1
|
Liu B, Zhang Y, Wang Y, Meng Q, Zhang D, Yang H, Li G, Wang Y, Zhou H. Pharmacological targeting of AMPK to restore glucose and fatty acid metabolism homeostasis attenuates transplanted kidney fibrosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167510. [PMID: 39278511 DOI: 10.1016/j.bbadis.2024.167510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 08/10/2024] [Accepted: 09/07/2024] [Indexed: 09/18/2024]
Abstract
Chronic fibrosis often occurs in transplanted kidneys, leading to progressive functional decline. The underlying mechanisms may involve disruption in the metabolism of renal tubular epithelial cells. The liver kinase B1 (LKB1)-AMPK pathway is a pivotal regulatory hub for glucose and fatty acid metabolism and may play a role in transplanted kidney fibrosis, but it has not been reported. In this study we administered fenofibrate, 2-deoxyglucose, or metformin to modulate metabolism in Brown Norway rat kidney transplants and investigated pathways involved in fibrosis using various assays. We identified an impaired LKB1-AMPK pathway within epithelial cells, resulting in perturbed glucose and fatty acid metabolism, collagen secretion, extracellular matrix remodeling, and epithelial-mesenchymal transition. ACOX1, a pivotal enzyme in the fatty acid peroxisomal β-oxidation pathway, played an important role in transplanted renal fibrosis. Furthermore, several metabolism-targeting drugs, particularly metformin, emerged as potent fibrosis inhibitors. Metformin attenuated fibrosis, improved renal function, and reduced inflammation and macrophage infiltration in the transplanted kidneys. These results provide new perspectives for understanding the complex molecular basis underlying transplanted renal fibrosis and developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Bin Liu
- Department of Urology II, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Yanghe Zhang
- Department of Urology II, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Yuxiong Wang
- Department of Urology II, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Qingfei Meng
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, Jilin, China
| | - Dan Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, Jilin, China
| | - Hongxia Yang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, Jilin, China
| | - Guangtao Li
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, Jilin, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, Jilin, China
| | - Honglan Zhou
- Department of Urology II, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| |
Collapse
|
2
|
Hamidi N, Feizi F, Azadmehr A, Zabihi E, Khafri S, Zarei-Behjani Z, Babazadeh Z. Disulfiram ameliorates bleomycin induced pulmonary inflammation and fibrosis in rats. Biotech Histochem 2023; 98:584-592. [PMID: 37779489 DOI: 10.1080/10520295.2023.2261367] [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: 10/03/2023] Open
Abstract
Bleomycin (BL) is a widely used anticancer drug that can cause pulmonary fibrosis due to increased fibroblast proliferation and increased secretion of extracellular matrix. RASSF1A is a tumor suppressor gene that is down-regulated by DNA methylation during fibrosis. Disulfiram (DSF), a noncytosine DNA methyltransferase inhibitor, can revert epigenetic biomarkers and re-express silenced genes. We investigated anti-inflammatory and anti-fibrotic effects of DSF on regulation of epigenetic molecules and histopathology in a rat model of BL induced pulmonary fibrosis. We used six groups of rats: sesame oil (SO) control (Co) group, BL group, BL + SO group and three BL + DSF groups administered 1 mg/kg DSF (BL + DSF), 10 mg/kg DSF (BL + DSF10) or 100 mg/kg DSF (BL + DSF100), respectively. BL was administered intratracheally to induce pulmonary fibrosis. DSF and SO were injected intraperitoneally (i.p.) 2 days before BL administration; these injections were continued for 3 weeks. At the end of the study, lung tissues were removed for molecular and histopathologic studies. Administration of 10 or 100 mg/kg DSF after BL induced pulmonary inflammation and fibrosis, and up-regulated RASSF1A and down-regulated TNF-α and IL-1 β compared to the BL and BL + SO groups. A RASSF1A unmethylated band was observed using the methylation-specific PCR technique in rats that had been administered 10 and 100 mg/kg DSF, which indicated partial DNA demethylation. Histopathologic evaluation revealed that fibrosis and all inflammatory scores were decreased significantly in the BL + DSF10 and BL + DSF100 groups compared to the BL group. Our findings indicate that DSF modified DNA methylation by up-regulating RASSF1A, which reduced inflammation and fibrosis in BL induced pulmonary inflammation and fibrosis.
Collapse
Affiliation(s)
- Negar Hamidi
- Cellular and Molecular Biology Research Center, Institute of Health, Babol University of Medical Sciences, Babol, Iran
- Department of Anatomical Science, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Farideh Feizi
- Cellular and Molecular Biology Research Center, Institute of Health, Babol University of Medical Sciences, Babol, Iran
- Department of Anatomical Science, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Abbas Azadmehr
- Cellular and Molecular Biology Research Center, Institute of Health, Babol University of Medical Sciences, Babol, Iran
- Department of Immunology, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Ebrahim Zabihi
- Cellular and Molecular Biology Research Center, Institute of Health, Babol University of Medical Sciences, Babol, Iran
| | - Soraya Khafri
- Department of Biostatistics and Epidemiology, Faculty of Medicine, Babol University of Medical Science, Babol, Iran
| | - Zeinab Zarei-Behjani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Babazadeh
- Cellular and Molecular Biology Research Center, Institute of Health, Babol University of Medical Sciences, Babol, Iran
- Department of Anatomical Science, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
3
|
Zhao J, Yang S, Xu Y, Qin S, Bie F, Chen L, Zhou F, Xie J, Liu X, Shu B, Qi S. Mechanical pressure-induced dedifferentiation of myofibroblasts inhibits scarring via SMYD3/ITGBL1 signaling. Dev Cell 2023:S1534-5807(23)00190-9. [PMID: 37192621 DOI: 10.1016/j.devcel.2023.04.014] [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: 08/18/2022] [Revised: 12/21/2022] [Accepted: 04/24/2023] [Indexed: 05/18/2023]
Abstract
Pressure therapy (PT) is an effective intervention for reducing scarring, but its underlying mechanism remains largely unclear. Here, we demonstrate that human scar-derived myofibroblasts dedifferentiate into normal fibroblasts in response to PT, and we identify how SMYD3/ITGBL1 contributes to the nuclear relay of mechanical signals. In clinical specimens, reductions in SMYD3 and ITGBL1 expression levels are strongly associated with the anti-scarring effects of PT. The integrin β1/ILK pathway is inhibited in scar-derived myofibroblasts upon PT, leading to decreased TCF-4 and subsequently to reductions in SMYD3 expression, which reduces the levels of H3K4 trimethylation (H3K4me3) and further suppresses ITGBL1 expression, resulting the dedifferentiation of myofibroblasts into fibroblasts. In animal models, blocking SMYD3 expression results in reductions of scarring, mimicking the positive effects of PT. Our results show that SMYD3 and ITGBL1 act as sensors and mediators of mechanical pressure to inhibit the progression of fibrogenesis and provide therapeutic targets for fibrotic diseases.
Collapse
Affiliation(s)
- Jingling Zhao
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Shuai Yang
- Department of Neurosurgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Yingbin Xu
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Shitian Qin
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Fan Bie
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Lei Chen
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Fei Zhou
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Julin Xie
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Xusheng Liu
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Bin Shu
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China.
| | - Shaohai Qi
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
4
|
Kumari J, Wagener FADTG, Kouwer PHJ. Novel Synthetic Polymer-Based 3D Contraction Assay: A Versatile Preclinical Research Platform for Fibrosis. ACS APPLIED MATERIALS & INTERFACES 2022; 14:19212-19225. [PMID: 35468292 PMCID: PMC9073832 DOI: 10.1021/acsami.2c02549] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The driving factors causing fibrosis and scar formation include fibroblast differentiation into myofibroblasts and hampered myofibroblast apoptosis, which ultimately results in collagen accumulation and tissue contraction. Currently, only very few drugs are available for fibrosis treatment, and there is an urgent demand for new pharmaceutical products. High-throughput in vitro fibrosis models are necessary to develop such drugs. In this study, we developed such a novel model based on synthetic polyisocyanide (PIC-RGD) hydrogels. The model not only measures contraction but also allows for subsequent molecular and cellular analysis. Fibroblasts were seeded in small (10 μL) PIC-RGD gels in the absence or presence of TGFβ1, the latter to induce myofibroblast differentiation. The contraction model clearly differentiates fibroblasts and myofibroblasts. Besides a stronger contraction, we also observed α-smooth muscle actin (αSMA) production and higher collagen deposition for the latter. The results were supported by mRNA expression experiments of αSMA, Col1α1, P53, and Ki67. As proof of principle, the effects of FDA-approved antifibrotic drugs nintedanib and pirfenidone were tested in our newly developed fibrosis model. Both drugs clearly reduce myofibroblast-induced contraction. Moreover, both drugs significantly decrease myofibroblast viability. Our low-volume synthetic PIC-RGD hydrogel platform is an attractive tool for high-throughput in vitro antifibrotic drug screening.
Collapse
Affiliation(s)
- Jyoti Kumari
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- Department
of Dentistry - Orthodontics and Craniofacial Biology, Radboud University Medical Centre, 6525 EX Nijmegen, The Netherlands
| | - Frank A. D. T. G. Wagener
- Department
of Dentistry - Orthodontics and Craniofacial Biology, Radboud University Medical Centre, 6525 EX Nijmegen, The Netherlands
- (F.A.D.T.G.W.)
| | - Paul H. J. Kouwer
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- (P.H.J.K.)
| |
Collapse
|
5
|
Mlodawska OW, Saini P, Parker JB, Wei JJ, Bulun SE, Simon MA, Chakravarti D. Epigenomic and enhancer dysregulation in uterine leiomyomas. Hum Reprod Update 2022; 28:518-547. [PMID: 35199155 PMCID: PMC9247409 DOI: 10.1093/humupd/dmac008] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/16/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Uterine leiomyomas, also known as uterine fibroids or myomas, are the most common benign gynecological tumors and are found in women of reproductive and postmenopausal age. There is an exceptionally high prevalence of this tumor in women by the age of 50 years. Black women are particularly affected, with an increased incidence, earlier age of onset, larger and faster growing fibroids and greater severity of symptoms as compared to White women. Although advances in identifying genetic and environmental factors to delineate these fibroids have already been made, only recently has the role of epigenomics in the pathogenesis of this disease been considered. OBJECTIVE AND RATIONALE Over recent years, studies have identified multiple epigenomic aberrations that may contribute to leiomyoma development and growth. This review will focus on the most recent discoveries in three categories of epigenomic changes found in uterine fibroids, namely aberrant DNA methylation, histone tail modifications and histone variant exchange, and their translation into altered target gene architecture and transcriptional outcome. The findings demonstrating how the altered 3D shape of the enhancer can regulate gene expression from millions of base pairs away will be discussed. Additionally, translational implications of these discoveries and potential roadblocks in leiomyoma treatment will be addressed. SEARCH METHODS A comprehensive PubMed search was performed to identify published articles containing keywords relevant to the focus of the review, such as: uterine leiomyoma, uterine fibroids, epigenetic alterations, epigenomics, stem cells, chromatin modifications, extracellular matrix [ECM] organization, DNA methylation, enhancer, histone post-translational modifications and dysregulated gene expression. Articles until September 2021 were explored and evaluated to identify relevant updates in the field. Most of the articles focused on in the discussion were published between 2015 and 2021, although some key discoveries made before 2015 were included for background information and foundational purposes. We apologize to the authors whose work was not included because of space restrictions or inadvertent omission. OUTCOMES Chemical alterations to the DNA structure and of nucleosomal histones, without changing the underlying DNA sequence, have now been implicated in the phenotypic manifestation of uterine leiomyomas. Genome-wide DNA methylation analysis has revealed subsets of either suppressed or overexpressed genes accompanied by aberrant promoter methylation. Furthermore, differential promoter access resulting from altered 3D chromatin structure and histone modifications plays a role in regulating transcription of key genes thought to be involved in leiomyoma etiology. The dysregulated genes function in tumor suppression, apoptosis, angiogenesis, ECM formation, a variety of cancer-related signaling pathways and stem cell differentiation. Aberrant DNA methylation or histone modification is also observed in altering enhancer architecture, which leads to changes in enhancer-promoter contact strength, producing novel explanations for the overexpression of high mobility group AT-hook 2 and gene dysregulation found in mediator complex subunit 12 mutant fibroids. While many molecular mechanisms and epigenomic features have been investigated, the basis for the racial disparity observed among those in the Black population remains unclear. WIDER IMPLICATIONS A comprehensive understanding of the exact pathogenesis of uterine leiomyoma is lacking and requires attention as it can provide clues for prevention and viable non-surgical treatment. These findings will widen our knowledge of the role epigenomics plays in the mechanisms related to uterine leiomyoma development and highlight novel approaches for the prevention and identification of epigenome targets for long-term non-invasive treatment options of this significantly common disease.
Collapse
Affiliation(s)
| | | | - J Brandon Parker
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jian-Jun Wei
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA
| | - Serdar E Bulun
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Melissa A Simon
- Department of Obstetrics and Gynecology, Center for Health Equity Transformation, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Debabrata Chakravarti
- Correspondence address. Department of Obstetrics and Gynecology, Northwestern University, Feinberg School of Medicine, 303 E Superior Street, Lurie 4-119, Chicago, IL 60611, USA. E-mail:
| |
Collapse
|
6
|
Wang S, Zuo A, Jiang W, Xie J, Lin H, Sun W, Zhao M, Xia J, Shao J, Zhao X, Liang D, Yang A, Sun J, Wang M. JMJD1A/NR4A1 Signaling Regulates the Procession of Renal Tubular Epithelial Interstitial Fibrosis Induced by AGEs in HK-2. Front Med (Lausanne) 2022; 8:807694. [PMID: 35186975 PMCID: PMC8850412 DOI: 10.3389/fmed.2021.807694] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
Diabetic kidney disease (DKD) is one of the most serious complications of diabetic patients. Advanced glycation end products (AGEs) induce epithelial-mesenchymal transformation (EMT) of renal tubular epithelial cells (HK-2), resulting in renal tubulointerstitial fibrosis. However, the underlying epigenetic mechanisms remain to be further investigated. In this work, we investigated the functional role of JMJD1A involved in DKD progression. The molecular mechanism study was performed in AGEs-induced HK-2 cells by gene expression analysis, RNA sequencing (RNA-seq), and JMJD1A lentiviral knockdown and overexpression particle transfection. The results showed that AGEs could upregulate JMJD1A, and the expressions of related fibrotic factor were also increased. At the same time, in the DKD animal model induced by unilateral nephrectomy plus streptozotocin (STZ), IHC immunohistochemical staining showed that compared with the control group, the expressions of JMJD1A, FN, and COL1 in the model group were all increased, masson staining results also show that the model group has typical fibrotic changes. This is consistent with the results of our in vitro experiments. In order to determine the downstream pathway, we screened out JMJD1A downstream transcription factors by RNA-seq. Further analysis showed that JMJD1A overexpression could accelerate the progression of AGEs-induced renal fibrosis by reducing the expression of NR4A1 in HK-2 cells. Meanwhile, NR4A1 inhibitor can promote the expression of fibrosis-related factors such as VIM, a-SMA in HK-2 cells, and aggravate the process of fibrosis. Taken together, JMJD1A/NR4A1 signaling can regulate the procession of renal tubular epithelial interstitial fibrosis induced by AGEs in HK-2.
Collapse
Affiliation(s)
- Shaoting Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Anna Zuo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Weiqiang Jiang
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiarun Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Haoyu Lin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Wei Sun
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Min Zhao
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jinjin Xia
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Junqiao Shao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiaoshan Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Donghui Liang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Aicheng Yang
- The Affiliated Jiangmen Traditional Chinese Medicine Hospital, Jinan University, Guangzhou, China
- Aicheng Yang
| | - Jia Sun
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Jia Sun
| | - Ming Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Ming Wang
| |
Collapse
|
7
|
Juillerat-Jeanneret L, Tafelmeyer P, Golshayan D. Regulation of Fibroblast Activation Protein-α Expression: Focus on Intracellular Protein Interactions. J Med Chem 2021; 64:14028-14045. [PMID: 34523930 DOI: 10.1021/acs.jmedchem.1c01010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The prolyl-specific peptidase fibroblast activation protein-α (FAP-α) is expressed at very low or undetectable levels in nondiseased human tissues but is selectively induced in activated (myo)fibroblasts at sites of tissue remodeling in fibrogenic processes. In normal regenerative processes involving transient fibrosis FAP-α+(myo)fibroblasts disappear from injured tissues, replaced by cells with a normal FAP-α- phenotype. In chronic uncontrolled pathological fibrosis FAP-α+(myo)fibroblasts permanently replace normal tissues. The mechanisms of regulation and elimination of FAP-α expression in(myo)fibroblasts are unknown. According to a yeast two-hybrid screen and protein databanks search, we propose that the intracellular (co)-chaperone BAG6/BAT3 can interact with FAP-α, mediated by the BAG6/BAT3 Pro-rich domain, inducing proteosomal degradation of FAP-α protein under tissue homeostasis. In this Perspective, we discuss our findings in the context of current knowledge on the regulation of FAP-α expression and comment potential therapeutic strategies for uncontrolled fibrosis, including small molecule degraders (PROTACs)-modified FAP-α targeted inhibitors.
Collapse
Affiliation(s)
- Lucienne Juillerat-Jeanneret
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), CH1011 Lausanne, Switzerland.,University Institute of Pathology, CHUV and UNIL, CH1011 Lausanne, Switzerland
| | - Petra Tafelmeyer
- Hybrigenics Services, Laboratories and Headquarters-Paris, 1 rue Pierre Fontaine, 91000 Evry, France.,Hybrigenics Corporation, Cambridge Innovation Center, 50 Milk Street, Cambridge, Massachusetts 02142, United States
| | - Dela Golshayan
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), CH1011 Lausanne, Switzerland
| |
Collapse
|
8
|
Hatanaka H, Mukai A, Ito E, Ueno M, Sotozono C, Kinoshita S, Hamuro J. Epigenetic regulation of the epithelial mesenchymal transition induced by synergistic action of TNF-α and TGF-β in retinal pigment epithelial cells. Biochem Biophys Res Commun 2021; 544:31-37. [PMID: 33516879 DOI: 10.1016/j.bbrc.2021.01.060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 01/19/2021] [Indexed: 12/30/2022]
Abstract
To clarify the influence of tumor necrosis factor (TNF)-α on fibrotic phenotypes induced by transforming growth factor (TGF)-β in retinal pigment epithelial cells (RPECs) by epigenetic regulation. Human primary retinal pigment epithelial cells (RPECs including ARPE19) were used in cultures in the presence or absence of TNF-α and/or TGF-β2. RT2 Profiler™ (Qiagen) was used for PCR Array for fibrosis and epithelial mesenchymal transition (EMT). Microarray analysis by 3D gene DNA chip was outsourced to Toray Industries Inc. Quantification of histone acetyl transferase (HAT)-related and histone deacetylase (HDAC) related gene expression were also analyzed. HDAC and HAT activity was measured using an EpiQuik HDAC and HAT Activity/Inhibition Assay Kit (Epigentek). CD44, MMP-9, HAT, and HDAC in RPECs were analyzed by western blotting. Analysis of expression of the EMT/fibrosis related CD44 and MMP-9 phenotypes induced by TNF-α+TGF-β2 revealed four alterations in RPECs: 1) abolition of TGF-β2-induced α-SMA by TNF-α; 2) synergy between TNF-α+TGF-β2 for induction of CD44 and MMP-9 phenotypes 3) no inhibition of HDAC activity by either TNF-α or TGF-β2; and 4) significant inhibition of HAT activity by either TNF-α or TGF-β2, but no synergy. The HDAC activation through HAT inhibition by TNF-α+TGF-β was counteracted by HDAC inhibitors, leading to the inhibition of EMT/fibrosis. EMT/fibrotic CD44 and MMP-9 phenotypes were epigenetically upregulated by concerted action of TNF-α and TGF-β in RPECs. The intervention in epigenetic regulation may hold potential in preventing intraocular proliferative diseases.
Collapse
Affiliation(s)
- Hiroki Hatanaka
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Atsushi Mukai
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Eiko Ito
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Morio Ueno
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Chie Sotozono
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Shigeru Kinoshita
- Department of Frontier Medical Science and Technology for Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Junji Hamuro
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan.
| |
Collapse
|
9
|
Bangert C, Rindler K, Krausgruber T, Alkon N, Thaler FM, Kurz H, Ayub T, Demirtas D, Fortelny N, Vorstandlechner V, Bauer WM, Quint T, Mildner M, Jonak C, Elbe-Bürger A, Griss J, Bock C, Brunner PM. Persistence of mature dendritic cells, T H2A, and Tc2 cells characterize clinically resolved atopic dermatitis under IL-4Rα blockade. Sci Immunol 2021; 6:eabe2749. [PMID: 33483337 DOI: 10.1126/sciimmunol.abe2749] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022]
Abstract
Therapeutic options for autoimmune diseases typically consist of broad and targeted immunosuppressive agents. However, sustained clinical benefit is rarely achieved, as the disease phenotype usually returns after cessation of treatment. To better understand tissue-resident immune memory in human disease, we investigated patients with atopic dermatitis (AD) who underwent short-term or long-term treatment with the IL-4Rα blocker dupilumab. Using multi-omics profiling with single-cell RNA sequencing and multiplex proteomics, we found significant decreases in overall skin immune cell counts and normalization of transcriptomic dysregulation in keratinocytes consistent with clearance of disease. However, we identified specific immune cell populations that persisted for up to a year after clinical remission while being absent from healthy controls. These populations included LAMP3 + CCL22+ mature dendritic cells, CRTH2 + CD161 + T helper ("TH2A") cells, and CRTAM + cytotoxic T cells, which expressed high levels of CCL17 (dendritic cells) and IL13 (T cells). TH2A cells showed a characteristic cytokine receptor constellation with IL17RB, IL1RL1 (ST2), and CRLF2 expression, suggesting that these cells are key responders to the AD-typical epidermal alarmins IL-25, IL-33, and TSLP, respectively. We thus identified disease-linked immune cell populations in resolved AD indicative of a persisting disease memory, facilitating a rapid response system of epidermal-dermal cross-talk between keratinocytes, dendritic cells, and T cells. This observation may help to explain the disease recurrence upon termination of immunosuppressive treatments in AD, and it identifies potential disease memory-linked cell types that may be targeted to achieve a more sustained therapeutic response.
Collapse
Affiliation(s)
- Christine Bangert
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Katharina Rindler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Natalia Alkon
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Felix M Thaler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Harald Kurz
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Tanya Ayub
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Denis Demirtas
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Nikolaus Fortelny
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Wolfgang M Bauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Tamara Quint
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Constanze Jonak
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | - Johannes Griss
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Patrick M Brunner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
10
|
Yamamoto Y, Mukai A, Ikushima T, Urata Y, Kinoshita S, Hamuro J, Ueno M, Sotozono C. Pluripotent epigenetic regulator OBP-801 maintains filtering blebs in glaucoma filtration surgery model. Sci Rep 2020; 10:20936. [PMID: 33262357 PMCID: PMC7708845 DOI: 10.1038/s41598-020-77811-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Inhibition of fibrosis is indispensable for maintaining filtering blebs after glaucoma filtration surgery (GFS). The purpose of this study was to investigate the ability of a pluripotent epigenetic regulator OBP-801 (OBP) to ameliorate extracellular matrix formation in a rabbit model of GFS. Rabbits that underwent GFS were treated with OBP. The gene expression profiles and intraocular pressure (IOP) were monitored until 30 postoperative days. The bleb tissues were evaluated for tissue fibrosis at 30 postoperative days. In in vitro models, OBP interfered the functions of diverse genes during the wound-healing process. In in vivo GFS models, the expressions of TGF-β3, MMP-2, TIMP-2 and 3, LOX, COL1A and SERPINH1 were significantly inhibited at 30 postoperative days in the OBP group compared with those in the vehicle control group. OBP treatment involving subconjunctival injection or eye drops showed no adverse effects, and reduced levels of α-SMA and collagen deposition at the surgical wound site. OBP maintained the long-lived bleb without scar formation, and IOP was lower at 30 postoperative days compared with the vehicle control group. These findings suggest that OBP is an effective and useful candidate low-molecular-weight agent for improving wound healing and surgical outcomes in a rabbit model of GFS.
Collapse
Affiliation(s)
- Yuji Yamamoto
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Atsushi Mukai
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Toru Ikushima
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yasuo Urata
- Oncolys BioPharma, Inc., Tokyo, 106-0032, Japan
| | - Shigeru Kinoshita
- Department of Frontier Medical Science and Technology for Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Junji Hamuro
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Morio Ueno
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Chie Sotozono
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
11
|
Ding Z, Wu X, Wang Y, Ji S, Zhang W, Kang J, Li J, Fei G. Melatonin prevents LPS-induced epithelial-mesenchymal transition in human alveolar epithelial cells via the GSK-3β/Nrf2 pathway. Biomed Pharmacother 2020; 132:110827. [PMID: 33065391 DOI: 10.1016/j.biopha.2020.110827] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Oxidative stress plays a critical role in pulmonary fibrosis after acute lung injury (ALI), and epithelial-mesenchymal transition (EMT) events are involved in this process. The purpose of this study was to investigate the protective effects of melatonin, a natural antioxidant, on lipopolysaccharide (LPS)-induced EMT in human alveolar epithelial cells. METHODS Human type II alveolar epithelial cell-derived A549 cells were incubated with LPS and melatonin alone or in combination for up to 24 h. The morphological changes of the treated cells were evaluated as well as indexes of oxidative stress. EMT-related proteins and the Nrf2 signaling pathway were detected by western blot analysis and immunofluorescence staining, respectively. To further investigate the underlying mechanisms, the effects of melatonin on cells transfected Nrf2 short hairpin RNA (shRNA) and the PI3K / GSK-3β signaling pathway were evaluated. RESULTS Treatment with melatonin upregulated Nrf2 expression, inhibited LPS-induced cell morphological change, reversed the expressions of EMT-related proteins, and reduced reactive oxygen species (ROS) production in A549 cells, as well as the levels of malondialdehyde (MDA) and anti-oxidative enzymes. Yet, the effects of melatonin were almost completely abolished in cells transfected Nrf2 shRNA. Furthermore, the data demonstrated that melatonin could activate the PI3K/AKT signaling pathway, resulting in phosphorylation of GSK-3β (Ser9) and upregulation of the Nrf2 protein in A549 cells, which ultimately attenuated LPS-induced EMT. CONCLUSION The present study is the first to demonstrate that melatonin can protect human alveolar epithelial cells against oxidative stress by effectively inhibiting LPS-induced EMT, which was mostly dependent on upregulation of the Nrf2 pathway via the PI3K/GSK-3β axis. Further studies are warranted to investigate the role of melatonin for the treatment of oxidative stress-associated diseases, as well as pulmonary fibrosis after ALI.
Collapse
Affiliation(s)
- Zhenxing Ding
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China
| | - Xu Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China
| | - Yueguo Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China
| | - Shuang Ji
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China
| | - Wenying Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China
| | - Jiaying Kang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China
| | - Jiajia Li
- Center Lab of The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China
| | - Guanghe Fei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China; Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, China.
| |
Collapse
|
12
|
Zhang Y, Distler JHW. Therapeutic molecular targets of SSc-ILD. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2020; 5:17-30. [DOI: 10.1177/2397198319899013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 11/26/2019] [Indexed: 12/16/2022]
Abstract
Systemic sclerosis is a fibrosing chronic connective tissue disease of unknown etiology. A major hallmark of systemic sclerosis is the uncontrolled and persistent activation of fibroblasts, which release excessive amounts of extracellular matrix, lead to organ dysfunction, and cause high mobility and motility of patients. Systemic sclerosis–associated interstitial lung disease is one of the most common fibrotic organ manifestations in systemic sclerosis and a major cause of death. Treatment options for systemic sclerosis–associated interstitial lung disease and other fibrotic manifestations, however, remain very limited. Thus, there is a huge medical need for effective therapies that target tissue fibrosis, vascular alterations, inflammation, and autoimmune disease in systemic sclerosis–associated interstitial lung disease. In this review, we discuss data suggesting therapeutic ways to target different genes in distinct tissues/organs that contribute to the development of SSc.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Internal Medicine 3—Rheumatology and Immunology, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Jörg HW Distler
- Department of Internal Medicine 3—Rheumatology and Immunology, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
13
|
Ligresti G, Caporarello N, Meridew JA, Jones DL, Tan Q, Choi KM, Haak AJ, Aravamudhan A, Roden AC, Prakash YS, Lomberk G, Urrutia RA, Tschumperlin DJ. CBX5/G9a/H3K9me-mediated gene repression is essential to fibroblast activation during lung fibrosis. JCI Insight 2019; 5:127111. [PMID: 31095524 DOI: 10.1172/jci.insight.127111] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Pulmonary fibrosis is a devastating disease characterized by accumulation of activated fibroblasts and scarring in the lung. While fibroblast activation in physiological wound repair reverses spontaneously, fibroblast activation in fibrosis is aberrantly sustained. Here we identified histone 3 lysine 9 methylation (H3K9me) as a critical epigenetic modification that sustains fibroblast activation by repressing the transcription of genes essential to returning lung fibroblasts to an inactive state. We show that the histone methyltransferase G9a (EHMT2) and chromobox homolog 5 (CBX5, also known as HP1α), which deposit H3K9me marks and assemble an associated repressor complex respectively, are essential to initiation and maintenance of fibroblast activation specifically through epigenetic repression of peroxisome proliferator-activated receptor gamma coactivator 1 alpha gene (PPARGC1A, encoding PGC1α). Both TGFβ and increased matrix stiffness potently inhibit PGC1α expression in lung fibroblasts through engagement of the CBX5/G9a pathway. Inhibition of CBX5/G9a pathway in fibroblasts elevates PGC1α, attenuates TGFβ- and matrix stiffness-promoted H3K9 methylation, and reduces collagen accumulation in the lungs following bleomycin injury. Our results demonstrate that epigenetic silencing mediated by H3K9 methylation is essential for both biochemical and biomechanical fibroblast activation, and that targeting this epigenetic pathway may provide therapeutic benefit by returning lung fibroblasts to quiescence.
Collapse
Affiliation(s)
| | | | | | | | - Qi Tan
- Department of Physiology and Biomedical Engineering
| | | | | | | | | | - Y S Prakash
- Department of Physiology and Biomedical Engineering.,Department of Anesthesiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gwen Lomberk
- Division of Research,Department of Surgery and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| | - Raul A Urrutia
- Division of Research,Department of Surgery and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| | | |
Collapse
|
14
|
Ricard-Blum S, Baffet G, Théret N. Molecular and tissue alterations of collagens in fibrosis. Matrix Biol 2018; 68-69:122-149. [DOI: 10.1016/j.matbio.2018.02.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/02/2018] [Accepted: 02/02/2018] [Indexed: 02/07/2023]
|
15
|
Vella S, Conaldi PG, Cova E, Meloni F, Liotta R, Cuzzocrea S, Martino L, Bertani A, Luca A, Vitulo P. Lung resident mesenchymal cells isolated from patients with the Bronchiolitis Obliterans Syndrome display a deregulated epigenetic profile. Sci Rep 2018; 8:11167. [PMID: 30042393 PMCID: PMC6057887 DOI: 10.1038/s41598-018-29504-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 07/09/2018] [Indexed: 12/13/2022] Open
Abstract
Bronchiolitis Obliterans Syndrome is the major determinant of the graft function loss after lung transplantation, but its pathogenesis is still incompletely understood and currently available therapeutic strategies are poorly effective. A deeper understanding of its pathogenic mechanisms is crucial for the development of new strategies to prevent and treat this devastating complication. In this study, we focused on the mesenchymal stromal cells, recently recognized as BOS key effectors, and our primary aim was to identify their epigenetic determinants, such as histone modifications and non-coding RNA regulation, which could contribute to their differentiation in myofibroblasts. Interestingly, we identified a deregulated expression of histone deacetylases and methyltransferases, and a microRNA-epigenetic regulatory network, which could represent novel targets for anti-fibrotic therapy. We validated our results in vitro, in a cell model of fibrogenesis, confirming the epigenetic involvement in this process and paving the way for a new application for epigenetic drugs.
Collapse
Affiliation(s)
- Serena Vella
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy.
- Anemocyte S.r.l, Gerenzano, Italy.
| | - Pier Giulio Conaldi
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Emanuela Cova
- Department of Respiratory Diseases, IRCCS San Matteo Foundation and University of Pavia, Pavia, Italy
| | - Federica Meloni
- Department of Respiratory Diseases, IRCCS San Matteo Foundation and University of Pavia, Pavia, Italy
| | - Rosa Liotta
- Department of Diagnostic and Therapeutic Services, Pathology Service, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Lavinia Martino
- Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Alessandro Bertani
- Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Angelo Luca
- Department of Diagnostic and Therapeutic Services, Radiology Service, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Patrizio Vitulo
- Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| |
Collapse
|
16
|
A novel indole compound MA-35 attenuates renal fibrosis by inhibiting both TNF-α and TGF-β 1 pathways. Sci Rep 2017; 7:1884. [PMID: 28507324 PMCID: PMC5432497 DOI: 10.1038/s41598-017-01702-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/03/2017] [Indexed: 01/03/2023] Open
Abstract
Renal fibrosis is closely related to chronic inflammation and is under the control of epigenetic regulations. Because the signaling of transforming growth factor-β1 (TGF-β1) and tumor necrosis factor-α (TNF-α) play key roles in progression of renal fibrosis, dual blockade of TGF-β1 and TNF-α is desired as its therapeutic approach. Here we screened small molecules showing anti-TNF-α activity in the compound library of indole derivatives. 11 out of 41 indole derivatives inhibited the TNF-α effect. Among them, Mitochonic Acid 35 (MA-35), 5-(3, 5-dimethoxybenzyloxy)-3-indoleacetic acid, showed the potent effect. The anti-TNF-α activity was mediated by inhibiting IκB kinase phosphorylation, which attenuated the LPS/GaIN-induced hepatic inflammation in the mice. Additionally, MA-35 concurrently showed an anti-TGF-β1 effect by inhibiting Smad3 phosphorylation, resulting in the downregulation of TGF-β1-induced fibrotic gene expression. In unilateral ureter obstructed mouse kidney, which is a renal fibrosis model, MA-35 attenuated renal inflammation and fibrosis with the downregulation of inflammatory cytokines and fibrotic gene expressions. Furthermore, MA-35 inhibited TGF-β1-induced H3K4me1 histone modification of the fibrotic gene promoter, leading to a decrease in the fibrotic gene expression. MA-35 affects multiple signaling pathways involved in the fibrosis and may recover epigenetic modification; therefore, it could possibly be a novel therapeutic drug for fibrosis.
Collapse
|
17
|
Wang HD, Liu L, Zhao HR, Hou QF, Yan JB, Shi WL, Guo QN, Wang L, Liao SX, Zhu BF. Detection of fetal epigenetic biomarkers through genome-wide DNA methylation study for non-invasive prenatal diagnosis. Mol Med Rep 2017; 15:3989-3998. [PMID: 28440505 PMCID: PMC5436219 DOI: 10.3892/mmr.2017.6506] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/13/2017] [Indexed: 12/20/2022] Open
Abstract
The discovery of cell-free DNA fetal (cff DNA) in maternal plasma during pregnancy provides a novel perspective for the development of non-invasive prenatal diagnosis (NIPD). Against the background of maternal DNA, the use of the relatively low concentration of cff DNA is limited in NIPD. Therefore, in order to overcome the complication of the background of maternal DNA and expand the scope of cff DNA application in clinical practice, it is necessary to identify novel universal fetal-specific DNA markers. The GeneChip Human Promoter 1.0R Array set was used in the present study to analyze the methylation status of 12 placental tissue and maternal peripheral blood whole-genome DNA samples. In total, 5 fetus differential hypermethylation regions and 6 fetus differential hypomethylation regions were identified. In order to verify the 11 selected methylation regions and detect the differential CpG sites in these regions, a bisulfate direct sequencing strategy was used. In total, 87 fetal differential methylation CpG sites were identified from 123 CpG sites. The detection of fetal differential methylation DNA regions and CpG sites may be instrumental in the development of efficient NIPD and in the expansion of its application in other disorders.
Collapse
Affiliation(s)
- Hong-Dan Wang
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Lin Liu
- Department of Cardiovascular Ultrasound, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Hui-Ru Zhao
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Qiao-Fang Hou
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Jing-Bin Yan
- Shanghai Children's Hospital, Shanghai Institute of Medical Genetics, Key Laboratory of Embryo Molecular Biology, Ministry of Health of China and Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai 200040, P.R. China
| | - Wei-Li Shi
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Qian-Nan Guo
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Li Wang
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Shi-Xiu Liao
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Bo-Feng Zhu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
18
|
Yang Y, Chen XX, Li WX, Wu XQ, Huang C, Xie J, Zhao YX, Meng XM, Li J. EZH2-mediated repression of Dkk1 promotes hepatic stellate cell activation and hepatic fibrosis. J Cell Mol Med 2017; 21:2317-2328. [PMID: 28332284 PMCID: PMC5618695 DOI: 10.1111/jcmm.13153] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 02/02/2017] [Indexed: 12/21/2022] Open
Abstract
EZH2, a histone H3 lysine‐27‐specific methyltransferase, is involved in diverse physiological and pathological processes including cell proliferation and differentiation. However, the role of EZH2 in liver fibrosis is largely unknown. In this study, it was identified that EZH2 promoted Wnt pathway‐stimulated fibroblasts in vitro and in vivo by repressing Dkk‐1, which is a Wnt pathway antagonist. The expression of EZH2 was increased in CCl4‐induced rat liver and primary HSCs as well as TGF‐β1‐treated HSC‐T6, whereas the expression of Dkk1 was reduced. Silencing of EZH2 prevented TGF‐β1‐induced proliferation of HSC‐T6 cells and the expression of α‐SMA. In addition, knockdown of Dkk1 promoted TGF‐β1‐induced activation of HSCs. Moreover, silencing of EZH2 could restore the repression of Dkk‐1 through trimethylation of H3K27me3 in TGF‐β1‐treated HSC‐T6 cells. Interestingly, inhibition of EZH2 had almost no effect on the activation of HSC when Dkk1 was silenced. Collectively, EZH2‐mediated repression of Dkk1 promotes the activation of Wnt/β‐catenin pathway, which is an essential event for HSC activation.
Collapse
Affiliation(s)
- Yang Yang
- The key of Laboratory Precision Medicine for Severe Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiao-Xia Chen
- The key of Laboratory Precision Medicine for Severe Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China.,Department of Pharmacy, Anhui No. 2 Province People's Hospital, Hefei, China
| | - Wan-Xia Li
- The key of Laboratory Precision Medicine for Severe Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiao-Qin Wu
- The key of Laboratory Precision Medicine for Severe Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Cheng Huang
- The key of Laboratory Precision Medicine for Severe Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Juan Xie
- The key of Laboratory Precision Medicine for Severe Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Yu-Xin Zhao
- The key of Laboratory Precision Medicine for Severe Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- The key of Laboratory Precision Medicine for Severe Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Jun Li
- The key of Laboratory Precision Medicine for Severe Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| |
Collapse
|
19
|
Inhibition of DNA Methylation in the COL1A2 Promoter by Anacardic Acid Prevents UV-Induced Decrease of Type I Procollagen Expression. J Invest Dermatol 2017; 137:1343-1352. [PMID: 28237615 DOI: 10.1016/j.jid.2017.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 01/13/2017] [Accepted: 02/01/2017] [Indexed: 12/22/2022]
Abstract
UV radiation decreases type I procollagen production mainly by inhibiting the transforming growth factor-β/Smad signaling pathway. Because further epigenetic regulatory mechanisms are unclear, we investigated the roles of DNA methylation and histone acetylation in UV-induced regulation of COL1A2 transcription in human dermal fibroblasts. Anacardic acid, a p300 histone acetyltransferase inhibitor, rescued the UV-induced decrease of type I procollagen expression in human dermal fibroblasts. Although UV irradiation induced global histone acetylation, it reduced the local recruitment of histone H3 acetylation as well as p300, and Smad2/3 to the p300 binding site (-1406/-1393), in the COL1A2 promoter as shown by chromatin immunoprecipitation. This effect was reversed by anacardic acid treatment. In contrast, pyrosequencing analysis showed that UV irradiation induced DNA methylation in the same region of the COL1A2 promoter, which was reversed by anacardic acid and a DNA methyltransferase inhibitor (5-AZA-2'-deoxycytidine). Inhibition of UV-induced DNA methylation led to an increase of UV-induced histone acetylation in the COL1A2 promoter and increased the recruitment of transcription factors, leading to up-regulation of type I collagen after UV irradiation. Collectively, our findings indicate that the epigenetic crosstalk between DNA methylation and histone acetylation plays a crucial role in COL1A2 transcription induced by UV irradiation.
Collapse
|
20
|
Zhou W, Mo X, Cui W, Zhang Z, Li D, Li L, Xu L, Yao H, Gao J. Nrf2 inhibits epithelial-mesenchymal transition by suppressing snail expression during pulmonary fibrosis. Sci Rep 2016; 6:38646. [PMID: 27982105 PMCID: PMC5159829 DOI: 10.1038/srep38646] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/10/2016] [Indexed: 12/17/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a phenotype conversion that plays a critical role in the development of pulmonary fibrosis (PF). It is known that snail could regulate the progression of EMT. Nuclear factor erythroid 2 related factor 2 (Nrf2), a key regulator of antioxidant defense system, protects cells against oxidative stress. However, it is not known whether Nrf2 regulates snail thereby modulating the development of PF. Here, bleomycin (BLM) was intratracheally injected into both Nrf2-knockout (Nrf2-/-) and wild-type mice to compare the development of PF. Rat type II alveolar epithelial cells (RLE-6TN) were treated with a specific Nrf2 activator sulforaphane, or transfected with Nrf2 and snail siRNAs to determine their effects on transforming growth factor β1 (TGF-β1)-induced EMT. We found that BLM-induced EMT and lung fibrosis were more severe in Nrf2-/- mice compared to wild-type mice. In vitro, sulforaphane treatment attenuated TGF-β1-induced EMT, accompanied by the down-regulation of snail. Inversely, silencing Nrf2 by siRNA enhanced TGF-β1-induced EMT along with increased expression of snail. Interestingly, when snail was silenced by siRNA, sulforaphane treatment was unable to reduce the progression of EMT in RLE-6TN cells. These findings suggest that Nrf2 attenuates EMT and fibrosis process by regulating the expression of snail in PF.
Collapse
Affiliation(s)
- Wencheng Zhou
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Xiaoting Mo
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Wenhui Cui
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, 230032, China.,The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Zhihui Zhang
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Delin Li
- Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Liucheng Li
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Liang Xu
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Hongwei Yao
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Jian Gao
- The Second Hospital of Dalian Medical University, Dalian, 116023, China
| |
Collapse
|
21
|
Yang C, Zheng SD, Wu HJ, Chen SJ. Regulatory Mechanisms of the Molecular Pathways in Fibrosis Induced by MicroRNAs. Chin Med J (Engl) 2016; 129:2365-72. [PMID: 27647197 PMCID: PMC5040024 DOI: 10.4103/0366-6999.190677] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE MicroRNAs (miRNAs or miRs) play critical roles in the fibrotic process in different organs. We summarized the latest research progress on the roles and mechanisms of miRNAs in the regulation of the molecular signaling pathways involved in fibrosis. DATA SOURCES Papers published in English from January 2010 to August 2015 were selected from the PubMed and Web of Science databases using the search terms "microRNA", "miR", "transforming growth factor β", "tgf β", "mitogen-activated protein kinase", "mapk", "integrin", "p38", "c-Jun NH2-terminal kinase", "jnk", "extracellular signal-regulated kinase", "erk", and "fibrosis". STUDY SELECTION Articles were obtained and reviewed to analyze the regulatory effects of miRNAs on molecular signaling pathways involved in the fibrosis. RESULTS Recent evidence has shown that miRNAs are involved in regulating fibrosis by targeting different substrates in the molecular processes that drive fibrosis, such as immune cell sensitization, effector cell activation, and extracellular matrix remodeling. Moreover, several important molecular signaling pathways involve in fibrosis, such as the transforming growth factor-beta (TGF-β) pathway, mitogen-activated protein kinase (MAPK) pathways, and the integrin pathway are regulated by miRNAs. Third, regulation of the fibrotic pathways induced by miRNAs is found in many other tissues in addition to the heart, lung, liver, and kidney. Interestingly, the actions of many drugs on the human body are also induced by miRNAs. It is encouraging that the fibrotic process can be blocked or reversed by targeting specific miRNAs and their signaling pathways, thereby protecting the structures and functions of different organs. CONCLUSIONS miRNAs not only regulate molecular signaling pathways in fibrosis but also serve as potential targets of novel therapeutic interventions for fibrosing diseases.
Collapse
Affiliation(s)
- Cui Yang
- Department of Cardiology, Huairou Hospital of Traditional Chinese Medicine, Beijing 101400, China
| | - Si-Dao Zheng
- Department of Cardiology, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, Beijing 100039, China
| | - Hong-Jin Wu
- Department of Cardiology, Beijing Haidian Hospital, Haidian Section of Peking University Third Hospital, Beijing 100191, China
| | - Shao-Jun Chen
- Department of Cardiology, Huairou Hospital of Traditional Chinese Medicine, Beijing 101400, China
| |
Collapse
|
22
|
Kurose H, Mangmool S. Myofibroblasts and inflammatory cells as players of cardiac fibrosis. Arch Pharm Res 2016; 39:1100-13. [PMID: 27515051 DOI: 10.1007/s12272-016-0809-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/01/2016] [Indexed: 01/18/2023]
Abstract
On myocardial infarction, many cells are injured or died owing to arterial occlusion. Intracellular molecules released from injured or dead cells initiate inflammatory responses that play important roles in cardiac remodeling including fibrosis. Fibrosis is an excess accumulation of extracellular collagen. Currently, drugs used to treat cardiac fibrosis are not commercially available. Myofibroblasts are responsible for the production and secretion of collagen. Infiltrating inflammatory cells interact with fibroblasts or other cells and promote myofibroblast formation. Inflammatory cells also modulate the activities of myofibroblasts. Regulation of collagen production is critical for modulating the progression of fibrosis. Hence, the manipulation of activities of inflammatory cells and myofibroblasts will provide promising therapeutic targets for treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Hitoshi Kurose
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
| |
Collapse
|
23
|
Yue L, Yao H. Mitochondrial dysfunction in inflammatory responses and cellular senescence: pathogenesis and pharmacological targets for chronic lung diseases. Br J Pharmacol 2016; 173:2305-18. [PMID: 27189175 DOI: 10.1111/bph.13518] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/04/2016] [Accepted: 05/05/2016] [Indexed: 12/16/2022] Open
Abstract
Mitochondria are dynamic organelles, which couple the various cellular processes that regulate metabolism, cell proliferation and survival. Environmental stress can cause mitochondrial dysfunction and dynamic changes including reduced mitochondrial biogenesis, oxidative phosphorylation and ATP production, as well as mitophagy impairment, which leads to increased ROS, inflammatory responses and cellular senescence. Oxidative stress, inflammation and cellular senescence all have important roles in the pathogenesis of chronic lung diseases, such as chronic obstructive pulmonary disease, pulmonary fibrosis and bronchopulmonary dysplasia. In this review, we discuss the current state on how mitochondrial dysfunction affects inflammatory responses and cellular senescence, the mechanisms of mitochondrial dysfunction underlying the pathogenesis of chronic lung diseases and the potential of mitochondrial transfer and replacement as treatments for these diseases.
Collapse
Affiliation(s)
- Li Yue
- Department of Orthopaedics and Rehabilitation, University of Rochester, Rochester, NY, USA
| | - Hongwei Yao
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University Alpert Medical School, Providence, RI, USA
| |
Collapse
|
24
|
|
25
|
Salminen A, Kauppinen A, Kaarniranta K. 2-Oxoglutarate-dependent dioxygenases are sensors of energy metabolism, oxygen availability, and iron homeostasis: potential role in the regulation of aging process. Cell Mol Life Sci 2015; 72:3897-914. [PMID: 26118662 PMCID: PMC11114064 DOI: 10.1007/s00018-015-1978-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 06/10/2015] [Accepted: 06/22/2015] [Indexed: 02/06/2023]
Abstract
Recent studies have revealed that the members of an ancient family of nonheme Fe(2+)/2-oxoglutarate-dependent dioxygenases (2-OGDO) are involved in the functions associated with the aging process. 2-Oxoglutarate and O2 are the obligatory substrates and Fe(2+) a cofactor in the activation of 2-OGDO enzymes, which can induce the hydroxylation of distinct proteins and the demethylation of DNA and histones. For instance, ten-eleven translocation 1-3 (TET1-3) are the demethylases of DNA, whereas Jumonji C domain-containing histone lysine demethylases (KDM2-7) are the major epigenetic regulators of chromatin landscape, known to be altered with aging. The functions of hypoxia-inducible factor (HIF) prolyl hydroxylases (PHD1-3) as well as those of collagen hydroxylases are associated with age-related degeneration. Moreover, the ribosomal hydroxylase OGFOD1 controls mRNA translation, which is known to decline with aging. 2-OGDO enzymes are the sensors of energy metabolism, since the Krebs cycle intermediate 2-oxoglutarate is an activator whereas succinate and fumarate are the potent inhibitors of 2-OGDO enzymes. In addition, O2 availability and iron redox homeostasis control the activities of 2-OGDO enzymes in tissues. We will briefly elucidate the catalytic mechanisms of 2-OGDO enzymes and then review the potential functions of the above-mentioned 2-OGDO enzymes in the control of the aging process.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| | - Anu Kauppinen
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
- Department of Ophthalmology, Kuopio University Hospital, P.O.B. 100, 70029, Kuopio, Finland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
- Department of Ophthalmology, Kuopio University Hospital, P.O.B. 100, 70029, Kuopio, Finland.
| |
Collapse
|
26
|
PNPO Deficiency and Cirrhosis: Expanding the Clinical Phenotype? JIMD Rep 2015; 25:71-75. [PMID: 26108646 DOI: 10.1007/8904_2015_456] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 05/06/2015] [Accepted: 05/08/2015] [Indexed: 12/13/2022] Open
Abstract
We report the case of a 4-year-old boy with pyridoxamine 5-phosphate oxidase deficiency, now the second reported case to develop hepatic cirrhosis. He presented with an encephalopathy in the first 1.5 h of life and received a first dose of PLP at 40 h of life. PNPO gene sequencing identified homozygosity for a novel variant in exon 7, c.637C>T (p.Pro213Ser). Persistent elevations in alanine transferase and aspartate transferase combined with an echogenic liver on ultrasound prompted performance of a liver biopsy which demonstrated hepatic cirrhosis. This is the second reported case of hepatic cirrhosis in PNPO deficiency. The pathogenesis is unclear but may be related to epigenetic activation of purinergic signaling in the hepatic stellate cells. PNPO deficiency may in time prove to be a suitable candidate for consideration of therapeutic orthotropic liver transplantation in select patients.
Collapse
|
27
|
Wang WP, Ho PY, Chen QX, Addepalli B, Limbach PA, Li MM, Wu WJ, Jilek JL, Qiu JX, Zhang HJ, Li T, Wun T, White RD, Lam KS, Yu AM. Bioengineering Novel Chimeric microRNA-34a for Prodrug Cancer Therapy: High-Yield Expression and Purification, and Structural and Functional Characterization. J Pharmacol Exp Ther 2015; 354:131-41. [PMID: 26022002 DOI: 10.1124/jpet.115.225631] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 05/27/2015] [Indexed: 12/18/2022] Open
Abstract
Development of anticancer treatments based on microRNA (miRNA/miR) such as miR-34a replacement therapy is limited to the use of synthetic RNAs with artificial modifications. Herein, we present a new approach to a high-yield and large-scale biosynthesis, in Escherichia coli using transfer RNA (tRNA) scaffold, of chimeric miR-34a agent, which may act as a prodrug for anticancer therapy. The recombinant tRNA fusion pre-miR-34a (tRNA/mir-34a) was quickly purified to a high degree of homogeneity (>98%) using anion-exchange fast protein liquid chromatography, whose primary sequence and post-transcriptional modifications were directly characterized by mass spectrometric analyses. Chimeric tRNA/mir-34a showed a favorable cellular stability while it was degradable by several ribonucleases. Deep sequencing and quantitative real-time polymerase chain reaction studies revealed that tRNA-carried pre-miR-34a was precisely processed to mature miR-34a within human carcinoma cells, and the same tRNA fragments were produced from tRNA/mir-34a and the control tRNA scaffold (tRNA/MSA). Consequently, tRNA/mir-34a inhibited the proliferation of various types of human carcinoma cells in a dose-dependent manner and to a much greater degree than the control tRNA/MSA, which was mechanistically attributable to the reduction of miR-34a target genes. Furthermore, tRNA/mir-34a significantly suppressed the growth of human non-small-cell lung cancer A549 and hepatocarcinoma HepG2 xenograft tumors in mice, compared with the same dose of tRNA/MSA. In addition, recombinant tRNA/mir-34a had no or minimal effect on blood chemistry and interleukin-6 level in mouse models, suggesting that recombinant RNAs were well tolerated. These findings provoke a conversation on producing biologic miRNAs to perform miRNA actions, and point toward a new direction in developing miRNA-based therapies.
Collapse
Affiliation(s)
- Wei-Peng Wang
- Department of Biochemistry and Molecular Medicine (W.-P.W., P.Y.H., Q.-X.C., M.-M.L., W.-J.W., J.L.J., K.S.L., A.-M.Y.), Division of Hematology Oncology (T.L., T.W., K.S.L.) and Department of Urology (R.D.W.), School of Medicine, University of California-Davis, Sacramento, California; Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio (B.A., P.A.L.); Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York (J.-X.Q.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W., H.-J.Z.)
| | - Pui Yan Ho
- Department of Biochemistry and Molecular Medicine (W.-P.W., P.Y.H., Q.-X.C., M.-M.L., W.-J.W., J.L.J., K.S.L., A.-M.Y.), Division of Hematology Oncology (T.L., T.W., K.S.L.) and Department of Urology (R.D.W.), School of Medicine, University of California-Davis, Sacramento, California; Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio (B.A., P.A.L.); Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York (J.-X.Q.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W., H.-J.Z.)
| | - Qiu-Xia Chen
- Department of Biochemistry and Molecular Medicine (W.-P.W., P.Y.H., Q.-X.C., M.-M.L., W.-J.W., J.L.J., K.S.L., A.-M.Y.), Division of Hematology Oncology (T.L., T.W., K.S.L.) and Department of Urology (R.D.W.), School of Medicine, University of California-Davis, Sacramento, California; Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio (B.A., P.A.L.); Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York (J.-X.Q.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W., H.-J.Z.)
| | - Balasubrahmanyam Addepalli
- Department of Biochemistry and Molecular Medicine (W.-P.W., P.Y.H., Q.-X.C., M.-M.L., W.-J.W., J.L.J., K.S.L., A.-M.Y.), Division of Hematology Oncology (T.L., T.W., K.S.L.) and Department of Urology (R.D.W.), School of Medicine, University of California-Davis, Sacramento, California; Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio (B.A., P.A.L.); Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York (J.-X.Q.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W., H.-J.Z.)
| | - Patrick A Limbach
- Department of Biochemistry and Molecular Medicine (W.-P.W., P.Y.H., Q.-X.C., M.-M.L., W.-J.W., J.L.J., K.S.L., A.-M.Y.), Division of Hematology Oncology (T.L., T.W., K.S.L.) and Department of Urology (R.D.W.), School of Medicine, University of California-Davis, Sacramento, California; Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio (B.A., P.A.L.); Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York (J.-X.Q.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W., H.-J.Z.)
| | - Mei-Mei Li
- Department of Biochemistry and Molecular Medicine (W.-P.W., P.Y.H., Q.-X.C., M.-M.L., W.-J.W., J.L.J., K.S.L., A.-M.Y.), Division of Hematology Oncology (T.L., T.W., K.S.L.) and Department of Urology (R.D.W.), School of Medicine, University of California-Davis, Sacramento, California; Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio (B.A., P.A.L.); Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York (J.-X.Q.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W., H.-J.Z.)
| | - Wen-Juan Wu
- Department of Biochemistry and Molecular Medicine (W.-P.W., P.Y.H., Q.-X.C., M.-M.L., W.-J.W., J.L.J., K.S.L., A.-M.Y.), Division of Hematology Oncology (T.L., T.W., K.S.L.) and Department of Urology (R.D.W.), School of Medicine, University of California-Davis, Sacramento, California; Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio (B.A., P.A.L.); Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York (J.-X.Q.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W., H.-J.Z.)
| | - Joseph L Jilek
- Department of Biochemistry and Molecular Medicine (W.-P.W., P.Y.H., Q.-X.C., M.-M.L., W.-J.W., J.L.J., K.S.L., A.-M.Y.), Division of Hematology Oncology (T.L., T.W., K.S.L.) and Department of Urology (R.D.W.), School of Medicine, University of California-Davis, Sacramento, California; Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio (B.A., P.A.L.); Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York (J.-X.Q.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W., H.-J.Z.)
| | - Jing-Xin Qiu
- Department of Biochemistry and Molecular Medicine (W.-P.W., P.Y.H., Q.-X.C., M.-M.L., W.-J.W., J.L.J., K.S.L., A.-M.Y.), Division of Hematology Oncology (T.L., T.W., K.S.L.) and Department of Urology (R.D.W.), School of Medicine, University of California-Davis, Sacramento, California; Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio (B.A., P.A.L.); Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York (J.-X.Q.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W., H.-J.Z.)
| | - Hong-Jian Zhang
- Department of Biochemistry and Molecular Medicine (W.-P.W., P.Y.H., Q.-X.C., M.-M.L., W.-J.W., J.L.J., K.S.L., A.-M.Y.), Division of Hematology Oncology (T.L., T.W., K.S.L.) and Department of Urology (R.D.W.), School of Medicine, University of California-Davis, Sacramento, California; Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio (B.A., P.A.L.); Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York (J.-X.Q.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W., H.-J.Z.)
| | - Tianhong Li
- Department of Biochemistry and Molecular Medicine (W.-P.W., P.Y.H., Q.-X.C., M.-M.L., W.-J.W., J.L.J., K.S.L., A.-M.Y.), Division of Hematology Oncology (T.L., T.W., K.S.L.) and Department of Urology (R.D.W.), School of Medicine, University of California-Davis, Sacramento, California; Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio (B.A., P.A.L.); Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York (J.-X.Q.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W., H.-J.Z.)
| | - Theodore Wun
- Department of Biochemistry and Molecular Medicine (W.-P.W., P.Y.H., Q.-X.C., M.-M.L., W.-J.W., J.L.J., K.S.L., A.-M.Y.), Division of Hematology Oncology (T.L., T.W., K.S.L.) and Department of Urology (R.D.W.), School of Medicine, University of California-Davis, Sacramento, California; Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio (B.A., P.A.L.); Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York (J.-X.Q.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W., H.-J.Z.)
| | - Ralph DeVere White
- Department of Biochemistry and Molecular Medicine (W.-P.W., P.Y.H., Q.-X.C., M.-M.L., W.-J.W., J.L.J., K.S.L., A.-M.Y.), Division of Hematology Oncology (T.L., T.W., K.S.L.) and Department of Urology (R.D.W.), School of Medicine, University of California-Davis, Sacramento, California; Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio (B.A., P.A.L.); Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York (J.-X.Q.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W., H.-J.Z.)
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine (W.-P.W., P.Y.H., Q.-X.C., M.-M.L., W.-J.W., J.L.J., K.S.L., A.-M.Y.), Division of Hematology Oncology (T.L., T.W., K.S.L.) and Department of Urology (R.D.W.), School of Medicine, University of California-Davis, Sacramento, California; Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio (B.A., P.A.L.); Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York (J.-X.Q.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W., H.-J.Z.)
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine (W.-P.W., P.Y.H., Q.-X.C., M.-M.L., W.-J.W., J.L.J., K.S.L., A.-M.Y.), Division of Hematology Oncology (T.L., T.W., K.S.L.) and Department of Urology (R.D.W.), School of Medicine, University of California-Davis, Sacramento, California; Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio (B.A., P.A.L.); Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York (J.-X.Q.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W., H.-J.Z.)
| |
Collapse
|