1
|
Zhang Z, Zhao Z, Qi C, Zhang X, Xiao Y, Chen C, Zou Y, Chen X, Gu L, Huang J, Huang K, Xiang M, Zhang T, Tong Q, Zhang Y. Butyrolactone I blocks the transition of acute kidney injury to chronic kidney disease in mice by targeting JAK1. MedComm (Beijing) 2025; 6:e70064. [PMID: 39845897 PMCID: PMC11751251 DOI: 10.1002/mco2.70064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025] Open
Abstract
Chronic kidney disease (CKD) is a disease that affects more than 850 million people. Acute kidney injury (AKI) is a common cause of CKD, and blocking the AKI-CKD transition shows promising therapeutic potential. Herein, we found that butyrolactone I (BLI), a natural product, exerts significant nephroprotective effects, including maintenance of kidney function, inhibition of inflammatory response, and prevention of fibrosis, in both folic acid- and ureteral obstruction-induced AKI-CKD transition mouse models. Notably, BLI showed greater blood urea nitrogen reduction and anti-inflammatory effects than telmisartan. Bioinformatics analysis and target confirmation assays suggested that BLI directly binds to JAK1, and kinase inhibition assay confirmed it is a potent JAK1inhibitor with an IC50 of 0.376 µM. Experiments in JAK1-knockdown mice also proved that BLI targets JAK1 to work. Furthermore, BLI demonstrated nephroprotective effects and safety comparable to ivarmacitinib, the well-known JAK1 inhibitor. Mechanistically, BLI targets JAK1 and inhibits its phosphorylation and JAK-STAT activation, subsequently regulating the downstream signaling pathways to inhibit reactive oxygen species production, inflammation, and ferroptosis, thereby preventing the occurrence of kidney fibrosis and blocking the AKI-CKD transition process. This study demonstrates for the first time that BLI is a JAK1 inhibitor and a promising candidate for delaying CKD progression, which warrants further investigation.
Collapse
Affiliation(s)
- Zijun Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource EvaluationSchool of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ziming Zhao
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource EvaluationSchool of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Changxing Qi
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource EvaluationSchool of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaotian Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource EvaluationSchool of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yang Xiao
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource EvaluationSchool of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chengjuan Chen
- State Key Laboratory of Bioactive Substances and Function of Natural MedicineInstitute of Materia MedicaChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Yu Zou
- Institute of Pharmaceutical ProcessHubei Province Key Laboratory of Occupational Hazard Identification and ControlSchool of MedicineWuhan University of Science and TechnologyWuhanChina
| | - Xia Chen
- Institute of Pharmaceutical ProcessHubei Province Key Laboratory of Occupational Hazard Identification and ControlSchool of MedicineWuhan University of Science and TechnologyWuhanChina
| | - Lianghu Gu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource EvaluationSchool of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jianzheng Huang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource EvaluationSchool of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Kun Huang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource EvaluationSchool of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ming Xiang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource EvaluationSchool of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Tiantai Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural MedicineInstitute of Materia MedicaChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Qingyi Tong
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource EvaluationSchool of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource EvaluationSchool of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
2
|
Mansoor S, Hamid S, Tuan TT, Park JE, Chung YS. Advance computational tools for multiomics data learning. Biotechnol Adv 2024; 77:108447. [PMID: 39251098 DOI: 10.1016/j.biotechadv.2024.108447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 09/01/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024]
Abstract
The burgeoning field of bioinformatics has seen a surge in computational tools tailored for omics data analysis driven by the heterogeneous and high-dimensional nature of omics data. In biomedical and plant science research multi-omics data has become pivotal for predictive analytics in the era of big data necessitating sophisticated computational methodologies. This review explores a diverse array of computational approaches which play crucial role in processing, normalizing, integrating, and analyzing omics data. Notable methods such similarity-based methods, network-based approaches, correlation-based methods, Bayesian methods, fusion-based methods and multivariate techniques among others are discussed in detail, each offering unique functionalities to address the complexities of multi-omics data. Furthermore, this review underscores the significance of computational tools in advancing our understanding of data and their transformative impact on research.
Collapse
Affiliation(s)
- Sheikh Mansoor
- Department of Plant Resources and Environment, Jeju National University, 63243, Republic of Korea
| | - Saira Hamid
- Watson Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Pulwama, J&K, India
| | - Thai Thanh Tuan
- Department of Plant Resources and Environment, Jeju National University, 63243, Republic of Korea; Multimedia Communications Laboratory, University of Information Technology, Ho Chi Minh city 70000, Vietnam; Multimedia Communications Laboratory, Vietnam National University, Ho Chi Minh city 70000, Vietnam
| | - Jong-Eun Park
- Department of Animal Biotechnology, College of Applied Life Science, Jeju National University, Jeju, Jeju-do, Republic of Korea.
| | - Yong Suk Chung
- Department of Plant Resources and Environment, Jeju National University, 63243, Republic of Korea.
| |
Collapse
|
3
|
Tan RJ, Liu Y. Matrix metalloproteinases in kidney homeostasis and diseases: an update. Am J Physiol Renal Physiol 2024; 327:F967-F984. [PMID: 39361724 PMCID: PMC11687849 DOI: 10.1152/ajprenal.00179.2024] [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: 06/20/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/05/2024] Open
Abstract
Matrix metalloproteinases (MMPs) are zinc-dependent endopeptidases with important roles in kidney homeostasis and pathology. While capable of collectively degrading each component of the extracellular matrix, MMPs also degrade nonmatrix substrates to regulate inflammation, epithelial plasticity, proliferation, apoptosis, and angiogenesis. More recently, intriguing mechanisms that directly alter podocyte biology have been described. There is now irrefutable evidence for MMP dysregulation in many types of kidney disease including acute kidney injury, diabetic and hypertensive nephropathy, polycystic kidney disease, and Alport syndrome. This updated review will detail the complex biology of MMPs in kidney disease.
Collapse
Affiliation(s)
- Roderick J Tan
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States
| | - Youhua Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
4
|
Cohen C, Mhaidly R, Croizer H, Kieffer Y, Leclere R, Vincent-Salomon A, Robley C, Anglicheau D, Rabant M, Sannier A, Timsit MO, Eddy S, Kretzler M, Ju W, Mechta-Grigoriou F. WNT-dependent interaction between inflammatory fibroblasts and FOLR2+ macrophages promotes fibrosis in chronic kidney disease. Nat Commun 2024; 15:743. [PMID: 38272907 PMCID: PMC10810789 DOI: 10.1038/s41467-024-44886-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 01/08/2024] [Indexed: 01/27/2024] Open
Abstract
Chronic kidney disease (CKD) is a public health problem driven by myofibroblast accumulation, leading to interstitial fibrosis. Heterogeneity is a recently recognized characteristic in kidney fibroblasts in CKD, but the role of different populations is still unclear. Here, we characterize a proinflammatory fibroblast population (named CXCL-iFibro), which corresponds to an early state of myofibroblast differentiation in CKD. We demonstrate that CXCL-iFibro co-localize with macrophages in the kidney and participate in their attraction, accumulation, and switch into FOLR2+ macrophages from early CKD stages on. In vitro, macrophages promote the switch of CXCL-iFibro into ECM-secreting myofibroblasts through a WNT/β-catenin-dependent pathway, thereby suggesting a reciprocal crosstalk between these populations of fibroblasts and macrophages. Finally, the detection of CXCL-iFibro at early stages of CKD is predictive of poor patient prognosis, which shows that the CXCL-iFibro population is an early player in CKD progression and demonstrates the clinical relevance of our findings.
Collapse
Affiliation(s)
- Camille Cohen
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Rana Mhaidly
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Hugo Croizer
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Yann Kieffer
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Renaud Leclere
- Department of Diagnostic and Theragnostic Medicine, Institut Curie Hospital Group, 26, rue d'Ulm, F-75248, Paris, France
| | - Anne Vincent-Salomon
- Department of Diagnostic and Theragnostic Medicine, Institut Curie Hospital Group, 26, rue d'Ulm, F-75248, Paris, France
| | - Catherine Robley
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Dany Anglicheau
- Department of Nephrology and Kidney Transplantation, Necker Hospital, AP-HP, Paris Cité University, Inserm U1151, 149 rue de Sèvres, 75015, Paris, France
| | - Marion Rabant
- Department of Pathology, Necker Hospital, AP-HP, Paris Cité University, 149 rue de Sèvres, 75015, Paris, France
| | - Aurélie Sannier
- Department of Pathology, AP-HP, Bichat-Claude Bernard Hospital, Paris Cité University, Inserm, U1148, 46, rue Henri Huchard, 75877, Paris, France
| | - Marc-Olivier Timsit
- Department of Urology, Européen George Pompidou Hospital, APHP, Paris Cité University, Paris, France
| | - Sean Eddy
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Matthias Kretzler
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Wenjun Ju
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Fatima Mechta-Grigoriou
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France.
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France.
| |
Collapse
|
5
|
Klinkhammer BM, Boor P. Kidney fibrosis: Emerging diagnostic and therapeutic strategies. Mol Aspects Med 2023; 93:101206. [PMID: 37541106 DOI: 10.1016/j.mam.2023.101206] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/25/2023] [Indexed: 08/06/2023]
Abstract
An increasing number of patients worldwide suffers from chronic kidney disease (CKD). CKD is accompanied by kidney fibrosis, which affects all compartments of the kidney, i.e., the glomeruli, tubulointerstitium, and vasculature. Fibrosis is the best predictor of progression of kidney diseases. Currently, there is no specific anti-fibrotic therapy for kidney patients and invasive renal biopsy remains the only option for specific detection and quantification of kidney fibrosis. Here we review emerging diagnostic approaches and potential therapeutic options for fibrosis. We discuss how translational research could help to establish fibrosis-specific endpoints for clinical trials, leading to improved patient stratification and potentially companion diagnostics, and facilitating and optimizing development of novel anti-fibrotic therapies for kidney patients.
Collapse
Affiliation(s)
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany; Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany; Division of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
6
|
Nørregaard R, Mutsaers HAM, Frøkiær J, Kwon TH. Obstructive nephropathy and molecular pathophysiology of renal interstitial fibrosis. Physiol Rev 2023; 103:2827-2872. [PMID: 37440209 PMCID: PMC10642920 DOI: 10.1152/physrev.00027.2022] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 07/14/2023] Open
Abstract
The kidneys play a key role in maintaining total body homeostasis. The complexity of this task is reflected in the unique architecture of the organ. Ureteral obstruction greatly affects renal physiology by altering hemodynamics, changing glomerular filtration and renal metabolism, and inducing architectural malformations of the kidney parenchyma, most importantly renal fibrosis. Persisting pathological changes lead to chronic kidney disease, which currently affects ∼10% of the global population and is one of the major causes of death worldwide. Studies on the consequences of ureteral obstruction date back to the 1800s. Even today, experimental unilateral ureteral obstruction (UUO) remains the standard model for tubulointerstitial fibrosis. However, the model has certain limitations when it comes to studying tubular injury and repair, as well as a limited potential for human translation. Nevertheless, ureteral obstruction has provided the scientific community with a wealth of knowledge on renal (patho)physiology. With the introduction of advanced omics techniques, the classical UUO model has remained relevant to this day and has been instrumental in understanding renal fibrosis at the molecular, genomic, and cellular levels. This review details key concepts and recent advances in the understanding of obstructive nephropathy, highlighting the pathophysiological hallmarks responsible for the functional and architectural changes induced by ureteral obstruction, with a special emphasis on renal fibrosis.
Collapse
Affiliation(s)
- Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | - Jørgen Frøkiær
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu, Korea
| |
Collapse
|
7
|
Mohammed Y, Goodlett D, Borchers CH. Bioinformatics Tools and Knowledgebases to Assist Generating Targeted Assays for Plasma Proteomics. Methods Mol Biol 2023; 2628:557-577. [PMID: 36781806 DOI: 10.1007/978-1-0716-2978-9_32] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
In targeted proteomics experiments, selecting the appropriate proteotypic peptides as surrogate for the target protein is a crucial pre-acquisition step. This step is largely a bioinformatics exercise that involves integrating information on the peptides and proteins and using various software tools and knowledgebases. We present here a few resources that automate and simplify the selection process to a great degree. These tools and knowledgebases were developed primarily to streamline targeted proteomics assay development and include PeptidePicker, PeptidePickerDB, MRMAssayDB, MouseQuaPro, and PeptideTracker. We have used these tools to develop and document thousands of targeted proteomics assays, many of them for plasma proteins with focus on human and mouse. An important aspect in all these resources is the integrative approach on which they are based. Using these tools in the first steps of designing a singleplexed or multiplexed targeted proteomic experiment can reduce the necessary experimental steps tremendously. All the tools and knowledgebases we describe here are Web-based and freely accessible so scientists can query the information conveniently from the browser. This chapter provides an overview of these software tools and knowledgebases, their content, and how to use them for targeted plasma proteomics. We further demonstrate how to use them with the results of the HUPO Human Plasma Proteome Project to produce a new database of 3.8 k targeted assays for known human plasma proteins. Upon experimental validation, these assays should help in the further quantitative characterizing of the plasma proteome.
Collapse
Affiliation(s)
- Yassene Mohammed
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, ZA, Netherlands. .,University of Victoria - Genome BC Proteomics Centre, Victoria, BC, Canada. .,Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada.
| | - David Goodlett
- University of Victoria - Genome BC Proteomics Centre, Victoria, BC, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada.,University of Gdansk, International Centre for Cancer Vaccine Science, Gdansk, Poland
| | - Christoph H Borchers
- Proteomics Centre, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada.,Gerald Bronfman Department of Oncology, Jewish General Hospital, Montreal, QC, Canada.,Division of Experimental Medicine, McGill University, Montreal, QC, Canada.,Department of Pathology, McGill University, Montreal, QC, Canada
| |
Collapse
|
8
|
Snead AA, Clark RD. The Biological Hierarchy, Time, and Temporal 'Omics in Evolutionary Biology: A Perspective. Integr Comp Biol 2022; 62:1872-1886. [PMID: 36057775 DOI: 10.1093/icb/icac138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 01/05/2023] Open
Abstract
Sequencing data-genomics, transcriptomics, epigenomics, proteomics, and metabolomics-have revolutionized biological research, enabling a more detailed study of processes, ranging from subcellular to evolutionary, that drive biological organization. These processes, collectively, are responsible for generating patterns of phenotypic variation and can operate over dramatically different timescales (milliseconds to billions of years). While researchers often study phenotypic variation at specific levels of biological organization to isolate processes operating at that particular scale, the varying types of sequence data, or 'omics, can also provide complementary inferences to link molecular and phenotypic variation to produce an integrated view of evolutionary biology, ranging from molecular pathways to speciation. We briefly describe how 'omics has been used across biological levels and then demonstrate the utility of integrating different types of sequencing data across multiple biological levels within the same study to better understand biological phenomena. However, single-time-point studies cannot evaluate the temporal dynamics of these biological processes. Therefore, we put forward temporal 'omics as a framework that can better enable researchers to study the temporal dynamics of target processes. Temporal 'omics is not infallible, as the temporal sampling regime directly impacts inferential ability. Thus, we also discuss the role the temporal sampling regime plays in deriving inferences about the environmental conditions driving biological processes and provide examples that demonstrate the impact of the sampling regime on biological inference. Finally, we forecast the future of temporal 'omics by highlighting current methodological advancements that will enable temporal 'omics to be extended across species and timescales. We extend this discussion to using temporal multi-omics to integrate across the biological hierarchy to evaluate and link the temporal dynamics of processes that generate phenotypic variation.
Collapse
Affiliation(s)
- Anthony A Snead
- Department of Biological Sciences, University of Alabama, 300 Hackberry Lane, Tuscaloosa, AL 35487, USA
| | - René D Clark
- Department of Ecology, Evolution and Natural Resources, Rutgers University, 14 College Farm Road, New Brunswick, NJ 08901, USA
| |
Collapse
|
9
|
Swanson ML, Regner KR, Moore BM, Park F. Cannabinoid Type 2 Receptor Activation Reduces the Progression of Kidney Fibrosis Using a Mouse Model of Unilateral Ureteral Obstruction. Cannabis Cannabinoid Res 2022; 7:790-803. [PMID: 35196117 DOI: 10.1089/can.2021.0127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Background: Kidney fibrosis is a hallmark consequence of all forms of chronic kidney disease with few available treatment modalities. Material and Methods: In this study, we performed the unilateral ureteral obstruction (UUO) procedure to investigate the effects of a selective cannabinoid type 2 (CB2) agonist receptor, SMM-295, as a nephroprotective therapy. Results: SMM-295 was demonstrated to exhibit 50-fold selectivity over the cannabinoid type 1 (CB1) receptor with an EC50 ∼2 nM. Four other off-targets were identified in the safety panel, but only at the highest concentration (5 mM) tested in the assay demonstrating the relative selectivity and safety of our compound. Administration of SMM-295 (12 mg/kg IP daily) in UUO mice led to a significant decrease of 33% in tubular damage compared to the vehicle-treated UUO mice after 7 days. Consistent with these findings, there was a significant decrease in α-smooth muscle actin and fibronectin, which are markers of tubulointerstitial fibrosis, as determined by Western blot analysis. DNA damage as detected by a classic marker, γ-H2AX, was significantly reduced by 50% in the SMM-295 treatment group compared to vehicle treatment. Genetic knockout of CB2 or administration of a CB2 inverse agonist did not exhibit any beneficial effect on tubulointerstitial fibrosis or kidney tubule injury. Conclusions: In conclusion, our study provides new evidence that SMM-295 can therapeutically target the CB2 receptor with few, if any, physiological off-target sites to reduce kidney tissue damage and slow the fibrotic progression in a mouse model of kidney fibrosis.
Collapse
Affiliation(s)
- Mallory L Swanson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Kevin R Regner
- Division of Nephrology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Bob M Moore
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Frank Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
10
|
Xu S, Yang X, Chen Q, Liu Z, Chen Y, Yao X, Xiao A, Tian J, Xie L, Zhou M, Hu Z, Zhu F, Xu X, Hou F, Nie J. Leukemia inhibitory factor is a therapeutic target for renal interstitial fibrosis. EBioMedicine 2022; 86:104312. [PMID: 36335669 PMCID: PMC9646860 DOI: 10.1016/j.ebiom.2022.104312] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The role of the IL6 family members in organ fibrosis, including renal interstitial fibrosis (TIF), has been widely explored. However, few studies have ever simultaneously examined them in the same cohort of patients. Besides, the role of leukemia inhibitory factor (LIF) in TIF remains unclear. METHODS RNA-seq data of kidney biopsies from chronic kidney disease (CKD) patients, in both public databases and our assays, were used to analyze transcript levels of IL6 family members. Two TIF mouse models, the unilateral ureteral obstruction (UUO) and the ischemia reperfusion injury (IRI), were employed to validate the finding. To assess the role of LIF in vivo, short hairpin RNA, lenti-GFP-LIF was used to knockdown LIF receptor (LIFR), overexpress LIF, respectively. LIF-neutralizing antibody was used in therapeutic studies. Whether urinary LIF could be used as a promising predictor for CKD progression was investigated in a prospective observation patient cohort. FINDINGS Among IL6 family members, LIF is the most upregulated one in both human and mouse renal fibrotic lesions. The mRNA level of LIF negatively correlated with eGFR with the strongest correlation and the smallest P value. Baseline urinary concentrations of LIF in CKD patients predict the risk of CKD progression to end-stage kidney disease by Kaplan-Meier analysis. In mouse TIF models, knockdown of LIFR alleviated TIF; conversely, overexpressing LIF exacerbated TIF. Most encouragingly, visible efficacy against TIF was observed by administering LIF-neutralizing antibodies to mice. Mechanistically, LIF-LIFR-EGR1 axis and Sonic Hedgehog signaling formed a vicious cycle between fibroblasts and proximal tubular cells to augment LIF expression and promote the pro-fibrotic response via ERK and STAT3 activation. INTERPRETATION This study discovered that LIF is a noninvasive biomarker for the progression of CKD and a potential therapeutic target of TIF. FUNDINGS Stated in the Acknowledgements section of the manuscript.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Fanfan Hou
- Corresponding author. Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Jing Nie
- Corresponding author. Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
11
|
Shu S, Liu H, Yang J, Tang H, Li H, Liu Z, Zhou M, Zhu F, Hu Z, Ding K, Lu X, Nie J. Targeted inhibition of ZAK ameliorates renal interstitial fibrosis. Transl Res 2022; 246:49-65. [PMID: 35276386 DOI: 10.1016/j.trsl.2022.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 10/18/2022]
Abstract
ZAK (sterile alpha motif and leucine zipper-containing kinase) is a newly discovered member of the subfamily of mitogen-activated protein kinase kinase kinases (MAP3Ks). The role of ZAK in kidney disease remains largely unknown. In this study, we systematically investigated the expression and function of ZAK in the progression of tubulointerstitial fibrosis (TIF). ZAK was induced, predominantly in tubular epithelium, in both fibrotic kidneys of human and mouse models with TIF. ZAK expression level was correlated with the extent of renal fibrosis and the decline of eGFR of CKD patients. Depleting ZAK attenuated TIF and inflammation induced by unilateral ureteral occlusion (UUO) together with decreased activation of p38 MAPK and Smads signaling. Moreover, we demonstrated that overexpressed ZAK was in complex with Smad2/3 and TGF-β receptor Ⅰ (TβRI). Whereas, silencing endogenous ZAK ameliorated the amount of Smad2/3 recruited to TβRI. Moreover, we discovered a novel small molecule inhibitor of ZAK, named 6p. In vitro, incubation with 6p inhibited TGF-β1-induced fibrogenic response in NRK52E cells. In vivo, intragastric administration of 6p ameliorated TIF and inflammation in UUO and unilateral ischemia-reperfusion injury model. Delayed administration of 6p was also effective in retarding the progression of the established TIF. In conclusion, ZAK is a novel therapeutic target for TIF, and 6p might be a potential therapeutic agent for TIF.
Collapse
Affiliation(s)
- Shuangshuang Shu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Organ Failure Research, Ministry of Education, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Han Liu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Organ Failure Research, Ministry of Education, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Jianzhang Yang
- School of Pharmacy, Jinan University, Guangzhou, P. R. China
| | - Haie Tang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Organ Failure Research, Ministry of Education, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Hao Li
- State Key Laboratory of Organ Failure Research, Key Laboratory of Organ Failure Research, Ministry of Education, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Zhuoliang Liu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Organ Failure Research, Ministry of Education, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Miaomiao Zhou
- State Key Laboratory of Organ Failure Research, Key Laboratory of Organ Failure Research, Ministry of Education, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Fengxin Zhu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Organ Failure Research, Ministry of Education, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Zheng Hu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Organ Failure Research, Ministry of Education, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Ke Ding
- School of Pharmacy, Jinan University, Guangzhou, P. R. China
| | - Xiaoyun Lu
- School of Pharmacy, Jinan University, Guangzhou, P. R. China
| | - Jing Nie
- State Key Laboratory of Organ Failure Research, Key Laboratory of Organ Failure Research, Ministry of Education, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China.
| |
Collapse
|
12
|
Su H, Liu H, Yang K, Chen W, Yuan D, Wang W, Zhu G, Hu B, Jiang K, Zhu J. Construction of a novel rabbit model of ureteral calculi implanted with flowable resin. BMC Urol 2022; 22:105. [PMID: 35850713 PMCID: PMC9295447 DOI: 10.1186/s12894-022-01056-x] [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: 05/06/2022] [Accepted: 07/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The purpose of this study was to characterize the pathophysiological changes of hydronephrosis caused by ureteral calculi obstruction in a new rabbit ureteral calculi model by implanting flowable resin. METHODS Forty New Zealand rabbits were randomly divided into two groups: the calculi group and the sham control group. In the calculi group (n = 20), rabbits were operated at left lower abdomen and the left ureter was exposed. Then flowable resin (flowable restorative dental materials) was injected into the left ureter using a 0.45 mm diameter intravenous infusion needle. Then light-cured for 40 s by means of a dental curing light to form calculi. In the sham control group, normal saline was injected into the ureter. Rabbits underwent X-ray and routine blood and urine tests preoperatively, as well as X-ray, CT, dissection, HE staining and routine blood and urine tests on 1, 3, 5 and 7 days postoperatively. Stone formation was assessed by X-ray and unenhanced CT scan after surgery. The pathophysiological changes were evaluated through dissection, HE staining and routine blood and urine tests. RESULTS Ureteral calculi models were successfully constructed in 17 rabbits. In calculi group, high-density shadows were observed in the left lower abdomen on postoperative day 1st, 3rd, 5th and 7th by X-ray and CT scan. Dissection found obstruction formation of the left ureters, dilatation of the renal pelvis and upper ureter during 7 days after surgery. The renal long-diameters of the left ureters increased only on the 1st postoperative day. HE staining found ureteral and kidney damage after surgery. In calculi group and sham group,the serum creatinine, urea nitrogen, white blood cells and urine red blood cells were raised at day 1 after surgery. However, the indicators returned to normal at day 3, 5, and 7. CONCLUSIONS This is a stable, less complicated operation and cost-effective ureteral calculi model by implanting flowable resin. And this novel model may allow us to further understand the pathophysiology changes caused by ureteral calculi obstruction.
Collapse
Affiliation(s)
- Hao Su
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou Province, China.,Zunyi Medical University, Zunyi, 563000, Guizhou Province, China
| | - Heng Liu
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou Province, China.,Zunyi Medical University, Zunyi, 563000, Guizhou Province, China
| | - Ke Yang
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou Province, China.,Guizhou Medical University, Guiyang, 550002, Guizhou Province, China
| | - Weiming Chen
- Guizhou Medical University, Guiyang, 550002, Guizhou Province, China
| | - Dongbo Yuan
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou Province, China.,Guizhou University School of Medicine, Guiyang, 550025, Guizhou Province, China
| | - Wei Wang
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou Province, China
| | - Guohua Zhu
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou Province, China
| | - Bin Hu
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou Province, China
| | - Kehua Jiang
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou Province, China.
| | - Jianguo Zhu
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou Province, China. .,Zunyi Medical University, Zunyi, 563000, Guizhou Province, China. .,Guizhou Medical University, Guiyang, 550002, Guizhou Province, China. .,Guizhou University School of Medicine, Guiyang, 550025, Guizhou Province, China.
| |
Collapse
|
13
|
Lidberg KA, Muthusamy S, Adil M, Mahadeo A, Yang J, Patel RS, Wang L, Bammler TK, Reichel J, Yeung CK, Himmelfarb J, Kelly EJ, Akilesh S. Serum Protein Exposure Activates a Core Regulatory Program Driving Human Proximal Tubule Injury. J Am Soc Nephrol 2022; 33:949-965. [PMID: 35197326 PMCID: PMC9063895 DOI: 10.1681/asn.2021060751] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 02/06/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The kidneys efficiently filter waste products while retaining serum proteins in the circulation. However, numerous diseases compromise this barrier function, resulting in spillage of serum proteins into the urine (proteinuria). Some studies of glomerular filtration suggest that tubules may be physiologically exposed to nephrotic-range protein levels. Therefore, whether serum components can directly injure the downstream tubular portions of the kidney, which in turn can lead to inflammation and fibrosis, remains controversial. METHODS We tested the effects of serum protein exposure in human kidney tubule microphysiologic systems and with orthogonal epigenomic approaches since animal models cannot directly assess the effect of serum components on tubules. RESULTS Serum, but not its major protein component albumin, induced tubular injury and secretion of proinflammatory cytokines. Epigenomic comparison of serum-injured tubules and intact kidney tissue revealed canonical stress-inducible regulation of injury-induced genes. Concordant transcriptional changes in microdissected tubulointerstitium were also observed in an independent cohort of patients with proteinuric kidney disease. CONCLUSIONS Our results demonstrate a causal role for serum proteins in tubular injury and identify regulatory mechanisms and novel pathways for intervention.
Collapse
Affiliation(s)
- Kevin A. Lidberg
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Selvaraj Muthusamy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Mohamed Adil
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Anish Mahadeo
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Jade Yang
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | | | - Lu Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Theo K. Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Jonathan Reichel
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Catherine K. Yeung
- Department of Pharmacy, University of Washington, Seattle, Washington
- Kidney Research Institute, Seattle, Washington
| | - Jonathan Himmelfarb
- Kidney Research Institute, Seattle, Washington
- Nephrology Division, Department of Medicine, University of Washington, Seattle, Washington
| | - Edward J. Kelly
- Department of Pharmaceutics, University of Washington, Seattle, Washington
- Kidney Research Institute, Seattle, Washington
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
- Kidney Research Institute, Seattle, Washington
| |
Collapse
|
14
|
A novel renal perivascular mesenchymal cell subset gives rise to fibroblasts distinct from classic myofibroblasts. Sci Rep 2022; 12:5389. [PMID: 35354870 PMCID: PMC8967907 DOI: 10.1038/s41598-022-09331-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
Perivascular mesenchymal cells (PMCs), which include pericytes, give rise to myofibroblasts that contribute to chronic kidney disease progression. Several PMC markers have been identified; however, PMC heterogeneity and functions are not fully understood. Here, we describe a novel subset of renal PMCs that express Meflin, a glycosylphosphatidylinositol-anchored protein that was recently identified as a marker of fibroblasts essential for cardiac tissue repair. Tracing the lineage of Meflin+ PMCs, which are found in perivascular and periglomerular areas and exhibit renin-producing potential, showed that they detach from the vasculature and proliferate under disease conditions. Although the contribution of Meflin+ PMCs to conventional α-SMA+ myofibroblasts is low, they give rise to fibroblasts with heterogeneous α-SMA expression patterns. Genetic ablation of Meflin+ PMCs in a renal fibrosis mouse model revealed their essential role in collagen production. Consistent with this, human biopsy samples showed that progressive renal diseases exhibit high Meflin expression. Furthermore, Meflin overexpression in kidney fibroblasts promoted bone morphogenetic protein 7 signals and suppressed myofibroblastic differentiation, implicating the roles of Meflin in suppressing tissue fibrosis. These findings demonstrate that Meflin marks a PMC subset that is functionally distinct from classic pericytes and myofibroblasts, highlighting the importance of elucidating PMC heterogeneity.
Collapse
|
15
|
Doke T, Huang S, Qiu C, Sheng X, Seasock M, Liu H, Ma Z, Palmer M, Susztak K. Genome-wide association studies identify the role of caspase-9 in kidney disease. SCIENCE ADVANCES 2021; 7:eabi8051. [PMID: 34739325 PMCID: PMC8570608 DOI: 10.1126/sciadv.abi8051] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/16/2021] [Indexed: 06/13/2023]
Abstract
Genome-wide association studies (GWAS) have identified hundreds of genetic risk regions for kidney dysfunction [estimated glomerular filtration rate (eGFR)]; however, the causal genes, cell types, and pathways are poorly understood. Integration of GWAS and human kidney expression of quantitative trait analysis using Bayesian colocations, transcriptome-wide association studies, and summary-based Mendelian randomization studies prioritized caspase-9 (CASP9) as a kidney disease risk gene. Human kidney single-cell epigenetic and immunostaining studies indicated kidney tubule cells as a disease-causing cell type. Mice with genetic deletion or pharmacological inhibition of CASP9 showed lower apoptosis while having improved mitophagy, resulting in dampened activation of cytosolic nucleotide sensing pathways (cGAS-STING), reduction of inflammation, and protection from acute kidney disease or renal fibrosis. In summary, here, we prioritized CASP9 as an eGFR GWAS target gene and demonstrated the causal role of CASP9 in kidney disease development via improving mitophagy and lowering inflammation and apoptosis.
Collapse
Affiliation(s)
- Tomohito Doke
- Renal Electrolyte and Hypertension Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shizheng Huang
- Renal Electrolyte and Hypertension Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chengxiang Qiu
- Renal Electrolyte and Hypertension Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xin Sheng
- Renal Electrolyte and Hypertension Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew Seasock
- Renal Electrolyte and Hypertension Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongbo Liu
- Renal Electrolyte and Hypertension Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ziyuan Ma
- Renal Electrolyte and Hypertension Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew Palmer
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
16
|
Sun CM, Zhang WY, Wang SY, Qian G, Pei DL, Zhang GM. Fer exacerbates renal fibrosis and can be targeted by miR-29c-3p. Open Med (Wars) 2021; 16:1378-1385. [PMID: 34595351 PMCID: PMC8439263 DOI: 10.1515/med-2021-0319] [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: 09/19/2020] [Revised: 06/05/2021] [Accepted: 06/17/2021] [Indexed: 11/15/2022] Open
Abstract
Aim Renal fibrosis (RF) is a common clinical condition leading to irreversible renal function loss. Tyrosine kinase proteins and microRNAs (miRs) are associated with pathogenesis and we aim to investigate the role of Fer and its partner miR(s) in RF. Method In silico reproduction of Mouse Kidney FibrOmics browser was performed to identify potential miR(s) and target gene(s). In vivo validation was performed in C57BL/6 mice with unilateral ureteral obstruction (UUO). In vitro validation was performed in rat kidney fibroblast NRK-49F cells. Mimics and inhibitors of miR-29c-3p were constructed. The target gene Fer was monitored by RT-PCR and western blotting. The levels of interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α in serum and media were measured by ELISA. Results The Fer expression and protein level were gradually increased during 14 days of UUO modeling. miR-29c-3p expression was strongly correlated with that of Fer. In vivo validation showed increased expressions of fibrosis-associated genes and increased phospoho-Smad3 level in the UUO model. Fer-knockdown (KD) significantly decreased expressions of fibrosis-associated genes. Pharmaceutical inhibition of Fer showed similar effects to miR-29c-3p, and miR inhibition showed a significant decrease of excretion of inflammatory factors. Conclusion Dysregulation of miR-29c-3p and Fer plays a role in RF. Pharmaceutical or genetic inhibition of Fer may serve as the potential treatment for RF.
Collapse
Affiliation(s)
- Chen-Min Sun
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Wen-Yi Zhang
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Shu-Yan Wang
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Gang Qian
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Dong-Liang Pei
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Guang-Ming Zhang
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| |
Collapse
|
17
|
Wu Q, Fenton RA. Urinary proteomics for kidney dysfunction: insights and trends. Expert Rev Proteomics 2021; 18:437-452. [PMID: 34187288 DOI: 10.1080/14789450.2021.1950535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Introduction: Kidney dysfunction poses a high burden on patients and health care systems. Early detection and accurate prediction of kidney disease progression remains a major challenge. Compared to existing clinical parameters, urinary proteomics has the potential to reveal molecular alterations within the kidney that may alter its function before the onset of clinical symptoms. Thus, urinary proteomics has greater prognostic potential for assessment of kidney dysfunction progression.Areas covered: Advances in urinary proteomics for major causes of kidney dysfunction are discussed. The application of urinary extracellular vesicles for studying kidney dysfunction are discussed. Technological advances in urinary proteomics are discussed. The literature was identified using a database search for titles containing 'proteom*' and 'urin*' and published within the past 5 years. Retrieved literature was manually filtered to retain kidney dysfunctions-related studies.Expert opinion: Despite major advances, diagnosis by urinary proteomics has not been fully applied in any clinical settings. This could be attributed to the complex nature of kidney diseases, in addition to the constraints on study power and feasibility of incorporating mass spectrometry techniques in daily routine analysis. Nevertheless, we are confident that advances in urinary proteomics will soon provide superior insights into kidney disease beyond existing clinical parameters.
Collapse
Affiliation(s)
- Qi Wu
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Robert A Fenton
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
18
|
Higgins CE, Tang J, Higgins SP, Gifford CC, Mian BM, Jones DM, Zhang W, Costello A, Conti DJ, Samarakoon R, Higgins PJ. The Genomic Response to TGF-β1 Dictates Failed Repair and Progression of Fibrotic Disease in the Obstructed Kidney. Front Cell Dev Biol 2021; 9:678524. [PMID: 34277620 PMCID: PMC8284093 DOI: 10.3389/fcell.2021.678524] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Tubulointerstitial fibrosis is a common and diagnostic hallmark of a spectrum of chronic renal disorders. While the etiology varies as to the causative nature of the underlying pathology, persistent TGF-β1 signaling drives the relentless progression of renal fibrotic disease. TGF-β1 orchestrates the multifaceted program of kidney fibrogenesis involving proximal tubular dysfunction, failed epithelial recovery or re-differentiation, capillary collapse and subsequent interstitial fibrosis eventually leading to chronic and ultimately end-stage disease. An increasing complement of non-canonical elements function as co-factors in TGF-β1 signaling. p53 is a particularly prominent transcriptional co-regulator of several TGF-β1 fibrotic-response genes by complexing with TGF-β1 receptor-activated SMADs. This cooperative p53/TGF-β1 genomic cluster includes genes involved in cellular proliferative control, survival, apoptosis, senescence, and ECM remodeling. While the molecular basis for this co-dependency remains to be determined, a subset of TGF-β1-regulated genes possess both p53- and SMAD-binding motifs. Increases in p53 expression and phosphorylation, moreover, are evident in various forms of renal injury as well as kidney allograft rejection. Targeted reduction of p53 levels by pharmacologic and genetic approaches attenuates expression of the involved genes and mitigates the fibrotic response confirming a key role for p53 in renal disorders. This review focuses on mechanisms underlying TGF-β1-induced renal fibrosis largely in the context of ureteral obstruction, which mimics the pathophysiology of pediatric unilateral ureteropelvic junction obstruction, and the role of p53 as a transcriptional regulator within the TGF-β1 repertoire of fibrosis-promoting genes.
Collapse
Affiliation(s)
- Craig E. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Jiaqi Tang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Stephen P. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Cody C. Gifford
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Badar M. Mian
- The Urological Institute of Northeastern New York, Albany, NY, United States
- Division of Urology, Department of Surgery, Albany Medical College, Albany, NY, United States
| | - David M. Jones
- Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, NY, United States
| | - Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Angelica Costello
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - David J. Conti
- Division of Transplantation Surgery, Department of Surgery, Albany Medical College, Albany, NY, United States
| | - Rohan Samarakoon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Paul J. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
- The Urological Institute of Northeastern New York, Albany, NY, United States
- Division of Urology, Department of Surgery, Albany Medical College, Albany, NY, United States
| |
Collapse
|
19
|
Dolivo D, Rodrigues A, Sun L, Li Y, Hou C, Galiano R, Hong SJ, Mustoe T. The Na x (SCN7A) channel: an atypical regulator of tissue homeostasis and disease. Cell Mol Life Sci 2021; 78:5469-5488. [PMID: 34100980 PMCID: PMC11072345 DOI: 10.1007/s00018-021-03854-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/15/2021] [Accepted: 05/08/2021] [Indexed: 12/15/2022]
Abstract
Within an articulately characterized family of ion channels, the voltage-gated sodium channels, exists a black sheep, SCN7A (Nax). Nax, in contrast to members of its molecular family, has lost its voltage-gated character and instead rapidly evolved a new function as a concentration-dependent sensor of extracellular sodium ions and subsequent signal transducer. As it deviates fundamentally in function from the rest of its family, and since the bulk of the impressive body of literature elucidating the pathology and biochemistry of voltage-gated sodium channels has been performed in nervous tissue, reports of Nax expression and function have been sparse. Here, we investigate available reports surrounding expression and potential roles for Nax activity outside of nervous tissue. With these studies as justification, we propose that Nax likely acts as an early sensor that detects loss of tissue homeostasis through the pathological accumulation of extracellular sodium and/or through endothelin signaling. Sensation of homeostatic aberration via Nax then proceeds to induce pathological tissue phenotypes via promotion of pro-inflammatory and pro-fibrotic responses, induced through direct regulation of gene expression or through the generation of secondary signaling molecules, such as lactate, that can operate in an autocrine or paracrine fashion. We hope that our synthesis of much of the literature investigating this understudied protein will inspire more research into Nax not simply as a biochemical oddity, but also as a potential pathophysiological regulator and therapeutic target.
Collapse
Affiliation(s)
- David Dolivo
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Adrian Rodrigues
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Lauren Sun
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Yingxing Li
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Chun Hou
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
- Department of Plastic and Cosmetic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Robert Galiano
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Seok Jong Hong
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA.
- , 300 E. Superior St., Chicago, IL, 60611, USA.
| | - Thomas Mustoe
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA.
- , 737 N. Michigan Ave., Chicago, IL, 60611, USA.
| |
Collapse
|
20
|
Patel K, Chandrasegaran S, Clark IM, Proctor CJ, Young DA, Shanley DP. TimiRGeN: R/Bioconductor package for time series microRNA-mRNA integration and analysis. Bioinformatics 2021; 37:3604-3609. [PMID: 33993215 PMCID: PMC8545325 DOI: 10.1093/bioinformatics/btab377] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/09/2021] [Accepted: 05/12/2021] [Indexed: 12/11/2022] Open
Abstract
Motivation The analysis of longitudinal datasets and construction of gene regulatory networks (GRNs) provide a valuable means to disentangle the complexity of microRNA (miRNA)–mRNA interactions. However, there are no computational tools that can integrate, conduct functional analysis and generate detailed networks from longitudinal miRNA–mRNA datasets. Results We present TimiRGeN, an R package that uses time point-based differential expression results to identify miRNA–mRNA interactions influencing signaling pathways of interest. miRNA–mRNA interactions can be visualized in R or exported to PathVisio or Cytoscape. The output can be used for hypothesis generation and directing in vitro or further in silico work such as GRN construction. Availability and implementation TimiRGeN is available for download on Bioconductor (https://bioconductor.org/packages/TimiRGeN) and requires R v4.0.2 or newer and BiocManager v3.12 or newer. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- K Patel
- Campus for Ageing and Vitality, Biosciences Institute, Newcastle University, Newcastle upon-Tyne, NE4 5PL, UK
| | - S Chandrasegaran
- Campus for Ageing and Vitality, Biosciences Institute, Newcastle University, Newcastle upon-Tyne, NE4 5PL, UK
| | - I M Clark
- School of Biological Sciences, University of East Anglia, Norwich, NR4 7TJ, UK
| | - C J Proctor
- Campus for Ageing and Vitality, Biosciences Institute, Newcastle University, Newcastle upon-Tyne, NE4 5PL, UK
| | - D A Young
- Life Science Centre, Biosciences Institute, Newcastle University, Newcastle, upon, UK Tyne, NE1 4EP
| | - D P Shanley
- Campus for Ageing and Vitality, Biosciences Institute, Newcastle University, Newcastle upon-Tyne, NE4 5PL, UK
| |
Collapse
|
21
|
Mohammed Y, Bhowmick P, Michaud SA, Sickmann A, Borchers CH. Mouse Quantitative Proteomics Knowledgebase: reference protein concentration ranges in 20 mouse tissues using 5000 quantitative proteomics assays. Bioinformatics 2021; 37:1900-1908. [PMID: 33483739 DOI: 10.1093/bioinformatics/btab018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/12/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022] Open
Abstract
Laboratory mouse is the most used animal model in biological research, largely due to its high conserved synteny with human. Researchers use mice to answer various questions ranging from determining a pathological effect of knocked out/in gene to understanding drug metabolism. Our group developed >5000 quantitative targeted proteomics assays for 20 mouse tissues and determined the concentration ranges of a total of more than 1600 proteins using heavy labelled internal standards. We describe here MouseQuaPro; a knowledgebase that hosts this collection of carefully curated experimental data. The Web-based application includes protein concentrations from >700 mouse tissue samples from three common research strains, corresponding to more than 200k experimentally determined concentrations. The knowledgebase integrates the assay and protein concentration information with their human orthologs, functional and molecular annotations, biological pathways, related human diseases, and known gene expressions. At its core are the protein concentration ranges, which provide insights into (dis)similarities between tissues, strains, and sexes. MouseQuaPro implements advanced search as well as filtering functionalities with a simple interface and interactive visualization. This information-rich resource provides an initial map of protein absolute concentration in mouse tissues and allows guided design of proteomics phenotyping experiments. The knowledgebase is available at mousequapro.proteincentre.com. (Reviewer access username and password: mousequapro_reviewer1234567).
Collapse
Affiliation(s)
- Yassene Mohammed
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada.,Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Pallab Bhowmick
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada
| | - Sarah A Michaud
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada
| | - Albert Sickmann
- Leibniz Institut für Analytische Wissenschaften-ISAS-e. V, Dortmund, Germany
| | - Christoph H Borchers
- University of Victoria, Victoria, BC, Canada.,Gerald Bronfman Department of Oncology, Jewish General Hospital, Montreal, Quebec, Canada
| |
Collapse
|
22
|
Kim JE, Han D, Jeong JS, Moon JJ, Moon HK, Lee S, Kim YC, Yoo KD, Lee JW, Kim DK, Kwon YJ, Kim YS, Yang SH. Multisample Mass Spectrometry-Based Approach for Discovering Injury Markers in Chronic Kidney Disease. Mol Cell Proteomics 2021; 20:100037. [PMID: 33453410 PMCID: PMC7950200 DOI: 10.1074/mcp.ra120.002159] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 11/15/2020] [Accepted: 12/20/2020] [Indexed: 12/16/2022] Open
Abstract
Urinary proteomics studies have primarily focused on identifying markers of chronic kidney disease (CKD) progression. Here, we aimed to determine urinary markers of CKD renal parenchymal injury through proteomics analysis in animal kidney tissues and cells and in the urine of patients with CKD. Label-free quantitative proteomics analysis based on liquid chromatography-tandem mass spectrometry was performed on urine samples obtained from 6 normal controls and 9, 11, and 10 patients with CKD stages 1, 3, and 5, respectively, and on kidney tissue samples from a rat CKD model by 5/6 nephrectomy. Tandem mass tag-based quantitative proteomics analysis was performed for glomerular endothelial cells (GECs) and proximal tubular epithelial cells (PTECs) before and after inducing 24-h hypoxia injury. Upon hierarchical clustering, out of 858 differentially expressed proteins (DEPs) in the urine of CKD patients, the levels of 416 decreased and 403 increased sequentially according to the disease stage, respectively. Among 2965 DEPs across 5/6 nephrectomized and sham-operated rat kidney tissues, 86 DEPs showed same expression patterns in the urine and kidney tissue. After cross-validation with two external animal proteome data sets, 38 DEPs were organized; only ten DEPs, including serotransferrin, gelsolin, poly ADP-ribose polymerase 1, neuroblast differentiation-associated protein AHNAK, microtubule-associated protein 4, galectin-1, protein S, thymosin beta-4, myristoylated alanine-rich C-kinase substrate, and vimentin, were finalized by screening human GECs and PTECs data. Among these ten potential candidates for universal CKD marker, validation analyses for protein S and galectin-1 were conducted. Galectin-1 was observed to have a significant inverse correlation with renal function as well as higher expression in glomerulus with chronic injury than protein S. This constitutes the first multisample proteomics study for identifying key renal-expressed proteins associated with CKD progression. The discovered proteins represent potential markers of chronic renal cell and tissue damage and candidate contributors to CKD pathophysiology.
Collapse
Affiliation(s)
- Ji Eun Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea; Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Dohyun Han
- Proteomics Core Facility, Seoul National University Hospital, Seoul, Korea; Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Jin Seon Jeong
- Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Korea
| | - Jong Joo Moon
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hyun Kyung Moon
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Sunhwa Lee
- Department of Internal Medicine, Kangwon National University Hospital, Gangwon-Do, Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kyung Don Yoo
- Department of Internal Medicine, Ulsan University Hospital, Ulsan, Korea
| | - Jae Wook Lee
- Nephrology Clinic, National Cancer Center, Goyang, Gyeonggi-do, Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea; Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Young Joo Kwon
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea; Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Hee Yang
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea; Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
23
|
Smith ER, Hewitson TD. TGF-β1 is a regulator of the pyruvate dehydrogenase complex in fibroblasts. Sci Rep 2020; 10:17914. [PMID: 33087819 PMCID: PMC7578649 DOI: 10.1038/s41598-020-74919-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 09/07/2020] [Indexed: 12/21/2022] Open
Abstract
TGF-β1 reprograms metabolism in renal fibroblasts, inducing a switch from oxidative phosphorylation to aerobic glycolysis. However, molecular events underpinning this are unknown. Here we identify that TGF-β1 downregulates acetyl-CoA biosynthesis via regulation of the pyruvate dehydrogenase complex (PDC). Flow cytometry showed that TGF-β1 reduced the PDC subunit PDH-E1α in fibroblasts derived from injured, but not normal kidneys. An increase in expression of PDH kinase 1 (PDK1), and reduction in the phosphatase PDP1, were commensurate with net phosphorylation and inactivation of PDC. Over-expression of mutant PDH-E1α, resistant to phosphorylation, ameliorated effects of TGF-β1, while inhibition of PDC activity with CPI-613 was sufficient to induce αSMA and pro-collagen I expression, markers of myofibroblast differentiation and fibroblast activation. The effect of TGF-β1 on PDC activity, acetyl-CoA, αSMA and pro-collagen I was also ameliorated by sodium dichloroacetate, a small molecule inhibitor of PDK. A reduction in acetyl-CoA, and therefore acetylation substrate, also resulted in a generalised loss of protein acetylation with TGF-β1. In conclusion, TGF-β1 in part regulates fibroblast activation via effects on PDC activity.
Collapse
Affiliation(s)
- Edward R Smith
- Department of Nephrology, The Royal Melbourne Hospital (RMH), Grattan Street, Parkville, VIC, 3050, Australia.,Department of Medicine - RMH, University of Melbourne, Parkville, VIC, Australia
| | - Timothy D Hewitson
- Department of Nephrology, The Royal Melbourne Hospital (RMH), Grattan Street, Parkville, VIC, 3050, Australia. .,Department of Medicine - RMH, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
24
|
Ricard-Blum S, Miele AE. Omic approaches to decipher the molecular mechanisms of fibrosis, and design new anti-fibrotic strategies. Semin Cell Dev Biol 2020; 101:161-169. [DOI: 10.1016/j.semcdb.2019.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/16/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022]
|
25
|
Hashemi Gheinani A, Bigger-Allen A, Wacker A, Adam RM. Systems analysis of benign bladder disorders: insights from omics analysis. Am J Physiol Renal Physiol 2020; 318:F901-F910. [PMID: 32116016 DOI: 10.1152/ajprenal.00496.2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The signaling pathways and effectors that drive the response of the bladder to nonmalignant insults or injury are incompletely defined. Interrogation of biological systems has been revolutionized by the ability to generate high-content data sets that capture information on a variety of biomolecules in cells and tissues, from DNA to RNA to proteins. In oncology, such an approach has led to the identification of cancer subtypes, improved prognostic capability, and has provided a basis for precision treatment of patients. In contrast, systematic molecular characterization of benign bladder disorders has lagged behind, such that our ability to uncover novel therapeutic interventions or increase our mechanistic understanding of such conditions is limited. Here, we discuss existing literature on the application of omics approaches, including transcriptomics and proteomics, to urinary tract conditions characterized by pathological tissue remodeling. We discuss molecular pathways implicated in remodeling, challenges in the field, and aspirations for omics-based research in the future.
Collapse
Affiliation(s)
- Ali Hashemi Gheinani
- Department of Urology, Boston Children's Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - Alexander Bigger-Allen
- Department of Urology, Boston Children's Hospital, Boston, Massachusetts.,Biological and Biomedical Sciences PhD Program, Harvard Medical School, Boston, Massachusetts
| | - Amanda Wacker
- Department of Urology, Boston Children's Hospital, Boston, Massachusetts.,Florida State University, Tallahassee, Florida
| | - Rosalyn M Adam
- Department of Urology, Boston Children's Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
26
|
Conesa A, Beck S. Making multi-omics data accessible to researchers. Sci Data 2019; 6:251. [PMID: 31672978 PMCID: PMC6823467 DOI: 10.1038/s41597-019-0258-4] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 10/15/2019] [Indexed: 12/28/2022] Open
Abstract
A special collection on multi-omics data sharing, launched today at Scientific Data , offers to the scientific community a compendium of multi-omics datasets ready for reuse, which showcase the diversity of multi-omics projects and highlights innovative approaches for preprocessing, quality control, hosting and access.
Collapse
Affiliation(s)
- Ana Conesa
- Microbiology and Cell Science Department, Institute for Food and Agricultural Research, Genetics Institute, University of Florida, Gainesville, Florida, USA.
| | - Stephan Beck
- UCL Cancer institute, University College London, London, UK
| |
Collapse
|
27
|
Pavkovic M, Pantano L, Gerlach CV, Brutus S, Boswell SA, Everley RA, Shah JV, Sui SH, Vaidya VS. Multi omics analysis of fibrotic kidneys in two mouse models. Sci Data 2019; 6:92. [PMID: 31201317 PMCID: PMC6570759 DOI: 10.1038/s41597-019-0095-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 05/07/2019] [Indexed: 12/12/2022] Open
Abstract
Kidney fibrosis represents an urgent unmet clinical need due to the lack of effective therapies and an inadequate understanding of the molecular pathogenesis. We have generated a comprehensive and combined multi-omics dataset (proteomics, mRNA and small RNA transcriptomics) of fibrotic kidneys that is searchable through a user-friendly web application: http://hbcreports.med.harvard.edu/fmm/ . Two commonly used mouse models were utilized: a reversible chemical-induced injury model (folic acid (FA) induced nephropathy) and an irreversible surgically-induced fibrosis model (unilateral ureteral obstruction (UUO)). mRNA and small RNA sequencing, as well as 10-plex tandem mass tag (TMT) proteomics were performed with kidney samples from different time points over the course of fibrosis development. The bioinformatics workflow used to process, technically validate, and combine the single omics data will be described. In summary, we present temporal multi-omics data from fibrotic mouse kidneys that are accessible through an interrogation tool (Mouse Kidney Fibromics browser) to provide a searchable transcriptome and proteome for kidney fibrosis researchers.
Collapse
Affiliation(s)
- Mira Pavkovic
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Medicine - Renal Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Lorena Pantano
- Bioinformatics Core, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Cory V Gerlach
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Medicine - Renal Division, Brigham and Women's Hospital, Boston, MA, USA
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Sergine Brutus
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Sarah A Boswell
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Robert A Everley
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Jagesh V Shah
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Medicine - Renal Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Shannan H Sui
- Bioinformatics Core, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Vishal S Vaidya
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA.
- Department of Medicine - Renal Division, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|