1
|
Deltourbe LG, Durand M, Costas A, Ingersoll MA. A bladder blueprint to build better models for understanding homeostasis and disease. Nat Rev Urol 2025:10.1038/s41585-025-01013-x. [PMID: 40140722 DOI: 10.1038/s41585-025-01013-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2025] [Indexed: 03/28/2025]
Abstract
The bladder is a complex organ that can be affected by various pathologies, such as cancer or infection. It has a specific tissue structure composed of many different cell types and layers, including urothelial and endothelial cells but also a muscle layer controlling stretch and contraction to void urine. The bladder has constitutive and induced immune responses to infection or damage and harbours a microbiome. Each of these features can be influenced by factors including age and biological sex, which makes modelling homeostasis and disease in the bladder complex and challenging. To model diseases that affect the bladder, mouse models are an invaluable tool to understand the bladder in situ. However, stark differences exist between mice and humans, and so mouse models of human disease have limitations. Thus, models that more closely approximate human physiology would be expected to contribute to improved understanding of bladder biology. As technology advances, improvements in model development and creation of 3D bladder structures are enabling scientists to recapitulate essential aspects of human bladder physiology to gain increased understanding of bladder homeostasis and diseases.
Collapse
Affiliation(s)
- Léa G Deltourbe
- Mucosal Inflammation and Immunity Team, Université Paris Cité, CNRS, Inserm, Institut Cochin and Department of Immunology, Institut Pasteur, Paris, France
| | - Méline Durand
- Mucosal Inflammation and Immunity Team, Université Paris Cité, CNRS, Inserm, Institut Cochin and Department of Immunology, Institut Pasteur, Paris, France
| | - Ariana Costas
- Mucosal Inflammation and Immunity Team, Université Paris Cité, CNRS, Inserm, Institut Cochin and Department of Immunology, Institut Pasteur, Paris, France
- Australian Institute for Microbiology and Infection, University of Technology Sydney, ULTIMO, Sydney, Australia
| | - Molly A Ingersoll
- Mucosal Inflammation and Immunity Team, Université Paris Cité, CNRS, Inserm, Institut Cochin and Department of Immunology, Institut Pasteur, Paris, France.
| |
Collapse
|
2
|
Zhou Q, Harding JC, Fan P, Spasojevic I, Kovacs A, Akk A, Mitchell A, Springer LE, Gaut JP, Rauch DA, Wickline SA, Pham CTN, Fuh K, Pan H. Safety Evaluations of Rapamycin Perfluorocarbon Nanoparticles in Ovarian Tumor-Bearing Mice. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1752. [PMID: 39513832 PMCID: PMC11547995 DOI: 10.3390/nano14211752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Nanomedicine holds great potential for revolutionizing medical treatment. Ongoing research and advancements in nanotechnology are continuously expanding the possibilities, promising significant advancements in healthcare. To fully harness the potential of nanotechnology in medical applications, it is crucial to conduct safety evaluations for the nanomedicines that offer effective benefits in the preclinical stage. Our recent efficacy studies indicated that rapamycin perfluorocarbon (PFC) nanoparticles showed promise in mitigating cisplatin-induced acute kidney injury (AKI). As cisplatin is routinely administered to ovarian cancer patients as their first-line chemotherapy, in this study, we focused on evaluating the safety of rapamycin PFC nanoparticles in mice bearing ovarian tumor xenografts. Specifically, this study evaluated the effects of repeat-dose rapamycin PFC nanoparticle treatment on vital organs, the immune system, and tumor growth and assessed pharmacokinetics and biodistribution. Our results indicated that rapamycin PFC nanoparticle treatment did not cause any detectable adverse effects on cardiac, renal, or hepatic functions or on splenocyte populations, but it reduced the splenocyte secretion of IL-10, TNFα, and IL12p70 upon IgM stimulation. The pharmacokinetics and biodistribution results revealed a significant enhancement in the delivery of rapamycin to tumors by rapamycin PFC nanoparticles, which, in turn, led to a significant reduction in ovarian tumor growth. Therefore, rapamycin PFC nanoparticles have the potential to be clinically beneficial in cisplatin-treated ovarian cancer patients.
Collapse
Affiliation(s)
- Qingyu Zhou
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - John C. Harding
- Molecular Oncology, Oncology Division, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ping Fan
- Medical Oncology, Department of Medicine, Duke University, Durham, NC 27708, USA
| | - Ivan Spasojevic
- Medical Oncology, Department of Medicine, Duke University, Durham, NC 27708, USA
| | - Attila Kovacs
- Cardiovascular Division, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Antonina Akk
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Adam Mitchell
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Luke E. Springer
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joseph P. Gaut
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel A. Rauch
- Molecular Oncology, Oncology Division, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samuel A. Wickline
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christine T. N. Pham
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
- John Cochran Veterans Affairs Medical Center, St. Louis, MO 63106, USA
| | - Katherine Fuh
- Division of Gynecologic Oncology, University of California, San Francisco, NC 90095, USA
| | - Hua Pan
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
3
|
Wegener J, Dennhardt S, Loeffler I, Coldewey SM. Transition from acute kidney injury to chronic kidney disease in a long-term murine model of Shiga toxin-induced hemolytic-uremic syndrome. Front Immunol 2024; 15:1469353. [PMID: 39450175 PMCID: PMC11499141 DOI: 10.3389/fimmu.2024.1469353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/05/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Up to 40% of patients with typical hemolytic-uremic syndrome (HUS), characterized by microangiopathic hemolytic anemia and acute kidney injury (AKI), develop long-term consequences, most prominently chronic kidney disease (CKD). The transition from AKI to CKD, particularly in the context of HUS, is not yet fully understood. The objective of this study was to establish and characterize a Shiga toxin (Stx)-induced long-term HUS model to facilitate the study of mechanisms underlying the AKI-to-CKD transition. Methods C57BL/6J mice were subjected to 5, 10, 15, or 20 ng/kg Stx on days 0, 3, and 6 of the experiment and were sacrificed on day 14 or day 21 to identify the critical time of turnover from the acute to the chronic state of HUS disease. Results Acute disease, indicated by weight loss, plasma neutrophil gelatinase-associated lipocalin (NGAL) and urea, and renal neutrophils, diminished after 14 days and returned to sham level after 21 days. HUS-associated hemolytic anemia transitioned to non-hemolytic microcytic anemia along with unchanged erythropoietin levels after 21 days. Renal cytokine levels indicated a shift towards pro-fibrotic signaling, and interstitial fibrosis developed concentration-dependently after 21 days. While Stx induced the intrarenal invasion of pro-inflammatory M1 and pro-fibrotic M2 macrophages after 14 days, pro-fibrotic M2 macrophages were the dominant phenotype after 21 days. Conclusion In conclusion, we established and characterized the first Stx-induced long-term model of HUS. This tool facilitates the study of underlying mechanisms in the early AKI-to-CKD transition following HUS and allows the testing of compounds that may protect patients with AKI from developing subsequent CKD.
Collapse
Affiliation(s)
- Jamila Wegener
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Sophie Dennhardt
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Ivonne Loeffler
- Department of Internal Medicine III, Jena University Hospital, Jena, Germany
| | - Sina M. Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| |
Collapse
|
4
|
Vo NDN, Gaßler N, Wolf G, Loeffler I. The Role of Collagen VIII in the Aging Mouse Kidney. Int J Mol Sci 2024; 25:4805. [PMID: 38732023 PMCID: PMC11084264 DOI: 10.3390/ijms25094805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
The gradual loss of kidney function due to increasing age is accompanied by structural changes such as fibrosis of the tissue. The underlying molecular mechanisms are complex, but not yet fully understood. Non-fibrillar collagen type VIII (COL8) could be a potential factor in the fibrosis processes of the aging kidney. A pathophysiological significance of COL8 has already been demonstrated in the context of diabetic kidney disease, with studies showing that it directly influences both the development and progression of renal fibrosis occurring. The aim of this study was to investigate whether COL8 impacts age-related micro-anatomical and functional changes in a mouse model. The kidneys of wild-type (Col8-wt) and COL8-knockout (Col8-ko) mice of different age and sex were characterized with regard to the expression of molecular fibrosis markers, the development of nephrosclerosis and renal function. The age-dependent regulation of COL8 mRNA expression in the wild-type revealed sex-dependent effects that were not observed with collagen IV (COL4). Histochemical staining and protein analysis of profibrotic cytokines TGF-β1 (transforming growth factor) and CTGF (connective tissue growth factor) in mouse kidneys showed significant age effects as well as interactions of the factors age, sex and Col8 genotype. There were also significant age and Col8 genotype effects in the renal function data analyzed by urinary cystatin C. In summary, the present study shows, for the first time, that COL8 is regulated in an age- and sex-dependent manner in the mouse kidney and that the expression of COL8 influences the severity of age-induced renal fibrosis and function.
Collapse
Affiliation(s)
- Ngoc Dong Nhi Vo
- Department of Internal Medicine III, University Hospital Jena, 07745 Jena, Germany; (N.D.N.V.); (G.W.)
| | - Nikolaus Gaßler
- Institute of Forensic Medicine, Section Pathology, University Hospital Jena, 07745 Jena, Germany;
| | - Gunter Wolf
- Department of Internal Medicine III, University Hospital Jena, 07745 Jena, Germany; (N.D.N.V.); (G.W.)
| | - Ivonne Loeffler
- Department of Internal Medicine III, University Hospital Jena, 07745 Jena, Germany; (N.D.N.V.); (G.W.)
| |
Collapse
|
5
|
El Husseini N, Mekonnen SA, Hall CL, Cole SJ, Carter JA, Belew AT, El-Sayed NM, Lee VT. Characterization of the Entner-Doudoroff pathway in Pseudomonas aeruginosa catheter-associated urinary tract infections. J Bacteriol 2024; 206:e0036123. [PMID: 38047680 PMCID: PMC10809998 DOI: 10.1128/jb.00361-23] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 12/05/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic nosocomial pathogen responsible for a subset of catheter-associated urinary tract infections (CAUTI). In a murine model of P. aeruginosa CAUTI, we previously demonstrated that urea within urine suppresses quorum sensing and induces the Entner-Doudoroff (E-D) pathway. The E-D pathway consists of the genes zwf, pgl, edd, and eda. Zwf and Pgl convert glucose-6-phosphate into 6-phosphogluconate. Edd hydrolyzes 6-phosphogluconate to 2-keto-3-deoxy-6-phosphogluconate (KDPG). Finally, Eda cleaves KDPG to glyceraldehyde-3-phosphate and pyruvate, which enters the citric acid cycle. Here, we generated in-frame E-D mutants in the strain PA14 and assessed their growth phenotypes on chemically defined and complex media. These E-D mutants have a growth defect when grown on glucose or gluconate as the sole carbon source, which is similar to results previously reported for PAO1 mutants lacking E-D genes. RNA-sequencing following short exposure to urine revealed minimal gene regulation differences compared to the wild type. In a murine CAUTI model, virulence testing of E-D mutants revealed that two mutants lacking zwf and pgl showed minor fitness defects. Infection with the ∆pgl strain exhibited a 20% increase in host survival, and the ∆zwf strain displayed decreased colonization of the catheter and kidneys. Consequently, our findings suggest that the E-D pathway in P. aeruginosa is dispensable in this model of CAUTI. IMPORTANCE Prior studies have shown that the Entner-Doudoroff pathway is up-regulated when Pseudomonas aeruginosa is grown in urine. Pseudomonads use the Entner-Doudoroff (E-D) pathway to metabolize glucose instead of glycolysis, which led us to ask whether this pathway is required for urinary tract infection. Here, single-deletion mutants of each gene in the pathway were tested for growth on chemically defined media with single-carbon sources as well as complex media. The effect of each mutant on global gene expression in laboratory media and urine was characterized. The virulence of these mutants in a murine model of catheter-associated urinary tract infection revealed that these mutants had similar levels of colonization indicating that glucose is not the primary carbon source utilized in the urinary tract.
Collapse
Affiliation(s)
- Nour El Husseini
- Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, Maryland, USA
| | - Solomon A. Mekonnen
- Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, Maryland, USA
| | - Cherisse L. Hall
- Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, Maryland, USA
| | - Stephanie J. Cole
- Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, Maryland, USA
| | - Jared A. Carter
- Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, Maryland, USA
| | - Ashton T. Belew
- Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, Maryland, USA
| | - Najib M. El-Sayed
- Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, Maryland, USA
- Center for Bioinformatics and Computational Biology, University of Maryland at College Park, College Park, Maryland, USA
| | - Vincent T. Lee
- Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, Maryland, USA
| |
Collapse
|
6
|
Husseini NE, Mekonnen SA, Hall CL, Cole SJ, Carter JA, Belew AT, El-Sayed N, Lee VT. Characterization of the Entner-Douderoff Pathway in Pseudomonas aeruginosa Catheter-associated Urinary Tract Infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.567044. [PMID: 38014081 PMCID: PMC10680737 DOI: 10.1101/2023.11.14.567044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Pseudomonas aeruginosa is an opportunistic nosocomial pathogen responsible for catheter-associated urinary tract infections (CAUTI). In a murine model of P. aeruginosa CAUTI, we previously demonstrated that urea within urine suppresses quorum sensing and induces the Entner-Douderoff (E-D) pathway. The E-D pathway consists of the genes zwf, pgl, edd, and eda. Zwf and Pgl convert glucose-6-phosphate into 6-phosphogluconate. Edd hydrolyzes 6-phosphogluconate to 2-keto-3-deoxy-6-phosphogluconate (KDPG). Finally, Eda cleaves KDPG to glyceraldehyde-3-phosphate and pyruvate, which enters the citric acid cycle. Here, we generated in-frame E-D mutants in strain PA14 and assessed their growth phenotypes on chemically defined media. These E-D mutants have a growth defect when grown on glucose or gluconate as sole carbon source which are similar to results previously reported for PAO1 mutants lacking E-D genes. RNA-sequencing following short exposure to urine revealed minimal gene regulation differences compared to the wild type. In a murine CAUTI model, virulence testing of E-D mutants revealed that two mutants lacking zwf and pgl showed minor fitness defects. Infection with the ∆pgl strain exhibited a 20% increase in host survival, and the ∆zwf strain displayed decreased colonization of the catheter and kidneys. Consequently, our findings suggest that the E-D pathway in P. aeruginosa is dispensable in this model of CAUTI.
Collapse
Affiliation(s)
- Nour El Husseini
- Cell Biology and Molecular Genetics, University of Maryland at College Park, MD 20742
| | - Solomon A. Mekonnen
- Cell Biology and Molecular Genetics, University of Maryland at College Park, MD 20742
| | - Cherisse L. Hall
- Cell Biology and Molecular Genetics, University of Maryland at College Park, MD 20742
| | - Stephanie J. Cole
- Cell Biology and Molecular Genetics, University of Maryland at College Park, MD 20742
| | - Jared A. Carter
- Cell Biology and Molecular Genetics, University of Maryland at College Park, MD 20742
| | - Ashton T. Belew
- Cell Biology and Molecular Genetics, University of Maryland at College Park, MD 20742
| | - Najib El-Sayed
- Cell Biology and Molecular Genetics, University of Maryland at College Park, MD 20742
- Center for Bioinformatics and Computational Biology, University of Maryland at College Park, College Park, MD 20742
| | - Vincent T. Lee
- Cell Biology and Molecular Genetics, University of Maryland at College Park, MD 20742
| |
Collapse
|
7
|
Zhang J, Wang X, Wang R, Chen G, Wang J, Feng J, Li Y, Yu Z, Xiao H. Rapamycin Treatment Alleviates Chronic GVHD-Induced Lupus Nephritis in Mice by Recovering IL-2 Production and Regulatory T Cells While Inhibiting Effector T Cells Activation. Biomedicines 2023; 11:biomedicines11030949. [PMID: 36979928 PMCID: PMC10045991 DOI: 10.3390/biomedicines11030949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
In this study, we test the therapeutic effects of rapamycin in a murine model of SLE-like experimental lupus nephritis induced by chronic graft-versus-host disease (cGVHD). Our results suggest that rapamycin treatment reduced autoantibody production, inhibited T lymphocyte and subsequent B cell activation, and reduced inflammatory cytokine and chemokine production, thereby protecting renal function and alleviating histological lupus nephritis by reducing the occurrence of albuminuria. To explore the potential mechanism of rapamycin's reduction of kidney damage in mice with lupus nephritis, a series of functional assays were conducted. As expected, rapamycin remarkably inhibited the lymphocytes' proliferation within the morbid mice. Interestingly, significantly increased proportions of peripheral CD4+FOXP3+ and CD4+CD25high T cells were observed in rapamycin-treated group animals, suggesting an up-regulation of regulatory T cells (Tregs) in the periphery by rapamycin treatment. Furthermore, consistent with the results regarding changes in mRNA abundance in kidney by real-time PCR analysis, intracellular cytokine staining demonstrated that rapamycin treatment remarkably diminished the secretion of Th1 and Th2 cytokines, including IFN-γ, IL-4 and IL-10, in splenocytes of the morbid mice. However, the production of IL-2 from splenocytes in rapamycin-treated mice was significantly higher than in the cells from control group animals. These findings suggest that rapamycin treatment might alleviate systemic lupus erythematosus (SLE)-like experimental lupus nephritis through the recovery of IL-2 production, which promotes the expansion of regulatory T cells while inhibiting effector T cell activation. Our studies demonstrated that, unlike other commonly used immunosuppressants, rapamycin does not appear to interfere with tolerance induction but permits the expansion and suppressive function of Tregs in vivo.
Collapse
Affiliation(s)
- Jilu Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- Department of Biomedicine, Institute of Frontier Medical Sciences, Jilin University, Changchun 130021, China
| | - Xun Wang
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Renxi Wang
- Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Ministry of Science and Technology, Beijing 100054, China
| | - Guojiang Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Jing Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Jiannan Feng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yan Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zuyin Yu
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - He Xiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
8
|
Greve S, Kuhn GA, Saenz-de-Juano MD, Ghosh A, von Meyenn F, Giller K. The major urinary protein gene cluster knockout mouse as a novel model for translational metabolism research. Sci Rep 2022; 12:13161. [PMID: 35915220 PMCID: PMC9343454 DOI: 10.1038/s41598-022-17195-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 07/21/2022] [Indexed: 11/09/2022] Open
Abstract
Scientific evidence suggests that not only murine scent communication is regulated by major urinary proteins, but that their expression may also vary in response to metabolism via a yet unknown mechanism. Major urinary proteins are expressed mainly in the liver, showing a sexually dimorphic pattern with substantially higher expression in males. Here, we investigate the metabolic implications of a major urinary protein knockout in twelve-week-old male and female C57BL/6N mice during ad libitum feeding. Despite both sexes of major urinary protein knockout mice displayed numerically increased body weight and visceral adipose tissue proportions compared to sex-matched wildtype mice, the main genotype-specific metabolic differences were observed exclusively in males. Male major urinary protein knockout mice exhibited plasma and hepatic lipid accumulation accompanied by a hepatic transcriptome indicating an activation of lipogenesis. These findings match the higher major urinary protein expression in male compared to female wildtype mice, suggesting a more distinct reduction in energy requirements in male compared to female major urinary protein knockout mice. The observed sex-specific anabolic phenotype confirms a role of major urinary protein in metabolism and, since major urinary proteins are not expressed in humans, suggests the major urinary protein knockout mouse as a potential alternative model for translational metabolism research which needs to be further elucidated.
Collapse
Affiliation(s)
- Sarah Greve
- Animal Nutrition, ETH Zurich, Universitaetstrasse 2, 8092, Zurich, Switzerland
| | - Gisela A Kuhn
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | | | - Adhideb Ghosh
- Laboratory of Nutrition and Metabolic Epigenetics, ETH Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland
| | - Ferdinand von Meyenn
- Laboratory of Nutrition and Metabolic Epigenetics, ETH Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland
| | - Katrin Giller
- Animal Nutrition, ETH Zurich, Universitaetstrasse 2, 8092, Zurich, Switzerland.
| |
Collapse
|
9
|
Poitras T, Singh V, Piragasam RS, Wang X, Mannaa AM, Chandrasekhar A, Martinez J, Fahlman R, Zochodne DW. Repurposed major urinary protein pheromones and adult sensory neurons: roles in neuron plasticity and experimental diabetes. Am J Physiol Endocrinol Metab 2022; 323:E53-E68. [PMID: 35635311 DOI: 10.1152/ajpendo.00001.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Major urinary proteins (MUPs), members of the broader lipocalin protein family, are classified as pheromones that are excreted in male rodent urine to define conspecific territoriality. In screening for differentially regulated mRNA transcripts in a mouse model of type 1 experimental diabetes mellitus (DM), we identified an unexpected upregulation of several closely related MUP transcripts within diabetic sensory dorsal root ganglia (DRG). Both sexes expressed overall MUP protein content as identified by an antibody widely targeting these upregulated family members, and immunohistochemistry identified expression within neurons, satellite glial cells, and Schwann cells. In dissociated adult sensory neurons, knockdown by an siRNA targeting upregulated MUP mRNAs, enhanced neurite outgrowth, indicating a growth-suppressive role, an impact that was synergistic with subnanomolar insulin neuronal signaling. While MUP knockdown did not generate rises in insulin signaling transcripts, the protein did bind to several mitochondrial and glial targets in DRG lysates. Analysis of a protein closely related to MUPs but that is expressed in humans, lipocalin-2, also suppressed growth, but its impact was unrelated to insulin. In a model of chronic type 1 DM, MUP siRNA knockdown improved electrophysiological and behavioral abnormalities of experimental neuropathy. MUPs have actions beyond pheromone signaling in rodents that involve suppression of growth plasticity of sensory neurons. Its hitherto unanticipated actions overlap with those of lipocalin-2 and may identify a common and widely mediated impact on neuron growth properties by members of the lipocalin family. Knockdown of MUP supports the trophic actions of insulin as a strategy that may improve features of type 1 experimental diabetic neuropathy.NEW & NOTEWORTHY New molecular mechanisms are important to unravel and understand diabetic polyneuropathy, a disorder prevalent in over half of persons with diabetes mellitus (DM). MUPs, members of the lipocalin family of molecules, have an unexpected impact on the plasticity of sensory neurons that are targeted in type 1 experimental diabetic neuropathy. This work explores this potential target in neuropathy in the context of the lipocalin family of molecules.
Collapse
Affiliation(s)
- Trevor Poitras
- Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Vandana Singh
- Division of Neurology, Department of Clinical Neuroscience and the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | | | - Xiuling Wang
- Southern Alberta Microarray Facility, Department Biochemistry and Molecular. Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Oncology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Atef M Mannaa
- INSERM U1192, Laboratoire Protéomique, Réponse Inflammatoire & Spectrométrie de Masse (PRISM), Université de Lille, Lille, France
- Higher Institute of Engineering and Technology, New Borg El-Arab City, Egypt
| | - Ambika Chandrasekhar
- Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jose Martinez
- Division of Neurology, Department of Clinical Neuroscience and the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Richard Fahlman
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Douglas W Zochodne
- Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Division of Neurology, Department of Clinical Neuroscience and the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|