1
|
Jia S, Si R, Liu G, Zhong Q. Diosgenin protects against cationic bovine serum albumin-induced membranous glomerulonephritis by attenuating oxidative stress and renal inflammation via the NF-κB pathway. PHARMACEUTICAL BIOLOGY 2024; 62:285-295. [PMID: 38516898 PMCID: PMC10962310 DOI: 10.1080/13880209.2024.2330602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 03/10/2024] [Indexed: 03/23/2024]
Abstract
CONTEXT Membranous glomerulonephritis (MGN) is a leading cause of nephrotic syndrome in adults. Diosgenin (DG) has been reported to exert antioxidative and anti-inflammatory effects. OBJECTIVE To investigate the renoprotective activity of DG in a cationic bovine serum albumin-induced rat model of MGN. MATERIALS AND METHODS Fourty male Sprague-Dawley rats were randomized into four groups. The MGN model was established and treated with a DG dose (10 mg/kg) and a positive control (TPCA1, 10 mg/kg), while normal control and MGN groups received distilled water by gavage for four consecutive weeks. At the end of the experiment, 24 h urinary protein, biochemical indices, oxidation and antioxidant levels, inflammatory parameters, histopathological examination, immunohistochemistry and immunoblotting were evaluated. RESULTS DG significantly ameliorated kidney dysfunction by decreasing urinary protein (0.56-fold), serum creatinine (SCr) (0.78-fold), BUN (0.71-fold), TC (0.66-fold) and TG (0.73-fold) levels, and increasing ALB (1.44-fold). DG also reduced MDA (0.82-fold) and NO (0.83-fold) levels while increasing the activity of SOD (1.56-fold), CAT (1.25-fold), glutathione peroxidase (GPx) (1.55-fold) and GSH (1.81-fold). Furthermore, DG reduced Keap1 (0.76-fold) expression, Nrf2 nuclear translocation (0.79-fold), and induced NQO1 (1.25-fold) and HO-1 (1.46-fold) expression. Additionally, DG decreased IL-2 (0.55-fold), TNF-α (0.80-fold) and IL-6 (0.75-fold) levels, and reduced protein expression of NF-κB p65 (0.80-fold), IKKβ (0.93-fold), p-IKKβ (0.89-fold), ICAM-1 (0.88-fold), VCAM-1 (0.91-fold), MCP-1 (0.88-fold) and E-selectin (0.87-fold), and also inhibited the nuclear translocation of NF-κB p65 (0.64-fold). DISCUSSION AND CONCLUSIONS The results suggest a potential therapeutic benefit of DG against MGN due to the inhibition of the NF-κB pathway, supporting the need for further clinical trials.
Collapse
Affiliation(s)
- Shiyan Jia
- Department of Anesthesiology, Anesthesia and Trauma Research Unit, Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, China
- College of Basic Medical Sciences, Shanxi University of Chinese Medicine, Jinzhong, China
- Department of Nephrology, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China
| | - Ruihua Si
- College of Basic Medical Sciences, Shanxi University of Chinese Medicine, Jinzhong, China
- Department of Nephrology, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China
| | - Guangzhen Liu
- Department of Nephrology, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China
| | - Qiming Zhong
- Department of Nephrology, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China
| |
Collapse
|
2
|
Montenegro F, Giannuzzi F, Picerno A, Cicirelli A, Stea ED, Di Leo V, Sallustio F. How Stem and Progenitor Cells Can Affect Renal Diseases. Cells 2024; 13:1460. [PMID: 39273032 PMCID: PMC11393889 DOI: 10.3390/cells13171460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Stem and progenitor cells have been observed to contribute to regenerative processes in acute renal failure and chronic kidney disease. Recent research has delved into the intricate mechanisms by which stem and progenitor cells exert their influence on kidney diseases. Understanding how these cells integrate with the existing renal architecture and their response to injury could pave the way for innovative treatment strategies aimed at promoting kidney repair and regeneration. Overall, the role of stem and progenitor cells in kidney diseases is multifaceted, with their ability to contribute to tissue regeneration, immune modulation, and the maintenance of renal homeostasis. Here, we review the studies that we have available today about the involvement of stem and progenitor cells both in regenerative therapies and in the causes of renal diseases, as well as in natural healing mechanisms, taking into account the main kidney disorders, such as IgA nephropathy, lupus nephritis, diabetic nephropathy, C3 glomerulopathy, focal segmental glomerulosclerosis, idiopathic membranous nephropathy, anti-glomerular basement membrane glomerulonephritis, and ANCA-associated crescentic glomerulonephritis. Moreover, based on the comprehensive data available in the framework of the specific kidney diseases on stem cells and renal progenitors, we hypothesize a possible role of adult renal progenitors in exacerbating or recovering the illness.
Collapse
Affiliation(s)
- Francesca Montenegro
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Francesca Giannuzzi
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Angela Picerno
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Antonella Cicirelli
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Emma Diletta Stea
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Vincenzo Di Leo
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Fabio Sallustio
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
3
|
Wu H, Yao Z, Li H, Zhang L, Zhao Y, Li Y, Wu Y, Zhang Z, Xie J, Ding F, Zhu H. Improving dermal fibroblast-to-epidermis communications and aging wound repair through extracellular vesicle-mediated delivery of Gstm2 mRNA. J Nanobiotechnology 2024; 22:307. [PMID: 38825668 PMCID: PMC11145791 DOI: 10.1186/s12951-024-02541-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 05/09/2024] [Indexed: 06/04/2024] Open
Abstract
Skin aging is characterized by the disruption of skin homeostasis and impaired skin injury repair. Treatment of aging skin has long been limited by the unclear intervention targets and delivery techniques. Engineering extracellular vesicles (EVs) as an upgraded version of natural EVs holds great potential in regenerative medicine. In this study, we found that the expression of the critical antioxidant and detoxification gene Gstm2 was significantly reduced in aging skin. Thus, we constructed the skin primary fibroblasts-derived EVs encapsulating Gstm2 mRNA (EVsGstm2), and found that EVsGstm2 could significantly improve skin homeostasis and accelerate wound healing in aged mice. Mechanistically, we found that EVsGstm2 alleviated oxidative stress damage of aging dermal fibroblasts by modulating mitochondrial oxidative phosphorylation, and promoted dermal fibroblasts to regulate skin epidermal cell function by paracrine secretion of Nascent Polypeptide-Associated Complex Alpha subunit (NACA). Furthermore, we confirmed that NACA is a novel skin epidermal cell protective molecule that regulates skin epidermal cell turnover through the ROS-ERK-ETS-Cyclin D pathway. Our findings demonstrate the feasibility and efficacy of EVs-mediated delivery of Gstm2 for aged skin treatment and unveil novel roles of GSTM2 and NACA for improving aging skin.
Collapse
Affiliation(s)
- Haiyan Wu
- Institute for Regenerative Medicine & Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zuochao Yao
- Department of Plastic and Reconstructive Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Hongkun Li
- Department of Cardiology, Changzhi Medical College Affiliated Heji Hospital, Shanxi, 046000, China
| | - Laihai Zhang
- Department of Cardiothoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yuying Zhao
- Institute for Regenerative Medicine & Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yongwei Li
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yating Wu
- Institute for Regenerative Medicine & Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zhenchun Zhang
- Institute for Regenerative Medicine & Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Jiali Xie
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Feixue Ding
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People Hospital, School of Medicine, JiaoTong University, Shanghai, 200001, China
| | - Hongming Zhu
- Institute for Regenerative Medicine & Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
4
|
Wu H, Zhai Y, Yu J, Wei L, Qi X. Transcriptome and proteome analyses reveal that upregulation of GSTM2 by allisartan improves cardiac remodeling and dysfunction in hypertensive rats. Exp Ther Med 2024; 27:220. [PMID: 38590561 PMCID: PMC11000455 DOI: 10.3892/etm.2024.12508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 02/20/2024] [Indexed: 04/10/2024] Open
Abstract
Long-term hypertension can lead to hypertensive heart disease, which ultimately progresses to heart failure. As an angiotensin receptor blocker antihypertensive drug, allisartan can control blood pressure, and improve cardiac remodeling and cardiac dysfunction caused by hypertension. The aim of the present study was to investigate the protective effects of allisartan on the heart of spontaneously hypertensive rats (SHRs) and the underlying mechanisms. SHRs were used as an animal model of hypertensive heart disease and were treated with allisartan orally at a dose of 25 mg/kg/day. The blood pressure levels of the rats were continuously monitored, their body and heart weights were measured, and their cardiac structure and function were evaluated using echocardiography. Wheat germ agglutinin staining and Masson trichrome staining were employed to assess the morphology of the myocardial tissue. In addition, transcriptome and proteome analyses were performed using the Solexa/Illumina sequencing platform and tandem mass tag technology, respectively. Immunofluorescence co-localization was conducted to analyze Nrf2 nuclear translocation, and TUNEL was performed to detect the levels of cell apoptosis. The protein expression levels of pro-collagen I, collagen III, phosphorylated (p)-AKT, AKT, p-PI3K and PI3K, and the mRNA expression levels of Col1a1 and Col3a1 were determined by western blotting and reverse transcription-quantitative PCR, respectively. Allisartan lowered blood pressure, attenuated cardiac remodeling and improved cardiac function in SHRs. In addition, allisartan alleviated cardiomyocyte hypertrophy and cardiac fibrosis. Allisartan also significantly affected the 'pentose phosphate pathway', 'fatty acid elongation', 'valine, leucine and isoleucine degradation', 'glutathione metabolism', and 'amino sugar and nucleotide sugar metabolism' pathways in the hearts of SHRs, and upregulated the expression levels of GSTM2. Furthermore, allisartan activated the PI3K-AKT-Nrf2 signaling pathway and inhibited cardiomyocyte apoptosis. In conclusion, the present study demonstrated that allisartan can effectively control blood pressure in SHRs, and improves cardiac remodeling and cardiac dysfunction. Allisartan may also upregulate the expression levels of GSTM2 in the hearts of SHRs and significantly affect glutathione metabolism, as determined by transcriptome and proteome analyses. The cardioprotective effect of allisartan may be mediated through activation of the PI3K-AKT-Nrf2 signaling pathway, upregulation of GSTM2 expression and reduction of cardiomyocyte apoptosis in SHRs.
Collapse
Affiliation(s)
- Hao Wu
- School of Medicine, Nankai University, Tianjin 300071, P.R. China
- Department of Cardiology, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Yajun Zhai
- Graduate School, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Jing Yu
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Liping Wei
- School of Medicine, Nankai University, Tianjin 300071, P.R. China
- Department of Cardiology, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Xin Qi
- School of Medicine, Nankai University, Tianjin 300071, P.R. China
- Department of Cardiology, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| |
Collapse
|
5
|
de Souza W, Gemini-Piperni S, Grenho L, Rocha LA, Granjeiro JM, Melo SA, Fernandes MH, Ribeiro AR. Titanium dioxide nanoparticles affect osteoblast-derived exosome cargos and impair osteogenic differentiation of human mesenchymal stem cells. Biomater Sci 2023; 11:2427-2444. [PMID: 36756939 DOI: 10.1039/d2bm01854c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Titanium (Ti) and its alloys are the most widely used metallic biomaterials in total joint replacement; however, increasing evidence supports the degradation of its surface due to corrosion and wear processes releasing debris (ions, and micro and nanoparticles) and contribute to particle-induced osteolysis and implant loosening. Cell-to-cell communication involving several cell types is one of the major biological processes occurring during bone healing and regeneration at the implant-bone interface. In addition to the internal response of cells to the uptake and intracellular localization of wear debris, a red flag is the ability of titanium dioxide nanoparticles (mimicking wear debris) to alter cellular communication with the tissue background, disturbing the balance between osseous tissue integrity and bone regenerative processes. This study aims to understand whether titanium dioxide nanoparticles (TiO2 NPs) alter osteoblast-derived exosome (Exo) biogenesis and whether exosomal protein cargos affect the communication of osteoblasts with human mesenchymal stem/stromal cells (HMSCs). Osteoblasts are derived from mesenchymal stem cells coexisting in the bone microenvironment during development and remodelling. We observed that TiO2 NPs stimulate immature osteoblast- and mature osteoblast-derived Exo secretion that present a distinct proteomic cargo. Functional tests confirmed that Exos derived from both osteoblasts decrease the osteogenic differentiation of HMSCs. These findings are clinically relevant since wear debris alter extracellular communication in the bone periprosthetic niche, contributing to particle-induced osteolysis and consequent prosthetic joint failure.
Collapse
Affiliation(s)
- Wanderson de Souza
- Directory of Metrology Applied to Life Sciences, National Institute of Metrology Quality and Technology, Rio de Janeiro, Brazil.,Postgraduate Program in Biotechnology, National Institute of Metrology Quality and Technology, Rio de Janeiro, Brazil
| | - S Gemini-Piperni
- Postgraduate Program in Biotechnology, National Institute of Metrology Quality and Technology, Rio de Janeiro, Brazil.,Postgraduate Program in Translational Biomedicine, University Grande Rio, Duque de Caxias, Brazil.,Lab∈n Group, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil
| | - Liliana Grenho
- Faculty of Dental Medicine, University of Porto, Porto, Portugal.,LAQV/REQUIMTE, University of Porto, Porto, Portugal
| | - Luís A Rocha
- Physics Department, Paulista State University, São Paulo, Brazil.,IBTN/Br - Brazilian Branch of the Institute of Biomaterials, Tribocorrosion and Nanomedicine, São Paulo State University, Bauru, São Paulo, Brazil
| | - José M Granjeiro
- Directory of Metrology Applied to Life Sciences, National Institute of Metrology Quality and Technology, Rio de Janeiro, Brazil.,Postgraduate Program in Biotechnology, National Institute of Metrology Quality and Technology, Rio de Janeiro, Brazil.,Postgraduate Program in Translational Biomedicine, University Grande Rio, Duque de Caxias, Brazil.,Dental School, Fluminense Federal University, Niterói, Brazil
| | - Sonia A Melo
- i3S-Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Maria H Fernandes
- Faculty of Dental Medicine, University of Porto, Porto, Portugal.,LAQV/REQUIMTE, University of Porto, Porto, Portugal
| | - Ana R Ribeiro
- Postgraduate Program in Biotechnology, National Institute of Metrology Quality and Technology, Rio de Janeiro, Brazil.,NanoSafety group, International Iberian Nanotechnology Laboratory - INL, 4715-330, Braga, Portugal.
| |
Collapse
|
6
|
Zhang J, Li Y, Zou J, Lai CT, Zeng T, Peng J, Zou WD, Cao B, Liu D, Zhu LY, Li H, Li YK. Comprehensive analysis of the glutathione S-transferase Mu (GSTM) gene family in ovarian cancer identifies prognostic and expression significance. Front Oncol 2022; 12:968547. [PMID: 35965498 PMCID: PMC9366399 DOI: 10.3389/fonc.2022.968547] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/04/2022] [Indexed: 12/11/2022] Open
Abstract
Background Ovarian cancer (OC) is one of the most common types of gynecologic tumor over the world. The Glutathione S-transferase Mu (GSTM) has five members, including GSTM1-5. These GSTMs is involved in cell metabolism and detoxification, but their role in OC remains unknown. Methods Data from multiple public databases associated with OC and GSTMs were collected. Expression, prognosis, function enrichment, immune infiltration, stemness index, and drug sensitivity analysis was utilized to identify the roles of GSTMs in OC progression. RT-qPCR analysis confirmed the effect of AICAR, AT-7519, PHA-793887 and PI-103 on the mRNA levels of GSTM3/4. Results GSTM1-5 were decreased in OC samples compared to normal ovary samples. GSTM1/5 were positively correlated with OC prognosis, but GSTM3 was negatively correlated with OC prognosis. Function enrichment analysis indicated GSTMs were involved in glutathione metabolism, drug metabolism, and drug resistance. Immune infiltration analysis indicated GSTM2/3/4 promoted immune escape in OC. GSTM5 was significantly correlated with OC stemness index. GSTM3/4 were remarkedly associated with OC chemoresistance, especially in AICAR, AT-7519, PHA-793887 and PI-103. Conclusion GSTM3 was negatively correlated with OC prognosis, and associated with OC chemoresistance and immune escape. This gene may serve as potential prognostic biomarkers and therapeutic target for OC patients.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Assisted Reproductive Centre, Zhuzhou central hospital, Xiangya hospital Zhuzhou central south university, Central south university, Zhuzhou, China
| | - Yan Li
- Department of Assisted Reproductive Centre, Zhuzhou central hospital, Xiangya hospital Zhuzhou central south university, Central south university, Zhuzhou, China
| | - Juan Zou
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
| | - Chun-tian Lai
- Department of Assisted Reproductive Centre, Zhuzhou central hospital, Xiangya hospital Zhuzhou central south university, Central south university, Zhuzhou, China
| | - Tian Zeng
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
| | - Juan Peng
- Department of Assisted Reproductive Centre, Zhuzhou central hospital, Xiangya hospital Zhuzhou central south university, Central south university, Zhuzhou, China
| | - Wen-da Zou
- Department of Assisted Reproductive Centre, Zhuzhou central hospital, Xiangya hospital Zhuzhou central south university, Central south university, Zhuzhou, China
| | - Bei Cao
- Department of Assisted Reproductive Centre, Zhuzhou central hospital, Xiangya hospital Zhuzhou central south university, Central south university, Zhuzhou, China
| | - Dan Liu
- Department of Assisted Reproductive Centre, Zhuzhou central hospital, Xiangya hospital Zhuzhou central south university, Central south university, Zhuzhou, China
| | - Li-yu Zhu
- Department of Assisted Reproductive Centre, Zhuzhou central hospital, Xiangya hospital Zhuzhou central south university, Central south university, Zhuzhou, China
| | - Hui Li
- Department of Assisted Reproductive Centre, Zhuzhou central hospital, Xiangya hospital Zhuzhou central south university, Central south university, Zhuzhou, China
- *Correspondence: Hui Li, ; Yu-kun Li,
| | - Yu-kun Li
- Department of Assisted Reproductive Centre, Zhuzhou central hospital, Xiangya hospital Zhuzhou central south university, Central south university, Zhuzhou, China
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
- *Correspondence: Hui Li, ; Yu-kun Li,
| |
Collapse
|
7
|
Quaglia M, Merlotti G, Fornara L, Colombatto A, Cantaluppi V. Extracellular Vesicles Released from Stem Cells as a New Therapeutic Strategy for Primary and Secondary Glomerulonephritis. Int J Mol Sci 2022; 23:ijms23105760. [PMID: 35628570 PMCID: PMC9142886 DOI: 10.3390/ijms23105760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/13/2022] [Accepted: 05/20/2022] [Indexed: 12/04/2022] Open
Abstract
Current treatment of primary and secondary glomerulopathies is hampered by many limits and a significant proportion of these disorders still evolves towards end-stage renal disease. A possible answer to this unmet challenge could be represented by therapies with stem cells, which include a variety of progenitor cell types derived from embryonic or adult tissues. Stem cell self-renewal and multi-lineage differentiation ability explain their potential to protect and regenerate injured cells, including kidney tubular cells, podocytes and endothelial cells. In addition, a broad spectrum of anti-inflammatory and immunomodulatory actions appears to interfere with the pathogenic mechanisms of glomerulonephritis. Of note, mesenchymal stromal cells have been particularly investigated as therapy for Lupus Nephritis and Diabetic Nephropathy, whereas initial evidence suggest their beneficial effects in primary glomerulopathies such as IgA nephritis. Extracellular vesicles mediate a complex intercellular communication network, shuttling proteins, nucleic acids and other bioactive molecules from origin to target cells to modulate their functions. Stem cell-derived extracellular vesicles recapitulate beneficial cytoprotective, reparative and immunomodulatory properties of parental cells and are increasingly recognized as a cell-free alternative to stem cell-based therapies for different diseases including glomerulonephritis, also considering the low risk for potential adverse effects such as maldifferentiation and tumorigenesis. We herein summarize the renoprotective potential of therapies with stem cells and extracellular vesicles derived from progenitor cells in glomerulonephritis, with a focus on their different mechanisms of actions. Technological progress and growing knowledge are paving the way for wider clinical application of regenerative medicine to primary and secondary glomerulonephritis: this multi-level, pleiotropic therapy may open new scenarios overcoming the limits and side effects of traditional treatments, although the promising results of experimental models need to be confirmed in the clinical setting.
Collapse
|
8
|
Chen P, Wu H, Yao H, Zhang J, Fan W, Chen Z, Su W, Wang Y, Li P. Multi-Omics Analysis Reveals the Systematic Relationship Between Oral Homeostasis and Chronic Sleep Deprivation in Rats. Front Immunol 2022; 13:847132. [PMID: 35432311 PMCID: PMC9009293 DOI: 10.3389/fimmu.2022.847132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
Sleep disorders were associated with oral health. Inflammation has especially been thought to be a key factor in linking oral diseases and sleep deficiency. However, how chronic sleep deprivation (CSD) affects oral homeostasis, particularly oral inflammation and oral microbiota, is still unknown. This study aimed to uncover the systematic relationship between oral homeostasis and CSD in rats. The metabolomics in serum, proteomics in the tongue tissues, and microbiome analysis in the oral cavity in CSD rats were performed. Multi-omics data integration analysis was performed to uncover the systematic relationship between oral homeostasis and CSD through the weighted correlation network analysis. We found that CSD could lead to oral inflammation in rats. CSD significantly increased systemic inflammation by enhancing the serum levels of IL-1β, IL-6 and inhibiting the serum level of IL-10. Serum levels of adrenocorticotropin hormone, corticosterone, and triiodothyronine were increased in CSD rats, and the steroid hormone biosynthesis pathway was also found to be involved in the perturbation resulting from CSD, together suggesting the activation of the hypothalamic-pituitary-adrenocortical and hypothalamic‐pituitary‐thyroid axis. CSD led to changes of oral microbiota composition, and g_Acinetobacter, Candidatus Chryseobacterium massiliae, and g_Moraxella were significantly correlated with multiple proteins in bacterial invasion of epithelial cells pathway, which may partially responsible for oral inflammation resulting from CSD. The changes of proteomic profiling expression caused by CSD in tongue tissues were mainly enriched in neurodegenerative diseases pathways and immune/inflammation-related pathways. Multi-omics analysis indicated that the inflammatory response-related modules were significantly correlated with the neurodegenerative disease-related module suggesting a possible link between neurodegenerative diseases and oral inflammation. Together, CSD induced oral inflammation and subtle changes on oral microbiota. Our study is helpful to further understand the role that oral homeostasis plays in the process by which CSD affects human health and disease.
Collapse
Affiliation(s)
- Pan Chen
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed Traditional Chinese Medicine (TCM), State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hao Wu
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed Traditional Chinese Medicine (TCM), State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hongliang Yao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
| | - Jiashuo Zhang
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed Traditional Chinese Medicine (TCM), State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Weiyang Fan
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed Traditional Chinese Medicine (TCM), State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhen Chen
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed Traditional Chinese Medicine (TCM), State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Weiwei Su
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed Traditional Chinese Medicine (TCM), State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yonggang Wang
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed Traditional Chinese Medicine (TCM), State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Peibo Li
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed Traditional Chinese Medicine (TCM), State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
9
|
Jin Y, Tan Y, Zhao P, Guo Y, Chen S, Wu J, Ren Z. Glutathione S-transferase Mu 2 inhibits hepatic steatosis via ASK1 suppression. Commun Biol 2022; 5:326. [PMID: 35388144 PMCID: PMC8986781 DOI: 10.1038/s42003-022-03251-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 03/11/2022] [Indexed: 12/31/2022] Open
Abstract
Hepatic steatosis is the main characteristic of some liver metabolism diseases. However, unclear molecular mechanism of hepatic steatosis impedes the therapy of this hepatic steatosis. Glutathione-S-transferase mu 2 (GSTM2), as a member of phase II drug metabolizing enzymes (DMEs), regulates cellular antioxidant and detoxificant. GSTM2 was highly up-regulated in hepatic steatosis tissues and high-fat diet (HFD) fed mice. Loss-of-function GSTM2 mouse model demonstrated that GSTM2 protected mice from excess fat accumulation. Mechanistically, GSTM2 interacted with ASK1 and suppressed its phosphorylation and the activation of subsequent downstream p38-JNK signalling. Moreover, GSTM2 overexpression in the liver effectively ameliorated hepatic lipid accumulation. Therefore, we identified GSTM2 as an important negative regulator in progression of hepatic steatosis via both its detoxification/antioxidant and inhibition of ASK1-p38/JNK signalling. This study showed potential therapeutic function of the DME in progression of hepatic steatosis. Jin et al. investigate the underlying mechanisms of hepatic steatosis and show that Glutathione-S-transferase mu 2 (GSTM2), which is a drug metabolizing enzyme (DME), is upregulated in hepatic steatosis tissue from mice fed with a high fat diet. They show that GSTM2 is a negative regulator of hepatic steatosis via both detoxification/antioxidant and inhibition of ASK1-p38/JNK signalling, which sheds light on its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yi Jin
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China.,Hubei Hongshan Laboratory, Wuhan, Hubei, 430070, PR China
| | - Yanjie Tan
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China
| | - Pengxiang Zhao
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China
| | - Yu Guo
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China
| | - Shilin Chen
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China
| | - Jian Wu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China.
| | - Zhuqing Ren
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China. .,Hubei Hongshan Laboratory, Wuhan, Hubei, 430070, PR China.
| |
Collapse
|
10
|
Lan T, Hu Y, Hu F, Li H, Chen Y, Zhang J, Yu Y, Jiang S, Weng Q, Tian S, Ma T, Yang G, Luo D, Wang L, Li K, Piao S, Rong X, Guo J. Hepatocyte glutathione S-transferase mu 2 prevents non-alcoholic steatohepatitis by suppressing ASK1 signaling. J Hepatol 2022; 76:407-419. [PMID: 34656650 DOI: 10.1016/j.jhep.2021.09.040] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 08/10/2021] [Accepted: 09/23/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND & AIMS Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide. The advanced stage of NAFLD, non-alcoholic steatohepatitis (NASH), has been recognized as a leading cause of end-stage liver injury for which there are no FDA-approved therapeutic options. Glutathione S-transferase Mu 2 (GSTM2) is a phase II detoxification enzyme. However, the roles of GSTM2 in NASH have not been elucidated. METHODS Multiple RNA-seq analyses were used to identify hepatic GSTM2 expression in NASH. In vitro and in vivo gain- or loss-of-function approaches were used to investigate the role and molecular mechanism of GSTM2 in NASH. RESULTS We identified GSTM2 as a sensitive responder and effective suppressor of NASH progression. GSTM2 was significantly downregulated during NASH progression. Hepatocyte GSTM2 deficiency markedly aggravated insulin resistance, hepatic steatosis, inflammation and fibrosis induced by a high-fat diet and a high-fat/high-cholesterol diet. Mechanistically, GSTM2 sustained MAPK pathway signaling by directly interacting with apoptosis signal-regulating kinase 1 (ASK1). GSTM2 directly bound to the N-terminal region of ASK1 and inhibited ASK1 N-terminal dimerization to subsequently repress ASK1 phosphorylation and the activation of its downstream JNK/p38 signaling pathway under conditions of metabolic dysfunction. CONCLUSIONS These data demonstrated that hepatocyte GSTM2 is an endogenous suppressor that protects against NASH progression by blocking ASK1 N-terminal dimerization and phosphorylation. Activating GSTM2 holds promise as a therapeutic strategy for NASH. CLINICAL TRIAL NUMBER IIT-2021-277. LAY SUMMARY New therapeutic strategies for non-alcoholic steatohepatitis are urgently needed. We identified that the protein GSTM2 exerts a protective effect in response to metabolic stress. Therapies that aim to increase the activity of GSTM2 could hold promise for the treatment of non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Tian Lan
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Yufeng Hu
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Fengjiao Hu
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Haonan Li
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Yinghua Chen
- Organ Transplant, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Jing Zhang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Yang Yu
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Shuo Jiang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Qiqing Weng
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Song Tian
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Tengfei Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guizhi Yang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Duosheng Luo
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Lexun Wang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Kunping Li
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Shenghua Piao
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Xianglu Rong
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Jiao Guo
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China.
| |
Collapse
|
11
|
Landi C, Liberatori G, Cotugno P, Sturba L, Vannuccini ML, Massari F, Miniero DV, Tursi A, Shaba E, Behnisch PA, Carleo A, Di Giuseppe F, Angelucci S, Bini L, Corsi I. First Attempt to Couple Proteomics with the AhR Reporter Gene Bioassay in Soil Pollution Monitoring and Assessment. TOXICS 2021; 10:toxics10010009. [PMID: 35051051 PMCID: PMC8779689 DOI: 10.3390/toxics10010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 11/16/2022]
Abstract
A topsoil sample obtained from a highly industrialized area (Taranto, Italy) was tested on the DR-CALUX® cell line and the exposed cells processed with proteomic and bioinformatics analyses. The presence of polyhalogenated compounds in the topsoil extracts was confirmed by GC-MS/MS analysis. Proteomic analysis of the cells exposed to the topsoil extracts identified 43 differential proteins. Enrichment analysis highlighted biological processes, such as the cellular response to a chemical stimulus, stress, and inorganic substances; regulation of translation; regulation of apoptotic process; and the response to organonitrogen compounds in light of particular drugs and compounds, extrapolated by bioinformatics all linked to the identified protein modifications. Our results confirm and reflect the complex epidemiological situation occurring among Taranto inhabitants and underline the need to further investigate the presence and sources of inferred chemicals in soils. The combination of bioassays and proteomics reveals a more complex scenario of chemicals able to affect cellular pathways and leading to toxicities rather than those identified by only bioassays and related chemical analysis. This combined approach turns out to be a promising tool for soil risk assessment and deserves further investigation and developments for soil monitoring and risk assessment.
Collapse
Affiliation(s)
- Claudia Landi
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; (C.L.); (E.S.)
| | - Giulia Liberatori
- Department of Physical, Earth and Environmental Sciences, University of Siena, 53100 Siena, Italy; (G.L.); (L.S.); (M.L.V.)
| | - Pietro Cotugno
- Department of Biology, University of Bari Aldo Moro, 70121 Bari, Italy; (P.C.); (F.M.); (D.V.M.); (A.T.)
| | - Lucrezia Sturba
- Department of Physical, Earth and Environmental Sciences, University of Siena, 53100 Siena, Italy; (G.L.); (L.S.); (M.L.V.)
| | - Maria Luisa Vannuccini
- Department of Physical, Earth and Environmental Sciences, University of Siena, 53100 Siena, Italy; (G.L.); (L.S.); (M.L.V.)
| | - Federica Massari
- Department of Biology, University of Bari Aldo Moro, 70121 Bari, Italy; (P.C.); (F.M.); (D.V.M.); (A.T.)
| | - Daniela Valeria Miniero
- Department of Biology, University of Bari Aldo Moro, 70121 Bari, Italy; (P.C.); (F.M.); (D.V.M.); (A.T.)
| | - Angelo Tursi
- Department of Biology, University of Bari Aldo Moro, 70121 Bari, Italy; (P.C.); (F.M.); (D.V.M.); (A.T.)
| | - Enxhi Shaba
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; (C.L.); (E.S.)
| | - Peter A. Behnisch
- BioDetection System BV (BDS) Amsterdam, 1098 XH Amsterdam, The Netherlands;
| | - Alfonso Carleo
- Department of Pulmonology, Hannover Medical School, 30625 Hannover, Germany;
| | - Fabrizio Di Giuseppe
- Department of Medical, Oral & Biotechnological Sciences, Dentistry and Biotechnology and Proteomics Unit, Centre of Advanced Studies and Technology, “G. D’Annunzio”, University of Chieti-Pescara, 66100 Chieti, Italy; (F.D.G.); (S.A.)
| | - Stefania Angelucci
- Department of Medical, Oral & Biotechnological Sciences, Dentistry and Biotechnology and Proteomics Unit, Centre of Advanced Studies and Technology, “G. D’Annunzio”, University of Chieti-Pescara, 66100 Chieti, Italy; (F.D.G.); (S.A.)
| | - Luca Bini
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; (C.L.); (E.S.)
- Correspondence: (L.B.); (I.C.); Tel.: +39-0577-234938 (L.B.); +39-0577-232169 (I.C.)
| | - Ilaria Corsi
- Department of Physical, Earth and Environmental Sciences, University of Siena, 53100 Siena, Italy; (G.L.); (L.S.); (M.L.V.)
- Correspondence: (L.B.); (I.C.); Tel.: +39-0577-234938 (L.B.); +39-0577-232169 (I.C.)
| |
Collapse
|
12
|
Marzec J, Ross-Adams H, Pirrò S, Wang J, Zhu Y, Mao X, Gadaleta E, Ahmad AS, North BV, Kammerer-Jacquet SF, Stankiewicz E, Kudahetti SC, Beltran L, Ren G, Berney DM, Lu YJ, Chelala C. The Transcriptomic Landscape of Prostate Cancer Development and Progression: An Integrative Analysis. Cancers (Basel) 2021; 13:345. [PMID: 33477882 PMCID: PMC7838904 DOI: 10.3390/cancers13020345] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 11/16/2022] Open
Abstract
Next-generation sequencing of primary tumors is now standard for transcriptomic studies, but microarray-based data still constitute the majority of available information on other clinically valuable samples, including archive material. Using prostate cancer (PC) as a model, we developed a robust analytical framework to integrate data across different technical platforms and disease subtypes to connect distinct disease stages and reveal potentially relevant genes not identifiable from single studies alone. We reconstructed the molecular profile of PC to yield the first comprehensive insight into its development, by tracking changes in mRNA levels from normal prostate to high-grade prostatic intraepithelial neoplasia, and metastatic disease. A total of nine previously unreported stage-specific candidate genes with prognostic significance were also found. Here, we integrate gene expression data from disparate sample types, disease stages and technical platforms into one coherent whole, to give a global view of the expression changes associated with the development and progression of PC from normal tissue through to metastatic disease. Summary and individual data are available online at the Prostate Integrative Expression Database (PIXdb), a user-friendly interface designed for clinicians and laboratory researchers to facilitate translational research.
Collapse
Affiliation(s)
- Jacek Marzec
- Bioinformatics Unit, Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (J.M.); (S.P.); (J.W.); (E.G.)
| | - Helen Ross-Adams
- Bioinformatics Unit, Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (J.M.); (S.P.); (J.W.); (E.G.)
| | - Stefano Pirrò
- Bioinformatics Unit, Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (J.M.); (S.P.); (J.W.); (E.G.)
| | - Jun Wang
- Bioinformatics Unit, Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (J.M.); (S.P.); (J.W.); (E.G.)
| | - Yanan Zhu
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (Y.Z.); (X.M.); (S.-F.K.-J.); (E.S.); (S.C.K.); (D.M.B.); (Y.-J.L.)
| | - Xueying Mao
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (Y.Z.); (X.M.); (S.-F.K.-J.); (E.S.); (S.C.K.); (D.M.B.); (Y.-J.L.)
| | - Emanuela Gadaleta
- Bioinformatics Unit, Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (J.M.); (S.P.); (J.W.); (E.G.)
| | - Amar S. Ahmad
- Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK; (A.S.A.); (B.V.N.)
| | - Bernard V. North
- Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK; (A.S.A.); (B.V.N.)
| | - Solène-Florence Kammerer-Jacquet
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (Y.Z.); (X.M.); (S.-F.K.-J.); (E.S.); (S.C.K.); (D.M.B.); (Y.-J.L.)
| | - Elzbieta Stankiewicz
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (Y.Z.); (X.M.); (S.-F.K.-J.); (E.S.); (S.C.K.); (D.M.B.); (Y.-J.L.)
| | - Sakunthala C. Kudahetti
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (Y.Z.); (X.M.); (S.-F.K.-J.); (E.S.); (S.C.K.); (D.M.B.); (Y.-J.L.)
| | - Luis Beltran
- Department of Pathology, Barts Health NHS, London E1 F1R, UK;
| | - Guoping Ren
- Department of Pathology, The First Affiliated Hospital, Zhejiang University Medical College, Hangzhou 310058, China;
| | - Daniel M. Berney
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (Y.Z.); (X.M.); (S.-F.K.-J.); (E.S.); (S.C.K.); (D.M.B.); (Y.-J.L.)
- Department of Pathology, Barts Health NHS, London E1 F1R, UK;
| | - Yong-Jie Lu
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (Y.Z.); (X.M.); (S.-F.K.-J.); (E.S.); (S.C.K.); (D.M.B.); (Y.-J.L.)
| | - Claude Chelala
- Bioinformatics Unit, Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (J.M.); (S.P.); (J.W.); (E.G.)
- Centre for Computational Biology, Life Sciences Initiative, Queen Mary University London, London EC1M 6BQ, UK
| |
Collapse
|
13
|
Liu Y, Xu X, Xu R, Zhang S. Renoprotective Effects Of Isoliquiritin Against Cationic Bovine Serum Albumin-Induced Membranous Glomerulonephritis In Experimental Rat Model Through Its Anti-Oxidative And Anti-Inflammatory Properties. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3735-3751. [PMID: 31802848 PMCID: PMC6826199 DOI: 10.2147/dddt.s213088] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 08/08/2019] [Indexed: 12/11/2022]
Abstract
Background Membranous glomerulonephritis (MGN) is a nephrotic syndrome which shows the symptoms of heavy proteinuria and immune complex deposition in glomerular sub-epithelial space and finally leads to chronic kidney disease. Isoliquiritin (ILQ) is a flavonoid with a wide range of pharmacological properties, including antioxidant and anti-inflammatory activity. The present study was undertaken to investigate the possible mechanisms by which ILQ ameliorates cationic bovine serum albumin (C-BSA) induced MGN in rat model. Methods The MGN condition was confirmed by the 24 hr proteinuria and ILQ (10 mg/kg/bw/day) or TPCA-1 (10 mg/kg/bw/day; IKKβ inhibitor) was administered to successfully induce rats for 4 weeks. Results The present study revealed that MGN rats treated with ILQ showed significantly ameliorated kidney dysfunction and histopathological changes in kidneys. ILQ treated MGN rats alleviated the oxidative stress and were presented with increased anti-oxidative status in kidneys. Furthermore, ILQ treatment to MGN rats showed anti-oxidative effects through the prominent stimulation of Nrf2 signaling pathway and inhibition of Keap1, which consequently increases the Nrf2 nuclear translocation and thereby induces expression of NQO1 and HO-1. In addition, ILQ-treated MGN rats demonstrated anti-inflammatory effects by inhibiting NF-κB signaling pathway through decreased mRNA and protein expressions of NF-κB p65, IKKβ, COX-2, iNOS, p38-MAPK, p-p38-MAPK, TNF-α, IL-1β, IL-8, ICAM-1, E-selectin and VCAM-1 and reduced the nuclear translocation of NF-κB p65. Conclusion The protective effect of ILQ on MGN can be explained by its anti-oxidative and anti-inflammatory activities, which in turn due to the activation of Nrf2 and downregulation of NF-κB pathway.
Collapse
Affiliation(s)
- Yingying Liu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China
| | - Xiaohua Xu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China
| | - Ruisi Xu
- Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China
| | - Siqi Zhang
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China
| |
Collapse
|
14
|
Malila Y, Thanatsang K, Arayamethakorn S, Uengwetwanit T, Srimarut Y, Petracci M, Strasburg GM, Rungrassamee W, Visessanguan W. Absolute expressions of hypoxia-inducible factor-1 alpha (HIF1A) transcript and the associated genes in chicken skeletal muscle with white striping and wooden breast myopathies. PLoS One 2019; 14:e0220904. [PMID: 31393948 PMCID: PMC6687142 DOI: 10.1371/journal.pone.0220904] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/25/2019] [Indexed: 01/29/2023] Open
Abstract
Development of white striping (WS) and wooden breast (WB) in broiler breast meat have been linked to hypoxia, but their etiologies are not fully understood. This study aimed at investigating absolute expression of hypoxia-inducible factor-1 alpha subunit (HIF1A) and genes involved in stress responses and muscle repair using a droplet digital polymerase chain reaction. Total RNA was isolated from pectoralis major collected from male 6-week-old medium (carcass weight ≤ 2.5 kg) and heavy (carcass weight > 2.5 kg) broilers. Samples were classified as “non-defective” (n = 4), “medium-WS” (n = 6), “heavy-WS” (n = 7) and “heavy-WS+WB” (n = 3) based on abnormality scores. The HIF1A transcript was up-regulated in all of the abnormal groups. Transcript abundances of genes encoding 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 (PFKFB4), lactate dehydrogenase-A (LDHA), and phosphorylase kinase beta subunit (PHKB) were increased in heavy-WS but decreased in heavy-WS+WB. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was up-regulated in non-defective samples. The muscle-specific mu-2 isoform of glutathione S-transferases (GSTM2) was up-regulated in the abnormal samples, particularly in the heavy groups. The genes encoding myogenic differentiation (MYOD1) and myosin light chain kinase (MYLK) exhibited similar expression pattern, of which medium-WS and heavy-WS significantly increased compared to non-defective whereas expression in heavy-WS+WB was not different from either non-defective or WS-affected group. The greatest and the lowest levels of calpain-3 (CAPN3) and delta-sarcoglycan (SCGD) were observed in heavy-WS and heavy-WS+WB, respectively. Based on micrographs, the abnormal muscles primarily comprised fibers with cross-sectional areas ranging from 2,000 to 3,000 μm2. Despite induced glycolysis at the transcriptional level, lower stored glycogen in the abnormal muscles corresponded with the reduced lactate and higher pH within their meats. The findings support hypoxia within the abnormal breasts, potentially associated with oversized muscle fibers. Between WS and WB, divergent glucose metabolism, cellular detoxification and myoregeneration at the transcriptional level could be anticipated.
Collapse
Affiliation(s)
- Yuwares Malila
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Khlong Nueng, Khlong Luang, Pathum Thani, Thailand
- * E-mail:
| | - Krittaporn Thanatsang
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Khlong Nueng, Khlong Luang, Pathum Thani, Thailand
| | - Sopacha Arayamethakorn
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Khlong Nueng, Khlong Luang, Pathum Thani, Thailand
| | - Tanaporn Uengwetwanit
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Khlong Nueng, Khlong Luang, Pathum Thani, Thailand
| | - Yanee Srimarut
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Khlong Nueng, Khlong Luang, Pathum Thani, Thailand
| | - Massimiliano Petracci
- Department of Agricultural and Food Sciences, Alma Mater Studiorum, University of Bologna, Cesena (FC), Italy
| | - Gale M. Strasburg
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States of America
| | - Wanilada Rungrassamee
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Khlong Nueng, Khlong Luang, Pathum Thani, Thailand
| | - Wonnop Visessanguan
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Khlong Nueng, Khlong Luang, Pathum Thani, Thailand
| |
Collapse
|
15
|
Zhao YJ, Liu XY, Guo R, Hu KR, Cao Y, Dai F. Comparative genomics and transcriptomics analysis reveals evolution patterns of selection in the Salix phylogeny. BMC Genomics 2019; 20:253. [PMID: 30925896 PMCID: PMC6440167 DOI: 10.1186/s12864-019-5627-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/20/2019] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Willows are widely distributed in the northern hemisphere and have good adaptability to different living environment. The increasing of genome and transcriptome data provides a chance for comparative analysis to study the evolution patterns with the different origin and geographical distributions in the Salix phylogeny. RESULTS Transcript sequences of 10 Salicaceae species were downloaded from public databases. All pairwise of orthologues were identified by comparative analysis in these species, from which we constructed a phylogenetic tree and estimated the rate of diverse. Divergence times were estimated in the 10 Salicaceae using comparative transcriptomic analysis. All of the fast-evolving positive selection sequences were identified, and some cold-, drought-, light-, universal-, and heat- resistance genes were discovered. CONCLUSIONS The divergence time of subgenus Vetrix and Salix was about 17.6-16.0 Mya during the period of Middle Miocene Climate Transition (21-14 Mya). Subgenus Vetrix diverged to migratory and resident groups when the climate changed to the cool and dry trend by 14 Mya. Cold- and light- stress genes were involved in positive selection among the resident Vetrix, and which would help them to adapt the cooling stage. Universal- stress genes exhibited positive selection among the migratory group and subgenus Salix. These data are useful for comprehending the adaptive evolution and speciation in the Salix lineage.
Collapse
Affiliation(s)
- You-jie Zhao
- Key Laboratory of Forestry and Ecological Big Data State Forestry Administration, Southwest Forestry University, Kunming, 650224 Yunnan People’s Republic of China
- College of Big data and Intelligent Engineering, Southwest Forestry University, Kunming, 650224 Yunnan People’s Republic of China
- Key Laboratory for Forest Resources Conservation and Utilization in the Southwest Mountains of China, Ministry of Education, Southwest Forestry University, Kunming, 650224 Yunnan People’s Republic of China
| | - Xin-yi Liu
- College of Big data and Intelligent Engineering, Southwest Forestry University, Kunming, 650224 Yunnan People’s Republic of China
| | - Ran Guo
- College of Big data and Intelligent Engineering, Southwest Forestry University, Kunming, 650224 Yunnan People’s Republic of China
| | - Kun-rong Hu
- College of Big data and Intelligent Engineering, Southwest Forestry University, Kunming, 650224 Yunnan People’s Republic of China
| | - Yong Cao
- College of Big data and Intelligent Engineering, Southwest Forestry University, Kunming, 650224 Yunnan People’s Republic of China
| | - Fei Dai
- College of Big data and Intelligent Engineering, Southwest Forestry University, Kunming, 650224 Yunnan People’s Republic of China
| |
Collapse
|
16
|
Abstract
The number of individuals affected by acute kidney injury (AKI) and chronic kidney disease (CKD) is constantly rising. In light of the limited availability of treatment options and their relative inefficacy, cell based therapeutic modalities have been studied. However, not many efforts are put into safety evaluation of such applications. The aim of this study was to review the existing published literature on adverse events reported in studies with genetically modified cells for treatment of kidney disease. A systematic review was conducted by searching PubMed and EMBASE for relevant articles published until June 2018. The search results were screened and relevant articles selected using pre-defined criteria, by two researchers independently. After initial screening of 6894 abstracts, a total number of 97 preclinical studies was finally included for full assessment. Of these, 61 (63%) presented an inappropriate study design for the evaluation of safety parameters. Only 4 studies (4%) had the optimal study design, while 32 (33%) showed sub-optimal study design with either direct or indirect evidence of adverse events. The high heterogeneity of studies included regarding cell type and number, genetic modification, administration route, and kidney disease model applied, combined with the consistent lack of appropriate control groups, makes a reliable safety evaluation of kidney cell-based therapies impossible. Only a limited number of relevant studies included looked into essential safety-related outcomes, such as inflammatory (48%), tumorigenic and teratogenic potential (12%), cell biodistribution (82%), microbiological safety with respect to microorganism contamination and latent viruses' reactivation (1%), as well as overall well-being and animal survival (19%). In conclusion, for benign cell-based therapies, well-designed pre-clinical studies, including all control groups required and good manufacturing processes securing safety, need to be done early in development. Preferably, this should be performed side by side with efficacy evaluation and according to the official guidelines of leading health organizations.
Collapse
|
17
|
Wen W, Lin Y, Ti Z. Antidiabetic, Antihyperlipidemic, Antioxidant, Anti-inflammatory Activities of Ethanolic Seed Extract of Annona reticulata L. in Streptozotocin Induced Diabetic Rats. Front Endocrinol (Lausanne) 2019; 10:716. [PMID: 31708869 PMCID: PMC6819323 DOI: 10.3389/fendo.2019.00716] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 10/03/2019] [Indexed: 12/22/2022] Open
Abstract
Annona reticulata L. (Bullock's heart) is a pantropic tree commonly known as custard apple, which is used therapeutically for a variety of maladies. The present research was carried out to evaluate the possible protective effects of Annona reticulata L. (A. reticulata) ethanolic seed extract on an experimentally induced type 2 diabetes rat model. Male Albino Wistar rats were randomly divided into five groups with six animals in each group viz., control rats in group I, diabetic rats in group II, diabetic rats with 50 and 100 mg/kg/bw of ethanolic seed extract of A. reticulata in groups III and IV, respectively, and diabetic rats with metformin in group V. Treatment was given for 42 consecutive days through oral route by oro-gastric gavage. Administration of A. reticulata seed extract to diabetes rats significantly restored the alterations in the levels of body weight, food and water intake, fasting blood glucose (FBG), insulin levels, insulin sensitivity, HbA1c, HOMA-IR, islet area and insulin positive cells. Furthermore, A. reticulata significantly decreased the levels of triglycerides, cholesterol, LDL, and significantly increased the HDL in diabetic rats. A. reticulata effectively ameliorated the enzymatic (ALT, AST, ALP, GGT) and modification of histopathological changes in diabetic rats. The serum levels of the BUN, creatinine levels, uric acid, urine volume, and urinary protein were significantly declined with a significant elevation in CCr in diabetic rats treated with A. reticulata. MDA and NO levels were significantly reduced with an enhancement in SOD, CAT, and GPx antioxidant enzyme activities in the kidney, liver, and pancreas of diabetic rats treated with A. reticulata. Diabetic rats treated with A. reticulata have shown up-regulation in mRNA expression levels of nuclear factor erythroid 2-related factor 2 (Nrf2), NAD(P)H:quinone oxidoreductase 1 (NQO1), Heme oxygenase-1 (HO-1) and protein expression level of Nrf2 with diminution in Keap1 mRNA expression level in pancreas, kidney, and liver. From the outcome of the current results, it can be inferred that seed extract of A. reticulata exhibits a protective effect in diabetic rats through its anti-diabetic, anti-hyperlipidemic, antioxidant and anti-inflammatory effects and could be considered as a promising treatment therapy in the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Wenbin Wen
- Department of Nephrology, Heji Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Yukiat Lin
- Innoscience Research Sdn Bhd, Subang Jaya, Malaysia
| | - Zhenyu Ti
- The Department of General Surgery, Xi'an No. 3 Hospital, Xi'an, China
- *Correspondence: Zhenyu Ti
| |
Collapse
|
18
|
Tahir W, Zafar S, Llorens F, Arora AS, Thüne K, Schmitz M, Gotzmann N, Kruse N, Mollenhauer B, Torres JM, Andréoletti O, Ferrer I, Zerr I. Molecular Alterations in the Cerebellum of Sporadic Creutzfeldt-Jakob Disease Subtypes with DJ-1 as a Key Regulator of Oxidative Stress. Mol Neurobiol 2016; 55:517-537. [PMID: 27975168 DOI: 10.1007/s12035-016-0294-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 11/08/2016] [Indexed: 12/15/2022]
Abstract
Cerebellar damage and granular and Purkinje cell loss in sporadic Creutzfeldt-Jakob disease (sCJD) highlight a critical involvement of the cerebellum during symptomatic progression of the disease. In this project, global proteomic alterations in the cerebellum of brain from the two most prevalent subtypes (MM1 and VV2) of sCJD were studied. Two-dimensional gel electrophoresis (2DE) coupled mass spectrometric identification revealed 40 proteins in MM1 and 43 proteins in VV2 subtype to be differentially expressed. Of those, 12 proteins showed common differential expression in their expression between two subtypes. Differentially expressed proteins mainly belonged to (i) cell cycle, gene expression and cell death; (ii) cellular stress response/oxidative stress (OS) and (iii) signal transduction and synaptic functions, related molecular functions. We verified 10 differentially expressed proteins at transcriptional and translational level as well. Interestingly, protein deglycase DJ-1 (an antioxidative protein) showed an increase in its messenger RNA (mRNA) expression in both MM1 and VV2 subtypes but protein expression only in VV2 subtype in cerebellum of sCJD patients. Nuclear translocalization of DJ-1 confirmed its expressional alteration due to OS in sCJD. Downstream experiments showed the activation of nuclear factor erythroid-2 related factor 2 (Nrf2)/antioxidative response element (ARE) pathway. DJ-1 protein concentration was significantly increased during the clinical phase in cerebrospinal fluid of sCJD patients and also at presymptomatic and symptomatic stages in cerebellum of humanized PrP transgenic mice inoculated with sCJD (MM1 and VV2) brain. These results suggest the implication of oxidative stress during the pathophysiology of sCJD.
Collapse
Affiliation(s)
- Waqas Tahir
- Department of Neurology, University Medical Center Goettingen (UMG) and German Center for Neurodegenerative Diseases (DZNE) Goettingen, Robert-Koch-Str., 40, 37075, Goettingen, Germany
| | - Saima Zafar
- Department of Neurology, University Medical Center Goettingen (UMG) and German Center for Neurodegenerative Diseases (DZNE) Goettingen, Robert-Koch-Str., 40, 37075, Goettingen, Germany.
| | - Franc Llorens
- Department of Neurology, University Medical Center Goettingen (UMG) and German Center for Neurodegenerative Diseases (DZNE) Goettingen, Robert-Koch-Str., 40, 37075, Goettingen, Germany
| | - Amandeep Singh Arora
- Department of Neurology, University Medical Center Goettingen (UMG) and German Center for Neurodegenerative Diseases (DZNE) Goettingen, Robert-Koch-Str., 40, 37075, Goettingen, Germany
| | - Katrin Thüne
- Department of Neurology, University Medical Center Goettingen (UMG) and German Center for Neurodegenerative Diseases (DZNE) Goettingen, Robert-Koch-Str., 40, 37075, Goettingen, Germany
| | - Matthias Schmitz
- Department of Neurology, University Medical Center Goettingen (UMG) and German Center for Neurodegenerative Diseases (DZNE) Goettingen, Robert-Koch-Str., 40, 37075, Goettingen, Germany
| | - Nadine Gotzmann
- Department of Neurology, University Medical Center Goettingen (UMG) and German Center for Neurodegenerative Diseases (DZNE) Goettingen, Robert-Koch-Str., 40, 37075, Goettingen, Germany
| | - Niels Kruse
- Institute of Neuropathology, University Medical Center Goettingen (UMG), Robert-Koch-Str. 40, 37075, Goettingen, Germany
| | - Brit Mollenhauer
- Institute of Neuropathology, University Medical Center Goettingen (UMG), Robert-Koch-Str. 40, 37075, Goettingen, Germany
| | - Juan Maria Torres
- Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Carretera de Algete a El Casar Km. 8,1 S/N, 28130, Valdeolmos, Madrid, Spain
| | - Olivier Andréoletti
- Institut National de la Recherche Agronomique/Ecole Nationale Vétérinaire, Toulouse, France
| | - Isidre Ferrer
- Institute of Neuropathology, Hospitalet de Llobregat, IDIBELL-University Hospital Bellvitge, University of Barcelona, Barcelona, Spain.,Network Center for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Ministry of Health, Institute Carlos III, Madrid, Spain
| | - Inga Zerr
- Department of Neurology, University Medical Center Goettingen (UMG) and German Center for Neurodegenerative Diseases (DZNE) Goettingen, Robert-Koch-Str., 40, 37075, Goettingen, Germany
| |
Collapse
|
19
|
Yang Y, Song HL, Zhang W, Wu BJ, Fu NN, Dong C, Shen ZY. Heme oxygenase-1-transduced bone marrow mesenchymal stem cells in reducing acute rejection and improving small bowel transplantation outcomes in rats. Stem Cell Res Ther 2016; 7:164. [PMID: 27866474 PMCID: PMC5116370 DOI: 10.1186/s13287-016-0427-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/22/2016] [Accepted: 10/20/2016] [Indexed: 12/13/2022] Open
Abstract
Background We determined whether bone marrow mesenchymal stem cells (BMMSCs) transduced with heme oxygenase-1 (HO-1), a cytoprotective and immune-protective factor, could improve outcomes for small bowel transplantation (SBTx) in rats. Methods We performed heterotopic SBTx from Brown Norway rats to Lewis rats, before infusing Ad/HO-1-transduced BMMSCs (Ad/HO-1/BMMSCs) through the superficial dorsal veins of the penis. Respective infusions with Ad/BMMSCs, BMMSCs, and normal saline served as controls. The animals were sacrificed after 1, 5, 7, or 10 days. At each time point, we measured small bowel histology and apoptosis, HO-1 protein and mRNA expression, natural killer (NK) cell activity, cytokine concentrations in serum and intestinal graft, and levels of regulatory T (Treg) cells. Results The saline-treated control group showed aggravated acute cellular rejection over time, with mucosal destruction, increased apoptosis, NK cell activation, and upregulation of proinflammatory and immune-related mediators. Both the Ad/BMMSC-treated group and the BMMSC-treated group exhibited attenuated acute cellular rejection at an early stage, but the effects receded 7 days after transplantation. Strikingly, the Ad/HO-1/BMMSC-treated group demonstrated significantly attenuated acute cellular rejection, reduced apoptosis and NK cell activity, and suppressed concentrations of inflammation and immune-related cytokines, and upregulated expression of anti-inflammatory cytokine mediators and increased Treg cell levels. Conclusion Our data suggest that Ad/HO-1-transduced BMMSCs have a reinforced effect on reducing acute rejection and protecting the outcome of SBTx in rats.
Collapse
Affiliation(s)
- Yang Yang
- Department of Organ Transplantation, Tianjin First Central Hospital, 24# Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China
| | - Hong Li Song
- Department of Organ Transplantation, Tianjin First Central Hospital, 24# Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China. .,Tianjin Key Laboratory of Organ Transplantation, 24# Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China.
| | - Wen Zhang
- Department of Organ Transplantation, Tianjin First Central Hospital, 24# Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China
| | - Ben Juan Wu
- Department of Organ Transplantation, Tianjin First Central Hospital, 24# Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China
| | - Nan Nan Fu
- Department of Organ Transplantation, Tianjin First Central Hospital, 24# Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China
| | - Chong Dong
- Department of Organ Transplantation, Tianjin First Central Hospital, 24# Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China
| | - Zhong Yang Shen
- Department of Organ Transplantation, Tianjin First Central Hospital, 24# Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China.
| |
Collapse
|
20
|
Anti-inflammatory effects of alpinone 3-acetate from Alpinia japonica seeds. J Nat Med 2016; 70:653-60. [PMID: 27137785 DOI: 10.1007/s11418-016-0997-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 04/09/2016] [Indexed: 12/26/2022]
Abstract
We aimed to investigate the bioactive components of Alpinia japonica as anti-inflammatory compounds using searches of the Alpinia genus, and subsequently demonstrated that alpinone 3-acetate markedly inhibits 12-O-tetradecanoyiphorbol 13-acetate-induced inflammation in a mouse model of ear edema. To assess other bioactivities of alpinone 3-acetate, we performed translatome analyses and compared them with those of hydrocortisone. Polysome-associated mRNAs were prepared from alpinone 3-acetate- or hydrocortisone-treated and control cells from 12-O-tetradecanoyiphorbol 13-acetate-induced THP-1-derived macrophages cultured in the presence of Escherichia coli O-111 lipopolysaccharide. Subsequent microarray analysis revealed that alpinone 3-acetate and hydrocortisone upregulated and downregulated the same 155 and 41 genes, respectively. Moreover, direct comparisons of translationally regulated genes indicated 5 and 10 gene probes that were upregulated and downregulated by alpinone 3-acetate and hydrocortisone, respectively. In conclusion, assays of 12-O-tetradecanoyiphorbol 13-acetate-induced inflammation ear edema in mice and polysome profiling of alpinone 3-acetate bioactivities indicated similar medicinal possibilities to those of hydrocortisone.
Collapse
|
21
|
Mohamed HE, Elswefy SE, Rashed LA, Younis NN, Shaheen MA, Ghanim AMH. Bone marrow-derived mesenchymal stem cells effectively regenerate fibrotic liver in bile duct ligation rat model. Exp Biol Med (Maywood) 2016; 241:581-91. [PMID: 26811102 DOI: 10.1177/1535370215627219] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 12/21/2015] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have attracted lots of attention for the treatment of acute liver failure and end-stage liver diseases. This study aimed at investigating the fundamental mechanism by which bone marrow-derived MSCs (BM-MSCs) induce liver regeneration of fibrotic liver in rats. Rats underwent bile duct ligation (BDL) surgery and four weeks later they were treated with either BM-MSCs (3 × 10(6) cells /rat, once, tail vein injection) or silymarin (100 mg/kg, daily, orally) for four weeks. Liver function tests and hepatic oxidative stress were determined. Hepatic injury and fibrosis were assessed by H and E, Sirus red staining and immunohistochemical expression of α-smooth muscle actin (α-SMA). Hepatocyte growth factor (HGF) and the gene expression of cytokeratin-19 (CK-19) and matrix metalloproteinase-2 (MMP-2) in liver tissue were determined. BDL induced cholestatic liver injury characterized by elevated ALT and AST activities, bilirubin and decreased albumin. The architecture damage was staged as Metavir score: F3, A3. Fibrosis increased around proliferating bile duct as indicated by sirus red staining and α-SMA immunostaining. Fibrogenesis was favored over fibrolysis and confirmed by decreased HGF with increased expression of CK-19, but decreased MMP-2 expression. BM-MSCs treatment restored deteriorated liver functions and restored the histological changes, resolved fibrosis by improving liver regenerative capabilities (P < 0.001), increases in HGF and MMP-2 mRNA and downregulating CK-19 mRNA. Sliymarin, however, induced similar but less prominent effects compared to BM-MSCs. In conclusion, liver regenerative capabilities can be stimulated by BM-MSCs via augmentation of HGF that subsequently up-regulate MMP-2 mRNA while downregulating CK-19 mRNA.
Collapse
Affiliation(s)
- Hoda E Mohamed
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Sahar E Elswefy
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Laila A Rashed
- Department of Medical Biochemistry, Unit of Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Nahla N Younis
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Mohamed A Shaheen
- Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Amal M H Ghanim
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
22
|
The challenges and promises of allogeneic mesenchymal stem cells for use as a cell-based therapy. Stem Cell Res Ther 2015; 6:234. [PMID: 26620426 PMCID: PMC4665863 DOI: 10.1186/s13287-015-0240-9] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are ideal for cell-based therapy in various inflammatory diseases because of their immunosuppressive and tissue repair properties. Moreover, their immunosuppressive properties and low immunogenicity contribute to a reduced or weakened immune response elicited by the implantation of allogeneic MSCs compared with other cell types. Therefore, implantation of allogeneic MSCs may be a promising cell-based therapy. In this review, we first summarize the unique advantages of allogeneic MSCs for therapeutic applications. Second, we critically analyze the factors influencing their therapeutic effects, including administration routes, detection time-points, disease models, differentiation of MSCs in vivo, and timing and dosage of MSC administration. Finally, current approaches to allogeneic MSC application are discussed. In conclusion, allogeneic MSCs are a promising option because of their low immunogenicity and immunosuppressive and tissue repair capabilities. Further investigations are needed to enhance the consistency and efficacy of MSCs when used as a cell-based therapy in inflammatory diseases as well as for tissue repair.
Collapse
|
23
|
Nowakowski A, Walczak P, Janowski M, Lukomska B. Genetic Engineering of Mesenchymal Stem Cells for Regenerative Medicine. Stem Cells Dev 2015; 24:2219-42. [PMID: 26140302 DOI: 10.1089/scd.2015.0062] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs), which can be obtained from various organs and easily propagated in vitro, are one of the most extensively used types of stem cells and have been shown to be efficacious in a broad set of diseases. The unique and highly desirable properties of MSCs include high migratory capacities toward injured areas, immunomodulatory features, and the natural ability to differentiate into connective tissue phenotypes. These phenotypes include bone and cartilage, and these properties predispose MSCs to be therapeutically useful. In addition, MSCs elicit their therapeutic effects by paracrine actions, in which the metabolism of target tissues is modulated. Genetic engineering methods can greatly amplify these properties and broaden the therapeutic capabilities of MSCs, including transdifferentiation toward diverse cell lineages. However, cell engineering can also affect safety and increase the cost of therapy based on MSCs; thus, the advantages and disadvantages of these procedures should be discussed. In this review, the latest applications of genetic engineering methods for MSCs with regenerative medicine purposes are presented.
Collapse
Affiliation(s)
- Adam Nowakowski
- 1 NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences , Warsaw, Poland
| | - Piotr Walczak
- 2 Division of Magnetic Resonance Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine , Baltimore, Maryland.,3 Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland.,4 Department of Radiology, Faculty of Medical Sciences, University of Warmia and Mazury , Olsztyn, Poland
| | - Miroslaw Janowski
- 1 NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences , Warsaw, Poland .,2 Division of Magnetic Resonance Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine , Baltimore, Maryland.,3 Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Barbara Lukomska
- 1 NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences , Warsaw, Poland
| |
Collapse
|
24
|
Srivastava AK, Kadayakkara DK, Bar-Shir A, Gilad AA, McMahon MT, Bulte JWM. Advances in using MRI probes and sensors for in vivo cell tracking as applied to regenerative medicine. Dis Model Mech 2015; 8:323-36. [PMID: 26035841 PMCID: PMC4381332 DOI: 10.1242/dmm.018499] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The field of molecular and cellular imaging allows molecules and cells to be visualized in vivo non-invasively. It has uses not only as a research tool but in clinical settings as well, for example in monitoring cell-based regenerative therapies, in which cells are transplanted to replace degenerating or damaged tissues, or to restore a physiological function. The success of such cell-based therapies depends on several critical issues, including the route and accuracy of cell transplantation, the fate of cells after transplantation, and the interaction of engrafted cells with the host microenvironment. To assess these issues, it is necessary to monitor transplanted cells non-invasively in real-time. Magnetic resonance imaging (MRI) is a tool uniquely suited to this task, given its ability to image deep inside tissue with high temporal resolution and sensitivity. Extraordinary efforts have recently been made to improve cellular MRI as applied to regenerative medicine, by developing more advanced contrast agents for use as probes and sensors. These advances enable the non-invasive monitoring of cell fate and, more recently, that of the different cellular functions of living cells, such as their enzymatic activity and gene expression, as well as their time point of cell death. We present here a review of recent advancements in the development of these probes and sensors, and of their functioning, applications and limitations.
Collapse
Affiliation(s)
- Amit K Srivastava
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Deepak K Kadayakkara
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Amnon Bar-Shir
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Assaf A Gilad
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Michael T McMahon
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA. Department of Chemical & Biomolecular Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
25
|
Greenhall GHB, Salama AD. What is new in the management of rapidly progressive glomerulonephritis? Clin Kidney J 2015; 8:143-50. [PMID: 25815169 PMCID: PMC4370308 DOI: 10.1093/ckj/sfv008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/23/2015] [Indexed: 12/20/2022] Open
Abstract
Rapidly progressive glomerulonephritis (RPGN) results from severe crescentic damage to glomeruli and leads to irreversible kidney failure if not diagnosed and managed in a timely fashion. Traditional treatment has relied on glucocorticoids and cyclophosphamide, with additional plasmapheresis for certain conditions. Here we describe updates in the management of RPGN, according to the underlying renal pathology. However, there remains a paucity of trials that have enrolled patients with more advanced renal disease, dialysis dependence or with RPGN, and we are therefore still reliant on extrapolation of data from studies of patients with a less severe form of disease. In addition, reporting bias results in publication of cases or cohorts showing benefit for newer agents in advanced disease or RPGN, but it remains unclear how many unsuccessful outcomes in these circumstances take place. Since clinical trials specifically in RPGN are unlikely, use of biologic registries or combination of sufficient sized cohort series may provide indications of benefit outside of a clinical trial setting and should be encouraged, in order to provide some evidence for the efficacy of therapeutic regimens in RPGN and advanced renal disease.
Collapse
Affiliation(s)
| | - Alan D Salama
- UCL Centre for Nephrology , Royal Free Hospital , London , UK
| |
Collapse
|
26
|
Li Y, Li W, Liu C, Yan M, Raman I, Du Y, Fang X, Zhou XJ, Mohan C, Li QZ. Delivering Oxidation Resistance-1 (OXR1) to Mouse Kidney by Genetic Modified Mesenchymal Stem Cells Exhibited Enhanced Protection against Nephrotoxic Serum Induced Renal Injury and Lupus Nephritis. ACTA ACUST UNITED AC 2014; 4. [PMID: 25995969 PMCID: PMC4435960 DOI: 10.4172/2157-7633.1000231] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To elucidate the role of oxidation resistance 1 (OXR1) gene. Oxidative stress plays a pivotal role in pathogenesis of immune-mediated nephritis. Recently we identified oxidation resistance 1 (OXR1) is conventionally expressed in eukaryotes and has an ability to prevent oxidative damage caused by various oxidative stresses. However the protective effect of OXR1 in immune-associated inflammatory response and oxidative damage is not clear and will be investigated in this study. METHODS We utilized mesenchymal stem cells (MSCs) as vehicles to carry OXR1 into the injured kidneys of nephritis model mice and investigated the influence of OXR1 on glomerulonephritis. Human OXR1 gene was integrated into genome of MSCs via lentiviral vector, and established hOXR1-MSC cell line which still maintains the differentiation property. 129/svj mice with anti-glomerular basement membrane (GBM) challenge and spontaneous lupus mice B6.Sle1.Sle2.Sle3 were injected with hOXR1-MSCs (i.v. injection) to evaluate the function of hOXR1. Immunohistochemistry was used to appraise the renal pathology and Tunel staining was applied to detect cell apoptosis. RESULTS Compared with control mice, hOXR1-MSCs administration showed significantly decreased blood urea nitrogen (BUN), proteinuria and ameliorated renal pathological damage. hOXR1-MSCs transplantation significantly reduced macrophage and T lymphocyte infiltration by inhibiting the expression of CCL2, CCL7, IL-1β, IL-6 and NFκB in mouse kidney. Moreover, hOXR1-MSCs prevented hydrogen peroxide (H2O2)-induced oxidative stress and its implantation reduced nitric oxide (NO) in mouse serum and urine to inhibit tubular cell apoptosis. CONCLUSION OXR1-MSCs transplantation may exert a certain protective effect on nephritis by suppressing inflammation and oxidative stress.
Collapse
Affiliation(s)
- Yajuan Li
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100029, China ; Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Wei Li
- Key Laboratory of Medical Genetics, Wenzhou Medical University School of Laboratory Medicine & Life Science, Wenzhou, 325035, China
| | - Chu Liu
- Key Laboratory of Medical Genetics, Wenzhou Medical University School of Laboratory Medicine & Life Science, Wenzhou, 325035, China
| | - Mei Yan
- Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Indu Raman
- Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yong Du
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Xiangdong Fang
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100029, China
| | - Xin J Zhou
- Renal Path Diagnostics, Pathologist BioMedical Laboratories, Lewisville, TX, 75067, USA
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Quan-Zhen Li
- Key Laboratory of Medical Genetics, Wenzhou Medical University School of Laboratory Medicine & Life Science, Wenzhou, 325035, China ; Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|