shanghai 200003, P.R. China
| | - Changlin Mei
- Department of Nephrology, Changzheng Hospital, Shanghai 200003, P.R. China
| |
Collapse
|
37
|
Abstract
Diabetes mellitus contributes greatly to morbidity, mortality, and overall health care costs. In major part, these outcomes derive from the high incidence of progressive kidney dysfunction in patients with diabetes making diabetic nephropathy a leading cause of end-stage renal disease. A better understanding of the molecular mechanism involved and of the early dysfunctions observed in the diabetic kidney may permit the development of new strategies to prevent diabetic nephropathy. Here we review the pathophysiological changes that occur in the kidney in response to hyperglycemia, including the cellular responses to high glucose and the responses in vascular, glomerular, podocyte, and tubular function. The molecular basis, characteristics, and consequences of the unique growth phenotypes observed in the diabetic kidney, including glomerular structures and tubular segments, are outlined. We delineate mechanisms of early diabetic glomerular hyperfiltration including primary vascular events as well as the primary role of tubular growth, hyperreabsorption, and tubuloglomerular communication as part of a "tubulocentric" concept of early diabetic kidney function. The latter also explains the "salt paradox" of the early diabetic kidney, that is, a unique and inverse relationship between glomerular filtration rate and dietary salt intake. The mechanisms and consequences of the intrarenal activation of the renin-angiotensin system and of diabetes-induced tubular glycogen accumulation are discussed. Moreover, we aim to link the changes that occur early in the diabetic kidney including the growth phenotype, oxidative stress, hypoxia, and formation of advanced glycation end products to mechanisms involved in progressive kidney disease.
Collapse
Affiliation(s)
- Volker Vallon
- Department of Medicine, University of California San Diego & VA San Diego Healthcare System, San Diego, California, USA.
| | | |
Collapse
|
38
|
Qufeng Tongluo Prescription () inhibits mesangial cell proliferation and promotes apoptosis through regulating cell cycle progression. Chin J Integr Med 2013; 19:927-34. [PMID: 24307313 DOI: 10.1007/s11655-013-1655-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To study the effects and possible underlying mechanism of Qufeng Tongluo Prescription (, QFTL) on the regulation of mesangial cells (MCs) proliferation and apoptosis. METHODS The MCs used in this experiment have undergone five passages induced by lipopolysaccharide (LPS). Changes in the proliferation, apoptosis, cell cycle regulatory proteins and mRNA expression levels of the MCs after administration of Benazepril or QFTL were measured by methyl thiazolyl tetrazolium (MTT) reduction assay, flow cytometry, Western blot and quantitative real-time polymerase chain reaction (qRT-PCR), respectively. RESULTS The addition of Benazepril or QFTL serum inhibited LPS-induced MC proliferation after treatment for 24, 48 and 72 h, respectively (P<0.05 or P<0.01). Moreover, the inhibitory effect is more significant in the QFTL group at 48 h (P<0.05). Compared with the control group, LPS-induced cell proliferation decreased the number of cells in G1 phase versus cells in S and G2/M phases, while the addition of QFTL and Benazepril serum increased the ratio of cells at G1 phase (P<0.05 or P<0.01) to cells at S phase (P<0.01), implicating the cell cycle inhibition effect exerted by QFTL. LPS decreased the level of MC apoptosis, compared with the control group (P<0.05), while QFTL and Benazepril serum increased the level of MC apoptosis (P<0.01). Moreover, the difference between the QFTL group and the Benazepril group was statistically significant (P<0.01). Compared with the control group, the protein and mRNA expression levels of cylinD1, cyclin dependent kinase 2 (CDK2) and p21 were significantly increased (P<0.05 or P<0.01), p27 was decreased but with no statistical significance (P>0.05); After being treated with QFTL and Benazepril serum, the protein and mRNA expression levels of cylinD1, CDK2, p21 were decreased and p27 increased significantly (P<0.05 or P<0.01); Compared with the Benazepril group, QFTL show better effects on protein and mRNA expression levels of cylinD1, CDK2 (P<0.05 or P<0.01) and p21 protein expression (P<0.05). CONCLUSION QFTL inhibits MCs proliferation, promotes MCs apoptosis through an underlying mechanism of down-regulating the protein and mRNA expression levels of cylinD1, CDK2, p21 and up-regulation of the expression level of p27.
Collapse
|
39
|
Activation of ERK accelerates repair of renal tubular epithelial cells, whereas it inhibits progression of fibrosis following ischemia/reperfusion injury. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1998-2008. [DOI: 10.1016/j.bbadis.2013.07.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 06/12/2013] [Accepted: 07/01/2013] [Indexed: 11/18/2022]
|
40
|
ZOU LIPING, WANG WENJUAN, XU ZUDE, ZHANG NONG, JIANG TAO. Aldose reductase regulates platelet-derived growth factor-induced proliferation through mediating cell cycle progression in rat mesangial cells. Int J Mol Med 2012; 30:409-16. [DOI: 10.3892/ijmm.2012.997] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Accepted: 04/02/2012] [Indexed: 11/06/2022] Open
|
41
|
Wang SH, Liang CJ, Weng YW, Chen YH, Hsu HY, Chien HF, Tsai JS, Tseng YC, Li CY, Chen YL. Ganoderma lucidum polysaccharides prevent platelet-derived growth factor-stimulated smooth muscle cell proliferation in vitro and neointimal hyperplasia in the endothelial-denuded artery in vivo. J Cell Physiol 2012; 227:3063-71. [DOI: 10.1002/jcp.23053] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
42
|
Reversible cell-cycle entry in adult kidney podocytes through regulated control of telomerase and Wnt signaling. Nat Med 2011; 18:111-9. [PMID: 22138751 PMCID: PMC3272332 DOI: 10.1038/nm.2550] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 10/09/2011] [Indexed: 01/23/2023]
Abstract
Mechanisms of epithelial cell renewal remain poorly understood in the mammalian kidney, particularly in the glomerulus, a site of cellular damage in chronic kidney disease. Within the glomerulus, podocytes--differentiated epithelial cells crucial for filtration--are thought to lack substantial capacity for regeneration. Here we show that podocytes rapidly lose differentiation markers and enter the cell cycle in adult mice in which the telomerase protein component TERT is conditionally expressed. Transgenic TERT expression in mice induces marked upregulation of Wnt signaling and disrupts glomerular structure, resulting in a collapsing glomerulopathy resembling those in human disease, including HIV-associated nephropathy (HIVAN). Human and mouse HIVAN kidneys show increased expression of TERT and activation of Wnt signaling, indicating that these are general features of collapsing glomerulopathies. Silencing transgenic TERT expression or inhibiting Wnt signaling through systemic expression of the Wnt inhibitor Dkk1 in either TERT transgenic mice or in a mouse model of HIVAN results in marked normalization of podocytes, including rapid cell-cycle exit, re-expression of differentiation markers and improved filtration barrier function. These data reveal an unexpected capacity of podocytes to reversibly enter the cell cycle, suggest that podocyte renewal may contribute to glomerular homeostasis and implicate the telomerase and Wnt-β-catenin pathways in podocyte proliferation and disease.
Collapse
|
43
|
Abstract
As an integral member of the filtration barrier in the kidney glomerulus, the podocyte is in a unique geographical position: It is exposed to chemical signals from the urinary space (Bowman's capsule), it receives and transmits chemical and mechanical signals to/from the glomerular basement membrane upon which it elaborates, and it receives chemical and mechanical signals from the vascular space with which it also communicates. As with every cell, the ability of the podocyte to receive signals from the surrounding environment and to translate them to the intracellular milieu is dependent largely on molecules residing on the cell membrane. These molecules are the first-line soldiers in the ongoing battle to sense the environment, to respond to friendly signals, and to defend against injurious foes. In this review, we take a membrane biologist's view of the podocyte, examining the many membrane receptors, channels, and other signaling molecules that have been implicated in podocyte biology. Although we attempt to be comprehensive, our goal is not to capture every membrane-mediated pathway but rather to emphasize that this approach may be fruitful in understanding the podocyte and its unique properties.
Collapse
Affiliation(s)
- Anna Greka
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA.
| | | |
Collapse
|
44
|
Zhang Y, Yuan L, Fu L, Liu C, Liu D, Mei C. Overexpression of p18INK⁴C in LLC-PK1 cells increases resistance to cisplatin-induced apoptosis. Pediatr Nephrol 2011; 26:1291-301. [PMID: 21494915 DOI: 10.1007/s00467-011-1877-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 03/10/2011] [Accepted: 03/17/2011] [Indexed: 01/25/2023]
Abstract
Studies have demonstrated that cyclin-dependent kinase inhibitors (CDKI) that inhibit cell-cycle progression have a protective effect against acute kidney injury (AKI). Most studies have focused on the CIP/KIP family members of CDKI; only a few have explored the role of INK4 family members in AKI. Because INK4 family members block the G1-S transition, we postulated that they should have protective effects against AKI. The most conserved INK4 member is p18, so we selected it to explore its effects on cisplatin-induced renal cell injury. We overexpressed p18 in renal tubular epithelial cells (LLC-PK1) by transient transfection and investigated its effects on the cell cycle and proliferation. After transfection, cell injury was induced by cisplatin (100 μM) incubation for 24 h in a standard medium. The effect of p18 was assayed by assessing cell necrosis and apoptosis in transfected cells. The endoplasmic reticulum stress (ERS) pathway was evaluated to interpret the possible mechanism of p18 action in cisplatin-induced renal cell injury. Overexpression of p18 arrested cell cycle progression in the G1 phase and inhibited proliferation. Compared with vehicle transfection, p18 overexpression did not affect cisplatin-induced necrosis, but it reduced the percentage of apoptotic cells significantly. The severity of ERS induced by cisplatin was also decreased by p18 overexpression. P18 protects against cisplatin-induced renal cell injury. The mechanism of p18 protection may lie in its effect on the cell death pathway.
Collapse
Affiliation(s)
- Yi Zhang
- Nephrology Department of Changzheng Hospital, The Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| | | | | | | | | | | |
Collapse
|
45
|
Effects of ion channels on proliferation in cultured human cardiac fibroblasts. J Mol Cell Cardiol 2011; 51:198-206. [DOI: 10.1016/j.yjmcc.2011.05.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 04/24/2011] [Accepted: 05/11/2011] [Indexed: 01/01/2023]
|
46
|
Zhuo L, Fu B, Bai X, Zhang B, Wu L, Cui J, Cui S, Wei R, Chen X, Cai G. NAD blocks high glucose induced mesangial hypertrophy via activation of the sirtuins-AMPK-mTOR pathway. Cell Physiol Biochem 2011; 27:681-90. [PMID: 21691086 DOI: 10.1159/000330077] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2011] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND/AIMS Since the discovery of NAD-dependent deacetylases, Sirtuins, it has been recognized that maintaining intracellular levels of NAD is crucial for the management of stress-response of cells. Here we show that high glucose(HG)-induced mesangial hypertrophy is associated with loss of intracellular levels of NAD. This study was designed to investigate the effect of NAD on HG-induced mesangial hypertrophy. METHODS The rat glomerular mesangial cells (MCs) were incubated in HG medium with or without NAD. Afterwards, NAD(+)/NADH ratio and enzyme activity of Sirtuins was determined. In addition, the expression analyses of AMPK-mTOR signaling were evaluated by Western blot analysis. RESULTS We showed that HG induced the NAD(+)/NADH ratio and the levels of SIRT1 and SIRT3 activity decreased as well as mesangial hypertrophy, but NAD was capable of maintaining intracellular NAD(+)/NADH ratio and levels of SIRT1 and SIRT3 activity as well as of blocking the HG-induced mesangial hypertrophy in vitro. Activating Sirtuins by NAD blocked the activation of pro-hypertrophic Akt signaling, and augmented the activity of the antihypertrophic AMPK signaling in MCs, which prevented the subsequent induction of mTOR-mediated protein synthesis. By AMPK knockdown, we showed it upregulated phosphorylation of mTOR. In such, the NAD inhibited HG-induced mesangial hypertrophy whereas NAD lost its inhibitory effect in the presence of AMPK siRNA. CONCLUSION These results reveal a novel role of NAD as an inhibitor of mesangial hypertrophic signaling, and suggest that prevention of NAD depletion may be critical in the treatment of mesangial hypertrophy.
Collapse
Affiliation(s)
- Li Zhuo
- Department of Nephrology, Kidney Center and Key Lab of the People's Liberation Army, General Hospital of People's Liberation Army, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Xue Y, Tie CR, Li J, Tian T, Li QX. Ligustrazine inhibits lipopolysaccharide-induced proliferation by affecting P27, Bcl-2 expression in rat mesangial cells. Eur J Pharmacol 2011; 665:8-12. [PMID: 21586279 DOI: 10.1016/j.ejphar.2011.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 04/18/2011] [Accepted: 05/03/2011] [Indexed: 11/26/2022]
Abstract
Ligustrazine has a renoprotective effect against nephritis. In the present study, we investigated the roles of ligustrazine on lipopolysaccharide-induced changes of proliferation, cell cycle in cultured rat mesangial cells. 3-(4,5-dimethyltiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay revealed that rat mesangial cells treated with lipopolysaccharide (10mg/l) underwent significant proliferation compared with control group. This effect was significantly inhibited by ligustrazine (400 to 2500 mg/l). Flow cytometric analysis revealed that cells treated with lipopolysaccharide showed significant reduction in the ratio of G0/G1 phase and significant elevation in the ratio of S+G2/M phase. The changes of cell cycle induced by lipopolysaccharide were reversed by ligustrazine. In addition, lipopolysaccharide suppressed P27 protein expression was significantly increased by ligustrazine (100, 500, 2500 mg/l). Moreover, rat mesangial cells treated with lipopolysaccharide showed scanty apoptosis with up-regulation of Bcl-2expression, while Bax protein expression was not changed. Ligustrazine (100, 500, 2500 mg/l) significantly reversed lipopolysaccharide-induced up-regulation of Bcl-2 protein and increased apoptotic cell death. In summary, ligustrazine displayed a significant inhibiting effect on lipopolysaccharide-induced proliferation through increasing P27 and decreasing Bcl-2 protein expression in rat mesangial cells.
Collapse
Affiliation(s)
- Ying Xue
- Department of Otorhinolaryngology, Zhongnan Hospital, Wuhan University, China
| | | | | | | | | |
Collapse
|
48
|
Nishihara K, Masuda S, Nakagawa S, Yonezawa A, Ichimura T, Bonventre JV, Inui KI. Impact of Cyclin B2 and Cell division cycle 2 on tubular hyperplasia in progressive chronic renal failure rats. Am J Physiol Renal Physiol 2010; 298:F923-34. [PMID: 20071461 DOI: 10.1152/ajprenal.00567.2009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To clarify the specific molecular events of progressive tubular damage in chronic renal failure (CRF), we conducted microarray analyses using isolated proximal tubules from subtotally nephrectomized (Nx) rats as a model of CRF. Our results clearly demonstrated time-dependent changes in gene expression profiles localized to proximal tubules. The expression of mitosis-specific genes Cyclin B2 and Cell division cycle 2 (Cdc2) was significantly and selectively increased in the proximal tubules during the compensated period but decreased to basal level in the end-stage period. Administration of everolimus, a potent inhibitor of mammalian target of rapamycin, markedly reduced compensatory hypertrophy and hyperplasia of epithelial cells, which was accompanied by complete abolishment of the expression of Cyclin B2 and Cdc2 enhancement; renal function was then severely decreased. Treatment with the Cdc2 inhibitor 2-cyanoethyl alsterpaullone clearly decreased epithelial cell hyperplasia, based on staining of phosphorylated histone H3 and Ki-67, while hypertrophy was not inhibited. In conclusion, we have demonstrated roles of Cyclin B2 and Cdc2 in the epithelial hyperplasia in response to Nx. These results advance the knowledge of the contribution of cell cycle regulators, especially M phase, in pathophysiology of tubular restoration and/or degeneration, and these two molecules are suggested to be a marker for the proliferation of proximal tubular cells in CRF.
Collapse
Affiliation(s)
- Kumiko Nishihara
- Department of Pharmacy, Kyoto University Hospital, Sakyo-ku, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
49
|
Liu DZ, Ander BP, Sharp FR. Cell cycle inhibition without disruption of neurogenesis is a strategy for treatment of central nervous system diseases. Neurobiol Dis 2009; 37:549-57. [PMID: 19944161 DOI: 10.1016/j.nbd.2009.11.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 11/10/2009] [Accepted: 11/18/2009] [Indexed: 12/12/2022] Open
Abstract
Classically, the cell cycle is regarded as the process leading to cellular proliferation. However, increasing evidence over the last decade supports the notion that neuronal cell cycle re-entry results in post-mitotic death. A mature neuron that re-enters the cell cycle can neither advance to a new G0 quiescent state nor revert to its earlier G0 state. This presents a critical dilemma to the neuron from which death may be an unavoidable but necessary outcome for adult neurons attempting to complete the cell cycle. In contrast, tumor cells that undergo aberrant cell cycle re-entry divide and can survive. Thus, cell cycle inhibition strategies are of interest in cancer treatment but may also represent an important means of protecting neurons. In this review, we put forth the concept of the "expanded cell cycle" and summarize the cell cycle proteins, signal transduction events and mitogenic molecules that can drive a neuron into the cell cycle in various CNS diseases. We also discuss the pharmacological approaches that interfere with the mitogenic pathways and prevent mature neurons from attempting cell cycle re-entry, protecting them from cell death. Lastly, future attempts at blocking the cell cycle to rescue mature neurons from injury should be designed so as to not block normal neurogenesis.
Collapse
Affiliation(s)
- Da-Zhi Liu
- Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, CA 95817, USA.
| | | | | |
Collapse
|
50
|
Lindoso RS, Verdoorn KS, Einicker-Lamas M. Renal recovery after injury: the role of Pax-2. Nephrol Dial Transplant 2009; 24:2628-33. [PMID: 19556301 DOI: 10.1093/ndt/gfp307] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|