1
|
Marola OJ, MacLean M, Cossette TL, Diemler CA, Hewes AA, Reagan AM, Kanyinda JN, Skelly DA, Howell GR. Genetic context modulates aging and degeneration in the murine retina. Mol Neurodegener 2025; 20:8. [PMID: 39833899 PMCID: PMC11744848 DOI: 10.1186/s13024-025-00800-9] [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: 04/16/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Age is the principal risk factor for neurodegeneration in both the retina and brain. The retina and brain share many biological properties; thus, insights into retinal aging and degeneration may shed light onto similar processes in the brain. Genetic makeup strongly influences susceptibility to age-related retinal disease. However, studies investigating retinal aging have not sufficiently accounted for genetic diversity. Therefore, examining molecular aging in the retina across different genetic backgrounds will enhance our understanding of human-relevant aging and degeneration in both the retina and brain-potentially improving therapeutic approaches to these debilitating conditions. METHODS Transcriptomics and proteomics were employed to elucidate retinal aging signatures in nine genetically diverse mouse strains (C57BL/6J, 129S1/SvlmJ, NZO/HlLtJ, WSB/EiJ, CAST/EiJ, PWK/PhK, NOD/ShiLtJ, A/J, and BALB/cJ) across lifespan. These data predicted human disease-relevant changes in WSB and NZO strains. Accordingly, B6, WSB, and NZO mice were subjected to human-relevant in vivo examinations at 4, 8, 12, and/or 18M, including: slit lamp, fundus imaging, optical coherence tomography, fluorescein angiography, and pattern/full-field electroretinography. Retinal morphology, vascular structure, and cell counts were assessed ex vivo. RESULTS We identified common molecular aging signatures across the nine mouse strains, which included genes associated with photoreceptor function and immune activation. Genetic background strongly modulated these aging signatures. Analysis of cell type-specific marker genes predicted age-related loss of photoreceptors and retinal ganglion cells (RGCs) in WSB and NZO, respectively. Fundus exams revealed retinitis pigmentosa-relevant pigmentary abnormalities in WSB retinas and diabetic retinopathy (DR)-relevant cotton wool spots and exudates in NZO retinas. Profound photoreceptor dysfunction and loss were confirmed in WSB. Molecular analyses indicated changes in photoreceptor-specific proteins prior to loss, suggesting photoreceptor-intrinsic dysfunction in WSB. In addition, age-associated RGC dysfunction, loss, and concomitant microvascular dysfunction were observed in NZO mice. Proteomic analyses revealed an early reduction in protective antioxidant processes, which may underlie increased susceptibility to DR-relevant pathology in NZO. CONCLUSIONS Genetic context is a strong determinant of retinal aging, and our multi-omics resource can aid in understanding age-related diseases of the eye and brain. Our investigations identified and validated WSB and NZO mice as improved preclinical models relevant to common retinal neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | - Cory A Diemler
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, 04469, USA
| | | | | | | | | | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA.
- School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA.
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, 04469, USA.
| |
Collapse
|
2
|
Jäckel Z, Adžemović A, Kloos B, Hardung S, Sanchez-Brandelik R, Coulon P, Diester I. A Retrospective Report of Carprofen Administration as Post-Operative Analgesia Reveals Negative Effects of Recommended Doses. Animals (Basel) 2024; 14:3157. [PMID: 39518880 PMCID: PMC11544877 DOI: 10.3390/ani14213157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/15/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Effective pain management in laboratory animals is crucial for both animal welfare and the reliability of scientific research. We retrospectively examined the effects of carprofen as post-operative analgesia in Sprague Dawley rats following stereotactic surgery. Our data indicate that administering carprofen twice daily (5 mg/kg), as currently recommended by Die Gesellschaft für Versuchstierkunde/Society for Laboratory Animal Science (GV-SOLAS), led to adverse effects such as reduced food and water intake, disrupted fecal excretion, and abdominal bloating consistent with peritonitis. Continued administration exacerbated these symptoms, with post-mortem findings of intestinal obstructions and ulcers. However, when the frequency was reduced to once daily, such adverse symptoms were not observed. These results are based on incidental data collected from various neuroscientific experiments, resulting in small and uneven sample groups across various experimental cohorts. The inherent imbalances in these groups present challenges for statistical interpretation. While the findings suggest that less frequent carprofen use may reduce adverse effects, the surgical interventions and concurrent use of other drugs in these experiments likely exacerbated these outcomes. Further investigation into the interactions between carprofen, surgical stress, and other perioperative factors is needed to refine analgesia protocols in laboratory animals. Despite these limitations, these observations contribute to understanding analgesia protocols and may assist in improving animal welfare practices.
Collapse
Affiliation(s)
- Zoë Jäckel
- Optophysiology, Faculty of Biology, University of Freiburg, 79110 Freiburg, Germany; (A.A.); (S.H.); (P.C.)
- Intelligent Machine Brain Interfacing Technology (IMBIT)//BrainLinks-BrainTools, University of Freiburg, 79110 Freiburg, Germany
| | - Ahmed Adžemović
- Optophysiology, Faculty of Biology, University of Freiburg, 79110 Freiburg, Germany; (A.A.); (S.H.); (P.C.)
- Intelligent Machine Brain Interfacing Technology (IMBIT)//BrainLinks-BrainTools, University of Freiburg, 79110 Freiburg, Germany
| | - Benedikt Kloos
- Center for Experimental Models and Transgenic Service, University Medical Center Freiburg, 79104 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany;
| | - Stefanie Hardung
- Optophysiology, Faculty of Biology, University of Freiburg, 79110 Freiburg, Germany; (A.A.); (S.H.); (P.C.)
- Intelligent Machine Brain Interfacing Technology (IMBIT)//BrainLinks-BrainTools, University of Freiburg, 79110 Freiburg, Germany
| | | | - Philippe Coulon
- Optophysiology, Faculty of Biology, University of Freiburg, 79110 Freiburg, Germany; (A.A.); (S.H.); (P.C.)
- Intelligent Machine Brain Interfacing Technology (IMBIT)//BrainLinks-BrainTools, University of Freiburg, 79110 Freiburg, Germany
| | - Ilka Diester
- Optophysiology, Faculty of Biology, University of Freiburg, 79110 Freiburg, Germany; (A.A.); (S.H.); (P.C.)
- Intelligent Machine Brain Interfacing Technology (IMBIT)//BrainLinks-BrainTools, University of Freiburg, 79110 Freiburg, Germany
- Bernstein Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
3
|
Barranco MM, Zecchinati F, Perdomo VG, Habib MJ, Rico MJ, Rozados VR, Salazar M, Fusini ME, Scharovsky OG, Villanueva SSM, Mainetti LE, García F. Intestinal ABC transporters: Influence on the metronomic cyclophosphamide-induced toxic effect in an obese mouse mammary cancer model. Toxicol Appl Pharmacol 2024; 492:117130. [PMID: 39426530 DOI: 10.1016/j.taap.2024.117130] [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: 01/18/2024] [Revised: 09/26/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
Metronomic chemotherapy (MCT) is a cancer therapeutic approach characterized by low dose drug chronic administration and limited or null toxicity. Obesity-induced metabolic alterations worsen cancer prognosis and influence the intestinal biochemical barrier, altering the Multidrug resistance-associated protein 2 (Mrp2) and Multidrug resistance protein-1 (Mdr-1), efflux pumps that transport chemotherapeutic drugs. Obesity and cancer are frequent co-morbidities; thus, our aim was to evaluate the effectiveness and toxicity of MCT with cyclophosphamide (Cy) in obese mice with metabolic alterations bearing a mammary adenocarcinoma. Simultaneously, the expression and activities of intestinal Mrp2 and Mdr-1 were assessed. CBi male mice, were fed with chow diet (C) or diet with 40 % of fat (HFD). After 16 weeks, metabolic alterations were confirmed by biochemical and morphological parameters. At that time-point, HFD group showed decreased expressions of Mrp2 mRNA (53 %) as well as Mdr-1a and Mdr-1b (42 % and 59 %, respectively), compared to C (P < 0.05). This result correlated with decreased intestinal Mrp2 and Mdr-1 efflux activities (64 % and 45 %, respectively), compared to C (P < 0.05). Ultimately, mice were challenged with M-406 mammary adenocarcinoma; when the tumor was palpable, mice were distributed into 4 groups. The % inhibition of tumor growth with Cy (30 mg/kg/day) in C + Cy was higher than that of HFD + Cy (P = 0.052). Besides, it was observed a 21 % diminution in body weight and leukopenia in the HFD + Cy group. Conclusion: Obesity-induced metabolic alterations impair intestinal Mrp2 and Mdr-1 functions, bringing about increments in Cy absorption, leading to toxicity; in addition, the antitumor effectiveness of MCT decreased in obese animals.
Collapse
MESH Headings
- Animals
- Cyclophosphamide/toxicity
- Mice
- Obesity/metabolism
- Male
- Female
- Administration, Metronomic
- Multidrug Resistance-Associated Protein 2
- Antineoplastic Agents, Alkylating/toxicity
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/chemically induced
- Mice, Obese
- Multidrug Resistance-Associated Proteins/metabolism
- Multidrug Resistance-Associated Proteins/genetics
- Intestinal Mucosa/drug effects
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Adenocarcinoma/pathology
- Adenocarcinoma/drug therapy
- Adenocarcinoma/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP-Binding Cassette Transporters/metabolism
- ATP-Binding Cassette Transporters/genetics
- Diet, High-Fat
Collapse
Affiliation(s)
- María Manuela Barranco
- Laboratorio de Fisiología Metabólica, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina.; CONICET-Rosario. Rosario, Santa Fe, Argentina
| | - Felipe Zecchinati
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Fisiología Experimental-CONICET. Rosario, Santa Fe, Argentina
| | - Virginia Gabriela Perdomo
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Facultad de Ciencias Bioquímicas y Farmacéuticas-Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Martín José Habib
- Laboratorio de Fisiología Metabólica, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - María José Rico
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Viviana Rosa Rozados
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Mario Salazar
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Laboratorio de Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Matías Ezequiel Fusini
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Olga Graciela Scharovsky
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina.; CIC-UNR, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | | | - Leandro Ernesto Mainetti
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Fabiana García
- Laboratorio de Fisiología Metabólica, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina.; CONICET-Rosario. Rosario, Santa Fe, Argentina..
| |
Collapse
|
4
|
Baschant U, Fuqua BK, Ledesma-Colunga M, Vulpe CD, McLachlan S, Hofbauer LC, Lusis AJ, Rauner M. Effects of dietary iron deficiency or overload on bone: Dietary details matter. Bone 2024; 184:117092. [PMID: 38575048 DOI: 10.1016/j.bone.2024.117092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/28/2024] [Accepted: 03/31/2024] [Indexed: 04/06/2024]
Abstract
PURPOSE Bone is susceptible to fluctuations in iron homeostasis, as both iron deficiency and overload are linked to poor bone strength in humans. In mice, however, inconsistent results have been reported, likely due to different diet setups or genetic backgrounds. Here, we assessed the effect of different high and low iron diets on bone in six inbred mouse strains (C57BL/6J, A/J, BALB/cJ, AKR/J, C3H/HeJ, and DBA/2J). METHODS Mice received a high (20,000 ppm) or low-iron diet (∼10 ppm) after weaning for 6-8 weeks. For C57BL/6J males, we used two dietary setups with similar amounts of iron, yet different nutritional compositions that were either richer ("TUD study") or poorer ("UCLA study") in minerals and vitamins. After sacrifice, liver, blood and bone parameters as well as bone turnover markers in the serum were analyzed. RESULTS Almost all mice on the UCLA study high iron diet had a significant decrease of cortical and trabecular bone mass accompanied by high bone resorption. Iron deficiency did not change bone microarchitecture or turnover in C57BL/6J, A/J, and DBA/2J mice, but increased trabecular bone mass in BALB/cJ, C3H/HeJ and AKR/J mice. In contrast to the UCLA study, male C57BL/6J mice in the TUD study did not display any changes in trabecular bone mass or turnover on high or low iron diet. However, cortical bone parameters were also decreased in TUD mice on the high iron diet. CONCLUSION Thus, these data show that cortical bone is more susceptible to iron overload than trabecular bone and highlight the importance of a nutrient-rich diet to potentially mitigate the negative effects of iron overload on bone.
Collapse
Affiliation(s)
- Ulrike Baschant
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Germany
| | - Brie K Fuqua
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Maria Ledesma-Colunga
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Germany
| | - Christopher D Vulpe
- Department of Physiological Sciences, University of Florida Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA
| | | | - Lorenz C Hofbauer
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Germany
| | - Aldons J Lusis
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Martina Rauner
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Germany.
| |
Collapse
|
5
|
Hönes GS, Geist D, Wenzek C, Pfluger PT, Müller TD, Aguilar-Pimentel JA, Amarie OV, Becker L, Dragano N, Garrett L, Hölter SM, Rathkolb B, Rozman J, Spielmann N, Treise I, Wolf E, Wurst W, Fuchs H, Gailus-Durner V, Hrabe de Angelis M, Führer D, Moeller LC. Comparative Phenotyping of Mice Reveals Canonical and Noncanonical Physiological Functions of TRα and TRβ. Endocrinology 2024; 165:bqae067. [PMID: 38889231 DOI: 10.1210/endocr/bqae067] [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] [Received: 12/26/2023] [Revised: 05/14/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024]
Abstract
Thyroid hormone (TH) effects are mediated through TH receptors (TRs), TRα1, TRβ1, and TRβ2. The TRs bind to the DNA and regulate expression of TH target genes (canonical signaling). In addition, they mediate activation of signaling pathways (noncanonical signaling). Whether noncanonical TR action contributes to the spectrum of TH effects is largely unknown. The aim of this study was to attribute physiological effects to the TR isoforms and their canonical and noncanonical signaling. We conducted multiparameter phenotyping in male and female TR knockout mice (TRαKO, TRβKO), mice with disrupted canonical signaling due to mutations in the TR DNA binding domain (TRαGS, TRβGS), and their wild-type littermates. Perturbations in senses, especially hearing (mainly TRβ with a lesser impact of TRα), visual acuity, retinal thickness (TRα and TRβ), and in muscle metabolism (TRα) highlighted the role of canonical TR action. Strikingly, selective abrogation of canonical TR action often had little phenotypic consequence, suggesting that noncanonical TR action sufficed to maintain the wild-type phenotype for specific effects. For instance, macrocytic anemia, reduced retinal vascularization, or increased anxiety-related behavior were only observed in TRαKO but not TRαGS mice. Noncanonical TRα action improved energy utilization and prevented hyperphagia observed in female TRαKO mice. In summary, by examining the phenotypes of TRα and TRβ knockout models alongside their DNA binding-deficient mutants and wild-type counterparts, we could establish that the noncanonical actions of TRα and TRβ play a crucial role in modulating sensory, behavioral, and metabolic functions and, thus, contribute to the spectrum of physiological TH effects.
Collapse
Affiliation(s)
- Georg Sebastian Hönes
- Department of Endocrinology, Diabetes and Metabolism and Division of Laboratory Research, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Daniela Geist
- Department of Endocrinology, Diabetes and Metabolism and Division of Laboratory Research, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Christina Wenzek
- Department of Endocrinology, Diabetes and Metabolism and Division of Laboratory Research, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Paul Thomas Pfluger
- Research Unit NeuroBiology of Diabetes, Helmholtz Zentrum München, Neuherberg 85764, Germany
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg 85764, Germany
- German Center for Diabetes Research, Neuherberg 85764, Germany
- Division of Neurobiology of Diabetes, TUM School of Medicine, Technical University of Munich, Munich 80333, Germany
| | - Timo Dirk Müller
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg 85764, Germany
- German Center for Diabetes Research, Neuherberg 85764, Germany
- Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University (LMU) Munich, Munich 80336, Germany
| | - Juan Antonio Aguilar-Pimentel
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Oana Veronica Amarie
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Lore Becker
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Natalia Dragano
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Lillian Garrett
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Sabine Maria Hölter
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Birgit Rathkolb
- German Center for Diabetes Research, Neuherberg 85764, Germany
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians University (LMU) Munich, Munich 81377, Germany
| | - Jan Rozman
- German Center for Diabetes Research, Neuherberg 85764, Germany
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Nadine Spielmann
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Irina Treise
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians University (LMU) Munich, Munich 81377, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich 80336, Germany
- Chair of Developmental Genetics, TUM School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Valerie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Martin Hrabe de Angelis
- German Center for Diabetes Research, Neuherberg 85764, Germany
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Chair of Experimental Genetics, TUM School of Life Science Weihenstephan, Technical University of Munich, Freising 85354, Germany
| | - Dagmar Führer
- Department of Endocrinology, Diabetes and Metabolism and Division of Laboratory Research, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Lars Christian Moeller
- Department of Endocrinology, Diabetes and Metabolism and Division of Laboratory Research, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| |
Collapse
|
6
|
Cai Q, Sahu R, Ueberschlag-Pitiot V, Souali-Crespo S, Charvet C, Silem I, Cottard F, Ye T, Taleb F, Metzger E, Schuele R, Billas IML, Laverny G, Metzger D, Duteil D. LSD1 inhibition circumvents glucocorticoid-induced muscle wasting of male mice. Nat Commun 2024; 15:3563. [PMID: 38670969 PMCID: PMC11053113 DOI: 10.1038/s41467-024-47846-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Synthetic glucocorticoids (GC), such as dexamethasone, are extensively used to treat chronic inflammation and autoimmune disorders. However, long-term treatments are limited by various side effects, including muscle atrophy. GC activities are mediated by the glucocorticoid receptor (GR), that regulates target gene expression in various tissues in association with cell-specific co-regulators. Here we show that GR and the lysine-specific demethylase 1 (LSD1) interact in myofibers of male mice, and that LSD1 connects GR-bound enhancers with NRF1-associated promoters to stimulate target gene expression. In addition, we unravel that LSD1 demethylase activity is required for triggering starvation- and dexamethasone-induced skeletal muscle proteolysis in collaboration with GR. Importantly, inhibition of LSD1 circumvents muscle wasting induced by pharmacological levels of dexamethasone, without affecting their anti-inflammatory activities. Thus, our findings provide mechanistic insights into the muscle-specific GC activities, and highlight the therapeutic potential of targeting GR co-regulators to limit corticotherapy-induced side effects.
Collapse
Affiliation(s)
- Qingshuang Cai
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400, Illkirch, France
| | - Rajesh Sahu
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400, Illkirch, France
| | | | - Sirine Souali-Crespo
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400, Illkirch, France
| | - Céline Charvet
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400, Illkirch, France
| | - Ilyes Silem
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400, Illkirch, France
| | - Félicie Cottard
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400, Illkirch, France
| | - Tao Ye
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400, Illkirch, France
| | - Fatima Taleb
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400, Illkirch, France
| | - Eric Metzger
- Klinik für Urologie und Zentrale Klinische Forschung, Klinikum der Albert-Ludwigs-Universität Freiburg, D-79106, Freiburg, Germany
| | - Roland Schuele
- Klinik für Urologie und Zentrale Klinische Forschung, Klinikum der Albert-Ludwigs-Universität Freiburg, D-79106, Freiburg, Germany
| | - Isabelle M L Billas
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400, Illkirch, France
| | - Gilles Laverny
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400, Illkirch, France
| | - Daniel Metzger
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400, Illkirch, France
| | - Delphine Duteil
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400, Illkirch, France.
| |
Collapse
|
7
|
Marola OJ, MacLean M, Cossette TL, Diemler CA, Hewes AA, Reagan AM, Skelly DA, Howell GR. Genetic context modulates aging and degeneration in the murine retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.589625. [PMID: 38659747 PMCID: PMC11042269 DOI: 10.1101/2024.04.16.589625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Background Age is the principal risk factor for neurodegeneration in both the retina and brain. The retina and brain share many biological properties; thus, insights into retinal aging and degeneration may shed light onto similar processes in the brain. Genetic makeup strongly influences susceptibility to age-related retinal disease. However, studies investigating retinal aging have not sufficiently accounted for genetic diversity. Therefore, examining molecular aging in the retina across different genetic backgrounds will enhance our understanding of human-relevant aging and degeneration in both the retina and brain-potentially improving therapeutic approaches to these debilitating conditions. Methods Transcriptomics and proteomics were employed to elucidate retinal aging signatures in nine genetically diverse mouse strains (C57BL/6J, 129S1/SvlmJ, NZO/HlLtJ, WSB/EiJ, CAST/EiJ, PWK/PhK, NOD/ShiLtJ, A/J, and BALB/cJ) across lifespan. These data predicted human disease-relevant changes in WSB and NZO strains. Accordingly, B6, WSB and NZO mice were subjected to human-relevant in vivo examinations at 4, 8, 12, and/or 18M, including: slit lamp, fundus imaging, optical coherence tomography, fluorescein angiography, and pattern/full-field electroretinography. Retinal morphology, vascular structure, and cell counts were assessed ex vivo. Results We identified common molecular aging signatures across the nine mouse strains, which included genes associated with photoreceptor function and immune activation. Genetic background strongly modulated these aging signatures. Analysis of cell type-specific marker genes predicted age-related loss of photoreceptors and retinal ganglion cells (RGCs) in WSB and NZO, respectively. Fundus exams revealed retinitis pigmentosa-relevant pigmentary abnormalities in WSB retinas and diabetic retinopathy (DR)-relevant cotton wool spots and exudates in NZO retinas. Profound photoreceptor dysfunction and loss were confirmed in WSB. Molecular analyses indicated changes in photoreceptor-specific proteins prior to loss, suggesting photoreceptor-intrinsic dysfunction in WSB. In addition, age-associated RGC dysfunction, loss, and concomitant microvascular dysfunction was observed in NZO mice. Proteomic analyses revealed an early reduction in protective antioxidant processes, which may underlie increased susceptibility to DR-relevant pathology in NZO. Conclusions Genetic context is a strong determinant of retinal aging, and our multi-omics resource can aid in understanding age-related diseases of the eye and brain. Our investigations identified and validated WSB and NZO mice as improved preclinical models relevant to common retinal neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | - Cory A. Diemler
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA
| | | | | | | | - Gareth R. Howell
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
- Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA
| |
Collapse
|
8
|
Ball RL, Bogue MA, Liang H, Srivastava A, Ashbrook DG, Lamoureux A, Gerring MW, Hatoum AS, Kim MJ, He H, Emerson J, Berger AK, Walton DO, Sheppard K, El Kassaby B, Castellanos F, Kunde-Ramamoorthy G, Lu L, Bluis J, Desai S, Sundberg BA, Peltz G, Fang Z, Churchill GA, Williams RW, Agrawal A, Bult CJ, Philip VM, Chesler EJ. GenomeMUSter mouse genetic variation service enables multitrait, multipopulation data integration and analysis. Genome Res 2024; 34:145-159. [PMID: 38290977 PMCID: PMC10903950 DOI: 10.1101/gr.278157.123] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
Hundreds of inbred mouse strains and intercross populations have been used to characterize the function of genetic variants that contribute to disease. Thousands of disease-relevant traits have been characterized in mice and made publicly available. New strains and populations including consomics, the collaborative cross, expanded BXD, and inbred wild-derived strains add to existing complex disease mouse models, mapping populations, and sensitized backgrounds for engineered mutations. The genome sequences of inbred strains, along with dense genotypes from others, enable integrated analysis of trait-variant associations across populations, but these analyses are hampered by the sparsity of genotypes available. Moreover, the data are not readily interoperable with other resources. To address these limitations, we created a uniformly dense variant resource by harmonizing multiple data sets. Missing genotypes were imputed using the Viterbi algorithm with a data-driven technique that incorporates local phylogenetic information, an approach that is extendable to other model organisms. The result is a web- and programmatically accessible data service called GenomeMUSter, comprising single-nucleotide variants covering 657 strains at 106.8 million segregating sites. Interoperation with phenotype databases, analytic tools, and other resources enable a wealth of applications, including multitrait, multipopulation meta-analysis. We show this in cross-species comparisons of type 2 diabetes and substance use disorder meta-analyses, leveraging mouse data to characterize the likely role of human variant effects in disease. Other applications include refinement of mapped loci and prioritization of strain backgrounds for disease modeling to further unlock extant mouse diversity for genetic and genomic studies in health and disease.
Collapse
Affiliation(s)
- Robyn L Ball
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA;
| | - Molly A Bogue
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | | - Anuj Srivastava
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032, USA
| | - David G Ashbrook
- University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | | | | - Alexander S Hatoum
- Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri 63130, USA
- Artificial Intelligence and the Internet of Things Institute, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Matthew J Kim
- University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Hao He
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | - Jake Emerson
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | | | | | | | | | | | | - Lu Lu
- University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - John Bluis
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | - Sejal Desai
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | | - Gary Peltz
- Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Zhuoqing Fang
- Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | - Robert W Williams
- University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Carol J Bult
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | | | |
Collapse
|
9
|
Ball RL, Bogue MA, Liang H, Srivastava A, Ashbrook DG, Lamoureux A, Gerring MW, Hatoum AS, Kim M, He H, Emerson J, Berger AK, Walton DO, Sheppard K, Kassaby BE, Castellanos F, Kunde-Ramamoorthy G, Lu L, Bluis J, Desai S, Sundberg BA, Peltz G, Fang Z, Churchill GA, Williams RW, Agrawal A, Bult CJ, Philip VM, Chesler EJ. GenomeMUSter mouse genetic variation service enables multi-trait, multi-population data integration and analyses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.08.552506. [PMID: 37609331 PMCID: PMC10441370 DOI: 10.1101/2023.08.08.552506] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Hundreds of inbred laboratory mouse strains and intercross populations have been used to functionalize genetic variants that contribute to disease. Thousands of disease relevant traits have been characterized in mice and made publicly available. New strains and populations including the Collaborative Cross, expanded BXD and inbred wild-derived strains add to set of complex disease mouse models, genetic mapping resources and sensitized backgrounds against which to evaluate engineered mutations. The genome sequences of many inbred strains, along with dense genotypes from others could allow integrated analysis of trait - variant associations across populations, but these analyses are not feasible due to the sparsity of genotypes available. Moreover, the data are not readily interoperable with other resources. To address these limitations, we created a uniformly dense data resource by harmonizing multiple variant datasets. Missing genotypes were imputed using the Viterbi algorithm with a data-driven technique that incorporates local phylogenetic information, an approach that is extensible to other model organism species. The result is a web- and programmatically-accessible data service called GenomeMUSter ( https://muster.jax.org ), comprising allelic data covering 657 strains at 106.8M segregating sites. Interoperation with phenotype databases, analytic tools and other resources enable a wealth of applications including multi-trait, multi-population meta-analysis. We demonstrate this in a cross-species comparison of the meta-analysis of Type 2 Diabetes and of substance use disorders, resulting in the more specific characterization of the role of human variant effects in light of mouse phenotype data. Other applications include refinement of mapped loci and prioritization of strain backgrounds for disease modeling to further unlock extant mouse diversity for genetic and genomic studies in health and disease.
Collapse
|
10
|
Ghosh P, Sinha S, Katkar GD, Vo D, Taheri S, Dang D, Das S, Sahoo D. Machine learning identifies signatures of macrophage reactivity and tolerance that predict disease outcomes. EBioMedicine 2023; 94:104719. [PMID: 37516087 PMCID: PMC10388732 DOI: 10.1016/j.ebiom.2023.104719] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND Single-cell transcriptomic studies have greatly improved organ-specific insights into macrophage polarization states are essential for the initiation and resolution of inflammation in all tissues; however, such insights are yet to translate into therapies that can predictably alter macrophage fate. METHOD Using machine learning algorithms on human macrophages, here we reveal the continuum of polarization states that is shared across diverse contexts. A path, comprised of 338 genes accurately identified both physiologic and pathologic spectra of "reactivity" and "tolerance", and remained relevant across tissues, organs, species, and immune cells (>12,500 diverse datasets). FINDINGS This 338-gene signature identified macrophage polarization states at single-cell resolution, in physiology and across diverse human diseases, and in murine pre-clinical disease models. The signature consistently outperformed conventional signatures in the degree of transcriptome-proteome overlap, and in detecting disease states; it also prognosticated outcomes across diverse acute and chronic diseases, e.g., sepsis, liver fibrosis, aging, and cancers. Crowd-sourced genetic and pharmacologic studies confirmed that model-rationalized interventions trigger predictable macrophage fates. INTERPRETATION These findings provide a formal and universally relevant definition of macrophage states and a predictive framework (http://hegemon.ucsd.edu/SMaRT) for the scientific community to develop macrophage-targeted precision diagnostics and therapeutics. FUNDING This work was supported by the National Institutes for Health (NIH) grant R01-AI155696 (to P.G, D.S and S.D). Other sources of support include: R01-GM138385 (to D.S), R01-AI141630 (to P.G), R01-DK107585 (to S.D), and UG3TR003355 (to D.S, S.D, and P.G). D.S was also supported by two Padres Pedal the Cause awards (Padres Pedal the Cause/RADY #PTC2017 and San Diego NCI Cancer Centers Council (C3) #PTC2017). S.S, G.D.K, and D.D were supported through The American Association of Immunologists (AAI) Intersect Fellowship Program for Computational Scientists and Immunologists. We also acknowledge support from the Padres Pedal the Cause #PTC2021 and the Torey Coast Foundation, La Jolla (P.G and D.S). D.S, P.G, and S.D were also supported by the Leona M. and Harry B. Helmsley Charitable Trust.
Collapse
Affiliation(s)
- Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, USA; Department of Medicine, University of California San Diego, USA; Moores Cancer Center, University of California San Diego, USA.
| | - Saptarshi Sinha
- Department of Cellular and Molecular Medicine, University of California San Diego, USA; Department of Pediatrics, University of California San Diego, USA
| | - Gajanan D Katkar
- Department of Cellular and Molecular Medicine, University of California San Diego, USA
| | - Daniella Vo
- Department of Pediatrics, University of California San Diego, USA
| | - Sahar Taheri
- Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, USA
| | - Dharanidhar Dang
- Department of Pediatrics, University of California San Diego, USA
| | - Soumita Das
- Moores Cancer Center, University of California San Diego, USA; Department of Pathology, University of California San Diego, USA
| | - Debashis Sahoo
- Moores Cancer Center, University of California San Diego, USA; Department of Pediatrics, University of California San Diego, USA; Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, USA.
| |
Collapse
|
11
|
Arwood ML, Sun IH, Patel CH, Sun IM, Oh MH, Bettencourt IA, Claiborne MD, Chan-Li Y, Zhao L, Waickman AT, Mavrothalassitis O, Wen J, Aja S, Powell JD. Serendipitous Discovery of T Cell-Produced KLK1b22 as a Regulator of Systemic Metabolism. Immunohorizons 2023; 7:493-507. [PMID: 37358498 PMCID: PMC10580127 DOI: 10.4049/immunohorizons.2300016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/05/2023] [Indexed: 06/27/2023] Open
Abstract
In order to study mechanistic/mammalian target of rapamycin's role in T cell differentiation, we generated mice in which Rheb is selectively deleted in T cells (T-Rheb-/- C57BL/6J background). During these studies, we noted that T-Rheb-/- mice were consistently heavier but had improved glucose tolerance and insulin sensitivity as well as a marked increase in beige fat. Microarray analysis of Rheb-/- T cells revealed a marked increase in expression of kallikrein 1-related peptidase b22 (Klk1b22). Overexpression of KLK1b22 in vitro enhanced insulin receptor signaling, and systemic overexpression of KLK1b22 in C57BL/6J mice also enhances glucose tolerance. Although KLK1B22 expression was markedly elevated in the T-Rheb-/- T cells, we never observed any expression in wild-type T cells. Interestingly, in querying the mouse Immunologic Genome Project, we found that Klk1b22 expression was also increased in wild-type 129S1/SVLMJ and C3HEJ mice. Indeed, both strains of mice demonstrate exceptionally improved glucose tolerance. This prompted us to employ CRISPR-mediated knockout of KLK1b22 in 129S1/SVLMJ mice, which in fact led to reduced glucose tolerance. Overall, our studies reveal (to our knowledge) a novel role for KLK1b22 in regulating systemic metabolism and demonstrate the ability of T cell-derived KLK1b22 to regulate systemic metabolism. Notably, however, further studies have revealed that this is a serendipitous finding unrelated to Rheb.
Collapse
Affiliation(s)
- Matthew L. Arwood
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Im-Hong Sun
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Chirag H. Patel
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Im-Meng Sun
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Min-Hee Oh
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ian A. Bettencourt
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Michael D. Claiborne
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Yee Chan-Li
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Liang Zhao
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Adam T. Waickman
- State University of New York Upstate Medical University, Syracuse, NY
| | - Orestes Mavrothalassitis
- Department of Anesthesia, University of California, San Francisco School of Medicine, San Francisco, CA
| | - Jiayu Wen
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins Medicine, Baltimore, MD
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jonathan D. Powell
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
12
|
Kohram F, Deng Z, Zhang Y, Al Reza A, Li E, Kolesnichenko OA, Shukla S, Ustiyan V, Gomez-Arroyo J, Acharya A, Shi D, Kalinichenko VV, Kenny AP. Demonstration of Safety in Wild Type Mice of npFOXF1, a Novel Nanoparticle-Based Gene Therapy for Alveolar Capillary Dysplasia with Misaligned Pulmonary Veins. Biologics 2023; 17:43-55. [PMID: 36969329 PMCID: PMC10031269 DOI: 10.2147/btt.s400006] [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: 12/02/2022] [Accepted: 03/14/2023] [Indexed: 03/22/2023]
Abstract
Introduction Alveolar Capillary Dysplasia with Misaligned Pulmonary Veins (ACDMPV) is a fatal congenital disease resulting from a pulmonary vascular endothelial deficiency of FOXF1, producing abnormal morphogenesis of alveolar capillaries, malpositioned pulmonary veins and disordered development of lung lobes. Affected neonates suffer from cyanosis, severe breathing insufficiency, pulmonary hypertension, and death typically within days to weeks after birth. Currently, no treatment exists for ACDMPV, although recent murine research in the Kalinichenko lab demonstrates nanoparticle delivery improves survival and reconstitutes normal alveolar-capillary architecture. The aim of the present study is to investigate the safety of intravenous administration of FOXF1-expressing PEI-PEG nanoparticles (npFOXF1), our pioneering treatment for ACDMPV. Methods npFOXF1 was constructed, validated, and subsequently administered in a single dose to postnatal day 14 (P14) mice via retro-orbital injection. Biochemical, serologic, and histologic safety were monitored at postnatal day 16 (P16) and postnatal day 21 (P21). Results With treatment we observed no lethality, and the general condition of mice revealed no obvious abnormalities. Serum chemistry, whole blood, and histologic toxicity was assayed on P16 and P21 and revealed no abnormality. Discussion In conclusion, npFOXF1 has a very good safety profile and combined with preceding studies showing therapeutic efficacy, npFOXF1 can be considered as a good candidate therapy for ACDMPV in human neonates.
Collapse
Affiliation(s)
- Fatemeh Kohram
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Zicheng Deng
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- The Materials Science and Engineering Program, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, USA
| | - Yufang Zhang
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Abid Al Reza
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Enhong Li
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Olena A Kolesnichenko
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Samriddhi Shukla
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Vladimir Ustiyan
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Jose Gomez-Arroyo
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Anusha Acharya
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Donglu Shi
- The Materials Science and Engineering Program, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, USA
| | - Vladimir V Kalinichenko
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Alan P Kenny
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
13
|
Pototschnig I, Feiler U, Diwoky C, Vesely PW, Rauchenwald T, Paar M, Bakiri L, Pajed L, Hofer P, Kashofer K, Sukhbaatar N, Schoiswohl G, Weichhart T, Hoefler G, Bock C, Pichler M, Wagner EF, Zechner R, Schweiger M. Interleukin-6 initiates muscle- and adipose tissue wasting in a novel C57BL/6 model of cancer-associated cachexia. J Cachexia Sarcopenia Muscle 2023; 14:93-107. [PMID: 36351437 PMCID: PMC9891934 DOI: 10.1002/jcsm.13109] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 08/23/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Cancer-associated cachexia (CAC) is a wasting syndrome drastically reducing efficacy of chemotherapy and life expectancy of patients. CAC affects up to 80% of cancer patients, yet the mechanisms underlying the disease are not well understood and no approved disease-specific medication exists. As a multiorgan disorder, CAC can only be studied on an organismal level. To cover the diverse aetiologies of CAC, researchers rely on the availability of a multifaceted pool of cancer models with varying degrees of cachexia symptoms. So far, no tumour model syngeneic to C57BL/6 mice exists that allows direct comparison between cachexigenic- and non-cachexigenic tumours. METHODS MCA207 and CHX207 fibrosarcoma cells were intramuscularly implanted into male or female, 10-11-week-old C57BL/6J mice. Tumour tissues were subjected to magnetic resonance imaging, immunohistochemical-, and transcriptomic analysis. Mice were analysed for tumour growth, body weight and -composition, food- and water intake, locomotor activity, O2 consumption, CO2 production, circulating blood cells, metabolites, and tumourkines. Mice were sacrificed with same tumour weights in all groups. Adipose tissues were examined using high-resolution respirometry, lipolysis measurements in vitro and ex vivo, and radioactive tracer studies in vivo. Gene expression was determined in adipose- and muscle tissues by quantitative PCR and Western blotting analyses. Muscles and cultured myotubes were analysed histologically and by immunofluorescence microscopy for myofibre cross sectional area and myofibre diameter, respectively. Interleukin-6 (Il-6) was deleted from cancer cells using CRISPR/Cas9 mediated gene editing. RESULTS CHX207, but not MCA207-tumour-bearing mice exhibited major clinical features of CAC, including systemic inflammation, increased plasma IL-6 concentrations (190 pg/mL, P ≤ 0.0001), increased energy expenditure (+28%, P ≤ 0.01), adipose tissue loss (-47%, P ≤ 0.0001), skeletal muscle wasting (-18%, P ≤ 0.001), and body weight reduction (-13%, P ≤ 0.01) 13 days after cancer cell inoculation. Adipose tissue loss resulted from reduced lipid uptake and -synthesis combined with increased lipolysis but was not associated with elevated beta-adrenergic signalling or adipose tissue browning. Muscle atrophy was evident by reduced myofibre cross sectional area (-21.8%, P ≤ 0.001), increased catabolic- and reduced anabolic signalling. Deletion of IL-6 from CHX207 cancer cells completely protected CHX207IL6KO -tumour-bearing mice from CAC. CONCLUSIONS In this study, we present CHX207 fibrosarcoma cells as a novel tool to investigate the mediators and metabolic consequences of CAC in C57BL/6 mice in comparison to non-cachectic MCA207-tumour-bearing mice. IL-6 represents an essential trigger for CAC development in CHX207-tumour-bearing mice.
Collapse
Affiliation(s)
| | - Ursula Feiler
- Institute of Molecular BiosciencesUniversity of GrazGrazAustria
| | - Clemens Diwoky
- Institute of Molecular BiosciencesUniversity of GrazGrazAustria
| | - Paul W. Vesely
- Diagnostic and Research Institute of PathologyMedical University of GrazGrazAustria
| | | | - Margret Paar
- Division of Physiological Chemistry, Otto‐Loewi Research CenterMedical University of GrazGrazAustria
| | - Latifa Bakiri
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
| | - Laura Pajed
- Institute of Molecular BiosciencesUniversity of GrazGrazAustria
| | - Peter Hofer
- Institute of Molecular BiosciencesUniversity of GrazGrazAustria
| | - Karl Kashofer
- Diagnostic and Research Institute of PathologyMedical University of GrazGrazAustria
| | | | | | - Thomas Weichhart
- Institute of Medical GeneticsMedical University of ViennaViennaAustria
| | - Gerald Hoefler
- Diagnostic and Research Institute of PathologyMedical University of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Institute of Artificial Intelligence, Center for Medical Statistics, Informatics, and Intelligent SystemsMedical University of ViennaViennaAustria
| | | | - Erwin F. Wagner
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
- Department of DermatologyMedical University of ViennaViennaAustria
| | - Rudolf Zechner
- Institute of Molecular BiosciencesUniversity of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
- Field of Excellence BioHealth ‐ University of GrazGrazAustria
| | - Martina Schweiger
- Institute of Molecular BiosciencesUniversity of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
- Field of Excellence BioHealth ‐ University of GrazGrazAustria
| |
Collapse
|
14
|
Gallage S, Avila JEB, Ramadori P, Focaccia E, Rahbari M, Ali A, Malek NP, Anstee QM, Heikenwalder M. A researcher's guide to preclinical mouse NASH models. Nat Metab 2022; 4:1632-1649. [PMID: 36539621 DOI: 10.1038/s42255-022-00700-y] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/26/2022] [Indexed: 12/24/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) and its inflammatory form, non-alcoholic steatohepatitis (NASH), have quickly risen to become the most prevalent chronic liver disease in the Western world and are risk factors for the development of hepatocellular carcinoma (HCC). HCC is not only one of the most common cancers but is also highly lethal. Nevertheless, there are currently no clinically approved drugs for NAFLD, and NASH-induced HCC poses a unique metabolic microenvironment that may influence responsiveness to certain treatments. Therefore, there is an urgent need to better understand the pathogenesis of this rampant disease to devise new therapies. In this line, preclinical mouse models are crucial tools to investigate mechanisms as well as novel treatment modalities during the pathogenesis of NASH and subsequent HCC in preparation for human clinical trials. Although, there are numerous genetically induced, diet-induced and toxin-induced models of NASH, not all of these models faithfully phenocopy and mirror the human pathology very well. In this Perspective, we shed some light onto the most widely used mouse models of NASH and highlight some of the key advantages and disadvantages of the various models with an emphasis on 'Western diets', which are increasingly recognized as some of the best models in recapitulating the human NASH pathology and comorbidities.
Collapse
Affiliation(s)
- Suchira Gallage
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- The M3 Research Institute, Eberhard Karls University Tübingen, Tuebingen, Germany.
| | - Jose Efren Barragan Avila
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pierluigi Ramadori
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Enrico Focaccia
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mohammad Rahbari
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Adnan Ali
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nisar P Malek
- The M3 Research Institute, Eberhard Karls University Tübingen, Tuebingen, Germany
- Department Internal Medicine I, Eberhard-Karls University, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
| | - Quentin M Anstee
- Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne Hospitals, NHS Foundation Trust, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- The M3 Research Institute, Eberhard Karls University Tübingen, Tuebingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
15
|
Dang DX, Li CJ, Li SH, Fan XY, Xu W, Cui Y, Li D. Ultra-early weaning alters growth performance, hematology parameters, and fecal microbiota in piglets with same genetic background. Front Microbiol 2022; 13:990905. [PMID: 36406459 PMCID: PMC9666885 DOI: 10.3389/fmicb.2022.990905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/10/2022] [Indexed: 01/24/2023] Open
Abstract
Piglets with the same genetic background were used to investigate the effects of different lengths of suckling period on growth performance, hematology parameters, and fecal microbiota. All piglets were born by a sow (Landrace×Yorkshire). On day 14 postpartum, a total of 16 piglets [Duroc×(Landrace×Yorkshire)] with a similar initial body weight (2.48 ± 0.25 kg) were randomly assigned into two groups with four replicates per group, two pigs per replicate pen (one barrow and one gilt). On day 14 of age, experiment started, piglets from the first group were weaned (14W), whereas the others continued to receive milk until day 28 of age (28W). The experiment completed on day 70 of age, last 56 days. Growth performance parameters including body weight, average daily gain, feed intake, feed efficiency, and growth rate and hematology parameters including immunoglobulin A (IgA), immunoglobulin G (IgG), immunoglobulin M (IgM), albumin, globulin, and total protein were measured in this study. Additionally, a technique of 16S rRNA gene sequencing was used to analyze fecal microbiota for revealing how the changes in the lengths of suckling period on intestinal microbiota. We found that ultra-early weaning impaired growth performance of piglets, whose worse body weight, average daily gain, feed intake, feed efficiency, and growth rate were observed in 14W group at all measured timepoints in comparison with those in 28W group (P < 0.05). Moreover, higher contents of serum IgA (P = 0.028), IgG (P = 0.041), and IgM (P = 0.047), as well as lower contents of serum albumin (P = 0.002), albumin-to-globulin ratio (P = 0.003), and total protein (P = 0.004), were observed in 14W group in comparison with those in 28W group on day 28 of age, but not on day 70 of age. High-throughput pyrosequencing of 16S rRNA indicated that the intestinal microbiota richness in 14W group was lower than that in 28W group (P < 0.05); moreover, in comparison with 28W group at all sampling timepoints, fecal microbiota in 14W group showed more beneficial bacteria and fewer pathogenic bacteria (P < 0.05). Therefore, we considered that ultra-early weaning had positive effects on immune status and fecal microbiota composition in piglets, but negative effects on growth performance and fecal microbiota abundance.
Collapse
Affiliation(s)
- De Xin Dang
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, China,Department of Animal Resource and Science, Dankook University, Cheonan, South Korea
| | - Cheng Ji Li
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, South Korea,Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, South Korea
| | - Shi Han Li
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, China
| | - Xin Yan Fan
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, China
| | - Weiguo Xu
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, China
| | - Yan Cui
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, China
| | - Desheng Li
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, China,*Correspondence: Desheng Li,
| |
Collapse
|
16
|
List EO, Berryman DE, Slyby J, Duran-Ortiz S, Funk K, Bisset ES, Howlett SE, Kopchick JJ. Disruption of Growth Hormone Receptor in Adipocytes Improves Insulin Sensitivity and Lifespan in Mice. Endocrinology 2022; 163:bqac129. [PMID: 35952979 PMCID: PMC9467438 DOI: 10.1210/endocr/bqac129] [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] [Received: 06/01/2022] [Indexed: 11/19/2022]
Abstract
Growth hormone receptor knockout (GHRKO) mice have been used for 25 years to uncover some of the many actions of growth hormone (GH). Since they are extremely long-lived with enhanced insulin sensitivity and protected from multiple age-related diseases, they are often used to study healthy aging. To determine the effect that adipose tissue has on the GHRKO phenotype, our laboratory recently created and characterized adipocyte-specific GHRKO (AdGHRKO) mice, which have increased adiposity but appear healthy with enhanced insulin sensitivity. To test the hypothesis that removal of GH action in adipocytes might partially replicate the increased lifespan and healthspan observed in global GHRKO mice, we assessed adiposity, cytokines/adipokines, glucose homeostasis, frailty, and lifespan in aging AdGHRKO mice of both sexes. Our results show that disrupting the GH receptor gene in adipocytes improved insulin sensitivity at advanced age and increased lifespan in male AdGHRKO mice. AdGHRKO mice also exhibited increased fat mass, reduced circulating levels of insulin, c-peptide, adiponectin, resistin, and improved frailty scores with increased grip strength at advanced ages. Comparison of published mean lifespan data from GHRKO mice to that from AdGHRKO and muscle-specific GHRKO mice suggests that approximately 23% of lifespan extension in male GHRKO is due to GHR disruption in adipocytes vs approximately 19% in muscle. Females benefited less from GHR disruption in these 2 tissues with approximately 19% and approximately 0%, respectively. These data indicate that removal of GH's action, even in a single tissue, is sufficient for observable health benefits that promote long-term health, reduce frailty, and increase longevity.
Collapse
Affiliation(s)
- Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
- Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Athens, Ohio 45701, USA
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens, Ohio 45701, USA
| | - Julie Slyby
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
| | | | - Kevin Funk
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
| | - Elise S Bisset
- Department of Pharmacology Dalhousie University Halifax, Halifax , Nova Scotia , Canada
| | - Susan E Howlett
- Department of Pharmacology Dalhousie University Halifax, Halifax , Nova Scotia , Canada
- Department of Medicine (Geriatric Medicine), Dalhousie University Halifax, Halifax , Nova Scotia , Canada
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens, Ohio 45701, USA
| |
Collapse
|
17
|
Abstract
As a muscular pump that contracts incessantly throughout life, the heart must constantly generate cellular energy to support contractile function and fuel ionic pumps to maintain electrical homeostasis. Thus, mitochondrial metabolism of multiple metabolic substrates such as fatty acids, glucose, ketones, and lactate is essential to ensuring an uninterrupted supply of ATP. Multiple metabolic pathways converge to maintain myocardial energy homeostasis. The regulation of these cardiac metabolic pathways has been intensely studied for many decades. Rapid adaptation of these pathways is essential for mediating the myocardial adaptation to stress, and dysregulation of these pathways contributes to myocardial pathophysiology as occurs in heart failure and in metabolic disorders such as diabetes. The regulation of these pathways reflects the complex interactions of cell-specific regulatory pathways, neurohumoral signals, and changes in substrate availability in the circulation. Significant advances have been made in the ability to study metabolic regulation in the heart, and animal models have played a central role in contributing to this knowledge. This review will summarize metabolic pathways in the heart and describe their contribution to maintaining myocardial contractile function in health and disease. The review will summarize lessons learned from animal models with altered systemic metabolism and those in which specific metabolic regulatory pathways have been genetically altered within the heart. The relationship between intrinsic and extrinsic regulators of cardiac metabolism and the pathophysiology of heart failure and how these have been informed by animal models will be discussed.
Collapse
Affiliation(s)
- Heiko Bugger
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Graz, Austria, Austria (H.B., N.J.B.)
| | - Nikole J Byrne
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Graz, Austria, Austria (H.B., N.J.B.)
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (E.D.A.)
| |
Collapse
|
18
|
Mouat MA, Wilkins BP, Ding E, Govindaraju H, Coleman JLJ, Graham RM, Turner N, Smith NJ. Metabolic Profiling of Mice with Deletion of the Orphan G Protein-Coupled Receptor, GPR37L1. Cells 2022; 11:cells11111814. [PMID: 35681509 PMCID: PMC9180194 DOI: 10.3390/cells11111814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/18/2022] [Accepted: 05/30/2022] [Indexed: 02/01/2023] Open
Abstract
Understanding the neurogenic causes of obesity may reveal novel drug targets to counter the obesity crisis and associated sequelae. Here, we investigate whether the deletion of GPR37L1, an astrocyte-specific orphan G protein-coupled receptor, affects whole-body energy homeostasis in mice. We subjected male Gpr37l1−/− mice and littermate wildtype (Gpr37l1+/+, C57BL/6J background) controls to either 12 weeks of high-fat diet (HFD) or chow feeding, or to 1 year of chow diet, with body composition quantified by EchoMRI, glucose handling by glucose tolerance test and metabolic rate by indirect calorimetry. Following an HFD, Gpr37l1−/− mice had similar glucose handling, body weight and fat mass compared with wildtype controls. Interestingly, we observed a significantly elevated respiratory exchange ratio in HFD- and chow-fed Gpr37l1−/− mice during daylight hours. After 1 year of chow feeding, we again saw no differences in glucose and insulin tolerance or body weight between genotypes, nor in energy expenditure or respiratory exchange ratio. However, there was significantly lower fat mass accumulation, and higher ambulatory activity in the Gpr37l1−/− mice during night hours. Overall, these results indicate that while GPR37L1 may play a minor role in whole-body metabolism, it is not a viable clinical target for the treatment of obesity.
Collapse
Affiliation(s)
- Margaret A. Mouat
- Orphan Receptor Laboratory, School of Medical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia; (M.A.M.); (B.P.W.)
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (J.L.J.C.); (R.M.G.)
| | - Brendan P. Wilkins
- Orphan Receptor Laboratory, School of Medical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia; (M.A.M.); (B.P.W.)
| | - Eileen Ding
- Mitochondrial Bioenergetics Laboratory, School of Medical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia; (E.D.); (H.G.)
| | - Hemna Govindaraju
- Mitochondrial Bioenergetics Laboratory, School of Medical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia; (E.D.); (H.G.)
| | - James L. J. Coleman
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (J.L.J.C.); (R.M.G.)
| | - Robert M. Graham
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (J.L.J.C.); (R.M.G.)
| | - Nigel Turner
- Mitochondrial Bioenergetics Laboratory, School of Medical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia; (E.D.); (H.G.)
- Cellular Bioenergetics Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- Correspondence: (N.T.); (N.J.S.)
| | - Nicola J. Smith
- Orphan Receptor Laboratory, School of Medical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia; (M.A.M.); (B.P.W.)
- Correspondence: (N.T.); (N.J.S.)
| |
Collapse
|
19
|
Addicks GC, Zhang H, Ryu D, Vasam G, Green AE, Marshall PL, Patel S, Kang BE, Kim D, Katsyuba E, Williams EG, Renaud JM, Auwerx J, Menzies KJ. GCN5 maintains muscle integrity by acetylating YY1 to promote dystrophin expression. J Cell Biol 2022; 221:e202104022. [PMID: 35024765 PMCID: PMC8931935 DOI: 10.1083/jcb.202104022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 11/04/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
Protein lysine acetylation is a post-translational modification that regulates protein structure and function. It is targeted to proteins by lysine acetyltransferases (KATs) or removed by lysine deacetylases. This work identifies a role for the KAT enzyme general control of amino acid synthesis protein 5 (GCN5; KAT2A) in regulating muscle integrity by inhibiting DNA binding of the transcription factor/repressor Yin Yang 1 (YY1). Here we report that a muscle-specific mouse knockout of GCN5 (Gcn5skm-/-) reduces the expression of key structural muscle proteins, including dystrophin, resulting in myopathy. GCN5 was found to acetylate YY1 at two residues (K392 and K393), disrupting the interaction between the YY1 zinc finger region and DNA. These findings were supported by human data, including an observed negative correlation between YY1 gene expression and muscle fiber diameter. Collectively, GCN5 positively regulates muscle integrity through maintenance of structural protein expression via acetylation-dependent inhibition of YY1. This work implicates the role of protein acetylation in the regulation of muscle health and for consideration in the design of novel therapeutic strategies to support healthy muscle during myopathy or aging.
Collapse
Affiliation(s)
- Gregory C Addicks
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Hongbo Zhang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| | - Dongryeol Ryu
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Goutham Vasam
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Alexander E Green
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology and the Éric Poulin Centre for Neuromuscular Disease, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Philip L Marshall
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Sonia Patel
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Baeki E Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Doyoun Kim
- Division of Therapeutics and Biotechnology, Korea Research Institute of Chemical Technology, Daejeon, South Korea
| | - Elena Katsyuba
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| | - Evan G Williams
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jean-Marc Renaud
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| | - Keir J Menzies
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology and the Éric Poulin Centre for Neuromuscular Disease, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
20
|
Wang L, Wang B, Gasek NS, Zhou Y, Cohn RL, Martin DE, Zuo W, Flynn WF, Guo C, Jellison ER, Kim T, Prata LGPL, Palmer AK, Li M, Inman CL, Barber LS, Al-Naggar IMA, Zhou Y, Du W, Kshitiz, Kuchel GA, Meves A, Tchkonia T, Kirkland JL, Robson P, Xu M. Targeting p21 Cip1 highly expressing cells in adipose tissue alleviates insulin resistance in obesity. Cell Metab 2022; 34:75-89.e8. [PMID: 34813734 PMCID: PMC8732323 DOI: 10.1016/j.cmet.2021.11.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/30/2021] [Accepted: 11/03/2021] [Indexed: 01/07/2023]
Abstract
Insulin resistance is a pathological state often associated with obesity, representing a major risk factor for type 2 diabetes. Limited mechanism-based strategies exist to alleviate insulin resistance. Here, using single-cell transcriptomics, we identify a small, critically important, but previously unexamined cell population, p21Cip1 highly expressing (p21high) cells, which accumulate in adipose tissue with obesity. By leveraging a p21-Cre mouse model, we demonstrate that intermittent clearance of p21high cells can both prevent and alleviate insulin resistance in obese mice. Exclusive inactivation of the NF-κB pathway within p21high cells, without killing them, attenuates insulin resistance. Moreover, fat transplantation experiments establish that p21high cells within fat are sufficient to cause insulin resistance in vivo. Importantly, a senolytic cocktail, dasatinib plus quercetin, eliminates p21high cells in human fat ex vivo and mitigates insulin resistance following xenotransplantation into immuno-deficient mice. Our findings lay the foundation for pursuing the targeting of p21high cells as a new therapy to alleviate insulin resistance.
Collapse
Affiliation(s)
- Lichao Wang
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
| | - Binsheng Wang
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
| | - Nathan S Gasek
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
| | - Yueying Zhou
- Xiangya Stomatological Hospital, Central South University, Changsha, 86-410000, China; Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT 06030, USA
| | - Rachel L Cohn
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
| | - Dominique E Martin
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Biomedical Science Graduate Program, UConn Health, Farmington, CT 06030, USA
| | - Wulin Zuo
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - William F Flynn
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Chun Guo
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA
| | - Evan R Jellison
- Department of Immunology, UConn Health, Farmington, CT 06030, USA
| | - Taewan Kim
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Biomedical Science Graduate Program, UConn Health, Farmington, CT 06030, USA
| | | | - Allyson K Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Ming Li
- Department of Dermatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Christina L Inman
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Lauren S Barber
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA
| | | | - Yanjiao Zhou
- Department of Medicine, UConn Health, Farmington, CT 06030, USA
| | - Wenqiang Du
- Department of Biomedical Engineering, UConn Health, Farmington, CT 06030, USA
| | - Kshitiz
- Department of Biomedical Engineering, UConn Health, Farmington, CT 06030, USA
| | - George A Kuchel
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA
| | - Alexander Meves
- Department of Dermatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Paul Robson
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA; The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Ming Xu
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA.
| |
Collapse
|
21
|
Templin AT, Schmidt C, Hogan MF, Esser N, Kitsis RN, Hull RL, Zraika S, Kahn SE. Loss of apoptosis repressor with caspase recruitment domain (ARC) worsens high fat diet-induced hyperglycemia in mice. J Endocrinol 2021; 251:125-135. [PMID: 34382577 PMCID: PMC8651217 DOI: 10.1530/joe-20-0612] [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] [Received: 07/20/2021] [Accepted: 08/11/2021] [Indexed: 11/08/2022]
Abstract
Apoptosis repressor with caspase recruitment domain (ARC) is an endogenous inhibitor of cell death signaling that is expressed in insulin-producing β cells. ARC has been shown to reduce β-cell death in response to diabetogenic stimuli in vitro, but its role in maintaining glucose homeostasis in vivo has not been fully established. Here we examined whether loss of ARC in FVB background mice exacerbates high fat diet (HFD)-induced hyperglycemia in vivo over 24 weeks. Prior to commencing 24-week HFD, ARC-/- mice had lower body weight than wild type (WT) mice. This body weight difference was maintained until the end of the study and was associated with decreased epididymal and inguinal adipose tissue mass in ARC-/- mice. Non-fasting plasma glucose was not different between ARC-/- and WT mice prior to HFD feeding, and ARC-/- mice displayed a greater increase in plasma glucose over the first 4 weeks of HFD. Plasma glucose remained elevated in ARC-/- mice after 16 weeks of HFD feeding, at which time it had returned to baseline in WT mice. Following 24 weeks of HFD, non-fasting plasma glucose in ARC-/- mice returned to baseline and was not different from WT mice. At this final time point, no differences were observed between genotypes in plasma glucose or insulin under fasted conditions or following intravenous glucose administration. However, HFD-fed ARC-/- mice exhibited significantly decreased β-cell area compared to WT mice. Thus, ARC deficiency delays, but does not prevent, metabolic adaptation to HFD feeding in mice, worsening transient HFD-induced hyperglycemia.
Collapse
Affiliation(s)
- Andrew T. Templin
- Department of Medicine, Division of Metabolism,
Endocrinology and Nutrition, Veteran Affairs Puget Sound Health Care System and
University of Washington, Seattle, WA, USA
| | - Christine Schmidt
- Department of Medicine, Division of Metabolism,
Endocrinology and Nutrition, Veteran Affairs Puget Sound Health Care System and
University of Washington, Seattle, WA, USA
| | - Meghan F. Hogan
- Department of Medicine, Division of Metabolism,
Endocrinology and Nutrition, Veteran Affairs Puget Sound Health Care System and
University of Washington, Seattle, WA, USA
| | - Nathalie Esser
- Department of Medicine, Division of Metabolism,
Endocrinology and Nutrition, Veteran Affairs Puget Sound Health Care System and
University of Washington, Seattle, WA, USA
| | - Richard N. Kitsis
- Departments of Medicine and Cell Biology and Wilf Family
Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY,
USA
| | - Rebecca L. Hull
- Department of Medicine, Division of Metabolism,
Endocrinology and Nutrition, Veteran Affairs Puget Sound Health Care System and
University of Washington, Seattle, WA, USA
| | - Sakeneh Zraika
- Department of Medicine, Division of Metabolism,
Endocrinology and Nutrition, Veteran Affairs Puget Sound Health Care System and
University of Washington, Seattle, WA, USA
| | - Steven E. Kahn
- Department of Medicine, Division of Metabolism,
Endocrinology and Nutrition, Veteran Affairs Puget Sound Health Care System and
University of Washington, Seattle, WA, USA
| |
Collapse
|
22
|
Valle AC, Yeh C, Huang Y. Near Infrared-Activatable Platinum-Decorated Gold Nanostars for Synergistic Photothermal/Ferroptotic Therapy in Combating Cancer Drug Resistance. Adv Healthc Mater 2020; 9:e2000864. [PMID: 32945131 DOI: 10.1002/adhm.202000864] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/15/2020] [Indexed: 12/15/2022]
Abstract
Ferroptotic cell death results from glutathione peroxidase 4 (GPX4) inactivation and/or glutathione (GSH) depletion. Elevated GSH levels are often found in multidrug-resistant (MDR) tumor cells, reducing their sensitivity to chemotherapeutic drugs and the efficacy of treatment. MDR cells also acquire a dependency on GPX4, reducing their oxidative stress and promoting their survival. Therefore, the depletion of GSH and inactivation of GPX4 has the potential to be a superior treatment strategy for MDR tumors. Platinum-decorated gold nanostars (Pt-AuNS) are presented as a novel metal nanoprodrug for ferroptotic therapy against MDR tumors. Under dark conditions, the synthesized Pt-AuNS exhibit negligible levels of toxicity. Upon exposure of the Pt-AuNS to near-infrared (NIR) light, active metallic (Pt and Au) species are released, subsequently inducing cytotoxicity. The mechanism of action is attributed to GSH depletion and GPX4 inactivation, accumulating lipid hydroperoxides, which in turn leads to ferroptosis. In in vivo xenograft, the MDR cancer model confirmed the NIR light-activation of Pt-AuNS prodrugs, resulting in efficient ferroptotic therapeutic action against MDR tumors without long-term side effects. The findings lay the groundwork for using Pt-AuNS prodrugs responsive to NIR light as ferroptosis-inducing agents in chemo-resistant cancer cells and demonstrate their potential for use in future clinical applications.
Collapse
Affiliation(s)
- Andrea C. Valle
- Department of Biomedical Engineering and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
| | - Chih‐Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
| | - Yu‐Fen Huang
- Department of Biomedical Engineering and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
- Institute of Analytical and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
| |
Collapse
|
23
|
Dhandapani PK, Lyyski AM, Paulin L, Khan NA, Suomalainen A, Auvinen P, Dufour E, Szibor M, Jacobs HT. Phenotypic effects of dietary stress in combination with a respiratory chain bypass in mice. Physiol Rep 2020; 7:e14159. [PMID: 31267687 PMCID: PMC6606514 DOI: 10.14814/phy2.14159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/28/2019] [Accepted: 06/10/2019] [Indexed: 12/12/2022] Open
Abstract
The alternative oxidase (AOX) from Ciona intestinalis was previously shown to be expressible in mice and to cause no physiological disturbance under unstressed conditions. Because AOX is known to become activated under some metabolic stress conditions, resulting in altered energy balance, we studied its effects in mice subjected to dietary stress. Wild‐type mice (Mus musculus, strain C57BL/6JOlaHsd) fed a high‐fat or ketogenic (high‐fat, low‐carbohydrate) diet show weight gain with increased fat mass, as well as loss of performance, compared with chow‐fed animals. Unexpectedly, AOX‐expressing mice fed on these metabolically stressful, fat‐rich diets showed almost indistinguishable patterns of weight gain and altered body composition as control animals. Cardiac performance was impaired to a similar extent by ketogenic diet in AOX mice as in nontransgenic littermates. AOX and control animals fed on ketogenic diet both showed wide variance in weight gain. Analysis of the gut microbiome in stool revealed a strong correlation with diet, rather than with genotype. The microbiome of the most and least obese outliers reared on the ketogenic diet showed no consistent trends compared with animals of normal body weight. We conclude that AOX expression in mice does not modify physiological responses to extreme diets.
Collapse
Affiliation(s)
- Praveen K Dhandapani
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Annina M Lyyski
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Lars Paulin
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Nahid A Khan
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anu Suomalainen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Petri Auvinen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Eric Dufour
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Marten Szibor
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Howard T Jacobs
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
24
|
Kollmus H, Fuchs H, Lengger C, Haselimashhadi H, Bogue MA, Östereicher MA, Horsch M, Adler T, Aguilar-Pimentel JA, Amarie OV, Becker L, Beckers J, Calzada-Wack J, Garrett L, Hans W, Hölter SM, Klein-Rodewald T, Maier H, Mayer-Kuckuk P, Miller G, Moreth K, Neff F, Rathkolb B, Rácz I, Rozman J, Spielmann N, Treise I, Busch D, Graw J, Klopstock T, Wolf E, Wurst W, Yildirim AÖ, Mason J, Torres A, Balling R, Mehaan T, Gailus-Durner V, Schughart K, Hrabě de Angelis M. A comprehensive and comparative phenotypic analysis of the collaborative founder strains identifies new and known phenotypes. Mamm Genome 2020; 31:30-48. [PMID: 32060626 PMCID: PMC7060152 DOI: 10.1007/s00335-020-09827-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/31/2020] [Indexed: 01/21/2023]
Abstract
The collaborative cross (CC) is a large panel of mouse-inbred lines derived from eight founder strains (NOD/ShiLtJ, NZO/HILtJ, A/J, C57BL/6J, 129S1/SvImJ, CAST/EiJ, PWK/PhJ, and WSB/EiJ). Here, we performed a comprehensive and comparative phenotyping screening to identify phenotypic differences and similarities between the eight founder strains. In total, more than 300 parameters including allergy, behavior, cardiovascular, clinical blood chemistry, dysmorphology, bone and cartilage, energy metabolism, eye and vision, immunology, lung function, neurology, nociception, and pathology were analyzed; in most traits from sixteen females and sixteen males. We identified over 270 parameters that were significantly different between strains. This study highlights the value of the founder and CC strains for phenotype-genotype associations of many genetic traits that are highly relevant to human diseases. All data described here are publicly available from the mouse phenome database for analyses and downloads.
Collapse
Affiliation(s)
- Heike Kollmus
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Inhoffenstr.7, 38124, Braunschweig, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Christoph Lengger
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Hamed Haselimashhadi
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | | | - Manuela A Östereicher
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Marion Horsch
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Thure Adler
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Juan Antonio Aguilar-Pimentel
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Oana Veronica Amarie
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Lore Becker
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Johannes Beckers
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Alte Akademie 8, 85354, Freising, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Julia Calzada-Wack
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Lillian Garrett
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Wolfgang Hans
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Sabine M Hölter
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Tanja Klein-Rodewald
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Holger Maier
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Philipp Mayer-Kuckuk
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Gregor Miller
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Kristin Moreth
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Frauke Neff
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Birgit Rathkolb
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University München, Feodor-Lynen Str. 25, 81377, Munich, Germany
| | - Ildikó Rácz
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Clinic of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Jan Rozman
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Nadine Spielmann
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Irina Treise
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Dirk Busch
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Trogerstrasse 30, 81675, Munich, Germany
| | - Jochen Graw
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Thomas Klopstock
- Department of Neurology, Friedrich-Baur-Institute, Klinikum Der Ludwig-Maximilians-Universität München, Ziemssenstr. 1a, 80336, Munich, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Site Munich, Feodor-Lynen-Str. 17, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 17, 81377, Munich, Germany
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University München, Feodor-Lynen Str. 25, 81377, Munich, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Site Munich, Feodor-Lynen-Str. 17, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 17, 81377, Munich, Germany
- Chair of Developmental Genetics, Technische Universität München-Weihenstephan, C/O Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Ali Önder Yildirim
- Institute of Lung Biology and Disease, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- German Center for Lung Research, Marburg, Germany
| | - Jeremy Mason
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Arturo Torres
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Rudi Balling
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg, Luxembourg
| | - Terry Mehaan
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Valerie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Klaus Schughart
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Inhoffenstr.7, 38124, Braunschweig, Germany.
- University of Veterinary Medicine Hannover, Hanover, Germany.
- University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Alte Akademie 8, 85354, Freising, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| |
Collapse
|
25
|
The glucocorticoid receptor agonistic modulators CpdX and CpdX-D3 do not generate the debilitating effects of synthetic glucocorticoids. Proc Natl Acad Sci U S A 2019; 116:14200-14209. [PMID: 31221758 DOI: 10.1073/pnas.1908264116] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Seventy years after the discovery of their anti-inflammatory properties, glucocorticoids (GCs) remain the mainstay treatment for major allergic and inflammatory disorders, such as atopic dermatitis, asthma, rheumatoid arthritis, colitis, and conjunctivitis, among others. However, their long-term therapeutical administration is limited by major debilitating side effects, e.g., skin atrophy, osteoporosis, Addison-like adrenal insufficiency, fatty liver, and type 2 diabetes syndrome, as well as growth inhibition in children. These undesirable side effects are mostly related to GC-induced activation of both the direct transactivation and the direct transrepression functions of the GC receptor (GR), whereas the activation of its GC-induced indirect tethered transrepression function results in beneficial anti-inflammatory effects. We have reported in the accompanying paper that the nonsteroidal compound CpdX as well as its deuterated form CpdX-D3 selectively activate the GR indirect transrepression function and are as effective as synthetic GCs at repressing inflammations generated in several mouse models of major pathologies. We now demonstrate that these CpdX compounds are bona fide selective GC receptor agonistic modulators (SEGRAMs) as none of the known GC-induced debilitating side effects were observed in the mouse upon 3-mo CpdX treatments. We notably report that, unlike that of GCs, the administration of CpdX to ovariectomized (OVX) mice does not induce a fatty liver nor type 2 diabetes, which indicates that CpdX could be used in postmenopausal women as an efficient "harmless" GC substitute.
Collapse
|
26
|
van der Sluis RJ, Depuydt MAC, Verwilligen RAF, Hoekstra M, Van Eck M. Elimination of adrenocortical apolipoprotein E production does not impact glucocorticoid output in wild-type mice. Mol Cell Endocrinol 2019; 490:21-27. [PMID: 30953750 DOI: 10.1016/j.mce.2019.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 01/07/2019] [Accepted: 04/02/2019] [Indexed: 12/26/2022]
Abstract
Apolipoprotein E (APOE) deficient mice exhibit unexplained hypercorticosteronemia. Given that APOE is also produced locally within the adrenals, we evaluated the effect of adrenal-specific APOE deficiency on the glucocorticoid function. Hereto, one adrenal containing or lacking APOE was transplanted into adrenalectomized wild-type mice. Adrenal APOE deficiency did not impact adrenal total cholesterol levels. Importantly, the ability of the two adrenal types to produce glucocorticoids was also not different as judged from the similar plasma corticosterone levels. Adrenal mRNA expression levels of HMG-CoA reductase and the LDL receptor were decreased by respectively 72% (p < 0.01) and 65% (p = 0.07), suggesting that cholesterol acquisition pathways were inhibited to possibly compensate the lack of APOE. In support, a parallel increase in the expression level of the cholesterol accumulation-associated ER stress marker CHOP was detected (+117%; p < 0.05). In conclusion, our studies show that elimination of adrenocortical APOE production does not impact glucocorticoid output in wild-type mice.
Collapse
Affiliation(s)
- Ronald J van der Sluis
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, 2333CC, Leiden, the Netherlands.
| | - Marie A C Depuydt
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, 2333CC, Leiden, the Netherlands
| | - Robin A F Verwilligen
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, 2333CC, Leiden, the Netherlands
| | - Menno Hoekstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, 2333CC, Leiden, the Netherlands
| | - Miranda Van Eck
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, 2333CC, Leiden, the Netherlands
| |
Collapse
|
27
|
Poeker SA, Lacroix C, de Wouters T, Spalinger MR, Scharl M, Geirnaert A. Stepwise Development of an in vitro Continuous Fermentation Model for the Murine Caecal Microbiota. Front Microbiol 2019; 10:1166. [PMID: 31191488 PMCID: PMC6548829 DOI: 10.3389/fmicb.2019.01166] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 05/07/2019] [Indexed: 12/24/2022] Open
Abstract
Murine models are valuable tools to study the role of gut microbiota in health or disease. However, murine and human microbiota differ in species composition, so further investigation of the murine gut microbiota is important to gain a better mechanistic understanding. Continuous in vitro fermentation models are powerful tools to investigate microbe-microbe interactions while circumventing animal testing and host confounding factors, but are lacking for murine gut microbiota. We therefore developed a novel continuous fermentation model based on the PolyFermS platform adapted to the murine caecum and inoculated with immobilized caecal microbiota. We followed a stepwise model development approach by adjusting parameters [pH, retention time (RT), growth medium] to reach fermentation metabolite profiles and marker bacterial levels similar to the inoculum. The final model had a stable and inoculum-alike fermentation profile during continuous operation. A lower pH during startup and continuous operation stimulated bacterial fermentation (115 mM short-chain fatty acids at pH 7 to 159 mM at pH 6.5). Adjustments to nutritive medium, a decreased pH and increased RT helped control the in vitro Enterobacteriaceae levels, which often bloom in fermentation models, to 6.6 log gene copies/mL in final model. In parallel, the Lactobacillus, Lachnospiraceae, and Ruminococcaceae levels were better maintained in vitro with concentrations of 8.5 log gene copies/mL, 8.8 log gene copies/mL and 7.5 log gene copies/mL, respectively, in the final model. An independent repetition with final model parameters showed reproducible results in maintaining the inoculum fermentation metabolite profile and its marker bacterial levels. Microbiota community analysis of the final model showed a decreased bacterial diversity and compositional differences compared to caecal inoculum microbiota. Most of the caecal bacterial families were represented in vitro, but taxa of the Muribaculaceae family were not maintained. Functional metagenomics prediction showed conserved metabolic and functional KEGG pathways between in vitro and caecal inoculum microbiota. To conclude, we showed that a rational and stepwise approach allowed us to model in vitro the murine caecal microbiota and functions. Our model is a first step to develop murine microbiota model systems and offers the potential to study microbiota functionality and structure ex vivo.
Collapse
Affiliation(s)
- Sophie A Poeker
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Tomas de Wouters
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Marianne R Spalinger
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Annelies Geirnaert
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
28
|
Vogel H, Kamitz A, Hallahan N, Lebek S, Schallschmidt T, Jonas W, Jähnert M, Gottmann P, Zellner L, Kanzleiter T, Damen M, Altenhofen D, Burkhardt R, Renner S, Dahlhoff M, Wolf E, Müller TD, Blüher M, Joost HG, Chadt A, Al-Hasani H, Schürmann A. A collective diabetes cross in combination with a computational framework to dissect the genetics of human obesity and Type 2 diabetes. Hum Mol Genet 2019; 27:3099-3112. [PMID: 29893858 PMCID: PMC6097155 DOI: 10.1093/hmg/ddy217] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/29/2018] [Indexed: 12/16/2022] Open
Abstract
To explore the genetic determinants of obesity and Type 2 diabetes (T2D), the German Center for Diabetes Research (DZD) conducted crossbreedings of the obese and diabetes-prone New Zealand Obese mouse strain with four different lean strains (B6, DBA, C3H, 129P2) that vary in their susceptibility to develop T2D. Genome-wide linkage analyses localized more than 290 quantitative trait loci (QTL) for obesity, 190 QTL for diabetes-related traits and 100 QTL for plasma metabolites in the outcross populations. A computational framework was developed that allowed to refine critical regions and to nominate a small number of candidate genes by integrating reciprocal haplotype mapping and transcriptome data. The efficiency of the complex procedure was demonstrated for one obesity QTL. The genomic interval of 35 Mb with 502 annotated candidate genes was narrowed down to six candidates. Accordingly, congenic mice retained the obesity phenotype owing to an interval that contains three of the six candidate genes. Among these the phospholipase PLA2G4A exhibited an elevated expression in adipose tissue of obese human subjects and is therefore a critical regulator of the obesity locus. Together, our broad and complex approach demonstrates that combined- and comparative-cross analysis exhibits improved mapping resolution and represents a valid tool for the identification of disease genes.
Collapse
Affiliation(s)
- Heike Vogel
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal D-14558, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany
| | - Anne Kamitz
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal D-14558, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany
| | - Nicole Hallahan
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal D-14558, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany
| | - Sandra Lebek
- German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany.,Medical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Heinrich Heine University, Düsseldorf D-40225, Germany
| | - Tanja Schallschmidt
- German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany.,Medical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Heinrich Heine University, Düsseldorf D-40225, Germany
| | - Wenke Jonas
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal D-14558, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany
| | - Markus Jähnert
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal D-14558, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany
| | - Pascal Gottmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal D-14558, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany
| | - Lisa Zellner
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal D-14558, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany
| | - Timo Kanzleiter
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal D-14558, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany
| | - Mareike Damen
- German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany.,Medical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Heinrich Heine University, Düsseldorf D-40225, Germany
| | - Delsi Altenhofen
- German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany.,Medical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Heinrich Heine University, Düsseldorf D-40225, Germany
| | - Ralph Burkhardt
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig D-04303, Germany
| | - Simone Renner
- German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany.,Chair for Molecular Animal Breeding and Biotechnology, Gene Center.,Department of Veterinary Sciences, Center for Innovative Medical Models (CiMM), LMU Munich, D-81377 Munich, Germany
| | - Maik Dahlhoff
- German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany.,Chair for Molecular Animal Breeding and Biotechnology, Gene Center.,Department of Veterinary Sciences, Center for Innovative Medical Models (CiMM), LMU Munich, D-81377 Munich, Germany
| | - Eckhard Wolf
- German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany.,Chair for Molecular Animal Breeding and Biotechnology, Gene Center.,Department of Veterinary Sciences, Center for Innovative Medical Models (CiMM), LMU Munich, D-81377 Munich, Germany
| | - Timo D Müller
- German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany.,Institute for Diabetes and Obesity, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg D-85764, Germany.,Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich D-80333, Germany
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig D-04103, Germany
| | - Hans-Georg Joost
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal D-14558, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany
| | - Alexandra Chadt
- German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany.,Medical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Heinrich Heine University, Düsseldorf D-40225, Germany
| | - Hadi Al-Hasani
- German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany.,Medical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Heinrich Heine University, Düsseldorf D-40225, Germany
| | - Annette Schürmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal D-14558, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg D-85764, Germany.,Institute of Nutritional Science, University of Potsdam, Nuthetal D-14558, Germany
| |
Collapse
|
29
|
Oh J, Magnuson A, Benoist C, Pittet MJ, Weissleder R. Age-related tumor growth in mice is related to integrin α 4 in CD8+ T cells. JCI Insight 2018; 3:122961. [PMID: 30385729 DOI: 10.1172/jci.insight.122961] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/27/2018] [Indexed: 11/17/2022] Open
Abstract
Cancer incidence increases with age, but paradoxically, cancers have been found to grow more quickly in young mice compared with aged ones. The cause of differential tumor growth has been debated and, over time, attributed to faster tumor cell proliferation, decreased tumor cell apoptosis, and/or increased angiogenesis in young animals. Despite major advances in our understanding of tumor immunity over the past 2 decades, little attention has been paid to comparing immune cell populations in young and aged mice. Using mouse colon adenocarcinoma model MC38 implanted in young and mature mice, we show that age substantially influences the number of tumor-infiltrating cytotoxic CD8+ T cells, which control cancer progression. The different tumor growth pace in young and mature mice was abrogated in RAG1null mice, which lack mature T and B lymphocytes, and upon selective depletion of endogenous CD8+ cells. Transcriptome analysis further indicated that young mice have decreased levels of the Itga4 gene (CD49d, VLA-4) in tumor-infiltrating lymphocytes when compared with mature mice. Hypothesizing that VLA-4 can have a tumor-protective effect, we depleted the protein, which resulted in accelerated tumor growth in mature mice. These observations may explain the paradoxical growth rates observed in murine cancers, point to the central role of VLA-4 in controlling tumor growth, and open new venues to therapeutic manipulation.
Collapse
Affiliation(s)
- Juhyun Oh
- Center for Systems Biology, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA
| | - Angela Magnuson
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Christophe Benoist
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Mikael J Pittet
- Center for Systems Biology, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA.,Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
30
|
van der Sluis RJ, Verwilligen RAF, Lendvai Z, Wever R, Hoekstra M, Van Eck M. HDL is essential for atherosclerotic lesion regression in Apoe knockout mice by bone marrow Apoe reconstitution. Atherosclerosis 2018; 278:240-249. [PMID: 30340108 DOI: 10.1016/j.atherosclerosis.2018.09.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/05/2018] [Accepted: 09/26/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND AND AIMS Although studies in mice have suggested that lesion regression is feasible, the underlying mechanisms remain largely unknown. Here we determined the impact of high-density lipoprotein (HDL) on atherosclerosis regression outcome. METHODS Atherosclerotic lesion dynamics were studied upon bone marrow transplantation-mediated re-introduction of apolipoprotein E (Apoe) in Apoe knockout mice. Probucol was used to pharmacologically deplete HDL. RESULTS Restoration of Apoe function was associated with an initial growth of atherosclerotic lesions and parallel decrease in lesional macrophage foam cell content (47 ± 4% at 4 weeks versus 72 ± 2% at baseline: p < 0.001), despite the fact that cholesterol levels were markedly reduced. Notably, significant lesion regression was detected from 4 weeks onwards, when plasma cholesterol levels had returned to the normolipidemic range. As a result, lesions were 41% smaller (p < 0.05) at 8 weeks than at 4 weeks after bone marrow transplantation. Regressed lesions contained an even lower level of macrophage foam cells (33 ± 5%: p < 0.001) and were rich in collagen. Probucol co-treatment was associated with a 3.2-fold lower (p < 0.05) plasma HDL-cholesterol level and a more pro-inflammatory (CCR2+) monocyte phenotype. Importantly, probucol-treated mice exhibited atherosclerotic lesions that were larger than those of regular chow diet-fed bone marrow transplanted mice at 8 weeks (186 ± 15*103 μm2 for probucol-treated versus 120 ± 19*103 μm2 for controls: p < 0.05). CONCLUSIONS We have shown that probucol-induced HDL deficiency impairs the ability of established lesions to regress in response to reversal of the genetic hypercholesterolemia in Apoe knockout mice. Our studies thus highlight a crucial role for HDL in the process of atherosclerosis regression.
Collapse
Affiliation(s)
- Ronald J van der Sluis
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands.
| | - Robin A F Verwilligen
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands
| | - Zsuzsanna Lendvai
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands
| | - Robbert Wever
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands
| | - Menno Hoekstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands
| | - Miranda Van Eck
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands
| |
Collapse
|
31
|
Two Novel Candidate Genes for Insulin Secretion Identified by Comparative Genomics of Multiple Backcross Mouse Populations. Genetics 2018; 210:1527-1542. [PMID: 30341086 DOI: 10.1534/genetics.118.301578] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 10/16/2018] [Indexed: 12/28/2022] Open
Abstract
To identify novel disease genes for type 2 diabetes (T2D) we generated two backcross populations of obese and diabetes-susceptible New Zealand Obese (NZO/HI) mice with the two lean mouse strains 129P2/OlaHsd and C3HeB/FeJ. Subsequent whole-genome linkage scans revealed 30 novel quantitative trait loci (QTL) for T2D-associated traits. The strongest association with blood glucose [12 cM, logarithm of the odds (LOD) 13.3] and plasma insulin (17 cM, LOD 4.8) was detected on proximal chromosome 7 (designated Nbg7p, NZO blood glucose on proximal chromosome 7) exclusively in the NZOxC3H crossbreeding, suggesting that the causal gene is contributed by the C3H genome. Introgression of the critical C3H fragment into the genetic NZO background by generating recombinant congenic strains and metabolic phenotyping validated the phenotype. For the detection of candidate genes in the critical region (30-46 Mb), we used a combined approach of haplotype and gene expression analysis to search for C3H-specific gene variants in the pancreatic islets, which appeared to be the most likely target tissue for the QTL. Two genes, Atp4a and Pop4, fulfilled the criteria from our candidate gene approaches. The knockdown of both genes in MIN6 cells led to decreased glucose-stimulated insulin secretion, indicating a regulatory role of both genes in insulin secretion, thereby possibly contributing to the phenotype linked to Nbg7p In conclusion, our combined- and comparative-cross analysis approach has successfully led to the identification of two novel diabetes susceptibility candidate genes, and thus has been proven to be a valuable tool for the discovery of novel disease genes.
Collapse
|
32
|
Bernard DJ, Pangilinan FJ, Cheng J, Molloy AM, Brody LC. Mice lacking the transcobalamin-vitamin B12 receptor, CD320, suffer from anemia and reproductive deficits when fed vitamin B12-deficient diet. Hum Mol Genet 2018; 27:3627-3640. [PMID: 30124850 PMCID: PMC6168973 DOI: 10.1093/hmg/ddy267] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/29/2018] [Accepted: 07/12/2018] [Indexed: 11/12/2022] Open
Abstract
In humans, poor nutrition, malabsorption and variation in cobalamin (vitamin B12) metabolic genes are associated with hematological, neurological and developmental pathologies. Cobalamin is transported from blood into tissues via the transcobalamin (TC) receptor encoded by the CD320 gene. We created mice carrying a targeted deletion of the mouse ortholog, Cd320. Knockout (KO) mice lacking this TC receptor have elevated levels of plasma methylmalonic acid and homocysteine but are otherwise healthy, viable, fertile and not anemic. To challenge the Cd320 KO mice we maintained them on a vitamin B12-deficient diet. After 5 weeks on this diet, reproductive failure develops in Cd320 KO females but not males. In vitro, homozygous Cd320 KO embryos from cobalamin-deficient Cd320 KO dams develop normally to embryonic day (E) 3.5, while in vivo, few uterine decidual implantation sites are observed at E7.5, suggesting that embryos perish around the time of implantation. Dietary restriction of vitamin B12 induces a severe macrocytic anemia in Cd320 KO mice after 10-12 months while control mice on this diet are anemia-free up to 2 years. Despite the severe anemia, cobalamin-deficient KO mice do not exhibit obvious neurological symptoms. Our results with Cd320 KO mice suggest that an alternative mechanism exists for mice to transport cobalamin independent of the Cd320 encoded receptor. Our findings with deficient diet are consistent with historical and epidemiological data suggesting that low vitamin B12 levels in humans are associated with infertility and developmental abnormalities. Our Cd320 KO mouse model is an ideal model system for studying vitamin B12 deficiency.
Collapse
Affiliation(s)
- David J Bernard
- Division of Intramural Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Faith J Pangilinan
- Division of Intramural Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jun Cheng
- Transgenic Mouse Core, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anne M Molloy
- School of Medicine, Trinity College, Dublin, Ireland
| | - Lawrence C Brody
- Division of Intramural Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
33
|
Haissaguerre M, Ferriere A, Clark S, Guzman-Quevedo O, Tabarin A, Cota D. NPV-BSK805, an Antineoplastic Jak2 Inhibitor Effective in Myeloproliferative Disorders, Causes Adiposity in Mice by Interfering With the Action of Leptin. Front Pharmacol 2018; 9:527. [PMID: 29867515 PMCID: PMC5962752 DOI: 10.3389/fphar.2018.00527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 05/02/2018] [Indexed: 11/23/2022] Open
Abstract
The pathophysiology of body weight gain that is observed in patients suffering from myeloproliferative neoplasms treated with inhibitors of the janus kinase (Jak) 1 and 2 pathway remains unknown. Here we hypothesized that this class of drugs interferes with the metabolic actions of leptin, as this hormone requires functional Jak2 signaling. To test this, C57BL/6J chow-fed mice received either chronic intraperitoneal (ip) or repeated intracerebroventricular (icv) administration of the selective Jak2 inhibitor NVP-BSK805, which was proven efficacious in treating polycythemia in rodents. Changes in food intake, body weight and body composition were recorded. Icv NVP-BSK805 was combined with ip leptin to evaluate ability to interfere with the action of this hormone on food intake and on induction of hypothalamic phosphorylation of signal transducer and activator of transcription 3 (STAT3). We found that chronic peripheral administration of NVP-BSK805 did not alter food intake, but increased fat mass and feed efficiency. The increase in fat mass was more pronounced during repeated icv administration of the compound, suggesting that metabolic effects were related to molecular interference in brain structures regulating energy balance. Accordingly, acute icv administration of NVP-BSK805 prevented the ability of leptin to decrease food intake and body weight by impeding STAT3 phosphorylation within the hypothalamus. Consequently, acute icv administration of NVP-BSK805 at higher dose induced hyperphagia and body weight gain. Our results provide evidence for a specific anabolic effect exerted by antineoplastic drugs targeting the Jak2 pathway, which is due to interference with the actions of leptin. Consequently, assessment of metabolic variables related to increased fat mass gain should be performed in patients treated with Jak2 inhibitors.
Collapse
Affiliation(s)
- Magalie Haissaguerre
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- Service d’Endocrinologie, Diabétologie et Nutrition, Hôpital Haut-Lévêque, CHU de Bordeaux, Pessac, France
| | - Amandine Ferriere
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- Service d’Endocrinologie, Diabétologie et Nutrition, Hôpital Haut-Lévêque, CHU de Bordeaux, Pessac, France
| | - Samantha Clark
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
| | - Omar Guzman-Quevedo
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- Facultad de Químico-Farmacobiología, Universidad Michoacána de San Nicolás de Hidalgo, Morelia, Mexico
| | - Antoine Tabarin
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- Service d’Endocrinologie, Diabétologie et Nutrition, Hôpital Haut-Lévêque, CHU de Bordeaux, Pessac, France
- *Correspondence: Antoine Tabarin, Daniela Cota,
| | - Daniela Cota
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- *Correspondence: Antoine Tabarin, Daniela Cota,
| |
Collapse
|
34
|
Qin K, Zhang N, Zhang Z, Nipper M, Zhu Z, Leighton J, Xu K, Musi N, Wang P. SIRT6-mediated transcriptional suppression of Txnip is critical for pancreatic beta cell function and survival in mice. Diabetologia 2018; 61:906-918. [PMID: 29322219 PMCID: PMC6203439 DOI: 10.1007/s00125-017-4542-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 12/04/2017] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS Better understanding of how genetic and epigenetic components control beta cell differentiation and function is key to the discovery of novel therapeutic approaches to prevent beta cell dysfunction and failure in the progression of type 2 diabetes. Our goal was to elucidate the role of histone deacetylase sirtuin 6 (SIRT6) in beta cell development and homeostasis. METHODS Sirt6 endocrine progenitor cell conditional knockout and beta cell-specific knockout mice were generated using the Cre-loxP system. Mice were assayed for islet morphology, glucose tolerance, glucose-stimulated insulin secretion and susceptibility to streptozotocin. Transcriptional regulatory functions of SIRT6 in primary islets were evaluated by RNA-Seq analysis. Reverse transcription-quantitative (RT-q)PCR and immunoblot were used to verify and investigate the gene expression changes. Chromatin occupancies of SIRT6, H3K9Ac, H3K56Ac and active RNA polymerase II were evaluated by chromatin immunoprecipitation. RESULTS Deletion of Sirt6 in pancreatic endocrine progenitor cells did not affect endocrine morphology, beta cell mass or insulin production but did result in glucose intolerance and defective glucose-stimulated insulin secretion in mice. Conditional deletion of Sirt6 in adult beta cells reproduced the insulin secretion defect. Loss of Sirt6 resulted in aberrant upregulation of thioredoxin-interacting protein (TXNIP) in beta cells. SIRT6 deficiency led to increased acetylation of histone H3 lysine residue at 9 (H3K9Ac), acetylation of histone H3 lysine residue at 56 (H3K56Ac) and active RNA polymerase II at the promoter region of Txnip. SIRT6-deficient beta cells exhibited a time-dependent increase in H3K9Ac, H3K56Ac and TXNIP levels. Finally, beta cell-specific SIRT6-deficient mice showed increased sensitivity to streptozotocin. CONCLUSIONS/INTERPRETATION Our results reveal that SIRT6 suppresses Txnip expression in beta cells via deacetylation of histone H3 and plays a critical role in maintaining beta cell function and viability. DATA AVAILABILITY Sequence data have been deposited in the National Institutes of Health (NIH) Gene Expression Omnibus (GEO) with the accession code GSE104161.
Collapse
Affiliation(s)
- Kunhua Qin
- Department of Molecular Medicine, University of Texas Health Science Centre at San Antonio, San Antonio, TX, USA
- Department of Cell Systems & Anatomy, University of Texas Health Science Centre at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Ning Zhang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Centre at San Antonio, San Antonio, TX, USA
| | - Zhao Zhang
- Department of Molecular Medicine, University of Texas Health Science Centre at San Antonio, San Antonio, TX, USA
| | - Michael Nipper
- Department of Cell Systems & Anatomy, University of Texas Health Science Centre at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Zhenxin Zhu
- Department of Cell Systems & Anatomy, University of Texas Health Science Centre at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Jake Leighton
- Department of Cell Systems & Anatomy, University of Texas Health Science Centre at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Kexin Xu
- Department of Molecular Medicine, University of Texas Health Science Centre at San Antonio, San Antonio, TX, USA
| | - Nicolas Musi
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Centre at San Antonio, San Antonio, TX, USA
| | - Pei Wang
- Department of Cell Systems & Anatomy, University of Texas Health Science Centre at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
| |
Collapse
|
35
|
Ryu D, Zhang H, Ropelle ER, Sorrentino V, Mázala DAG, Mouchiroud L, Marshall PL, Campbell MD, Ali AS, Knowels GM, Bellemin S, Iyer SR, Wang X, Gariani K, Sauve AA, Cantó C, Conley KE, Walter L, Lovering RM, Chin ER, Jasmin BJ, Marcinek DJ, Menzies KJ, Auwerx J. NAD+ repletion improves muscle function in muscular dystrophy and counters global PARylation. Sci Transl Med 2017; 8:361ra139. [PMID: 27798264 DOI: 10.1126/scitranslmed.aaf5504] [Citation(s) in RCA: 200] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 09/12/2016] [Indexed: 12/25/2022]
Abstract
Neuromuscular diseases are often caused by inherited mutations that lead to progressive skeletal muscle weakness and degeneration. In diverse populations of normal healthy mice, we observed correlations between the abundance of mRNA transcripts related to mitochondrial biogenesis, the dystrophin-sarcoglycan complex, and nicotinamide adenine dinucleotide (NAD+) synthesis, consistent with a potential role for the essential cofactor NAD+ in protecting muscle from metabolic and structural degeneration. Furthermore, the skeletal muscle transcriptomes of patients with Duchene's muscular dystrophy (DMD) and other muscle diseases were enriched for various poly[adenosine 5'-diphosphate (ADP)-ribose] polymerases (PARPs) and for nicotinamide N-methyltransferase (NNMT), enzymes that are major consumers of NAD+ and are involved in pleiotropic events, including inflammation. In the mdx mouse model of DMD, we observed significant reductions in muscle NAD+ levels, concurrent increases in PARP activity, and reduced expression of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme for NAD+ biosynthesis. Replenishing NAD+ stores with dietary nicotinamide riboside supplementation improved muscle function and heart pathology in mdx and mdx/Utr-/- mice and reversed pathology in Caenorhabditis elegans models of DMD. The effects of NAD+ repletion in mdx mice relied on the improvement in mitochondrial function and structural protein expression (α-dystrobrevin and δ-sarcoglycan) and on the reductions in general poly(ADP)-ribosylation, inflammation, and fibrosis. In combination, these studies suggest that the replenishment of NAD+ may benefit patients with muscular dystrophies or other neuromuscular degenerative conditions characterized by the PARP/NNMT gene expression signatures.
Collapse
Affiliation(s)
- Dongryeol Ryu
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Hongbo Zhang
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Eduardo R Ropelle
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.,Laboratory of Molecular Biology of Exercise, School of Applied Science, University of Campinas, CEP 13484-350 Limeira, São Paulo, Brazil
| | - Vincenzo Sorrentino
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Davi A G Mázala
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD 20742, USA
| | - Laurent Mouchiroud
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Philip L Marshall
- Interdisciplinary School of Health Sciences, University of Ottawa Brain and Mind Research Institute and Centre for Neuromuscular Disease, Ottawa, Ontario K1H 8M5, Canada
| | - Matthew D Campbell
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| | - Amir Safi Ali
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| | - Gary M Knowels
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| | - Stéphanie Bellemin
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, Université Claude Bernard Lyon 1, CNRS UMR 5534, 69622 Villeurbanne, France
| | - Shama R Iyer
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Xu Wang
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Karim Gariani
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Anthony A Sauve
- Department of Pharmacology, Weill Cornell Medical School, New York, NY 10065, USA
| | - Carles Cantó
- Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Kevin E Conley
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| | - Ludivine Walter
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, Université Claude Bernard Lyon 1, CNRS UMR 5534, 69622 Villeurbanne, France
| | - Richard M Lovering
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Eva R Chin
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD 20742, USA
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - David J Marcinek
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| | - Keir J Menzies
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland. .,Interdisciplinary School of Health Sciences, University of Ottawa Brain and Mind Research Institute and Centre for Neuromuscular Disease, Ottawa, Ontario K1H 8M5, Canada
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
36
|
Thaete LG, Qu XW, Neerhof MG, Hirsch E, Jilling T. Fetal Growth Restriction Induced by Transient Uterine Ischemia-Reperfusion: Differential Responses in Different Mouse Strains. Reprod Sci 2017; 25:1083-1092. [PMID: 28946817 DOI: 10.1177/1933719117732160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We characterized fetal and placental growth and uterine and placental inflammation in pregnant C3H/HeOuJ and C57BL/6J mice (strains with different sensitivities to metabolic and circulatory pathologies), using different uterine ischemia/reperfusion (I/R) protocols, to establish and refine a murine model of I/R-induced fetal growth restriction (FGR). Pregnant C3H/HeOuJ mice on gestation day 15 were subjected to unilateral uterine I/R by (1) total blood flow restriction (TFR) by occlusion of the right ovarian and uterine arteries for 30 minutes, (2) partial flow restriction (PFR) by occlusion of only the right ovarian artery for 30 minutes, or (3) sham surgery. Pregnant C57BL/6J mice were treated the same, but on gestation day 14 and with TFR for only 5 minutes due to high sensitivity of C57BL/6J mice to I/R. Four days post-I/R, the animals were euthanized to determine fetal and placental weight and fetal loss and to assay placental myeloperoxidase (MPO) activity. In C3H/HeOuJ mice, TFR/30 minutes induced significantly ( P < .05) lower fetal and placental weights and higher placental MPO activity, compared to controls. The PFR/30 minutes produced the same effects except placental weights were not reduced. In contrast, in C57BL/6J mice, TFR for only 5 minutes was sufficient to induce FGR and increase fetal loss; while PFR/30 minutes lowered fetal but not placental weights and increased fetal loss but not placental MPO activity. In summary, we present the first published model of I/R-induced FGR in mice. We find that mice of different strains have differing sensitivities to uterine I/R, therefore differing I/R response mechanisms.
Collapse
Affiliation(s)
- Larry G Thaete
- 1 Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, USA.,2 Department of Obstetrics and Gynecology, The University of Chicago, Pritzker School of Medicine, Chicago, IL, USA
| | - Xiao-Wu Qu
- 1 Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, USA
| | - Mark G Neerhof
- 1 Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, USA.,2 Department of Obstetrics and Gynecology, The University of Chicago, Pritzker School of Medicine, Chicago, IL, USA
| | - Emmet Hirsch
- 1 Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, USA.,2 Department of Obstetrics and Gynecology, The University of Chicago, Pritzker School of Medicine, Chicago, IL, USA
| | - Tamas Jilling
- 3 Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
37
|
Iron deposition is associated with differential macrophage infiltration and therapeutic response to iron chelation in prostate cancer. Sci Rep 2017; 7:11632. [PMID: 28912459 PMCID: PMC5599545 DOI: 10.1038/s41598-017-11899-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/24/2017] [Indexed: 02/06/2023] Open
Abstract
Immune cells such as macrophages are drivers and biomarkers of most cancers. Scoring macrophage infiltration in tumor tissue provides a prognostic assessment that is correlated with disease outcome and therapeutic response, but generally requires invasive biopsy. Routine detection of hemosiderin iron aggregates in macrophages in other settings histologically and in vivo by MRI suggests that similar assessments in cancer can bridge a gap in our ability to assess tumor macrophage infiltration. Quantitative histological and in vivo MRI assessments of non-heme cellular iron revealed that preclinical prostate tumor models could be differentiated according to hemosiderin iron accumulation-both in tumors and systemically. Monitoring cellular iron levels during "off-label" administration of the FDA-approved iron chelator deferiprone evidenced significant reductions in tumor size without extensive perturbation to these iron deposits. Spatial profiling of the iron-laden infiltrates further demonstrated that higher numbers of infiltrating macrophage iron deposits was associated with lower anti-tumor chelation therapy response. Imaging macrophages according to their innate iron status provides a new phenotypic window into the immune tumor landscape and reveals a prognostic biomarker associated with macrophage infiltration and therapeutic outcome.
Collapse
|
38
|
Cases O, Obry A, Ben-Yacoub S, Augustin S, Joseph A, Toutirais G, Simonutti M, Christ A, Cosette P, Kozyraki R. Impaired vitreous composition and retinal pigment epithelium function in the FoxG1::LRP2 myopic mice. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1242-1254. [DOI: 10.1016/j.bbadis.2017.03.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 03/09/2017] [Accepted: 03/29/2017] [Indexed: 01/12/2023]
|
39
|
Montoliu I, Cominetti O, Boulangé CL, Berger B, Siddharth J, Nicholson J, Martin FPJ. Modeling Longitudinal Metabonomics and Microbiota Interactions in C57BL/6 Mice Fed a High Fat Diet. Anal Chem 2016; 88:7617-26. [PMID: 27396289 DOI: 10.1021/acs.analchem.6b01343] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Longitudinal studies aim typically at following populations of subjects over time and are important to understand the global evolution of biological processes. When it comes to longitudinal omics data, it will often depend on the overall objective of the study, and constraints imposed by the data, to define the appropriate modeling tools. Here, we report the use of multilevel simultaneous component analysis (MSCA), orthogonal projection on latent structures (OPLS), and regularized canonical correlation analysis (rCCA) to study associations between specific longitudinal urine metabonomics data and microbiome data in a diet-induced obesity model using C57BL/6 mice. (1)H NMR urine metabolic profiling was performed on samples collected weekly over a period of 13 weeks, and stool microbial composition was assessed using 16S rRNA gene sequencing at three specific time periods (baseline, first week response, end of study). MSCA and OPLS allowed us to explore longitudinal urine metabonomics data in relation to the dietary groups, as well as dietary effects on body weight. In addition, we report a data integration strategy based on regularized CCA and correlation analyses of urine metabonomics data and 16S rRNA gene sequencing data to investigate the functional relationships between metabolites and gut microbial composition. Thanks to this workflow enabling the breakdown of this data set complexity, the most relevant patterns could be extracted to further explore physiological processes at an anthropometric, cellular, and molecular level.
Collapse
Affiliation(s)
- Ivan Montoliu
- Nestlé Institute of Health Sciences SA , EPFL Innovation Park, Building H, 1015 Lausanne, Switzerland.,Department of Biomolecular Medicine, Division of Surgery, Oncology, Reproductive Biology and Anaesthetics, Faculty of Medicine, Imperial College London , Sir Alexander Fleming Building, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Ornella Cominetti
- Nestlé Institute of Health Sciences SA , EPFL Innovation Park, Building H, 1015 Lausanne, Switzerland
| | - Claire L Boulangé
- Department of Biomolecular Medicine, Division of Surgery, Oncology, Reproductive Biology and Anaesthetics, Faculty of Medicine, Imperial College London , Sir Alexander Fleming Building, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Bernard Berger
- Nestlé Research Center , Vers-chez-les-Blanc, 1000 Lausanne 26, Switzerland
| | - Jay Siddharth
- Nestlé Institute of Health Sciences SA , EPFL Innovation Park, Building H, 1015 Lausanne, Switzerland
| | - Jeremy Nicholson
- Department of Biomolecular Medicine, Division of Surgery, Oncology, Reproductive Biology and Anaesthetics, Faculty of Medicine, Imperial College London , Sir Alexander Fleming Building, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - François-Pierre J Martin
- Nestlé Institute of Health Sciences SA , EPFL Innovation Park, Building H, 1015 Lausanne, Switzerland
| |
Collapse
|
40
|
Study of the pathogenesis and treatment of diabetes mellitus through animal models. ACTA ACUST UNITED AC 2016; 63:345-53. [PMID: 27246633 DOI: 10.1016/j.endonu.2016.03.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 12/16/2022]
Abstract
Most research in diabetes mellitus (DM) has been conducted in animals, and their replacement is currently a chimera. As compared to when they started to be used by modern science in the 17th century, a very high number of animal models of diabetes is now available, and they provide new insights into almost every aspect of diabetes. Approaches combining human, in vitro, and animal studies are probably the best strategy to improve our understanding of the underlying mechanisms of diabetes, and the choice of the best model to achieve such objective is crucial. Traditionally classified based on pathogenesis as spontaneous or induced models, each has its own advantages and disadvantages. The most common animal models of diabetes are described, and in addition to non-obese diabetic mice, biobreeding diabetes-prone (BB-DP) rats, streptozotocin-induced models, or high-fat diet-induced diabetic C57Bl/6J mice, new valuable models, such as dogs and cats with spontaneous diabetes, are described.
Collapse
|
41
|
Zhang H, Ryu D, Wu Y, Gariani K, Wang X, Luan P, D'Amico D, Ropelle ER, Lutolf MP, Aebersold R, Schoonjans K, Menzies KJ, Auwerx J. NAD⁺ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science 2016; 352:1436-43. [PMID: 27127236 DOI: 10.1126/science.aaf2693] [Citation(s) in RCA: 880] [Impact Index Per Article: 97.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/13/2016] [Indexed: 12/12/2022]
Abstract
Adult stem cells (SCs) are essential for tissue maintenance and regeneration yet are susceptible to senescence during aging. We demonstrate the importance of the amount of the oxidized form of cellular nicotinamide adenine dinucleotide (NAD(+)) and its effect on mitochondrial activity as a pivotal switch to modulate muscle SC (MuSC) senescence. Treatment with the NAD(+) precursor nicotinamide riboside (NR) induced the mitochondrial unfolded protein response and synthesis of prohibitin proteins, and this rejuvenated MuSCs in aged mice. NR also prevented MuSC senescence in the mdx (C57BL/10ScSn-Dmd(mdx)/J) mouse model of muscular dystrophy. We furthermore demonstrate that NR delays senescence of neural SCs and melanocyte SCs and increases mouse life span. Strategies that conserve cellular NAD(+) may reprogram dysfunctional SCs and improve life span in mammals.
Collapse
Affiliation(s)
- Hongbo Zhang
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Dongryeol Ryu
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Yibo Wu
- Department of Biology, Institute of Molecular Systems Biology, Eidgenössische Technische Hochschule Zürich (ETHZ), 8093 Zurich, Switzerland
| | - Karim Gariani
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Xu Wang
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Peiling Luan
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Davide D'Amico
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Eduardo R Ropelle
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland. Laboratory of Molecular Biology of Exercise, School of Applied Science, University of Campinas, CEP 13484-350 Limeira, São Paulo, Brazil
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, EPFL, 1015 Lausanne, Switzerland
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, Eidgenössische Technische Hochschule Zürich (ETHZ), 8093 Zurich, Switzerland. Faculty of Science, University of Zurich, 8057 Zurich, Switzerland
| | | | - Keir J Menzies
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland. Interdisciplinary School of Health Sciences, University of Ottawa Brain and Mind Research Institute, 451 Smyth Road, K1H 8M5 Ottawa, Ontario, Canada.
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
42
|
Jha P, Wang X, Auwerx J. Analysis of Mitochondrial Respiratory Chain Supercomplexes Using Blue Native Polyacrylamide Gel Electrophoresis (BN-PAGE). ACTA ACUST UNITED AC 2016; 6:1-14. [PMID: 26928661 DOI: 10.1002/9780470942390.mo150182] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Mitochondria are cellular organelles that harvest energy in the form of ATP through a process termed oxidative phosphorylation (OXPHOS), which occurs via the protein complexes of the electron transport chain (ETC). In recent years it has become unequivocally clear that mitochondrial complexes of the ETC are not static entities in the inner mitochondrial membrane. These complexes are dynamic and in mammals they aggregate in different stoichiometric combinations to form supercomplexes (SCs) or respirasomes. It has been proposed that the net respiration is more efficient via SCs than via isolated complexes. However, it still needs to be determined whether the activity of a particular SC is associated with a disease etiology. Here we describe a simplified method to visualize and assess in-gel activity of SCs and the individual complexes with good resolution using blue native polyacrylamide gel electrophoresis (BN-PAGE).
Collapse
Affiliation(s)
- Pooja Jha
- Laboratory for Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Xu Wang
- Laboratory for Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory for Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
43
|
Arbogast T, Ouagazzal AM, Chevalier C, Kopanitsa M, Afinowi N, Migliavacca E, Cowling BS, Birling MC, Champy MF, Reymond A, Herault Y. Reciprocal Effects on Neurocognitive and Metabolic Phenotypes in Mouse Models of 16p11.2 Deletion and Duplication Syndromes. PLoS Genet 2016; 12:e1005709. [PMID: 26872257 PMCID: PMC4752317 DOI: 10.1371/journal.pgen.1005709] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/06/2015] [Indexed: 11/18/2022] Open
Abstract
The 16p11.2 600 kb BP4-BP5 deletion and duplication syndromes have been associated with developmental delay; autism spectrum disorders; and reciprocal effects on the body mass index, head circumference and brain volumes. Here, we explored these relationships using novel engineered mouse models carrying a deletion (Del/+) or a duplication (Dup/+) of the Sult1a1-Spn region homologous to the human 16p11.2 BP4-BP5 locus. On a C57BL/6N inbred genetic background, Del/+ mice exhibited reduced weight and impaired adipogenesis, hyperactivity, repetitive behaviors, and recognition memory deficits. In contrast, Dup/+ mice showed largely opposite phenotypes. On a F1 C57BL/6N × C3B hybrid genetic background, we also observed alterations in social interaction in the Del/+ and the Dup/+ animals, with other robust phenotypes affecting recognition memory and weight. To explore the dosage effect of the 16p11.2 genes on metabolism, Del/+ and Dup/+ models were challenged with high fat and high sugar diet, which revealed opposite energy imbalance. Transcriptomic analysis revealed that the majority of the genes located in the Sult1a1-Spn region were sensitive to dosage with a major effect on several pathways associated with neurocognitive and metabolic phenotypes. Whereas the behavioral consequence of the 16p11 region genetic dosage was similar in mice and humans with activity and memory alterations, the metabolic defects were opposite: adult Del/+ mice are lean in comparison to the human obese phenotype and the Dup/+ mice are overweight in comparison to the human underweight phenotype. Together, these data indicate that the dosage imbalance at the 16p11.2 locus perturbs the expression of modifiers outside the CNV that can modulate the penetrance, expressivity and direction of effects in both humans and mice.
Collapse
Affiliation(s)
- Thomas Arbogast
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Abdel-Mouttalib Ouagazzal
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Claire Chevalier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Maksym Kopanitsa
- Synome Ltd, Moneta Building, Babraham Research Campus, Cambridge, United Kingdom
| | - Nurudeen Afinowi
- Synome Ltd, Moneta Building, Babraham Research Campus, Cambridge, United Kingdom
| | - Eugenia Migliavacca
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Belinda S. Cowling
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Marie-Christine Birling
- PHENOMIN, Institut Clinique de la Souris, ICS; CNRS, INSERM, University of Strasbourg, Illkirch-Graffenstaden, France
| | - Marie-France Champy
- PHENOMIN, Institut Clinique de la Souris, ICS; CNRS, INSERM, University of Strasbourg, Illkirch-Graffenstaden, France
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
- Université de Strasbourg, Illkirch, France
- PHENOMIN, Institut Clinique de la Souris, ICS; CNRS, INSERM, University of Strasbourg, Illkirch-Graffenstaden, France
- * E-mail:
| |
Collapse
|
44
|
Jo YS, Ryu D, Maida A, Wang X, Evans RM, Schoonjans K, Auwerx J. Phosphorylation of the nuclear receptor corepressor 1 by protein kinase B switches its corepressor targets in the liver in mice. Hepatology 2015; 62:1606-18. [PMID: 25998209 PMCID: PMC4618256 DOI: 10.1002/hep.27907] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 05/17/2015] [Indexed: 01/22/2023]
Abstract
UNLABELLED Nuclear receptor corepressor 1 (NCoR1) is a transcriptional coregulator that has wide-ranging effects on gene expression patterns. In the liver, NCoR1 represses lipid synthesis in the fasting state, whereas it inhibits activation of peroxisome proliferator-activated receptor alpha (PPARα) upon feeding, thereby blunting ketogenesis. Here, we show that insulin by activation of protein kinase B induces phosphorylation of NCoR1 on serine 1460, which selectively favors its interaction with PPARα and estrogen-related receptor alpha (ERRα) over liver X receptor alpha (LXRα). Phosphorylation of NCoR1 on S1460 selectively derepresses LXRα target genes, resulting in increased lipogenesis, whereas, at the same time, it inhibits PPARα and ERRα targets, thereby attenuating oxidative metabolism in the liver. Phosphorylation-gated differential recruitment of NCoR1 to different nuclear receptors explains the apparent paradox that liver-specific deletion of NCoR1 concurrently induces both lipogenesis and oxidative metabolism owing to a global derepression of LXRα, PPARα, and ERRα activity. CONCLUSION Phosphorylation-mediated recruitment switch of NCoR1 between nuclear receptor subsets provides a mechanism by which corepressors can selectively modulate liver energy metabolism during the fasting-feeding transition.
Collapse
Affiliation(s)
- Young Suk Jo
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
- Division of Endocrinology, Department of Internal Medicine, Yonsei University College of Medicine, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, South Korea
| | - Dongryeol Ryu
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Adriano Maida
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Xu Wang
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Ronald M. Evans
- Gene Expression Laboratory, Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Kristina Schoonjans
- Metabolic Signaling, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|
45
|
Cavey T, Ropert M, de Tayrac M, Bardou-Jacquet E, Island ML, Leroyer P, Bendavid C, Brissot P, Loréal O. Mouse genetic background impacts both on iron and non-iron metals parameters and on their relationships. Biometals 2015; 28:733-43. [PMID: 26041486 DOI: 10.1007/s10534-015-9862-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 05/20/2015] [Indexed: 12/17/2022]
Abstract
Iron is reported to interact with other metals. In addition, it has been shown that genetic background may impact iron metabolism. Our objective was to characterize, in mice of three genetic backgrounds, the links between iron and several non-iron metals. Thirty normal mice (C57BL/6, Balb/c and DBA/2; n = 10 for each group), fed with the same diet, were studied. Quantification of iron, zinc, cobalt, copper, manganese, magnesium and rubidium was performed by ICP/MS in plasma, erythrocytes, liver and spleen. Transferrin saturation was determined. Hepatic hepcidin1 mRNA level was evaluated by quantitative RT-PCR. As previously reported, iron parameters were modulated by genetic background with significantly higher values for plasma iron parameters and liver iron concentration in DBA/2 and Balb/c strains. Hepatic hepcidin1 mRNA level was lower in DBA/2 mice. No iron parameter was correlated with hepcidin1 mRNA levels. Principal component analysis of the data obtained for non-iron metals indicated that metals parameters stratified the mice according to their genetic background. Plasma and tissue metals parameters that are dependent or independent of genetic background were identified. Moreover, relationships were found between plasma and tissue content of iron and some other metals parameters. Our data: (i) confirms the impact of the genetic background on iron parameters, (ii) shows that genetic background may also play a role in the metabolism of non-iron metals, (iii) identifies links between iron and other metals parameters which may have implications in the understanding and, potentially, the modulation of iron metabolism.
Collapse
|
46
|
Rathkolb B, Klempt M, Sabrautzki S, Michel D, Klaften M, Laufs J, Sedlmeier R, Hans W, Fuchs H, Muckenthaler MU, Horsch M, Campagna DR, Fleming M, Hrabé de Angelis M, Wolf E, Aigner B. Screen for alterations of iron related parameters in N-ethyl-N-nitrosourea-treated mice identified mutant lines with increased plasma ferritin levels. Biometals 2015; 28:293-306. [DOI: 10.1007/s10534-015-9824-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 01/21/2015] [Indexed: 11/28/2022]
|
47
|
Boutant M, Joffraud M, Kulkarni SS, García-Casarrubios E, García-Roves PM, Ratajczak J, Fernández-Marcos PJ, Valverde AM, Serrano M, Cantó C. SIRT1 enhances glucose tolerance by potentiating brown adipose tissue function. Mol Metab 2014; 4:118-31. [PMID: 25685699 PMCID: PMC4314542 DOI: 10.1016/j.molmet.2014.12.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 12/11/2014] [Accepted: 12/13/2014] [Indexed: 10/24/2022] Open
Abstract
OBJECTIVE SIRT1 has been proposed to be a key signaling node linking changes in energy metabolism to transcriptional adaptations. Although SIRT1 overexpression is protective against diverse metabolic complications, especially in response to high-fat diets, studies aiming to understand the etiology of such benefits are scarce. Here, we aimed to identify the key tissues and mechanisms implicated in the beneficial effects of SIRT1 on glucose homeostasis. METHODS We have used a mouse model of moderate SIRT1 overexpression, under the control of its natural promoter, to evaluate glucose homeostasis and thoroughly characterize how different tissues could influence insulin sensitivity. RESULTS Mice with moderate overexpression of SIRT1 exhibit better glucose tolerance and insulin sensitivity even on a low fat diet. Euglycemic-hyperinsulinemic clamps and in-depth tissue analyses revealed that enhanced insulin sensitivity was achieved through a higher brown adipose tissue activity and was fully reversed by housing the mice at thermoneutrality. SIRT1 did not influence brown adipocyte differentiation, but dramatically enhanced the metabolic transcriptional responses to β3-adrenergic stimuli in differentiated adipocytes. CONCLUSIONS Our work demonstrates that SIRT1 improves glucose homeostasis by enhancing BAT function. This is not consequent to an alteration in the brown adipocyte differentiation process, but as a result of potentiating the response to β3-adrenergic stimuli.
Collapse
Affiliation(s)
- Marie Boutant
- Nestlé Institute of Health Sciences (NIHS) SA, EPFL Campus, Quartier de l'Innovation, Bâtiment G, Lausanne CH-1015, Switzerland
| | - Magali Joffraud
- Nestlé Institute of Health Sciences (NIHS) SA, EPFL Campus, Quartier de l'Innovation, Bâtiment G, Lausanne CH-1015, Switzerland
| | - Sameer S Kulkarni
- Nestlé Institute of Health Sciences (NIHS) SA, EPFL Campus, Quartier de l'Innovation, Bâtiment G, Lausanne CH-1015, Switzerland
| | - Ester García-Casarrubios
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain ; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
| | - Pablo M García-Roves
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain ; Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Joanna Ratajczak
- Nestlé Institute of Health Sciences (NIHS) SA, EPFL Campus, Quartier de l'Innovation, Bâtiment G, Lausanne CH-1015, Switzerland ; Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | | | - Angela M Valverde
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain ; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
| | - Manuel Serrano
- Spanish National Cancer Research Center (CNIO), Madrid E28029, Spain
| | - Carles Cantó
- Nestlé Institute of Health Sciences (NIHS) SA, EPFL Campus, Quartier de l'Innovation, Bâtiment G, Lausanne CH-1015, Switzerland
| |
Collapse
|
48
|
Zhou P, Hummel AD, Pywell CM, Dong XC, Duffield GE. High fat diet rescues disturbances to metabolic homeostasis and survival in the Id2 null mouse in a sex-specific manner. Biochem Biophys Res Commun 2014; 451:374-81. [PMID: 25108156 DOI: 10.1016/j.bbrc.2014.07.106] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 07/23/2014] [Indexed: 12/24/2022]
Abstract
Inhibitor of DNA binding 2 (ID2) is a helix-loop-helix transcriptional repressor rhythmically expressed in many adult tissues. Our previous studies have demonstrated that Id2 null mice have altered expression of circadian genes involved in lipid metabolism, altered circadian feeding behavior, and sex-specific enhancement of insulin sensitivity and elevated glucose uptake in skeletal muscle and brown adipose tissue. Here we further characterized the Id2-/- mouse metabolic phenotype in a sex-specific context and under low and high fat diets, and examined metabolic and endocrine parameters associated with lipid and glucose metabolism. Under the low-fat diet Id2-/- mice showed decreased weight gain, reduced gonadal fat mass, and a lower survival rate. Under the high-fat diet, body weight and gonadal fat gain of Id2-/- male mice was comparable to control mice and survival rate improved markedly. Furthermore, the high-fat diet treated Id2-/- male mice lost the enhanced glucose tolerance feature observed in the other Id2-/- groups, and there was a sex-specific difference in white adipose tissue storage of Id2-/- mice. Additionally, a distinct pattern of hepatic lipid accumulation was observed in Id2-/- males: low lipids on the low-fat diet and steatosis on the high-fat diet. In summary, these data provides valuable insights into the impact of Id2 deficiency on metabolic homeostasis of mice in a sex-specific manner.
Collapse
Affiliation(s)
- Peng Zhou
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Alyssa D Hummel
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Cameron M Pywell
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | - Giles E Duffield
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
49
|
Evaluation of growth patterns and body composition in C57Bl/6J mice using dual energy X-ray absorptiometry. BIOMED RESEARCH INTERNATIONAL 2014; 2014:253067. [PMID: 25110666 PMCID: PMC4119710 DOI: 10.1155/2014/253067] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 05/29/2014] [Accepted: 06/16/2014] [Indexed: 11/17/2022]
Abstract
The normal growth pattern of female C57BL/6J mice, from 5 to 30 weeks of age, has been investigated in a longitudinal study. Weight, body surface area (BS), and body mass index (BMI) were evaluated in forty mice. Lean mass and fat mass, bone mineral content (BMC), and bone mineral density (BMD) were monitored by dual energy X-ray absorptiometry (DEXA). Weight and BS increased linearly (16.15 ± 0.64-27.64 ± 1.42 g; 51.13 ± 0.74-79.57 ± 2.15 cm(2), P < 0.01), more markedly from 5 to 9 weeks of age (P < 0.001). BMD showed a peak at 17 weeks (0.0548 ± 0.0011 g/cm(2) ∗ m, P < 0.01). Lean mass showed an evident gain at 9 (15.8 ± 0.8 g, P < 0.001) and 25 weeks (20.5 ± 0.3 g, P < 0.01), like fat mass from 13 to 17 weeks (2.0 ± 0.4-3.6 ± 0.7 g, P < 0.01). BMI and lean mass index (LMI) reached the highest value at 21 weeks (3.57 ± 0.02-0.284 ± 0.010 g/cm(2), resp.), like fat mass index (FMI) at 17 weeks (0.057 ± 0.009 g/cm(2)) (P < 0.01). BMI, weight, and BS showed a moderate positive correlation (0.45-0.85) with lean mass from 5 to 21 weeks. Mixed linear models provided a good prediction for lean mass, fat mass, and BMD. This study may represent a baseline reference for a future comparison of wild-type C57BL/6J mice with models of altered growth.
Collapse
|
50
|
Lim MA, Bence KK, Sandesara I, Andreux P, Auwerx J, Ishibashi J, Seale P, Kalb RG. Genetically altering organismal metabolism by leptin-deficiency benefits a mouse model of amyotrophic lateral sclerosis. Hum Mol Genet 2014; 23:4995-5008. [PMID: 24833719 DOI: 10.1093/hmg/ddu214] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal, neurodegenerative disease that causes death of motor neurons. ALS patients and mouse models of familial ALS display organismal level metabolic dysfunction, which includes increased energy expenditure despite decreased lean mass. The pathophysiological relevance of abnormal energy homeostasis to motor neuron disease remains unclear. Leptin is an adipocyte-derived hormone that regulates whole-animal energy expenditure. Here, we report that placing mutant superoxide dismutase 1 (SOD1) mice in a leptin-deficient background improves energy homeostasis and slows disease progression. Leptin-deficient mutant SOD1 mice possess increased bodyweight and fat mass, as well as decreased energy expenditure. These observations coincide with enhanced survival, improved strength and decreased motor neuron loss. These results suggest that altering whole-body energy metabolism in mutant SOD1 mice can mitigate disease progression. We propose that manipulations that increase fat mass and reduce energy expenditure will be beneficial in the setting of motor neuron disease.
Collapse
Affiliation(s)
- Maria A Lim
- Division of Neurology, Department of Pediatrics, Research Institute, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA, Neuroscience Graduate Group
| | - Kendra K Bence
- Neuroscience Graduate Group, Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ishani Sandesara
- Division of Neurology, Department of Pediatrics, Research Institute, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Pénélope Andreux
- Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Jeff Ishibashi
- Institute for Diabetes, Obesity and Metabolism, Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity and Metabolism, Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert G Kalb
- Division of Neurology, Department of Pediatrics, Research Institute, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA, Neuroscience Graduate Group, Department of Neurology and
| |
Collapse
|