1
|
Zhang S, Xie R, Wang L, Fu G, Zhang C, Zhang Y, Yu J. TMEM252 inhibits epithelial-mesenchymal transition and progression in papillary thyroid carcinoma by regulating Notch1 expression. Head Neck 2024. [PMID: 39152570 DOI: 10.1002/hed.27922] [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: 03/28/2024] [Revised: 07/23/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Papillary thyroid carcinoma (PTC) accounts for about 85% of thyroid cancer cases. Transmembrane protein 252 (TMEM252) is a gene encoding a transmembrane protein that has only been reported to be associated with triple-negative breast cancer. Herein, we first elucidated the physiological roles and possible regulatory proteins of TMEM252 in PTC pathogenesis. METHODS Quantitative real-time polymerase chain reaction, western blot, and immunohistochemical analyses were utilized to ascertain the relative TMEM252 expression in PTC and surrounding normal tissues. Functional investigations involved CCK-8 viability assay, EdU incorporation assay for proliferation, transwell assays for migration and invasion, and an in vivo tumor development assessment to evaluate the TMEM252-mediated regulation of tumor formation. RESULTS Our results first revealed diminished TMEM252 transcript and protein expressions in PTC tissues and cell lines. TMEM252 overexpression suppressed cell proliferation through reducing p53, p21, and p16 expression. Conversely, TMEM252 depletion has opposite effects in PTC cells both in vivo. Additionally, the upregulation of TMEM252 demonstrated cell migration and invasion suppression by impeding the epithelial-mesenchymal transition (EMT) process via inhibition of the Notch pathway. Furthermore, overexpression of TMEM252 suppressed tumor growth in vivo. CONCLUSION Our study elucidates that TMEM252 suppresses PTC progression by modulating the Notch pathway. These findings underscore TMEM252 is a potential therapeutic target in managing PTC.
Collapse
Affiliation(s)
- Shuyong Zhang
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rong Xie
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Liuhuan Wang
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guoxue Fu
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chenxi Zhang
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yang Zhang
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jichun Yu
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
2
|
Tommasini D, Fox R, Ngo KJ, Hinman JD, Fogel BL. Alterations in oligodendrocyte transcriptional networks reveal region-specific vulnerabilities to neurological disease. iScience 2023; 26:106358. [PMID: 36994077 PMCID: PMC10040735 DOI: 10.1016/j.isci.2023.106358] [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: 07/20/2022] [Revised: 12/22/2022] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Neurological disease is characterized the by dysfunction of specific neuroanatomical regions. To determine whether region-specific vulnerabilities have a transcriptional basis at cell-type-specific resolution, we analyzed gene expression in mouse oligodendrocytes across various brain regions. Oligodendrocyte transcriptomes cluster in an anatomical arrangement along the rostrocaudal axis. Moreover, regional oligodendrocyte populations preferentially regulate genes implicated in diseases that target their region of origin. Systems-level analyses identify five region-specific co-expression networks representing distinct molecular pathways in oligodendrocytes. The cortical network exhibits alterations in mouse models of intellectual disability and epilepsy, the cerebellar network in ataxia, and the spinal network in multiple sclerosis. Bioinformatic analyses reveal potential molecular regulators of these networks, which were confirmed to modulate network expression in vitro in human oligodendroglioma cells, including reversal of the disease-associated transcriptional effects of a pathogenic Spinocerebellar ataxia type 1 allele. These findings identify targetable region-specific vulnerabilities to neurological disease mediated by oligodendrocytes.
Collapse
Affiliation(s)
- Dario Tommasini
- Department of Neurology, UCLA David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rachel Fox
- Department of Human Genetics, UCLA David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kathie J. Ngo
- Department of Neurology, UCLA David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jason D. Hinman
- Department of Neurology, UCLA David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Brent L. Fogel
- Department of Neurology, UCLA David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, UCLA David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
3
|
Allograft inflammatory factor-1 enhances inflammation and oxidative stress via the NF-κB pathway in diabetic kidney disease. Biochem Biophys Res Commun 2022; 614:63-69. [DOI: 10.1016/j.bbrc.2022.04.089] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/13/2022] [Accepted: 04/20/2022] [Indexed: 12/31/2022]
|
4
|
Panvini AR, Gvritishvili A, Galvan H, Nashine S, Atilano SR, Kenney MC, Tombran-Tink J. Differential mitochondrial and cellular responses between H vs. J mtDNA haplogroup-containing human RPE transmitochondrial cybrid cells. Exp Eye Res 2022; 219:109013. [PMID: 35283109 PMCID: PMC9949352 DOI: 10.1016/j.exer.2022.109013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/08/2022] [Accepted: 02/23/2022] [Indexed: 02/02/2023]
Abstract
Mitochondrial dysfunction is associated with several retinal degenerative diseases including Age-related Macular Degeneration (AMD). Human mitochondrial DNA (mtDNA) haplogroups are inherited from a common ancestral clan and are defined by specific sets of genetic differences. The purpose of this study was to determine and compare the effects of mtDNA haplogroups H and J on transcriptome regulation and cellular resilience to oxidative stress in human RPE cytoplasmic hybrid (cybrid) cell lines in vitro. ARPE-19 cybrid cell lines containing mtDNA haplogroups H and J were created by fusing platelets obtained from normal individuals containing H and J haplogroups with mitochondria-deficient (Rho0) ARPE-19 cell lines. These cybrids were exposed to oxidative stress using 300 μM hydrogen peroxide (H2O2), following which mitochondrial structural dynamics was studied at varying time points using the mitochondrial markers - TOMM20 (Translocase of Outer Mitochondrial Membrane 20) and Mitotracker. To evaluate mitochondrial function, levels of ROS, ΔΨm and [Ca2+]m were measured using flow cytometry, and ATP levels were measured using luminescence. The H and J cybrid cell transcriptomes were compared using RNAseq to determine how changes in mtDNA regulate gene expression. Inflammatory and angiogenic markers were measured using Luminex assay to understand how these mtDNAs influenced cellular response to oxidative stress. Actin filaments' morphology was examined using confocal microscopy. Following exposure to H2O2 stress, the J cybrids showed increased mitochondrial swelling and perinuclear localization, disturbed fission and fusion, increased calcium uptake (p < 0.05), and higher secreted levels of TNF-α and VEGF (p < 0.001), compared to the H cybrids. Calcium uptake by J cybrids was reduced using an IP3R inhibitor. Thirteen genes involved in mitochondrial complex I and V function, fusion/fission events, cellular energy homeostasis, antioxidant defenses, and inflammatory responses, were significantly downregulated with log2 fold changes ranging between -1.5 and -5.1. Actin levels were also significantly reduced in stressed J cybrids (p ≤ 0.001) and disruption in actin filaments was observed. Thirty-eight genes involved in mitochondrial and cellular support functions, were upregulated with log2 fold changes of +1.5 to +5.9 in J cybrids compared to H cybrids. Our results demonstrate significant structural and functional differences between mtDNA haplogroups H vs. J -containing cybrid cells. Our study suggests that the J mtDNA haplogroup can alter the transcriptome to increase cellular susceptibility to stress and retinal degenerations.
Collapse
Affiliation(s)
- Ana Rubin Panvini
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Anzor Gvritishvili
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Hannah Galvan
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Sonali Nashine
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, 92697, USA
| | - Shari R. Atilano
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, 92697, USA
| | - M. Cristina Kenney
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, 92697, USA
| | - Joyce Tombran-Tink
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, 17033, USA; Department of Ophthalmology, Penn State College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
5
|
The analysis of a subset of HLA region associations in type 1 diabetes and multiple sclerosis suggests the involvement mechanisms other than antigen presentation in the pathogenesis. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2021.100831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
6
|
Wang Y, Niu A, Pan Y, Cao S, Terker AS, Wang S, Fan X, Toth CL, Ramirez Solano MA, Michell DL, Contreras D, Allen RM, Zhu W, Sheng Q, Fogo AB, Vickers KC, Zhang MZ, Harris RC. Profile of Podocyte Translatome During Development of Type 2 and Type 1 Diabetic Nephropathy Using Podocyte-Specific TRAP mRNA RNA-seq. Diabetes 2021; 70:2377-2390. [PMID: 34233930 PMCID: PMC8576501 DOI: 10.2337/db21-0110] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/29/2021] [Indexed: 12/22/2022]
Abstract
Podocyte injury is important in development of diabetic nephropathy (DN). Although several studies have reported single-cell-based RNA sequencing (RNA-seq) of podocytes in type 1 DN (T1DN), the podocyte translating mRNA profile in type 2 DN (T2DN) has not previously been compared with that of T1DN. We analyzed the podocyte translatome in T2DN in podocin-Cre; Rosa26fsTRAP; eNOS-/-; db/db mice and compared it with that of streptozotocin-induced T1DN in podocin-Cre; Rosa26fsTRAP; eNOS-/- mice using translating ribosome affinity purification (TRAP) and RNA-seq. More than 125 genes were highly enriched in the podocyte ribosome. More podocyte TRAP genes were differentially expressed in T2DN than in T1DN. TGF-β signaling pathway genes were upregulated, while MAPK pathway genes were downregulated only in T2DN, while ATP binding and cAMP-mediated signaling genes were downregulated only in T1DN. Genes regulating actin filament organization and apoptosis increased, while genes regulating VEGFR signaling and glomerular basement membrane components decreased in both type 1 and type 2 diabetic podocytes. A number of diabetes-induced genes not previously linked to podocyte injury were confirmed in both mouse and human DN. On the basis of differences and similarities in the podocyte translatome in T2DN and T1DN, investigators can identify factors underlying the pathophysiology of DN and novel therapeutic targets to treat diabetes-induced podocyte injury.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetic Nephropathies/genetics
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/pathology
- Gene Expression Profiling
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Organ Specificity/genetics
- Podocytes/metabolism
- Podocytes/pathology
- Protein Biosynthesis/genetics
- Proteome/analysis
- Proteome/genetics
- Proteome/metabolism
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA-Seq
- Sequence Analysis, RNA
- Streptozocin
- Transcriptome
Collapse
Affiliation(s)
- Yinqiu Wang
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Aolei Niu
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Yu Pan
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Shirong Cao
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Andrew S Terker
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Suwan Wang
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Xiaofeng Fan
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Cynthia L Toth
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Marisol A Ramirez Solano
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Danielle L Michell
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Danielle Contreras
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Ryan M Allen
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Wanying Zhu
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Quanhu Sheng
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Agnes B Fogo
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Kasey C Vickers
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Ming-Zhi Zhang
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Raymond C Harris
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
- Department of Veterans Affairs, Nashville, TN
| |
Collapse
|
7
|
Wang WJ, Jiang X, Gao CC, Chen ZW. Salusin‑β participates in high glucose‑induced HK‑2 cell ferroptosis in a Nrf‑2‑dependent manner. Mol Med Rep 2021; 24:674. [PMID: 34296310 PMCID: PMC8335735 DOI: 10.3892/mmr.2021.12313] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Ferroptosis is critically involved in the pathophysiology of diabetic nephropathy (DN). As a bioactive peptide, salusin‑β is abundantly expressed in the kidneys. However, it is unclear whether salusin‑β participates in the pathologies of diabetic kidney damage by regulating ferroptosis. The present study found that high glucose (HG) treatment upregulated the protein expressions of salusin‑β in a dose‑ and time‑dependent manner. Genetic knockdown of salusin‑β retarded, whereas overexpression of salusin‑β aggravated, HG‑triggered iron overload, antioxidant capability reduction, massive reactive oxygen species production and lipid peroxidation in HK‑2 cells. Mechanistically, salusin‑β inactivated nuclear factor erythroid‑derived 2‑like 2 (Nrf‑2) signaling, thus contributing to HG‑induced ferroptosis‑related changes in HK‑2 cells. Notably, the protein expression of salusin‑β was upregulated by ferroptosis activators, such as erastin, RSL3, FIN56 and buthionine sulfoximine. Pretreatment with ferrostatin‑1 (a ferroptosis inhibitor) prevented the upregulated protein expression of salusin‑β in HK‑2 cells exposed to HG. Taken together, these results suggested that a positive feedback loop between salusin‑β and ferroptosis primes renal tubular cells for injury in diabetes.
Collapse
Affiliation(s)
- Wen-Juan Wang
- Department of Nephrology, Center of Blood Purification, The Second People's Hospital of Nantong, Nantong, Jiangsu 226002, P.R. China
| | - Xia Jiang
- Department of Nephrology, Center of Blood Purification, The Second People's Hospital of Nantong, Nantong, Jiangsu 226002, P.R. China
| | - Chang-Chun Gao
- Department of Nephrology, Center of Blood Purification, The Second People's Hospital of Nantong, Nantong, Jiangsu 226002, P.R. China
| | - Zhi-Wei Chen
- Department of Nephrology, Center of Blood Purification, The Second People's Hospital of Nantong, Nantong, Jiangsu 226002, P.R. China
| |
Collapse
|
8
|
Klimontov VV, Saik OV, Korbut AI. Glucose Variability: How Does It Work? Int J Mol Sci 2021; 22:ijms22157783. [PMID: 34360550 PMCID: PMC8346105 DOI: 10.3390/ijms22157783] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 02/07/2023] Open
Abstract
A growing body of evidence points to the role of glucose variability (GV) in the development of the microvascular and macrovascular complications of diabetes. In this review, we summarize data on GV-induced biochemical, cellular and molecular events involved in the pathogenesis of diabetic complications. Current data indicate that the deteriorating effect of GV on target organs can be realized through oxidative stress, glycation, chronic low-grade inflammation, endothelial dysfunction, platelet activation, impaired angiogenesis and renal fibrosis. The effects of GV on oxidative stress, inflammation, endothelial dysfunction and hypercoagulability could be aggravated by hypoglycemia, associated with high GV. Oscillating hyperglycemia contributes to beta cell dysfunction, which leads to a further increase in GV and completes the vicious circle. In cells, the GV-induced cytotoxic effect includes mitochondrial dysfunction, endoplasmic reticulum stress and disturbances in autophagic flux, which are accompanied by reduced viability, activation of apoptosis and abnormalities in cell proliferation. These effects are realized through the up- and down-regulation of a large number of genes and the activity of signaling pathways such as PI3K/Akt, NF-κB, MAPK (ERK), JNK and TGF-β/Smad. Epigenetic modifications mediate the postponed effects of glucose fluctuations. The multiple deteriorative effects of GV provide further support for considering it as a therapeutic target in diabetes.
Collapse
Affiliation(s)
- Vadim V. Klimontov
- Laboratory of Endocrinology, Research Institute of Clinical and Experimental Lymphology—Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (RICEL—Branch of IC&G SB RAS), 630060 Novosibirsk, Russia; (O.V.S.); (A.I.K.)
- Correspondence:
| | - Olga V. Saik
- Laboratory of Endocrinology, Research Institute of Clinical and Experimental Lymphology—Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (RICEL—Branch of IC&G SB RAS), 630060 Novosibirsk, Russia; (O.V.S.); (A.I.K.)
- Laboratory of Computer Proteomics, Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (IC&G SB RAS), 630090 Novosibirsk, Russia
| | - Anton I. Korbut
- Laboratory of Endocrinology, Research Institute of Clinical and Experimental Lymphology—Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (RICEL—Branch of IC&G SB RAS), 630060 Novosibirsk, Russia; (O.V.S.); (A.I.K.)
| |
Collapse
|
9
|
The effect of methylethylpiridinol addition to the therapy on the level of pigment epithelium-derived factor and oxidative status in patients with diabetic nephropathy: randomized controlled open-label clinical study. J Diabetes Metab Disord 2021; 20:709-717. [PMID: 34222086 DOI: 10.1007/s40200-021-00802-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
Purpose The diabetic nephropathy is associated with oxidative stress and increases in pigment epithelium-derived factor (PEDF) level in the patient's blood. For the first time, authors investigated the effect of methylethylpiridinol addition to the therapy on oxidative status and pigment epithelium-derived factor concentrations, and examined the relationship between these indicators and clinical markers of pathology development. Methods Study design: open label randomized controlled trial study. Authors assessed the effect of methylethylpiridinol addition to the therapy vs basic treatment on antioxidant and NADPH-generating enzymes activity, glutathione's concentration and free radical-induced oxidation's intensity using a spectrophotometric method and iron-induced biochemiluminescence. The pigment epithelium-derived factor concentration in the serum was measured by enzyme-linked immunosorbent assay. Results Patients receiving combination therapy with methylethylpiridinol showed a more substantial increase in activity of glutathione peroxidase (Δ = 0.04 ± 0.11, p = 0.002), glutathione transferase (Δ = 0.12 ± 0.08, p < 0.001) and the concentration of reduced glutathione (Δ = 0.30 ± 0.17, p = 0.039). In addition, there was a significant decrease in PEDF level (Δ = -6.4 ± 5.4, p = 0.004). Correlation analysis showed a negative link between Δ postprandial glucose and Δ NADP-isocitrate dehydrogenase (-0.39, p = 0.033), Δ reduced glutathione and Δ postprandial glucose (-0.372, p = 0.043), Δ glutathione transferase and Δ PEDF (-0.37, p = 0.044). Conclusions The methylethylpiridinol addition to the therapy had a more potent stimulating effect on the patients' oxidative status in comparison with standard treatment, and reliably decreased pigment epithelium-derived factor level in patients' serum. The observed differences seem to be associated with the antioxidant activity of methylethylpiridinol which contributing to the mitigation of oxidative stress reducing at diabetes mellitus.
Collapse
|
10
|
Levin A, Reznichenko A, Witasp A, Liu P, Greasley PJ, Sorrentino A, Blondal T, Zambrano S, Nordström J, Bruchfeld A, Barany P, Ebefors K, Erlandsson F, Patrakka J, Stenvinkel P, Nyström J, Wernerson A. Novel insights into the disease transcriptome of human diabetic glomeruli and tubulointerstitium. Nephrol Dial Transplant 2021; 35:2059-2072. [PMID: 32853351 PMCID: PMC7716805 DOI: 10.1093/ndt/gfaa121] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 03/19/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is the most common cause of end-stage renal disease, affecting ∼30% of the rapidly growing diabetic population, and strongly associated with cardiovascular risk. Despite this, the molecular mechanisms of disease remain unknown. METHODS RNA sequencing (RNAseq) was performed on paired, micro-dissected glomerular and tubulointerstitial tissue from patients diagnosed with DN [n = 19, 15 males, median (range) age: 61 (30-85) years, chronic kidney disease stages 1-4] and living kidney donors [n = 20, 12 males, median (range) age: 56 (30-70) years]. RESULTS Principal component analysis showed a clear separation between glomeruli and tubulointerstitium transcriptomes. Differential expression analysis identified 1550 and 4530 differentially expressed genes, respectively (adjusted P < 0.01). Gene ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses highlighted activation of inflammation and extracellular matrix (ECM) organization pathways in glomeruli, and immune and apoptosis pathways in tubulointerstitium of DN patients. Specific gene modules were associated with renal function in weighted gene co-expression network analysis. Increased messengerRNA (mRNA) expression of renal damage markers lipocalin 2 (LCN) and hepatitis A virus cellular receptor1 (HAVCR1) in the tubulointerstitial fraction was observed alongside higher urinary concentrations of the corresponding proteins neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1) in DN patients. CONCLUSIONS Here we present the first RNAseq experiment performed on paired glomerular and tubulointerstitial samples from DN patients. We show that prominent disease-specific changes occur in both compartments, including relevant cellular processes such as reorganization of ECM and inflammation (glomeruli) as well as apoptosis (tubulointerstitium). The results emphasize the potential of utilizing high-throughput transcriptomics to decipher disease pathways and treatment targets in this high-risk patient population.
Collapse
Affiliation(s)
- Anna Levin
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Reznichenko
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Witasp
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peidi Liu
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Peter J Greasley
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | | | - Sonia Zambrano
- KI/AZ Integrated Cardio Metabolic Center, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital, Stockholm, Sweden
| | - Johan Nordström
- Division of Transplantation, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Annette Bruchfeld
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peter Barany
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kerstin Ebefors
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Erlandsson
- Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jaakko Patrakka
- KI/AZ Integrated Cardio Metabolic Center, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital, Stockholm, Sweden
| | - Peter Stenvinkel
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jenny Nyström
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Annika Wernerson
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Wang L, Han Y, Wang X. The relationship between plasma serglycin levels and the diagnosis of diabetic retinopathy. J Clin Lab Anal 2020; 35:e23663. [PMID: 33314317 PMCID: PMC7957973 DOI: 10.1002/jcla.23663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/04/2020] [Accepted: 11/10/2020] [Indexed: 12/28/2022] Open
Abstract
Background Diabetic retinopathy (DR), a microvascular complication which is closely related to diabetes, remains the leading cause of vision loss around the world among older adults. Serglycin (SRGN) was known as a hematopoietic cell granule proteoglycan, exerting its function in the formation of mast cell secretory granules and mediates the storage of various compounds in secretory vesicles. The present study illustrates the potential clinical value and experimental mechanisms of SRGN in the DR. Methods Firstly, the mRNA expression and protein expression of SRGN in plasma samples from NPDR, PDR patients, type 2 diabetes mellitus (T2Dm) cases, and healthy controls were measured by qPCR and Western blotting assays, respectively. Then, the potentials of SRGN functioning as a diagnostic indicator in DR were verified by the receiver operating characteristic (ROC) analysis. We established in vitro DR model of human retinal endothelial cells through high‐glucose treatment. The expression of SRGN and its mechanisms of regulating cellular processes were illustrated subsequently. Results Our data revealed that SRGN was dramatically upregulated in NPDR and PDR cases compared with healthy controls and T2DM patients; meanwhile, the expression of SRGN was further increased in the PDR group with regard to the NPDR group. Furthermore, the ROC analysis demonstrated that SRGN could distinguish the DR cases from type 2 diabetes mellitus (T2DM) patients and healthy controls with the area under the curve (AUC) of 0.7680 (95% CI = 0.6780 ~ 0.8576, sensitivity = 47.27%, specificity = 100%, cutoff value = 1.4727) and 0.8753 (95% CI = 0.8261 ~ 0.9244, sensitivity = 69.23%, specificity = 100%, cutoff value = 1.6923), respectively. In vitro high‐glucose treatment showed that the SRGN expressions were dramatically increased. The loss of SRGN could partially counteract the inhibition of HREC proliferation caused by high‐glucose stimulation. Meanwhile, SRGN knockdown could reverse the promotion of HREC apoptosis induced by high glucose as well. Conclusions Consequently, our study implied that SRGN might serve as a promising biomarker with high specificity and sensitivity in the DR diagnosis and progression.
Collapse
Affiliation(s)
| | - Yin Han
- Ningbo Eye Hospital, Ningbo, China
| | | |
Collapse
|
12
|
Zhang Y, Jiang H, Dou S, Zhang B, Qi X, Li J, Zhou Q, Li W, Chen C, Wang Q, Xie L. Comprehensive analysis of differentially expressed microRNAs and mRNAs involved in diabetic corneal neuropathy. Life Sci 2020; 261:118456. [PMID: 32956661 DOI: 10.1016/j.lfs.2020.118456] [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: 07/23/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 11/26/2022]
Abstract
AIMS Corneal nerve fibers are derived from the ophthalmic division of the trigeminal ganglion (TG). Here, by sequencing of microRNAs (miRNAs) and messenger RNAs (mRNAs) from diabetic and normal TG tissues, we aimed to uncover potential miRNAs, mRNAs, and the network of their interactions involved in the pathogenesis of diabetic corneal neuropathy. MAIN METHODS We performed RNA sequencing to systematically screen out differentially expressed miRNAs and mRNAs in TG tissues from diabetic and normal mice. Functional enrichment analyses were performed to illustrate the biological functions of differentially expressed mRNAs (DEmRNAs). Following this, miRNA-mRNA regulatory networks were built by means of bioinformatics methods to suggest regulatory role for miRNAs in the pathogenesis of diabetic corneal neuropathy. Finally, the credibility of the sequencing-based results was validated using qRT-PCR. KEY FINDINGS Sequencing analyses disclosed that 68 miRNAs and 114 mRNAs were differentially expressed in diabetic TG tissues compared with normal TG samples. The functional analyses showed that DEmRNAs participated in diabetes-related biological processes. After applying an optimized approach to predict miRNA-mRNA pairs, a miRNA-mRNA interacting network was inferred. Subsequently, the expression and correlation of miR-350-5p and Mup20, miR-592-5p and Angptl7 as well as miR-351-5p and Elovl6 were preliminarily validated. SIGNIFICANCE Our study provides a systematic characterization of miRNA and mRNA expression in the TG during diabetic corneal neuropathy and will contribute to the development of clinical diagnostic and therapeutic strategies for diabetic corneal neuropathy.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan 430060, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Hui Jiang
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao 266071, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Shengqian Dou
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao 266071, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Bin Zhang
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao 266071, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Xia Qi
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao 266071, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Jing Li
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Qingjun Zhou
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao 266071, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Weina Li
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan 430060, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Chen Chen
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Qun Wang
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao 266071, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China.
| | - Lixin Xie
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao 266071, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China.
| |
Collapse
|
13
|
Grujic M, Hellman L, Gustafson AM, Akula S, Melo FR, Pejler G. Protective role of mouse mast cell tryptase Mcpt6 in melanoma. Pigment Cell Melanoma Res 2020; 33:579-590. [PMID: 31894627 PMCID: PMC7317424 DOI: 10.1111/pcmr.12859] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 12/17/2019] [Accepted: 12/27/2019] [Indexed: 12/11/2022]
Abstract
Tryptase-positive mast cells populate melanomas, but it is not known whether tryptase impacts on melanoma progression. Here we addressed this and show that melanoma growth is significantly higher in tryptase-deficient (Mcpt6-/- ) versus wild-type mice. Histochemical analysis showed that mast cells were frequent in the tumor stroma of both wild-type and Mcpt6-/- mice, and also revealed their presence within the tumor parenchyma. Confocal microscopy analysis revealed that tryptase was taken up by the tumor cells. Further, tryptase-positive granules were released from mast cells and were widely distributed within the tumor tissue, suggesting that tryptase could impact on the tumor microenvironment. Indeed, gene expression analysis showed that the absence of Mcpt6 caused decreased expression of numerous genes, including Cxcl9, Tgtp2, and Gbp10, while the expression of 5p-miR3098 was enhanced. The levels of CXCL9 were lower in serum from Mcpt6-/- versus wild-type mice. In further support of a functional impact of tryptase on melanoma, recombinant tryptase (Mcpt6) was taken up by cultured melanoma cells and caused reduced proliferation. Altogether, our results indicate a protective role of mast cell tryptase in melanoma growth.
Collapse
Affiliation(s)
- Mirjana Grujic
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Lars Hellman
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Ann-Marie Gustafson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Srinivas Akula
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Fabio Rabelo Melo
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.,Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
14
|
Altered Expression of Long Noncoding and Messenger RNAs in Diabetic Nephropathy following Treatment with Rosiglitazone. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1360843. [PMID: 32025515 PMCID: PMC6983290 DOI: 10.1155/2020/1360843] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/09/2019] [Indexed: 02/07/2023]
Abstract
Diabetic nephropathy (DN) is characterized by metabolic disorder and inflammation. However, the regulatory effects that long noncoding RNAs (lncRNAs) have on the pathogenesis of DN and on the efficacy of rosiglitazone treatment have yet to be clearly defined. Herein, we performed unbiased RNA sequencing to characterize the transcriptomic profiles in db/db diabetic mouse model with or without rosiglitazone treatment that served to improve the phenotypes of DN. Moreover, RNA-seq profiling revealed that the development of DN caused an upregulation in the expression of 1176 mRNAs and a downregulation in the expression of 1010 mRNAs compared to controls, with the expression of 251 mRNAs being returned to normal following treatment with rosiglitazone. Further, 88 upregulated and 68 downregulated lncRNAs were identified in db/db mice compared to controls, 10 of which had their normal expression restored following treatment with rosiglitazone. Bioinformatic analysis revealed that the primary pathways involved in the pathogenesis of DN, and subsequently in the therapeutic effects of PPARγ, are related to inflammatory and metabolic processes. From bioinformatics analysis, lncRNA-AI838599 emerged as a novel molecular mechanism for rosiglitazone treatment in DN through TNFα-NFκb pathway. These findings may indicate a new molecular regulatory approach for the development of DN therapeutic agents.
Collapse
|
15
|
Lv J, Wu Y, Mai Y, Bu S. Noncoding RNAs in Diabetic Nephropathy: Pathogenesis, Biomarkers, and Therapy. J Diabetes Res 2020; 2020:3960857. [PMID: 32656264 PMCID: PMC7327582 DOI: 10.1155/2020/3960857] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/27/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022] Open
Abstract
The correlation between diabetes and systematic well-being on human life has long established. As a common complication of diabetes, the prevalence of diabetic nephropathy (DN) has been increasing globally. DN is known to be a major cause of end-stage kidney disease (ESKD). Till now, the molecular mechanisms for DN have not been fully explored and the effective therapies are still lacking. Noncoding RNAs are a class of RNAs produced by genome transcription that cannot be translated into proteins. It has been documented that ncRNAs participate in the pathogenesis of DN by regulating inflammation, apoptosis, autophagy, cell proliferation, and other pathological processes. In this review, the pathological roles and diagnostic and therapeutic potential of three types of ncRNAs (microRNA, long noncoding RNA, and circular RNA) in the progression of DN are summarized and illustrated.
Collapse
Affiliation(s)
- Jiarong Lv
- Diabetes Research Center, Medical School of Ningbo University, Ningbo, 315000 Zhejiang, China
| | - Yu Wu
- Diabetes Research Center, Medical School of Ningbo University, Ningbo, 315000 Zhejiang, China
| | - Yifeng Mai
- The Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315000 Zhejiang, China
| | - Shizhong Bu
- Diabetes Research Center, Medical School of Ningbo University, Ningbo, 315000 Zhejiang, China
| |
Collapse
|
16
|
Li W, Sargsyan D, Wu R, Li S, Wang L, Cheng D, Kong AN. DNA Methylome and Transcriptome Alterations in High Glucose-Induced Diabetic Nephropathy Cellular Model and Identification of Novel Targets for Treatment by Tanshinone IIA. Chem Res Toxicol 2019; 32:1977-1988. [PMID: 31525975 DOI: 10.1021/acs.chemrestox.9b00117] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Diabetic nephropathy (DN) is a diabetes complication that comes from overactivation of Renin-Angiotensin System, excessive pro-inflammatory factors, reactive oxygen species (ROS) overproduction, and potential epigenetic changes. Tanshinone IIA (TIIA), a diterpene quinone phytochemical, has been shown to possess powerful antioxidant, anti-inflammatory, epigenetics, and protective effects against different diseases including DN by inhibiting ROS induced by high glucose (HG). However, epigenomic and transcriptomic study of DN and the protective effect of TIIA are lacking. In this study, next-generation sequencing of RNA and DNA methylation profiles on the potential underlying mechanisms of a DN model in mouse kidney mesangial mes13 cells challenged with HG and treatment with TIIA were conducted. Bioinformatic analysis coupled with Ingenuity Pathway analysis of RNA-seq was performed, and 1780 genes from HG/LG and 1416 genes from TIIA/HG were significantly altered. Several pro-inflammatory pathways like leukotriene biosynthesis and eicosanoid signaling pathways were activated by HG stimulation, while TIIA treatment would enhance glutathione-mediated detoxification pathway to overcome the excess oxidative stress and inflammation triggered by HG. Combination analysis of RNA-seq and Methyl-seq data sets, DNA methylation, and RNA expression of a list of DN associated genes, Nmu, Fgl2, Glo, and Kcnip2, were found to be altered in HG-induced mes13 DN model, and TIIA treatment would effectively restore the alterations. Taken together, these findings provide novel insights into the understanding of how epigenetic/epigenomic modifications could affect the progression of DN and the potential preventive effect of TIIA in DN.
Collapse
Affiliation(s)
- Wenji Li
- Department of Pharmaceutics, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 160 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States.,Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, P. R. China,Jiangsu Key laboratory of integrated traditional Chinese and Western
Medicine for prevention and treatment of Senile Diseases, Yangzhou University, Yangzhou 225001, P. R. China
| | - Davit Sargsyan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 160 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States.,Graduate Program in Pharmaceutical Sciences , Ernest Mario School of Pharmacy, The State University of New Jersey , Piscataway , New Jersey 08854 , United States
| | - Renyi Wu
- Department of Pharmaceutics, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 160 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| | - Shanyi Li
- Department of Pharmaceutics, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 160 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States.,Graduate Program in Pharmaceutical Sciences , Ernest Mario School of Pharmacy, The State University of New Jersey , Piscataway , New Jersey 08854 , United States
| | - Lujing Wang
- Department of Pharmaceutics, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 160 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States.,Graduate Program in Pharmaceutical Sciences , Ernest Mario School of Pharmacy, The State University of New Jersey , Piscataway , New Jersey 08854 , United States
| | - David Cheng
- Department of Pharmaceutics, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 160 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States.,Graduate Program in Pharmaceutical Sciences , Ernest Mario School of Pharmacy, The State University of New Jersey , Piscataway , New Jersey 08854 , United States
| | - Ah-Ng Kong
- Department of Pharmaceutics, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 160 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| |
Collapse
|
17
|
Balmer LA, Whiting R, Rudnicka C, Gallo LA, Jandeleit KA, Chow Y, Chow Z, Richardson KL, Forbes JM, Morahan G. Genetic characterization of early renal changes in a novel mouse model of diabetic kidney disease. Kidney Int 2019; 96:918-926. [PMID: 31420193 DOI: 10.1016/j.kint.2019.04.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/09/2019] [Accepted: 04/22/2019] [Indexed: 01/13/2023]
Abstract
Genetic factors influence susceptibility to diabetic kidney disease. Here we mapped genes mediating renal hypertrophic changes in response to diabetes. A survey of 15 mouse strains identified variation in diabetic kidney hypertrophy. Strains with greater (FVB/N(FVB)) and lesser (C57BL/6 (B6)) responses were crossed and diabetic F2 progeny were characterized. Kidney weights of diabetic F2 mice were broadly distributed. Quantitative trait locus analyses revealed diabetic mice with kidney weights in the upper quartile shared alleles on chromosomes (chr) 6 and 12; these loci were designated as Diabetic kidney hypertrophy (Dkh)-1 and -2. To confirm these loci, reciprocal congenic mice were generated with defined FVB chromosome segments on the B6 strain background (B6.Dkh1/2f) or vice versa (FVB.Dkh1/2b). Diabetic mice of the B6.Dkh1/2f congenic strain developed diabetic kidney hypertrophy, while the reciprocal FVB.Dkh1/2b congenic strain was protected. The chr6 locus contained the candidate gene; Ark1b3, coding aldose reductase; the FVB allele has a missense mutation in this gene. Microarray analysis identified differentially expressed genes between diabetic B6 and FVB mice. Thus, since the two loci identified by quantitative trait locus mapping are syntenic with regions identified for human diabetic kidney disease, the congenic strains we describe provide a valuable new resource to study diabetic kidney disease and test agents that may prevent it.
Collapse
Affiliation(s)
- Lois A Balmer
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, the University of Western Australia, Perth, Western Australia, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup, Perth, Western Australia, Australia
| | - Rhiannon Whiting
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, the University of Western Australia, Perth, Western Australia, Australia
| | - Caroline Rudnicka
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, the University of Western Australia, Perth, Western Australia, Australia
| | - Linda A Gallo
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia; School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | | | - Yan Chow
- Glenferrie Private Hospital, Ramsay Health Care, Donvale, Victoria, Australia
| | - Zenia Chow
- ENT Doctors, Northpark Private Hospital, Bundoora, Victoria, Australia
| | - Kirsty L Richardson
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Western Australia, Australia
| | - Josephine M Forbes
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia; Mater Clinical School, University of Queensland, Brisbane, Queensland, Australia
| | - Grant Morahan
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, the University of Western Australia, Perth, Western Australia, Australia.
| |
Collapse
|
18
|
Lv X, He M, Zhao Y, Zhang L, Zhu W, Jiang L, Yan Y, Fan Y, Zhao H, Zhou S, Ma H, Sun Y, Li X, Xu H, Wei M. Identification of potential key genes and pathways predicting pathogenesis and prognosis for triple-negative breast cancer. Cancer Cell Int 2019; 19:172. [PMID: 31297036 PMCID: PMC6599314 DOI: 10.1186/s12935-019-0884-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023] Open
Abstract
Background Triple negative breast cancer (TNBC) is a specific subtype of breast cancer with a poor prognosis due to its aggressive biological behaviour and lack of therapeutic targets. We aimed to explore some novel genes and pathways related to TNBC prognosis through bioinformatics methods as well as potential initiation and progression mechanisms. Methods Breast cancer mRNA data were obtained from The Cancer Genome Atlas database (TCGA). Differential expression analysis of cancer and adjacent cancer, as well as, triple negative breast cancer and non-triple negative breast cancer were performed using R software. The key genes related to the pathogenesis were identified by functional and pathway enrichment analysis and protein-protein interaction network analysis. Based on univariate and multivariate Cox proportional hazards model analyses, a gene signature was established to predict overall survival. Receiver operating characteristic curve was used to evaluate the prognostic performance of our model. Results Based on mRNA expression profiling of breast cancer patients from the TCGA database, 755 differentially expressed overlapping mRNAs were detected between TNBC/non-TNBC samples and normal tissue. We found eight hub genes associated with the cell cycle pathway highly expressed in TNBC. Additionally, a novel six-gene (TMEM252, PRB2, SMCO1, IVL, SMR3B and COL9A3) signature from the 755 differentially expressed mRNAs was constructed and significantly associated with prognosis as an independent prognostic signature. TNBC patients with high-risk scores based on the expression of the 6-mRNAs had significantly shorter survival times compared to patients with low-risk scores (P < 0.0001). Conclusions The eight hub genes we identified might be tightly correlated with TNBC pathogenesis. The 6-mRNA signature established might act as an independent biomarker with a potentially good performance in predicting overall survival.
Collapse
Affiliation(s)
- Xuemei Lv
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Miao He
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Yanyun Zhao
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Liwen Zhang
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Wenjing Zhu
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Longyang Jiang
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Yuanyuan Yan
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Yue Fan
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Hongliang Zhao
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Shuqi Zhou
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Heyao Ma
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Yezhi Sun
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Xiang Li
- 3Department of Breast Cancer, Cancer Hospital of China Medical University, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042 China
| | - Hong Xu
- 3Department of Breast Cancer, Cancer Hospital of China Medical University, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042 China
| | - Minjie Wei
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| |
Collapse
|
19
|
Quantitative Trait Locus and Integrative Genomics Revealed Candidate Modifier Genes for Ectopic Mineralization in Mouse Models of Pseudoxanthoma Elasticum. J Invest Dermatol 2019; 139:2447-2457.e7. [PMID: 31207231 DOI: 10.1016/j.jid.2019.04.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/28/2019] [Accepted: 04/26/2019] [Indexed: 02/06/2023]
Abstract
Pseudoxanthoma elasticum, a prototype of heritable multisystem ectopic mineralization disorders, is caused by mutations in the ABCC6 gene encoding a putative efflux transporter, ABCC6. The phenotypic spectrum of pseudoxanthoma elasticum varies, and the correlation between genotype and phenotype has not been established. To identify genetic modifiers, we performed quantitative trait locus analysis in inbred mouse strains that carry the same hypomorphic allele in Abcc6 yet with highly variable ectopic mineralization phenotypes of pseudoxanthoma elasticum. Abcc6 was confirmed as a major determinant for ectopic mineralization in multiple tissues. Integrative analysis using functional genomics tools that included GeneWeaver, String, and Mouse Genome Informatics identified a total of nine additional candidate modifier genes that could influence the organ-specific ectopic mineralization phenotypes. Integration of the candidate genes into the existing ectopic mineralization gene network expands the current knowledge on the complexity of the network that, as a whole, governs ectopic mineralization in soft connective tissues.
Collapse
|
20
|
Park J, Guan Y, Sheng X, Gluck C, Seasock MJ, Hakimi AA, Qiu C, Pullman J, Verma A, Li H, Palmer M, Susztak K. Functional methylome analysis of human diabetic kidney disease. JCI Insight 2019; 4:128886. [PMID: 31167971 DOI: 10.1172/jci.insight.128886] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 04/23/2019] [Indexed: 12/16/2022] Open
Abstract
In patients with diabetes mellitus, poor metabolic control has a long-lasting impact on kidney disease development. Epigenetic changes, including cytosine methylation, have been proposed as potential mediators of the long-lasting effect of adverse metabolic events. Our understanding of the presence and contribution of methylation changes to disease development is limited because of the lack of comprehensive base-resolution methylome information of human kidney tissue samples and site-specific methylation editing. Base resolution, whole-genome bisulfite sequencing methylome maps of human diabetic kidney disease (DKD) tubule samples, and associated gene expression measured by RNA sequencing highlighted widespread methylation changes in DKD. Pathway analysis highlighted coordinated (methylation and gene expression) changes in immune signaling, including tumor necrosis factor alpha (TNF). Changes in TNF methylation correlated with kidney function decline. dCas9-Tet1-based lowering of the cytosine methylation level of the TNF differentially methylated region resulted in an increase in the TNF transcript level, indicating that methylation of this locus plays an important role in controlling TNF expression. Increasing the TNF level in diabetic mice increased disease severity, such as albuminuria. In summary, our results indicate widespread methylation differences in DKD kidneys and highlights epigenetic changes in the TNF locus and its contribution to the development of nephropathy in patients with diabetes mellitus.
Collapse
Affiliation(s)
- Jihwan Park
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yuting Guan
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xin Sheng
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Caroline Gluck
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Matthew J Seasock
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - A. Ari Hakimi
- Department of Surgery, Urology Service, Memorial Sloan Kettering Medical Center, New York, New York, USA
| | - Chengxiang Qiu
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James Pullman
- Department of Pathology, Montefiore Medical Center, New York, New York, USA
| | - Amit Verma
- Department of Oncology and Developmental Biology, Albert Einstein College of Medicine, New York, New York, USA
| | - Hongzhe Li
- Department of Epidemiology and Biostatistics and
| | - Matthew Palmer
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
Transcriptome Changes of Skeletal Muscle RNA-Seq Speculates the Mechanism of Postprandial Hyperglycemia in Diabetic Goto-Kakizaki Rats During the Early Stage of T2D. Genes (Basel) 2019; 10:genes10060406. [PMID: 31141985 PMCID: PMC6627578 DOI: 10.3390/genes10060406] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 12/14/2022] Open
Abstract
To address how skeletal muscle contributes to postprandial hyperglycemia, we performed skeletal muscle transcriptome analysis of diabetic Goto-Kakizaki (GK) and control Wistar rats by RNA sequencing (RNA-Seq). We obtained 600 and 1785 differentially expressed genes in GK rats compared to those Wistar rats at three and four weeks of age, respectively. Specifically, Tbc1d4, involved in glucose uptake, was significantly downregulated in the skeletal muscle of GK aged both three and four weeks compared to those of age-matched Wistar rats. Pdk4, related to glucose uptake and oxidation, was significantly upregulated in the skeletal muscle of GK aged both three and four weeks compared to that of age-matched Wistar rats. Genes (Acadl, Acsl1 and Fabp4) implicated in fatty acid oxidation were significantly upregulated in the skeletal muscle of GK aged four weeks compared to those of age-matched Wistar rats. The overexpression or knockout of Tbc1d4, Pdk4, Acadl, Acsl1 and Fabp4 has been reported to change glucose uptake and fatty acid oxidation directly in rodents. By taking the results of previous studies into consideration, we speculated that dysregulation of key dysregulated genes (Tbc1d4, Pdk4, Acadl, Acsl1 and Fabp4) may lead to a decrease in glucose uptake and oxidation, and an increase in fatty acid oxidation in GK skeletal muscle at three and four weeks, which may, in turn, contribute to postprandial hyperglycemia. Our research revealed transcriptome changes in GK skeletal muscle at three and four weeks. Tbc1d4, Acadl, Acsl1 and Fabp4 were found to be associated with early diabetes in GK rats for the first time, which may provide a new scope for pathogenesis of postprandial hyperglycemia.
Collapse
|
22
|
Bai W, Zhang C, Chen H. Transcriptomic analysis of Momordica charantia polysaccharide on streptozotocin-induced diabetic rats. Gene 2018; 675:208-216. [DOI: 10.1016/j.gene.2018.06.106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 06/11/2018] [Accepted: 06/29/2018] [Indexed: 10/28/2022]
|
23
|
Autophagy Is Indispensable for the Self-Renewal and Quiescence of Ovarian Cancer Spheroid Cells with Stem Cell-Like Properties. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7010472. [PMID: 30319732 PMCID: PMC6167563 DOI: 10.1155/2018/7010472] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/26/2018] [Accepted: 08/01/2018] [Indexed: 02/07/2023]
Abstract
Epithelial ovarian cancer has the highest mortality rate of all gynecologic cancers. Cancer stem cells are considered to be the initiating cells of tumors. It is known that spheroid culture promotes ovarian cancer cells to acquire stem cell characteristics and to become stem cell-like. But the mechanisms remain largely unclear. Our data show that autophagy is sustainably activated in ovarian cancer spheroid cells. Inhibition of autophagy by knockdown of ATG5 abolishes the self-renewal ability of ovarian cancer spheroid cells. Knockdown of ATG5 prevents ovarian cancer spheroid cells to enter quiescent state. Autophagy is critical for quiescent ovarian cancer spheroid cells to reenter the cell cycle because rapamycin can promote quiescent ovarian cancer spheroid cells to form colonies on soft agar and knockdown of ATG5 can arrest ovarian cancer cells in G0/G1. Autophagy and NRF2 form a positive feedback regulation loop to regulate reactive oxygen species (ROS) levels in ovarian cancer spheroid cells. The optimal ROS level, neither too high nor too low, facilitates the self-renewal marker, NOTCH1, to reach to the highest level. Bafilomycin A1 can impair the self-renewal of ovarian cancer spheroid cells by disturbing ROS levels.
Collapse
|
24
|
Ge Q, Zhang S, Chen L, Tang M, Liu L, Kang M, Gao L, Ma S, Yang Y, Lv P, Kong M, Yao Q, Feng F, Chen K. Mulberry Leaf Regulates Differentially Expressed Genes in Diabetic Mice Liver Based on RNA-Seq Analysis. Front Physiol 2018. [PMID: 30131712 DOI: 10.3389/fphys.2018.01051/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The pathogenesis of diabetes mellitus is a complicated process involving much gene regulation. The molecular mechanism of mulberry (Morus alba L.) leaf in the treatment of diabetes is not fully understood. In this study, we used the Illumina HiSeq™ 2,500 platform to explore the liver transcriptome of normal mice, STZ-induced diabetic mice, and mulberry leaf-treated diabetic mice, and we obtained 52,542,956, 52,626,414, and 52,780,196 clean reads, respectively. We identified differentially expressed genes (DEGs) during the pathogenesis of diabetes in mice. The functional properties of DEGs were characterized by comparison with the GO and KEGG databases, and the results show that DEGs are mainly involved in the metabolic pathway. qRT-PCR was used to analyse 27 differential genes involved in liver expression in different groups of diabetic mice. Among the DEGs, the expression of Scube1, Spns3, Ly6a, Igf2, and other genes between the control (C) and diabetic control (DC) groups was significantly upregulated; the expression of Grb10, Mup2, and Fasn was significantly downregulated; the expression of the Sqle, Lss, and Irs2 genes between the C group and diabetic group treated with mulberry (DD) was significantly upregulated; the expression of Fabp2, Ly6a, and Grb10 was significantly downregulated; and the expression of Sqle and Lss was significantly upregulated in the DC and DD groups, but Tap1, Igf2, and Spns3 were significantly downregulated. The results of Western blot validation showed that dynamic changes in proteins, such as IGF2, Ly6a, Grb10, and UBD, occurred to regulate the incidence of diabetes by influencing the insulin receptor substrate (IRS) signaling pathway.
Collapse
Affiliation(s)
- Qi Ge
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Shu Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Liang Chen
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Min Tang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Lanlan Liu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Mengna Kang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Lu Gao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Shangshang Ma
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yanhua Yang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Peng Lv
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Ming Kong
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Qin Yao
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Fan Feng
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Keping Chen
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| |
Collapse
|
25
|
Ge Q, Zhang S, Chen L, Tang M, Liu L, Kang M, Gao L, Ma S, Yang Y, Lv P, Kong M, Yao Q, Feng F, Chen K. Mulberry Leaf Regulates Differentially Expressed Genes in Diabetic Mice Liver Based on RNA-Seq Analysis. Front Physiol 2018; 9:1051. [PMID: 30131712 PMCID: PMC6090096 DOI: 10.3389/fphys.2018.01051] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 07/16/2018] [Indexed: 12/25/2022] Open
Abstract
The pathogenesis of diabetes mellitus is a complicated process involving much gene regulation. The molecular mechanism of mulberry (Morus alba L.) leaf in the treatment of diabetes is not fully understood. In this study, we used the Illumina HiSeq™ 2,500 platform to explore the liver transcriptome of normal mice, STZ-induced diabetic mice, and mulberry leaf-treated diabetic mice, and we obtained 52,542,956, 52,626,414, and 52,780,196 clean reads, respectively. We identified differentially expressed genes (DEGs) during the pathogenesis of diabetes in mice. The functional properties of DEGs were characterized by comparison with the GO and KEGG databases, and the results show that DEGs are mainly involved in the metabolic pathway. qRT-PCR was used to analyse 27 differential genes involved in liver expression in different groups of diabetic mice. Among the DEGs, the expression of Scube1, Spns3, Ly6a, Igf2, and other genes between the control (C) and diabetic control (DC) groups was significantly upregulated; the expression of Grb10, Mup2, and Fasn was significantly downregulated; the expression of the Sqle, Lss, and Irs2 genes between the C group and diabetic group treated with mulberry (DD) was significantly upregulated; the expression of Fabp2, Ly6a, and Grb10 was significantly downregulated; and the expression of Sqle and Lss was significantly upregulated in the DC and DD groups, but Tap1, Igf2, and Spns3 were significantly downregulated. The results of Western blot validation showed that dynamic changes in proteins, such as IGF2, Ly6a, Grb10, and UBD, occurred to regulate the incidence of diabetes by influencing the insulin receptor substrate (IRS) signaling pathway.
Collapse
Affiliation(s)
- Qi Ge
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Shu Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Liang Chen
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Min Tang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Lanlan Liu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Mengna Kang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Lu Gao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Shangshang Ma
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yanhua Yang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Peng Lv
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Ming Kong
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Qin Yao
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Fan Feng
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Keping Chen
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| |
Collapse
|
26
|
Lyu LL, Feng Y, Liu BC. Urinary Biomarkers for Chronic Kidney Disease with a Focus on Gene Transcript. Chin Med J (Engl) 2018; 130:2251-2256. [PMID: 28875962 PMCID: PMC5598339 DOI: 10.4103/0366-6999.213965] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective: In the upcoming era of precision medicine, searching for the early, noninvasive biomarkers has been the cornerstone and major challenge in the management of chronic kidney disease (CKD). Urine contains rich biological information which could be the ideal source for noninvasive biomarkers of CKD. This review will discuss the recent advance in urinary biomarker. Data Sources: This review was based on data in articles published in the PubMed databases up to June 20, 2017, with the following keywords: “Chronic kidney disease”, “Biomarker”, and “Urine”. Study Selection: Original articles and important reviews on urinary biomarker were selected for this review. Results: Urinary biomarker studies of CKD mainly focused on urine sediment, supernatant, and urinary extracellular vesicles. The gene transcript (microRNA [miRNA], messenger RNA [mRNA]) biomarkers have been recently shown with diagnostic potential for CKD reflecting kidney function and histological change. However, challenges regarding technique and data analysis need to be resolved before translation to clinic. Conclusions: Different fractions of urine contain rich information for biomarker discovery, among which urine (extracellular vesicles) mRNA, miRNA, might represent promising biomarker for CKD.
Collapse
Affiliation(s)
- Lin-Li Lyu
- Department of Nephrology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, China
| | - Ye Feng
- Department of Nephrology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, China
| | - Bi-Cheng Liu
- Department of Nephrology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
27
|
Halperin Kuhns VL, Pluznick JL. Novel differences in renal gene expression in a diet-induced obesity model. Am J Physiol Renal Physiol 2017; 314:F517-F530. [PMID: 29141937 DOI: 10.1152/ajprenal.00345.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Obesity is a significant risk factor for both chronic kidney disease and end-stage renal disease. To better understand disease development, we sought to identify novel genes differentially expressed early in disease progression. We first confirmed that mice fed a high-fat (HF) diet exhibit early signs of renal injury including hyperfiltration. We then performed RNA-Seq using renal cortex RNA from C57BL6/J male mice fed either HF or control (Ctrl) diet. We identified 1,134 genes differentially expressed in the cortex on HF vs. Ctrl, of which 31 genes were selected for follow-up analysis. This included the 9 most upregulated, the 11 most downregulated, and 11 genes of interest (primarily sensory receptors and G proteins). Quantitative (q)RT-PCR for these 31 genes was performed on additional male renal cortex and medulla samples, and 11 genes (including all 9 upregulated genes) were selected for further study based on qRT-PCR. We then examined expression of these 11 genes in Ctrl and HF male heart and liver samples, which demonstrated that these changes are relatively specific to the renal cortex. These 11 genes were also examined in female renal cortex, where we found that the expression changes seen in males on a HF diet are not replicated in females, even when the females are started on the diet sooner to match weight gain of the males. In sum, these data demonstrate that in a HF-diet model of early disease, novel transcriptional changes occur that are both sex specific and specific to the renal cortex.
Collapse
Affiliation(s)
| | - Jennifer L Pluznick
- Department of Physiology, Johns Hopkins University School of Medicine , Baltimore, Maryland
| |
Collapse
|