1
|
Capelli I, Lerario S, Ciurli F, Berti GM, Aiello V, Provenzano M, La Manna G. Investigational agents for autosomal dominant polycystic kidney disease: preclinical and early phase study insights. Expert Opin Investig Drugs 2024; 33:469-484. [PMID: 38618918 DOI: 10.1080/13543784.2024.2342327] [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: 12/01/2023] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
INTRODUCTION Autosomal Dominant Polycystic Kidney Disease (ADPKD) is the most common inherited kidney condition caused by a single-gene mutation. It leads patients to kidney failure in more than 50% of cases by the age of 60, and, given the dominant inheritance, this disease is present in the family history in more than 90% of cases. AREAS COVERED This review aims to analyze the set of preclinical and early-phase studies to provide a general view of the current progress on ADPKD therapeutic options. Articles from PubMed and the current status of the trials listed in clinicaltrials.gov were examined for the review. EXPERT OPINION Many potential therapeutic targets are currently under study for the treatment of ADPKD. A few drugs have reached the clinical phase, while many are currently still in the preclinical phase. Organoids could be a novel approach to the study of drugs in this phase. Other than pharmacological options, very important developing approaches are represented by gene therapy and the use of MiRNA inhibitors.
Collapse
Affiliation(s)
- Irene Capelli
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Sarah Lerario
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Francesca Ciurli
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Gian Marco Berti
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Valeria Aiello
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Michele Provenzano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Gaetano La Manna
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
2
|
Abstract
Imaging mass spectrometry is a well-established technology that can easily and succinctly communicate the spatial localization of molecules within samples. This review communicates the recent advances in the field, with a specific focus on matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) applied on tissues. The general sample preparation strategies for different analyte classes are explored, including special considerations for sample types (fresh frozen or formalin-fixed,) strategies for various analytes (lipids, metabolites, proteins, peptides, and glycans) and how multimodal imaging strategies can leverage the strengths of each approach is mentioned. This work explores appropriate experimental design approaches and standardization of processes needed for successful studies, as well as the various data analysis platforms available to analyze data and their strengths. The review concludes with applications of imaging mass spectrometry in various fields, with a focus on medical research, and some examples from plant biology and microbe metabolism are mentioned, to illustrate the breadth and depth of MALDI IMS.
Collapse
Affiliation(s)
- Jessica L Moore
- Department of Proteomics, Discovery Life Sciences, Huntsville, Alabama 35806, United States
| | - Georgia Charkoftaki
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, Connecticut 06520, United States
| |
Collapse
|
3
|
Richardson LT, Neumann EK, Caprioli RM, Spraggins JM, Solouki T. Referenced Kendrick Mass Defect Annotation and Class-Based Filtering of Imaging MS Lipidomics Experiments. Anal Chem 2022; 94:5504-5513. [PMID: 35344335 PMCID: PMC10124143 DOI: 10.1021/acs.analchem.1c03715] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Because of their diverse functionalities in cells, lipids are of primary importance when characterizing molecular profiles of physiological and disease states. Imaging mass spectrometry (IMS) provides the spatial distributions of lipid populations in tissues. Referenced Kendrick mass defect (RKMD) analysis is an effective mass spectrometry (MS) data analysis tool for classification and annotation of lipids. Herein, we extend the capabilities of RKMD analysis and demonstrate an integrated method for lipid annotation and chemical structure-based filtering for IMS datasets. Annotation of lipid features with lipid molecular class, radyl carbon chain length, and degree of unsaturation allows image reconstruction and visualization based on each structural characteristic. We show a proof-of-concept application of the method to a computationally generated IMS dataset and validate that the RKMD method is highly specific for lipid components in the presence of confounding background ions. Moreover, we demonstrate an application of the RKMD-based annotation and filtering to matrix-assisted laser desorption/ionization (MALDI) IMS lipidomic data from human kidney tissue analysis.
Collapse
Affiliation(s)
- Luke T Richardson
- Department of Chemistry and Biochemistry, Baylor University, 101 Bagby Avenue, Waco, Texas 76706, United States
| | - Elizabeth K Neumann
- Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, Tennessee 37205, United States.,Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States
| | - Richard M Caprioli
- Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, Tennessee 37205, United States.,Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States.,Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Station B 351822, Nashville, Tennessee 37235, United States.,Department of Medicine, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States.,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jeffrey M Spraggins
- Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, Tennessee 37205, United States.,Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States.,Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Station B 351822, Nashville, Tennessee 37235, United States.,Department of Cell and Development Biology, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States
| | - Touradj Solouki
- Department of Chemistry and Biochemistry, Baylor University, 101 Bagby Avenue, Waco, Texas 76706, United States
| |
Collapse
|
4
|
Kruse ARS, Spraggins JM. Uncovering Molecular Heterogeneity in the Kidney With Spatially Targeted Mass Spectrometry. Front Physiol 2022; 13:837773. [PMID: 35222094 PMCID: PMC8874197 DOI: 10.3389/fphys.2022.837773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/04/2022] [Indexed: 02/06/2023] Open
Abstract
The kidney functions through the coordination of approximately one million multifunctional nephrons in 3-dimensional space. Molecular understanding of the kidney has relied on transcriptomic, proteomic, and metabolomic analyses of kidney homogenate, but these approaches do not resolve cellular identity and spatial context. Mass spectrometry analysis of isolated cells retains cellular identity but not information regarding its cellular neighborhood and extracellular matrix. Spatially targeted mass spectrometry is uniquely suited to molecularly characterize kidney tissue while retaining in situ cellular context. This review summarizes advances in methodology and technology for spatially targeted mass spectrometry analysis of kidney tissue. Profiling technologies such as laser capture microdissection (LCM) coupled to liquid chromatography tandem mass spectrometry provide deep molecular coverage of specific tissue regions, while imaging technologies such as matrix assisted laser desorption/ionization imaging mass spectrometry (MALDI IMS) molecularly profile regularly spaced tissue regions with greater spatial resolution. These technologies individually have furthered our understanding of heterogeneity in nephron regions such as glomeruli and proximal tubules, and their combination is expected to profoundly expand our knowledge of the kidney in health and disease.
Collapse
Affiliation(s)
- Angela R. S. Kruse
- Department of Biochemistry, Vanderbilt University, Nashville, TN, United States
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, United States
| | - Jeffrey M. Spraggins
- Department of Biochemistry, Vanderbilt University, Nashville, TN, United States
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
- *Correspondence: Jeffrey M. Spraggins,
| |
Collapse
|
5
|
Ma G, Zhao X, Guo M, Liu Y, Shi K, Guo C, Pan Y. 6-Glycosylaminoquinoline-assisted LDI MS for detection and imaging of small molecules with enhanced detection selectivity and sensitivity. Anal Chim Acta 2022; 1201:339620. [DOI: 10.1016/j.aca.2022.339620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/28/2022] [Accepted: 02/14/2022] [Indexed: 12/20/2022]
|
6
|
Vasileva VY, Sultanova RF, Sudarikova AV, Ilatovskaya DV. Insights Into the Molecular Mechanisms of Polycystic Kidney Diseases. Front Physiol 2021; 12:693130. [PMID: 34566674 PMCID: PMC8456103 DOI: 10.3389/fphys.2021.693130] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/10/2021] [Indexed: 12/18/2022] Open
Abstract
Autosomal dominant (AD) and autosomal recessive (AR) polycystic kidney diseases (PKD) are severe multisystem genetic disorders characterized with formation and uncontrolled growth of fluid-filled cysts in the kidney, the spread of which eventually leads to the loss of renal function. Currently, there are no treatments for ARPKD, and tolvaptan is the only FDA-approved drug that alleviates the symptoms of ADPKD. However, tolvaptan has only a modest effect on disease progression, and its long-term use is associated with many side effects. Therefore, there is still a pressing need to better understand the fundamental mechanisms behind PKD development. This review highlights current knowledge about the fundamental aspects of PKD development (with a focus on ADPKD) including the PC1/PC2 pathways and cilia-associated mechanisms, major molecular cascades related to metabolism, mitochondrial bioenergetics, and systemic responses (hormonal status, levels of growth factors, immune system, and microbiome) that affect its progression. In addition, we discuss new information regarding non-pharmacological therapies, such as dietary restrictions, which can potentially alleviate PKD.
Collapse
Affiliation(s)
| | - Regina F Sultanova
- Saint-Petersburg State Chemical Pharmaceutical University, St. Petersburg, Russia.,Department of Physiology, Augusta University, Augusta, GA, United States
| | | | | |
Collapse
|
7
|
Angelotti ML, Antonelli G, Conte C, Romagnani P. Imaging the kidney: from light to super-resolution microscopy. Nephrol Dial Transplant 2021; 36:19-28. [PMID: 31325314 PMCID: PMC7771978 DOI: 10.1093/ndt/gfz136] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Indexed: 12/13/2022] Open
Abstract
The important achievements in kidney physiological and pathophysiological mechanisms can largely be ascribed to progress in the technology of microscopy. Much of what we know about the architecture of the kidney is based on the fundamental descriptions of anatomic microscopists using light microscopy and later by ultrastructural analysis provided by electron microscopy. These two techniques were used for the first classification systems of kidney diseases and for their constant updates. More recently, a series of novel imaging techniques added the analysis in further dimensions of time and space. Confocal microscopy allowed us to sequentially visualize optical sections along the z-axis and the availability of specific analysis software provided a three-dimensional rendering of thicker tissue specimens. Multiphoton microscopy permitted us to simultaneously investigate kidney function and structure in real time. Fluorescence-lifetime imaging microscopy allowed to study the spatial distribution of metabolites. Super-resolution microscopy increased sensitivity and resolution up to nanoscale levels. With cryo-electron microscopy, researchers could visualize the individual biomolecules at atomic levels directly in the tissues and understand their interaction at subcellular levels. Finally, matrix-assisted laser desorption/ionization imaging mass spectrometry permitted the measuring of hundreds of different molecules at the same time on tissue sections at high resolution. This review provides an overview of available kidney imaging strategies, with a focus on the possible impact of the most recent technical improvements.
Collapse
Affiliation(s)
- Maria Lucia Angelotti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.,Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| | - Giulia Antonelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.,Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| | - Carolina Conte
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.,Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| | - Paola Romagnani
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.,Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| |
Collapse
|
8
|
Lukowski JK, Pamreddy A, Velickovic D, Zhang G, Pasa-Tolic L, Alexandrov T, Sharma K, Anderton CR. Storage Conditions of Human Kidney Tissue Sections Affect Spatial Lipidomics Analysis Reproducibility. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2020; 31:2538-2546. [PMID: 32897710 PMCID: PMC8162764 DOI: 10.1021/jasms.0c00256] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Lipids often are labile, unstable, and tend to degrade overtime, so it is of the upmost importance to study these molecules in their most native state. We sought to understand the optimal storage conditions for spatial lipidomic analysis of human kidney tissue sections. Specifically, we evaluated human kidney tissue sections on several different days throughout the span of a week using our established protocol for elucidating lipids using high mass resolution matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI). We studied kidney tissue sections stored under five different conditions: open stored at -80 °C, vacuumed sealed and stored at -80 °C, with matrix preapplied before storage at -80 °C, under a nitrogen atmosphere and stored at -80 °C, and at room temperature in a desiccator. Results were compared to data obtained from kidney tissue sections that were prepared and analyzed immediately after cryosectioning. Data was processed using METASPACE. After a week of storage, the sections stored at room temperature showed the largest amount of lipid degradation, while sections stored under nitrogen and at -80 °C retained the greatest number of overlapping annotations in relation to freshly cut tissue. Overall, we found that molecular degradation of the tissue sections was unavoidable over time, regardless of storage conditions, but storing tissue sections in an inert gas at low temperatures can curtail molecular degradation within tissue sections.
Collapse
Affiliation(s)
- Jessica K Lukowski
- Environmental Molecular Sciences Division, Pacific Northwest National Laboratory, Richland, Washington99352, United States
| | - Annapurna Pamreddy
- Center for Renal Precision Medicine, Division of Nephrology, Department of Medicine, The University of Texas Health, San Antonio, Texas 78284, United States
| | - Dusan Velickovic
- Environmental Molecular Sciences Division, Pacific Northwest National Laboratory, Richland, Washington99352, United States
| | - Guanshi Zhang
- Center for Renal Precision Medicine, Division of Nephrology, Department of Medicine, The University of Texas Health, San Antonio, Texas 78284, United States
- Audie L. Murphy Memorial VA Hospital, South Texas Veterans Health Care System, San Antonio, Texas 78284, United States
| | - Ljiljana Pasa-Tolic
- Environmental Molecular Sciences Division, Pacific Northwest National Laboratory, Richland, Washington99352, United States
| | - Theodore Alexandrov
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| | - Kumar Sharma
- Center for Renal Precision Medicine, Division of Nephrology, Department of Medicine, The University of Texas Health, San Antonio, Texas 78284, United States
- Audie L. Murphy Memorial VA Hospital, South Texas Veterans Health Care System, San Antonio, Texas 78284, United States
| | - Christopher R Anderton
- Environmental Molecular Sciences Division, Pacific Northwest National Laboratory, Richland, Washington99352, United States
- Center for Renal Precision Medicine, Division of Nephrology, Department of Medicine, The University of Texas Health, San Antonio, Texas 78284, United States
| |
Collapse
|
9
|
Barrientos RC, Zhang Q. Recent advances in the mass spectrometric analysis of glycosphingolipidome - A review. Anal Chim Acta 2020; 1132:134-155. [PMID: 32980104 PMCID: PMC7525043 DOI: 10.1016/j.aca.2020.05.051] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 05/17/2020] [Accepted: 05/21/2020] [Indexed: 12/30/2022]
Abstract
Aberrant expression of glycosphingolipids has been implicated in a myriad of diseases, but our understanding of the strucural diversity, spatial distribution, and biological function of this class of biomolecules remains limited. These challenges partly stem from a lack of sensitive tools that can detect, identify, and quantify glycosphingolipids at the molecular level. Mass spectrometry has emerged as a powerful tool poised to address most of these challenges. Here, we review the recent developments in analytical glycosphingolipidomics with an emphasis on sample preparation, mass spectrometry and tandem mass spectrometry-based structural characterization, label-free and labeling-based quantification. We also discuss the nomenclature of glycosphingolipids, and emerging technologies like ion mobility spectrometry in differentiation of glycosphingolipid isomers. The intrinsic advantages and shortcomings of each method are carefully critiqued in line with an individual's research goals. Finally, future perspectives on analytical sphingolipidomics are stated, including a need for novel and more sensive methods in isomer separation, low abundance species detection, and profiling the spatial distribution of glycosphingolipid molecular species in cells and tissues using imaging mass spectrometry.
Collapse
Affiliation(s)
- Rodell C Barrientos
- Department of Chemistry and Biochemistry, The University of North Carolina at Greensboro, Greensboro, NC, 27412, United States; UNCG Center for Translational Biomedical Research, NC Research Campus, Kannapolis, NC, 28081, United States
| | - Qibin Zhang
- Department of Chemistry and Biochemistry, The University of North Carolina at Greensboro, Greensboro, NC, 27412, United States; UNCG Center for Translational Biomedical Research, NC Research Campus, Kannapolis, NC, 28081, United States.
| |
Collapse
|
10
|
Nowling TK, Rodgers J, Thiyagarajan T, Wolf B, Bruner E, Sundararaj K, Molano I, Gilkeson G. Targeting glycosphingolipid metabolism as a potential therapeutic approach for treating disease in female MRL/lpr lupus mice. PLoS One 2020; 15:e0230499. [PMID: 32187230 PMCID: PMC7080257 DOI: 10.1371/journal.pone.0230499] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 03/03/2020] [Indexed: 12/19/2022] Open
Abstract
Glycosphingolipids (GSLs) hexosylceramides and lactosylceramides are elevated in lupus mice and human patients with nephritis. Whereas other renal diseases characterized by increased GSL levels are thought to be a result of upregulated GSL synthesis, our results suggest elevated hexosylceramides and lactosylceramides in lupus nephritis is a result of increased catabolism of ganglioside GM3 due to significantly increased neuraminidase (NEU) activity. Thus, we hypothesized GM3 would be decreased in lupus nephritis kidneys and blocking NEU activity would reduce GSLs and improve disease in lupus mice. Female MRL/lpr lupus mice were treated with water or the NEU inhibitor oseltamivir phosphate at the onset of proteinuria to block GSL catabolism. Age-matched (non-nephritic) female MRL/MpJ lupus mice served as controls. Renal GM3 levels were significantly higher in the nephritic MRL/lpr water-treated mice compared to non-nephritic MRL/MpJ mice, despite significantly increased renal NEU activity. Blocking GSL catabolism increased, rather than decreased, renal and urine GSL levels and disease was not significantly impacted. A pilot study treating MRL/lpr females with GlcCer synthase inhibitor Genz-667161 to block GSL synthesis resulted in a strong significant negative correlation between Genz-667161 dose and renal GSL hexosylceramide and GM3 levels. Splenomegaly was negatively correlated and serum IgG levels were marginally correlated with increasing Genz-667161 dose. These results suggest accumulation of renal GM3 may be due to dysregulation of one or more of the GSL ganglioside pathways and inhibiting GSL synthesis, but not catabolism, may be a therapeutic approach for treating lupus nephritis.
Collapse
Affiliation(s)
- Tamara K. Nowling
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- * E-mail:
| | - Jessalyn Rodgers
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Thirumagal Thiyagarajan
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Bethany Wolf
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Evelyn Bruner
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Kamala Sundararaj
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Ivan Molano
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Gary Gilkeson
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| |
Collapse
|
11
|
Natoli TA, Modur V, Ibraghimov-Beskrovnaya O. Glycosphingolipid metabolism and polycystic kidney disease. Cell Signal 2020; 69:109526. [PMID: 31911181 DOI: 10.1016/j.cellsig.2020.109526] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 12/21/2022]
Abstract
Sphingolipids and glycosphingolipids are classes of structurally and functionally important lipids that regulate multiple cellular processes, including membrane organization, proliferation, cell cycle regulation, apoptosis, transport, migration, and inflammatory signalling pathways. Imbalances in sphingolipid levels or subcellular localization result in dysregulated cellular processes and lead to the development and progression of multiple disorders, including polycystic kidney disease. This review will describe metabolic pathways of glycosphingolipids with a focus on the evidence linking glycosphingolipid mediated regulation of cell signalling, lipid microdomains, cilia, and polycystic kidney disease. We will discuss molecular mechanisms of glycosphingolipid dysregulation and their impact on cystogenesis. We will further highlight how modulation of sphingolipid metabolism can be translated into new approaches for the treatment of polycystic kidney disease and describe current clinical studies with glucosylceramide synthase inhibitors in Autosomal Dominant Polycystic Kidney Disease.
Collapse
Affiliation(s)
- Thomas A Natoli
- Rare and Neurological Disease Research, Sanofi-Genzyme, 49 New York Ave., Framingham, MA 01701, USA
| | - Vijay Modur
- Rare Disease Development, Sanofi-Genzyme, 50 Binney St., Cambridge, MA 02142, USA
| | | |
Collapse
|
12
|
Zhang G, Zhang J, DeHoog RJ, Pennathur S, Anderton CR, Venkatachalam MA, Alexandrov T, Eberlin LS, Sharma K. DESI-MSI and METASPACE indicates lipid abnormalities and altered mitochondrial membrane components in diabetic renal proximal tubules. Metabolomics 2020; 16:11. [PMID: 31925564 PMCID: PMC7301343 DOI: 10.1007/s11306-020-1637-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 01/04/2020] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Diabetic kidney disease (DKD) is the most prevalent complication in diabetic patients, which contributes to high morbidity and mortality. Urine and plasma metabolomics studies have been demonstrated to provide valuable insights for DKD. However, limited information on spatial distributions of metabolites in kidney tissues have been reported. OBJECTIVES In this work, we employed an ambient desorption electrospray ionization-mass spectrometry imaging (DESI-MSI) coupled to a novel bioinformatics platform (METASPACE) to characterize the metabolome in a mouse model of DKD. METHODS DESI-MSI was performed for spatial untargeted metabolomics analysis in kidneys of mouse models (F1 C57BL/6J-Ins2Akita male mice at 17 weeks of age) of type 1 diabetes (T1D, n = 5) and heathy controls (n = 6). RESULTS Multivariate analyses (i.e., PCA and PLS-DA (a 2000 permutation test: P < 0.001)) showed clearly separated clusters for the two groups of mice on the basis of 878 measured m/z's in kidney cortical tissues. Specifically, mice with T1D had increased relative abundances of pseudouridine, accumulation of free polyunsaturated fatty acids (PUFAs), and decreased relative abundances of cardiolipins in cortical proximal tubules when compared with healthy controls. CONCLUSION Results from the current study support potential key roles of pseudouridine and cardiolipins for maintaining normal RNA structure and normal mitochondrial function, respectively, in cortical proximal tubules with DKD. DESI-MSI technology coupled with METASPACE could serve as powerful new tools to provide insight on fundamental pathways in DKD.
Collapse
Affiliation(s)
- Guanshi Zhang
- Center for Renal Precision Medicine, Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Audie L. Murphy Memorial VA Hospital, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Jialing Zhang
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
| | - Rachel J DeHoog
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
| | - Subramaniam Pennathur
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Christopher R Anderton
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | | | - Theodore Alexandrov
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Livia S Eberlin
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
| | - Kumar Sharma
- Center for Renal Precision Medicine, Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Audie L. Murphy Memorial VA Hospital, South Texas Veterans Health Care System, San Antonio, TX, USA.
| |
Collapse
|
13
|
Tortorella S, Tiberi P, Bowman AP, Claes BSR, Ščupáková K, Heeren RMA, Ellis SR, Cruciani G. LipostarMSI: Comprehensive, Vendor-Neutral Software for Visualization, Data Analysis, and Automated Molecular Identification in Mass Spectrometry Imaging. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2020; 31:155-163. [PMID: 32881505 DOI: 10.1021/jasms.9b00034] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Mass Spectrometry Imaging (MSI) is an established and powerful MS technique that enables molecular mapping of tissues and cells finding widespread applications in academic, medical, and pharmaceutical industries. As both the applications and MSI technology have undergone rapid growth and improvement, the challenges associated both with analyzing large datasets and identifying the many detected molecular species have become apparent. The lack of readily available and comprehensive software covering all necessary data analysis steps has further compounded this challenge. To address this issue we developed LipostarMSI, comprehensive and vendor-neutral software for targeted and untargeted MSI data analysis. Through user-friendly implementation of image visualization and co-registration, univariate and multivariate image and spectral analysis, and for the first time, advanced lipid, metabolite, and drug metabolite (MetID) automated identification, LipostarMSI effectively streamlines biochemical interpretation of the data. Here, we introduce LipostarMSI and case studies demonstrating the versatility and many capabilities of the software.
Collapse
Affiliation(s)
- Sara Tortorella
- Molecular Horizon Srl, Via Montelino 30, 06084 Bettona, Perugia, Italy
- Consortium for Computational Molecular and Materials Sciences (CMS)2, Via Elce di Sotto 8, 06123 Perugia, Italy
| | - Paolo Tiberi
- Molecular Discovery Ltd., Centennial Park, WD6 3FG Borehamwood, Hertfordshire, United Kingdom
| | - Andrew P Bowman
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Britt S R Claes
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Klára Ščupáková
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Ron M A Heeren
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Shane R Ellis
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Gabriele Cruciani
- Consortium for Computational Molecular and Materials Sciences (CMS)2, Via Elce di Sotto 8, 06123 Perugia, Italy
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| |
Collapse
|
14
|
Cetraro N, Cody RB, Yew JY. Carbon-carbon double bond position elucidation in fatty acids using ozone-coupled direct analysis in real time mass spectrometry. Analyst 2019; 144:5848-5855. [PMID: 31482871 DOI: 10.1039/c9an01059a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The carbon-carbon double bond positions of unsaturated fatty acids can have markedly different effects on biological function and also serve as biomarkers of disease pathology, dietary history, and species identity. As such, there is great interest in developing methods for the facile determination of double bond position for natural product chemistry, the pharmaceutical industry, and forensics. We paired ozonolysis with direct analysis in real time mass spectrometry (DART MS) to cleave and rapidly identify carbon-carbon double bond position in fatty acids, fatty alcohols, wax esters, and crude fatty acid extracts. In addition, ozone exposure time and DART ion source temperature were investigated to identify optimal conditions. Our results reveal that brief, offline exposure to ozone-generated aldehyde and carboxylate products that are indicative of carbon-carbon double bond position. The relative abundance of diagnostic fragments quantitatively reflects the ratios of isobaric fatty acid positional isomers in a mixture with a correlation coefficient of 0.99. Lastly, the unsaturation profile generated from unfractionated, fatty acid extracts can be used to differentiate insect species and populations. The ability to rapidly elucidate lipid double bond position by combining ozonolysis with DART MS will be useful for lipid structural elucidation, assessing isobaric purity, and potentially distinguishing between animals fed on different diets or belonging to different ecological populations.
Collapse
Affiliation(s)
- Nicolas Cetraro
- Pacific Biosciences Research Center, University of Hawai'i at Mānoa, 1993 East West Road, Honolulu, USA 96822.
| | - Robert B Cody
- JEOL USA, Inc., 11 Dearborn Rd, Peabody, MA, USA 01960
| | - Joanne Y Yew
- Pacific Biosciences Research Center, University of Hawai'i at Mānoa, 1993 East West Road, Honolulu, USA 96822.
| |
Collapse
|
15
|
Bestard-Escalas J, Maimó-Barceló A, Pérez-Romero K, Lopez DH, Barceló-Coblijn G. Ins and Outs of Interpreting Lipidomic Results. J Mol Biol 2019; 431:5039-5062. [PMID: 31422112 DOI: 10.1016/j.jmb.2019.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 12/20/2022]
Abstract
Membrane lipids are essential for life; however, research on how cells regulate cell lipid composition has been falling behind for quite some time. One reason was the difficulty in establishing analytical methods able to cope with the cell lipid repertoire. Development of a diversity of mass spectrometry-based technologies, including imaging mass spectrometry, has helped to demonstrate beyond doubt that the cell lipidome is not only greatly cell type dependent but also highly sensitive to any pathophysiological alteration such as differentiation or tumorigenesis. Interestingly, the current popularization of metabolomic studies among numerous disciplines has led many researchers to rediscover lipids. Hence, it is important to underscore the peculiarities of these metabolites and their metabolism, which are both radically different from protein and nucleic acid metabolism. Once differences in lipid composition have been established, researchers face a rather complex scenario, to investigate the signaling pathways and molecular mechanisms accounting for their results. Thus, a detail often overlooked, but of crucial relevance, is the complex networks of enzymes involved in controlling the level of each one of the lipid species present in the cell. In most cases, these enzymes are redundant and promiscuous, complicating any study on lipid metabolism, since the modification of one particular lipid enzyme impacts simultaneously on many species. Altogether, this review aims to describe the difficulties in delving into the regulatory mechanisms tailoring the lipidome at the activity, genetic, and epigenetic level, while conveying the numerous, stimulating, and sometimes unexpected research opportunities afforded by this type of studies.
Collapse
Affiliation(s)
- Joan Bestard-Escalas
- Lipids in Human Pathology, Institut d'Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
| | - Albert Maimó-Barceló
- Lipids in Human Pathology, Institut d'Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
| | - Karim Pérez-Romero
- Lipids in Human Pathology, Institut d'Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
| | - Daniel H Lopez
- Lipids in Human Pathology, Institut d'Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
| | - Gwendolyn Barceló-Coblijn
- Lipids in Human Pathology, Institut d'Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain.
| |
Collapse
|
16
|
Bezençon J, Beaudoin JJ, Ito K, Fu D, Roth SE, Brock WJ, Brouwer KLR. Altered Expression and Function of Hepatic Transporters in a Rodent Model of Polycystic Kidney Disease. Drug Metab Dispos 2019; 47:899-906. [PMID: 31160314 PMCID: PMC6657211 DOI: 10.1124/dmd.119.086785] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/30/2019] [Indexed: 12/18/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a common form of inherited polycystic kidney disease (PKD) and is a leading cause of kidney failure. Fluid-filled cysts develop in the kidneys of patients with ADPKD, and cysts often form in their liver and other organs. Previous data have shown that bile acids are increased in the liver of polycystic kidney (PCK) rats, a rodent model of PKD; these changes may be associated with alterations in liver transporter expression and function. However, the impact of PKD on hepatic transporters has not been characterized. Therefore, this preclinical study was designed to investigate hepatic transporter expression and function in PCK compared with wild-type (WT) Sprague-Dawley rats. Transporter gene expression was measured by quantitative polymerase chain reaction, and protein levels were quantified by Western blot and liquid chromatography-tandem mass spectroscopy (LC-MS/MS)-based proteomic analysis in rat livers. Transporter function was assessed in isolated perfused livers (IPLs), and biliary and hepatic total glutathione content was measured. Protein expression of Mrp2 and Oatp1a4 was decreased 3.0-fold and 2.9-fold, respectively, in PCK rat livers based on Western blot analysis. Proteomic analysis confirmed a decrease in Mrp2 and a decrease in Oatp1a1 expression (PCK/WT ratios, 0.368 ± 0.098 and 0.563 ± 0.038, respectively; mean ± S.D.). The biliary excretion of 5(6)-carboxy-2',7'-dichlorofluorescein, a substrate of Oatp1a1, Mrp2, and Mrp3, was decreased 28-fold in PCK compared with WT rat IPLs. Total glutathione was significantly reduced in the bile of PCK rats. Differences in hepatic transporter expression and function may contribute to altered disposition of Mrp2 and Oatp substrates in PKD.
Collapse
Affiliation(s)
- Jacqueline Bezençon
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.B., J.J.B., K.I., D.F., K.L.R.B.); DMPK Research Department, Teijin Pharma Limited, Hino, Tokyo, Japan (K.I.); Otsuka Pharmaceutical Development & Commercialization, Inc., Rockville, Maryland (S.E.R.); and Brock Scientific Consulting, Montgomery Village, Maryland (W.J.B.)
| | - James J Beaudoin
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.B., J.J.B., K.I., D.F., K.L.R.B.); DMPK Research Department, Teijin Pharma Limited, Hino, Tokyo, Japan (K.I.); Otsuka Pharmaceutical Development & Commercialization, Inc., Rockville, Maryland (S.E.R.); and Brock Scientific Consulting, Montgomery Village, Maryland (W.J.B.)
| | - Katsuaki Ito
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.B., J.J.B., K.I., D.F., K.L.R.B.); DMPK Research Department, Teijin Pharma Limited, Hino, Tokyo, Japan (K.I.); Otsuka Pharmaceutical Development & Commercialization, Inc., Rockville, Maryland (S.E.R.); and Brock Scientific Consulting, Montgomery Village, Maryland (W.J.B.)
| | - Dong Fu
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.B., J.J.B., K.I., D.F., K.L.R.B.); DMPK Research Department, Teijin Pharma Limited, Hino, Tokyo, Japan (K.I.); Otsuka Pharmaceutical Development & Commercialization, Inc., Rockville, Maryland (S.E.R.); and Brock Scientific Consulting, Montgomery Village, Maryland (W.J.B.)
| | - Sharin E Roth
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.B., J.J.B., K.I., D.F., K.L.R.B.); DMPK Research Department, Teijin Pharma Limited, Hino, Tokyo, Japan (K.I.); Otsuka Pharmaceutical Development & Commercialization, Inc., Rockville, Maryland (S.E.R.); and Brock Scientific Consulting, Montgomery Village, Maryland (W.J.B.)
| | - William J Brock
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.B., J.J.B., K.I., D.F., K.L.R.B.); DMPK Research Department, Teijin Pharma Limited, Hino, Tokyo, Japan (K.I.); Otsuka Pharmaceutical Development & Commercialization, Inc., Rockville, Maryland (S.E.R.); and Brock Scientific Consulting, Montgomery Village, Maryland (W.J.B.)
| | - Kim L R Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.B., J.J.B., K.I., D.F., K.L.R.B.); DMPK Research Department, Teijin Pharma Limited, Hino, Tokyo, Japan (K.I.); Otsuka Pharmaceutical Development & Commercialization, Inc., Rockville, Maryland (S.E.R.); and Brock Scientific Consulting, Montgomery Village, Maryland (W.J.B.)
| |
Collapse
|
17
|
Gessel MM, Spraggins JM, Voziyan PA, Abrahamson DR, Caprioli RM, Hudson BG. Two Specific Sulfatide Species Are Dysregulated during Renal Development in a Mouse Model of Alport Syndrome. Lipids 2019; 54:411-418. [PMID: 31197846 DOI: 10.1002/lipd.12171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 01/26/2023]
Abstract
Alport syndrome is caused by mutations in collagen IV that alter the morphology of renal glomerular basement membrane. Mutations result in proteinuria, tubulointerstitial fibrosis, and renal failure but the pathogenic mechanisms are not fully understood. Using imaging mass spectrometry, we aimed to determine whether the spatial and/or temporal patterns of renal lipids are perturbed during the development of Alport syndrome in the mouse model. Our results show that most sulfatides are present at similar levels in both the wild-type (WT) and the Alport kidneys, with the exception of two specific sulfatide species, SulfoHex-Cer(d18:2/24:0) and SulfoHex-Cer(d18:2/16:0). In the Alport but not in WT kidneys, the levels of these species mirror the previously described abnormal laminin expression in Alport syndrome. The presence of these sulfatides in renal tubules but not in glomeruli suggests that this specific aberrant lipid pattern may be related to the development of tubulointerstitial fibrosis in Alport disease.
Collapse
Affiliation(s)
- Megan M Gessel
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeffrey M Spraggins
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA.,Department of Biochemistry, Vanderbilt University Nashville, Nashville, TN 37232, USA.,Department of Chemistry, Vanderbilt University Nashville, Nashville, TN 37232, USA
| | - Paul A Voziyan
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Dale R Abrahamson
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Richard M Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA.,Department of Biochemistry, Vanderbilt University Nashville, Nashville, TN 37232, USA.,Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Billy G Hudson
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Department of Biochemistry, Vanderbilt University Nashville, Nashville, TN 37232, USA.,Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
18
|
Meprin β metalloproteases associated with differential metabolite profiles in the plasma and urine of mice with type 1 diabetes and diabetic nephropathy. BMC Nephrol 2019; 20:141. [PMID: 31023251 PMCID: PMC6485094 DOI: 10.1186/s12882-019-1313-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/27/2019] [Indexed: 02/07/2023] Open
Abstract
Background Meprin metalloproteases are abundantly expressed in the brush border membranes of kidney proximal tubules and small intestines. Meprins are also expressed in podocytes and leukocytes (monocytes and macrophages). Meprins are implicated in the pathophysiology of diabetic nephropathy (DN) but underlying mechanisms are not fully understood. Single nucleotide polymophisms (SNPs) in the meprin β gene were associated with DKD in human subjects. Furthermore, meprin α and β double deficiency resulted in more severe kidney injury and higher mortality rates in mice with Streptozotocin (STZ)-induced type 1 diabetes. Identification of meprin substrates has provided insights on how meprins could modulate kidney injury. Meprin targets in the kidney include extracellular matrix (ECM) proteins, modulators of inflammation, and proteins involved in the protein kinase A (PKA) and PKC signaling pathways. The current study used a global metabolomics approach to determine how meprin β expression impacts the metabolite milieu in diabetes and DKD. Methods Low dose STZ was used to induce type 1 diabetes in 8-week old wild-type (WT) and meprin β knockout (βKO) mice. Blood and urine samples were obtained at 4 and 8 weeks post-STZ injection. Assays for albumin, creatinine, neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule − 1 (KIM-1), and cystatin C were used for biochemical assessment of kidney injury. Data for biomarkers of kidney injury utilized two-way ANOVA. Metabolomics data analysis utilized UPLC-QTOF MS and multivariate statistics. Results The number of metabolites with diabetes-associated changes in levels were significantly higher in the WT mice when compared to meprin βKO counterparts. Annotated meprin β expression-associated metabolites with strong variable importance in projection (VIP) scores play roles in lipid metabolism (LysoPC(16:1(9Z)), taurocholic acid), amino acid metabolism (indoxyl sulfate, hippuric acid), and neurotransmitter/stress hormone synthesis (cortisol, 3-methoxy-4-hydroxyphenylethylene glycolsulfate, homovanillic acid sulfate). Metabolites that associated with meprin β deficiency include; 3,5-dihydroxy-3′,4′-dimethoxy-6,7-methylenedioxyflavone 3-glucuronide, pantothenic acid, and indoxyl glucuronide (all decreased in plasma). Conclusion Taken together, the annotated metabolites suggest that meprin β impacts complications of diabetes such as DKD by altering distinct metabolite profiles. Electronic supplementary material The online version of this article (10.1186/s12882-019-1313-2) contains supplementary material, which is available to authorized users.
Collapse
|
19
|
Kidney Lipidomics by Mass Spectrometry Imaging: A Focus on the Glomerulus. Int J Mol Sci 2019; 20:ijms20071623. [PMID: 30939806 PMCID: PMC6480965 DOI: 10.3390/ijms20071623] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/22/2019] [Accepted: 03/28/2019] [Indexed: 12/13/2022] Open
Abstract
Lipid disorders have been associated with glomerulopathies, a distinct type of renal pathologies, such as nephrotic syndrome. Global analyses targeting kidney lipids in this pathophysiologic context have been extensively performed, but most often regardless of the architectural and functional complexity of the kidney. The new developments in mass spectrometry imaging technologies have opened a promising field in localized lipidomic studies focused on this organ. In this article, we revisit the main works having employed the Matrix Assisted Laser Desorption Ionization Time of Flight (MALDI-TOF) technology, and the few reports on the use of TOF-Secondary Ion Mass Spectrometry (TOF-SIMS). We also present a first analysis of mouse kidney cortex sections by cluster TOF-SIMS. The latter represents a good option for high resolution lipid imaging when frozen unfixed histological samples are available. The advantages and drawbacks of this developing field are discussed.
Collapse
|
20
|
Brock WJ, Beaudoin JJ, Slizgi JR, Su M, Jia W, Roth SE, Brouwer KLR. Bile Acids as Potential Biomarkers to Assess Liver Impairment in Polycystic Kidney Disease. Int J Toxicol 2019; 37:144-154. [PMID: 29587557 DOI: 10.1177/1091581818760746] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Polycystic kidney disease is characterized by the progressive development of kidney cysts and declining renal function with frequent development of cysts in other organs including the liver. The polycystic kidney (PCK) rat is a rodent model of polycystic liver disease that has been used to study hepatorenal disease progression and evaluate pharmacotherapeutic interventions. Biomarkers that describe the cyst progression, liver impairment, and/or hepatic cyst burden could provide clinical utility for this disease. In the present study, hepatic cyst volume was measured by magnetic resonance imaging in PCK rats at 12, 16, and 20 weeks. After 20 weeks, Sprague Dawley (n = 4) and PCK (n = 4) rats were sacrificed and 42 bile acids were analyzed in the liver, bile, serum, and urine by liquid chromatography coupled to tandem mass spectrometry. Bile acid profiling revealed significant increases in total bile acids (molar sum of all measured bile acids) in the liver (13-fold), serum (6-fold), and urine (3-fold) in PCK rats, including those speciated bile acids usually associated with hepatotoxicity. Total serum bile acids correlated with markers of liver impairment (liver weight, total liver bile acids, total hepatotoxic liver bile acids, and cyst volume [ r > 0.75; P < 0.05]). Based on these data, serum bile acids may be useful biomarkers of liver impairment in polycystic hepatorenal disease.
Collapse
Affiliation(s)
- William J Brock
- 1 Brock Scientific Consulting, LLC, Montgomery Village, MD, USA
| | - James J Beaudoin
- 2 Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jason R Slizgi
- 2 Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mingming Su
- 3 Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Wei Jia
- 3 Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, HI, USA
| | | | - Kim L R Brouwer
- 2 Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
21
|
Moreno-Gordaliza E, Esteban-Fernández D, Lázaro A, Aboulmagd S, Humanes B, Tejedor A, Linscheid MW, Gómez-Gómez MM. Lipid imaging for visualizing cilastatin amelioration of cisplatin-induced nephrotoxicity. J Lipid Res 2018; 59:1561-1574. [PMID: 30049708 PMCID: PMC6121926 DOI: 10.1194/jlr.m080465] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 07/12/2018] [Indexed: 12/26/2022] Open
Abstract
Nephrotoxicity is a major limitation to cisplatin antitumor therapies. Cilastatin, an inhibitor of renal dehydropeptidase-I, was recently proposed as a promising nephroprotector against cisplatin toxicity, preventing apoptotic cell death. In this work, cilastatin nephroprotection was further investigated in a rat model, with a focus on its effect on 76 renal lipids altered by cisplatin, including 13 new cisplatin-altered mitochondrial cardiolipin species. Lipid imaging was performed with MALDI mass spectrometry imaging (MALDI-MSI) in kidney sections from treated rats. Cilastatin was proved to significantly diminish the lipid distribution alterations caused by cisplatin, lipid levels being almost completely recovered to those of control samples. The extent of recovery of cisplatin-altered lipids by cilastatin turned out to be relevant for discriminating direct or secondary lipid alterations driven by cisplatin. Lipid peroxidation induced by cisplatin was also shown to be reduced when cilastatin was administered. Importantly, significant groups separation was achieved during multivariate analysis of cortex and outer-medullary lipids, indicating that damaged kidney can be discerned from the nephroprotected and healthy groups and classified according to lipid distribution. Therefore, we propose MALDI-MSI as a powerful potential tool offering multimolecule detection possibilities to visualize and evaluate nephrotoxicity and nephroprotection based on lipid analysis.
Collapse
Affiliation(s)
- Estefanía Moreno-Gordaliza
- Department of Analytical Chemistry, Faculty of Chemistry, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Diego Esteban-Fernández
- Department of Chemistry, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
| | - Alberto Lázaro
- Humboldt Universität zu Berlin, 12489 Berlin, Germany; and Renal Pathophysiology Laboratory, Department of Nephrology, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; Department of Physiology, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Sarah Aboulmagd
- Department of Chemistry, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
| | - Blanca Humanes
- Humboldt Universität zu Berlin, 12489 Berlin, Germany; and Renal Pathophysiology Laboratory, Department of Nephrology, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
| | - Alberto Tejedor
- Humboldt Universität zu Berlin, 12489 Berlin, Germany; and Renal Pathophysiology Laboratory, Department of Nephrology, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; Department of Medicine, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Michael W Linscheid
- Department of Chemistry, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
| | - M Milagros Gómez-Gómez
- Department of Analytical Chemistry, Faculty of Chemistry, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
| |
Collapse
|
22
|
Imaging Mass Microscopy of Kidneys from Azithromycin-Treated Rats with Phospholipidosis. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1993-2003. [PMID: 29981744 DOI: 10.1016/j.ajpath.2018.05.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 05/11/2018] [Accepted: 05/29/2018] [Indexed: 11/23/2022]
Abstract
Drug-induced phospholipidosis is a lysosomal storage disorder characterized by the excess accumulation of tissue phospholipids. Although azithromycin can be used to induce phospholipidosis, no experimental studies evaluating the relationship between drug accumulation and phospholipid localization have been performed. In this study, azithromycin was orally administered to rats for 7 days, and the relationship between drug and phospholipid accumulation was performed using imaging mass microscopy. The administration of azithromycin induced tubular epithelial vacuolation in the inner stripe of the outer medulla of the kidney, consistent with the lamellar bodies that are typical manifestations of drug-induced phospholipidosis. Azithromycin and phospholipid tissue levels were extensively elevated in the kidneys of azithromycin-treated rats. Imaging mass microscopy revealed that both azithromycin and its metabolites were found in the kidneys of azithromycin-treated rats but not in control animals. The vacuolated areas of the kidneys were primarily found in the inner stripe of the outer medulla, consistent with the areas of high azithromycin concentration. Azithromycin was colocalized with several phospholipids-phosphatidylinositol (18:0/20:4), phosphatidylethanolamine (18:0/20:4 and 16:0/20:4), and possibly didocosahexaenoyl (C22:6)-bis(monoacylglycerol) phosphate, a putative biomarker of drug-induced phospholipidosis. In summary, we found correlations between regions of kidney damage and the accumulation of azithromycin, its metabolites, and phospholipids using imaging mass microscopy. Such analyses may help reveal the mechanism and identify putative biomarkers of drug-induced phospholipidosis.
Collapse
|
23
|
Grove KJ, Lareau NM, Voziyan PA, Zeng F, Harris RC, Hudson BG, Caprioli RM. Imaging mass spectrometry reveals direct albumin fragmentation within the diabetic kidney. Kidney Int 2018; 94:292-302. [PMID: 29779708 DOI: 10.1016/j.kint.2018.01.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 01/24/2018] [Accepted: 01/25/2018] [Indexed: 12/26/2022]
Abstract
Albumin degradation in the renal tubules is impaired in diabetic nephropathy such that levels of the resulting albumin fragments increase with the degree of renal injury. However, the mechanism of albumin degradation is unknown. In particular, fragmentation of the endogenous native albumin has not been demonstrated in the kidney and the enzymes that may contribute to fragmentation have not been identified. To explore this we utilized matrix-assisted laser desorption/ionization imaging mass spectrometry for molecular profiling of specific renal regions without disturbing distinct tissue morphology. Changes in protein expression were measured in kidney sections of eNOS-/-db/db mice, a model of diabetic nephropathy, by high spatial resolution imaging allowing molecular localizations at the level of single glomeruli and tubules. Significant increases were found in the relative abundances of several albumin fragments in the kidney of the mice with diabetic nephropathy compared with control nondiabetic mice. The relative abundance of fragments detected correlated positively with the degree of nephropathy. Furthermore, specific albumin fragments accumulating in the lumen of diabetic renal tubules were identified and predicted the enzymatic action of cathepsin D based on cleavage specificity and in vitro digestions. Importantly, this was demonstrated directly in the renal tissue with the endogenous nonlabeled murine albumin. Thus, our results provide molecular insights into the mechanism of albumin degradation in diabetic nephropathy.
Collapse
Affiliation(s)
- Kerri J Grove
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Nichole M Lareau
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Paul A Voziyan
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Fenghua Zeng
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Raymond C Harris
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Billy G Hudson
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| | - Richard M Caprioli
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| |
Collapse
|
24
|
Rzagalinski I, Hainz N, Meier C, Tschernig T, Volmer DA. MALDI Mass Spectral Imaging of Bile Acids Observed as Deprotonated Molecules and Proton-Bound Dimers from Mouse Liver Sections. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2018; 29:711-722. [PMID: 29417494 PMCID: PMC5889423 DOI: 10.1007/s13361-017-1886-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 12/24/2017] [Accepted: 12/24/2017] [Indexed: 05/07/2023]
Abstract
Bile acids (BAs) play two vital roles in living organisms, as they are involved in (1) the secretion of cholesterol from liver, and (2) the lipid digestion/absorption in the intestine. Abnormal bile acid synthesis or secretion can lead to severe liver disorders. Even though there is extensive literature on the mass spectrometric determination of BAs in biofluids and tissue homogenates, there are no reports on the spatial distribution in the biliary network of the liver. Here, we demonstrate the application of high mass resolution/mass accuracy matrix-assisted laser desorption/ionization (MALDI)-Fourier-transform ion cyclotron resonance (FTICR) to MS imaging (MSI) of BAs at high spatial resolutions (pixel size, 25 μm). The results show chemical heterogeneity of the mouse liver sections with a number of branching biliary and blood ducts. In addition to ion signals from deprotonation of the BA molecules, MALDI-MSI generated several further intense signals at larger m/z for the BAs. These signals were spatially co-localized with the deprotonated molecules and easily misinterpreted as additional products of BA biotransformations. In-depth analysis of accurate mass shifts and additional electrospray ionization and MALDI-FTICR experiments, however, confirmed them as proton-bound dimers. Interestingly, dimers of bile acids, but also unusual mixed dimers of different taurine-conjugated bile acids and free taurine, were identified. Since formation of these complexes will negatively influence signal intensities of the desired [M - H]- ions and significantly complicate mass spectral interpretations, two simple broadband techniques were proposed for non-selective dissociation of dimers that lead to increased signals for the deprotonated BAs. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Ignacy Rzagalinski
- Institute of Bioanalytical Chemistry, Saarland University, 66123, Saarbrücken, Germany
| | - Nadine Hainz
- Institute of Anatomy and Cell Biology, Saarland University, 66421, Homburg, Germany
| | - Carola Meier
- Institute of Anatomy and Cell Biology, Saarland University, 66421, Homburg, Germany
| | - Thomas Tschernig
- Institute of Anatomy and Cell Biology, Saarland University, 66421, Homburg, Germany
| | - Dietrich A Volmer
- Institute of Bioanalytical Chemistry, Saarland University, 66123, Saarbrücken, Germany.
- Department of Chemistry, Humboldt University of Berlin, 12489, Berlin, Germany.
| |
Collapse
|
25
|
Prentice BM, Caprioli RM, Vuiblet V. Label-free molecular imaging of the kidney. Kidney Int 2017; 92:580-598. [PMID: 28750926 PMCID: PMC6193761 DOI: 10.1016/j.kint.2017.03.052] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 12/25/2022]
Abstract
In this review, we will highlight technologies that enable scientists to study the molecular characteristics of tissues and/or cells without the need for antibodies or other labeling techniques. Specifically, we will focus on matrix-assisted laser desorption/ionization imaging mass spectrometry, infrared spectroscopy, and Raman spectroscopy.
Collapse
Affiliation(s)
- Boone M Prentice
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Richard M Caprioli
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, Tennessee, USA; Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee, USA.
| | - Vincent Vuiblet
- Biophotonic Laboratory, UMR CNRS 7369 URCA, Reims, France; Nephropathology, Department of Biopathology Laboratory, CHU de Reims, Reims, France; Nephrology and Renal Transplantation department, CHU de Reims, Reims, France.
| |
Collapse
|
26
|
MALDI-LTQ-Orbitrap mass spectrometry imaging for lipidomic analysis in kidney under cisplatin chemotherapy. Talanta 2017; 164:16-26. [DOI: 10.1016/j.talanta.2016.11.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 11/06/2016] [Accepted: 11/12/2016] [Indexed: 01/06/2023]
|
27
|
Spatial Metabolite Profiling by Matrix-Assisted Laser Desorption Ionization Mass Spectrometry Imaging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 965:291-321. [DOI: 10.1007/978-3-319-47656-8_12] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
28
|
Quanico J, Franck J, Wisztorski M, Salzet M, Fournier I. Progress and Potential of Imaging Mass Spectrometry Applied to Biomarker Discovery. Methods Mol Biol 2017; 1598:21-43. [PMID: 28508356 DOI: 10.1007/978-1-4939-6952-4_2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Mapping provides a direct means to assess the impact of protein biomarkers and puts into context their relevance in the type of cancer being examined. To this end, mass spectrometry imaging (MSI) was developed to provide the needed spatial information which is missing in traditional liquid-based mass spectrometric proteomics approaches. Aptly described as a "molecular histology" technique, MSI gives an additional dimension in characterizing tumor biopsies, allowing for mapping of hundreds of molecules in a single analysis. A decade of developments focused on improving and standardizing MSI so that the technique can be translated into the clinical setting. This review describes the progress made in addressing the technological development that allows to bridge local protein detection by MSI to its identification and to illustrate its potential in studying various aspects of cancer biomarker discovery.
Collapse
Affiliation(s)
- Jusal Quanico
- Université de Lille 1, INSERM, U1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), 59000, Lille, France
| | - Julien Franck
- Université de Lille 1, INSERM, U1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), 59000, Lille, France
| | - Maxence Wisztorski
- Université de Lille 1, INSERM, U1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), 59000, Lille, France
| | - Michel Salzet
- Université de Lille 1, INSERM, U1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), 59000, Lille, France
| | - Isabelle Fournier
- Université de Lille 1, INSERM, U1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), 59000, Lille, France.
| |
Collapse
|
29
|
Fülöp A, Sammour DA, Erich K, von Gerichten J, van Hoogevest P, Sandhoff R, Hopf C. Molecular imaging of brain localization of liposomes in mice using MALDI mass spectrometry. Sci Rep 2016; 6:33791. [PMID: 27650487 PMCID: PMC5030664 DOI: 10.1038/srep33791] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/02/2016] [Indexed: 12/21/2022] Open
Abstract
Phospholipids have excellent biocompatibility and are therefore often used as main components of liposomal drug carriers. In traditional bioanalytics, the in-vivo distribution of liposomal drug carriers is assessed using radiolabeled liposomal constituents. This study presents matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) as an alternative, label-free method for ex-vivo molecular imaging of liposomal drug carriers in mouse tissue. To this end, indocyanine green as cargo and two liposomal markers, 1,2-dipalmitoyl-sn-glycero-3-phosphoglycerol (DPPG) and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine conjugated with monodisperse polyethylene glycol (PEG36-DSPE) were incorporated into liposomal carriers and administered to mice. We used MALDI MSI of the two lipid markers in both positive and negative ion mode for visualization of liposome integrity and distribution in mouse organs. Additional MSI of hemoglobin in the same tissue slice and pixel-by-pixel computational analysis of co-occurrence of lipid markers and hemoglobin served as indicator of liposome localization either in parenchyma or in blood vessels. Our proof-of-concept study suggests that liposomal components and indocyanine green distributed into all investigated organs.
Collapse
Affiliation(s)
- Annabelle Fülöp
- Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS). Paul-Wittsack-Str. 10, 68163 Mannheim, Germany.,Instrumental Analytics and Bioanalytics, Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163 Mannheim, Germany
| | - Denis A Sammour
- Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS). Paul-Wittsack-Str. 10, 68163 Mannheim, Germany.,Instrumental Analytics and Bioanalytics, Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163 Mannheim, Germany
| | - Katrin Erich
- Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS). Paul-Wittsack-Str. 10, 68163 Mannheim, Germany.,Instrumental Analytics and Bioanalytics, Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163 Mannheim, Germany
| | - Johanna von Gerichten
- Lipid Pathobiochemistry, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Peter van Hoogevest
- Phospholipid Research Center, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Roger Sandhoff
- Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS). Paul-Wittsack-Str. 10, 68163 Mannheim, Germany.,Lipid Pathobiochemistry, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Carsten Hopf
- Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS). Paul-Wittsack-Str. 10, 68163 Mannheim, Germany.,Instrumental Analytics and Bioanalytics, Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163 Mannheim, Germany.,Institute of Medical Technology, University of Heidelberg and Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163 Mannheim, Germany
| |
Collapse
|
30
|
Jung JW, Lee MS, Choi HJ, Jung S, Lee YJ, Hwang GS, Kwon TH. Mass spectrometric imaging of metabolites in kidney tissues from rats treated with furosemide. Am J Physiol Renal Physiol 2016; 310:F1317-27. [PMID: 26962105 DOI: 10.1152/ajprenal.00524.2015] [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: 11/20/2015] [Accepted: 03/07/2016] [Indexed: 12/16/2022] Open
Abstract
In the kidney, metabolic processes are different among the cortex (COR), outer medulla (OM), and inner medulla (IM). Using matrix-assisted laser desorption/ionization (MALDI) and imaging mass spectrometry (IMS), we examined the change of metabolites in the COR, OM, and IM of the rat kidney after furosemide treatment compared with vehicle-treated controls. Osmotic minipumps were implanted in male Sprague-Dawley rats to deliver 12 mg·day(-1)·rat(-1) of furosemide. Vehicle-treated (n = 14) and furosemide-treated (furosemide rats, n = 15) rats in metabolic cages received a fixed amount of rat chow (15 g·220 g body wt(-1)·day(-1) for each rat) with free access to water intake for 6 days. At day 6, higher urine output (32 ± 4 vs. 9 ± 1 ml/day) and lower urine osmolality (546 ± 44 vs. 1,677 ± 104 mosmol/kgH2O) were observed in furosemide rats. Extracts of COR, OM, and IM were analyzed by ultraperformance liquid chromatography coupled with quadrupole time-of-flight (TOF) mass spectrometry, where multivariate analysis revealed significant differences between the two groups. Several metabolites, including acetylcarnitine, betaine, carnitine, choline, and glycerophosphorylcholine (GPC), were significantly changed. The changes of metabolites were further identified by MALDI-TOF/TOF and IMS. Their spatial distribution and relative quantitation in the kidneys were analyzed by IMS. Carnitine compounds were increased in COR and IM, whereas carnitine and acetylcarnitine were decreased in OM. Choline compounds were increased in COR and OM but decreased in IM from furosemide rats. Betaine and GPC were decreased in OM and IM. Taken together, MALDI-TOF/TOF and IMS successfully provide the spatial distribution and relative quantitation of metabolites in the kidney.
Collapse
Affiliation(s)
- Jin Woo Jung
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Korea
| | - Mi Suk Lee
- Department of Biochemistry and Cell Biology, Korea; and BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Taegu, Korea
| | - Hyo-Jung Choi
- Department of Biochemistry and Cell Biology, Korea; and
| | - Sunhee Jung
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Korea
| | - Yu-Jung Lee
- Department of Biochemistry and Cell Biology, Korea; and BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Taegu, Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Korea
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, Korea; and BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Taegu, Korea
| |
Collapse
|
31
|
Muller L, Kailas A, Jackson SN, Roux A, Barbacci DC, Schultz JA, Balaban CD, Woods AS. Lipid imaging within the normal rat kidney using silver nanoparticles by matrix-assisted laser desorption/ionization mass spectrometry. Kidney Int 2015; 88:186-92. [PMID: 25671768 PMCID: PMC4527327 DOI: 10.1038/ki.2015.3] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 11/24/2014] [Accepted: 12/05/2014] [Indexed: 02/07/2023]
Abstract
The well-characterized cellular and structural components of the kidney show distinct regional compositions and distribution of lipids. In order to more fully analyze the renal lipidome we developed a matrix-assisted laser desorption/ionization mass spectrometry approach for imaging that may be used to pinpoint sites of changes from normal in pathological conditions. This was accomplished by implanting sagittal cryostat rat kidney sections with a stable, quantifiable and reproducible uniform layer of silver using a magnetron sputtering source to form silver nanoparticles. Thirty-eight lipid species including seven ceramides, eight diacylglycerols, 22 triacylglycerols, and cholesterol were detected and imaged in positive ion mode. Thirty-six lipid species consisting of seven sphingomyelins, 10 phosphatidylethanolamines, one phosphatidylglycerol, seven phosphatidylinositols, and 11 sulfatides were imaged in negative ion mode for a total of seventy-four high-resolution lipidome maps of the normal kidney. Thus, our approach is a powerful tool not only for studying structural changes in animal models of disease, but also for diagnosing and tracking stages of disease in human kidney tissue biopsies.
Collapse
Affiliation(s)
- Ludovic Muller
- 1] Structural Biology Unit, NIDA IRP, NIH, Baltimore, Maryland, USA [2] Departments of Otolaryngology, Neurobiology, Communication Sciences & Disorders, and Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ajay Kailas
- Structural Biology Unit, NIDA IRP, NIH, Baltimore, Maryland, USA
| | | | - Aurelie Roux
- Structural Biology Unit, NIDA IRP, NIH, Baltimore, Maryland, USA
| | - Damon C Barbacci
- 1] Structural Biology Unit, NIDA IRP, NIH, Baltimore, Maryland, USA [2] Ionwerks, Houston, Texas, USA
| | | | - Carey D Balaban
- Departments of Otolaryngology, Neurobiology, Communication Sciences & Disorders, and Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amina S Woods
- Structural Biology Unit, NIDA IRP, NIH, Baltimore, Maryland, USA
| |
Collapse
|
32
|
Niehoff AC, Kettling H, Pirkl A, Chiang YN, Dreisewerd K, Yew JY. Analysis of Drosophila Lipids by Matrix-Assisted Laser Desorption/Ionization Mass Spectrometric Imaging. Anal Chem 2014; 86:11086-92. [DOI: 10.1021/ac503171f] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Ann-Christin Niehoff
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604
- NRW
Graduate School of Chemistry, University of Münster, Wilhelm-Klemm-Strasse
10, 48149 Münster, Germany
- Institute
of Inorganic and Analytical Chemistry, University of Münster, Corrensstrasse
30, 48149 Münster, Germany
- Institute
for Hygiene, University of Münster, Robert-Koch-Strasse 41, 48149 Münster, Germany
| | - Hans Kettling
- Institute
for Hygiene, University of Münster, Robert-Koch-Strasse 41, 48149 Münster, Germany
- Interdisciplinary
Center for Clinical Research (IZKF), University of Münster, Domagkstrasse
3, 48149 Münster, Germany
| | - Alexander Pirkl
- Institute
for Hygiene, University of Münster, Robert-Koch-Strasse 41, 48149 Münster, Germany
| | - Yin Ning Chiang
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604
| | - Klaus Dreisewerd
- Institute
for Hygiene, University of Münster, Robert-Koch-Strasse 41, 48149 Münster, Germany
- Interdisciplinary
Center for Clinical Research (IZKF), University of Münster, Domagkstrasse
3, 48149 Münster, Germany
| | - Joanne Y. Yew
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604
- Department
of Biological Sciences, National University of Singapore, 14 Science
Drive 4, Singapore 117543
- Pacific Biosciences
Research Center, University of Hawaìi at Ma̅noa, 1993
East-West Road, Honolulu, Hawaii 96822, United States
| |
Collapse
|
33
|
Turiák L, Shao C, Meng L, Khatri K, Leymarie N, Wang Q, Pantazopoulos H, Leon DR, Zaia J. Workflow for combined proteomics and glycomics profiling from histological tissues. Anal Chem 2014; 86:9670-8. [PMID: 25203838 DOI: 10.1021/ac5022216] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Extracellular matrixes comprise glycoproteins, glycosaminoglycans and proteoglycans that order the environment through which cells receive signals and communicate. Proteomic and glycomic molecular signatures from tissue surfaces can add diagnostic power to the immunohistochemistry workflows. Acquired in a spatially resolved manner, such proteomic and glycomic information can help characterize disease processes and be easily applied in a clinical setting. Our aim toward obtaining integrated omics datasets was to develop the first workflow applicable for simultaneous analysis of glycosaminoglycans, N-glycans and proteins/peptides from tissue surface areas as small as 1.5 mm in diameter. Targeting small areas is especially important in the case of glycans, as their distribution can be very heterogeneous between different tissue regions. We first established reliable and reproducible digestion protocols for the individual compound classes by applying standards on the tissue using microwave irradiation to achieve reduced digestion times. Next, we developed a multienzyme workflow suitable for analysis of the different compound classes. Applicability of the workflow was demonstrated on serial mouse brain and liver sections, both fresh frozen and formalin-fixed. The glycomics data from the 1.5 mm diameter tissue surface area was consistent with data published on bulk mouse liver and brain tissues, which demonstrates the power of the workflow in obtaining combined molecular signatures from very small tissue regions.
Collapse
Affiliation(s)
- Lilla Turiák
- Center for Biomedical Mass Spectrometry, Department of Biochemistry, Boston University School of Medicine , Boston, Massachusetts 02118, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Jones EE, Dworski S, Canals D, Casas J, Fabrias G, Schoenling D, Levade T, Denlinger C, Hannun YA, Medin JA, Drake RR. On-tissue localization of ceramides and other sphingolipids by MALDI mass spectrometry imaging. Anal Chem 2014; 86:8303-11. [PMID: 25072097 PMCID: PMC4139181 DOI: 10.1021/ac501937d] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
A novel MALDI-FTICR imaging mass
spectrometry (MALDI-IMS) workflow
is described for on-tissue detection, spatial localization, and structural
confirmation of low abundance bioactive ceramides and other sphingolipids.
Increasingly, altered or elevated levels of sphingolipids, sphingolipid
metabolites, and sphingolipid metabolizing enzymes have been associated
with a variety of disorders such as diabetes, obesity, lysosomal storage
disorders, and cancer. Ceramide, which serves as a metabolic hub in
sphingolipid metabolism, has been linked to cancer signaling pathways
and to metabolic regulation with involvement in autophagy, cell-cycle
arrest, senescence, and apoptosis. Using kidney tissues from a new
Farber disease mouse model in which ceramides of all acyl chain lengths
and other sphingolipid metabolites accumulate in tissues, specific
ceramides and sphingomyelins were identified by on-tissue isolation
and fragmentation, coupled with an on-tissue digestion by ceramidase
or sphingomyelinase. Multiple glycosphingolipid species were also
detected. The newly generated library of sphingolipid ions was then
applied to MALDI-IMS of human lung cancer tissues. Multiple tumor
specific ceramide and sphingomyelin species were detected and confirmed
by on-tissue enzyme digests and structural confirmation. High-resolution
MALDI-IMS in combination with novel on-tissue ceramidase and sphingomyelinase
enzyme digestions makes it now possible to rapidly visualize the distribution
of bioactive ceramides and sphingomyelin in tissues.
Collapse
Affiliation(s)
- E Ellen Jones
- Department of Cell and Molecular Pharmacology and MUSC Proteomics Center, Medical University of South Carolina , 173 Ashley Avenue, Charleston, South Carolina 29425, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Patterson NH, Thomas A, Chaurand P. Monitoring time-dependent degradation of phospholipids in sectioned tissues by MALDI imaging mass spectrometry. JOURNAL OF MASS SPECTROMETRY : JMS 2014; 49:622-7. [PMID: 25044847 DOI: 10.1002/jms.3382] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 05/24/2023]
Abstract
Imaging mass spectrometry (IMS) is useful for visualizing the localization of phospholipids on biological tissue surfaces creating great opportunities for IMS in lipidomic investigations. With advancements in IMS of lipids, there is a demand for large-scale tissue studies necessitating stable, efficient and well-defined sample handling procedures. Our work within this article shows the effects of different storage conditions on the phospholipid composition of sectioned tissues from mouse organs. We have taken serial sections from mouse brain, kidney and liver thaw mounted unto ITO-coated glass slides and stored them under various conditions later analyzing them at fixed time points. A global decrease in phospholipid signal intensity is shown to occur and to be a function of time and temperature. Contrary to the global decrease, oxidized phospholipid and lysophospholipid species are found to increase within 2 h and 24 h, respectively, when mounted sections are kept at ambient room conditions. Imaging experiments reveal that degradation products increase globally across the tissue. Degradation is shown to be inhibited by cold temperatures, with sample integrity maintained up to a week after storage in -80 °C freezer under N2 atmosphere. Overall, the results demonstrate a timeline of the effects of lipid degradation specific to sectioned tissues and provide several lipid species which can serve as markers of degradation. Importantly, the timeline demonstrates oxidative sample degradation begins appearing within the normal timescale of IMS sample preparation of lipids (i.e. 1-2 h) and that long-term degradation is global. Taken together, these results strengthen the notion that standardized procedures are required for phospholipid IMS of large sample sets, or in studies where many serial sections are prepared together but analyzed over time such as in 3-D IMS reconstruction experiments.
Collapse
|
36
|
Grove KJ, Voziyan PA, Spraggins JM, Wang S, Paueksakon P, Harris RC, Hudson BG, Caprioli RM. Diabetic nephropathy induces alterations in the glomerular and tubule lipid profiles. J Lipid Res 2014; 55:1375-85. [PMID: 24864273 DOI: 10.1194/jlr.m049189] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Indexed: 12/19/2022] Open
Abstract
Diabetic nephropathy (DN) is a major life-threatening complication of diabetes. Renal lesions affect glomeruli and tubules, but the pathogenesis is not completely understood. Phospholipids and glycolipids are molecules that carry out multiple cell functions in health and disease, and their role in DN pathogenesis is unknown. We employed high spatial resolution MALDI imaging MS to determine lipid changes in kidneys of eNOS(-/-) db/db mice, a robust model of DN. Phospholipid and glycolipid structures, localization patterns, and relative tissue levels were determined in individual renal glomeruli and tubules without disturbing tissue morphology. A significant increase in the levels of specific glomerular and tubular lipid species from four different classes, i.e., gangliosides, sulfoglycosphingolipids, lysophospholipids, and phosphatidylethanolamines, was detected in diabetic kidneys compared with nondiabetic controls. Inhibition of nonenzymatic oxidative and glycoxidative pathways attenuated the increase in lipid levels and ameliorated renal pathology, even though blood glucose levels remained unchanged. Our data demonstrate that the levels of specific phospho- and glycolipids in glomeruli and/or tubules are associated with diabetic renal pathology. We suggest that hyperglycemia-induced DN pathogenic mechanisms require intermediate oxidative steps that involve specific phospholipid and glycolipid species.
Collapse
Affiliation(s)
- Kerri J Grove
- Departments of Chemistry, Vanderbilt University Medical Center, Nashville, TN 37232 Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, TN 37232 Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Paul A Voziyan
- Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN 37232 Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN 37232 Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jeffrey M Spraggins
- Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, TN 37232 Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Suwan Wang
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN 37232 Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Paisit Paueksakon
- Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Raymond C Harris
- Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN 37232 Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN 37232 Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Billy G Hudson
- Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN 37232 Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN 37232 Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Richard M Caprioli
- Departments of Chemistry, Vanderbilt University Medical Center, Nashville, TN 37232 Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, TN 37232 Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|