1
|
Shimizu K, Yoshida Y, Iwasa K, Fujii Y, Jacob UM, Fritsch T, Abdelhameed AS, Calabrese V, Osakabe N. A semi-automated observation approach to quantify mouse skeletal muscle differentiation using immunohistochemistry. Physiol Rep 2025; 13:e70330. [PMID: 40223406 PMCID: PMC11994891 DOI: 10.14814/phy2.70330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 04/15/2025] Open
Abstract
Histological analysis is vital for understanding skeletal muscle diseases. However, quantifying data requires much effort, so automation is expected to reduce workload. The present study proposes a semi-automated method to quantify expressed paired box protein (Pax-7) /bromodeoxyuridine (BrdU)-positive cells. Soleus muscle was harvested from mice 2 weeks after oral administration of the epicatechin tetramer cinnamantanin A2 (A2), known to induce skeletal muscle hypertrophy. Before the necropsy, mice were treated with BrdU to facilitate cell tracking. For histological examination, frozen sections were stained with hematoxylin and eosin (HE) to measure cell size by cross-sectional area (CSA) and were immunostained with anti-BrdU and anti-Pax-7 antibodies. Treatment with A2 caused a shift in the CSA distribution curve towards larger values, thus revealing an increase in muscle size. The analysis of BrdU/Pax-7 positive cells, performed both manually and semi-automatically, revealed a slight increase with A2 treatment, while Pax-7 positive cells remained unchanged. Correlation between manual and semi-automated analysis showed a coefficient of determination of 0.7132, indicating a significant reduction in analysis time by approximately 20 times. This study highlights the effectiveness of semi-automated histological analysis in skeletal muscle research and provides a practical solution to increase the efficiency of muscle regeneration evaluation.
Collapse
Affiliation(s)
- Kenta Shimizu
- Systems Engineering and Science, Graduate School of Engineering and ScienceShibaura Institute of TechnologySaitamaJapan
| | - Yamato Yoshida
- Systems Engineering and Science, Graduate School of Engineering and ScienceShibaura Institute of TechnologySaitamaJapan
| | - Kenshin Iwasa
- Department of Bioscience and Engineering, Faculty of System Science and EngineeringShibaura Institute of TechnologySaitamaJapan
| | - Yasuyuki Fujii
- Systems Engineering and Science, Graduate School of Engineering and ScienceShibaura Institute of TechnologySaitamaJapan
| | | | | | - Ali S. Abdelhameed
- Department of Pharmaceutical ChemistryCollege of Pharmacy, King Saud UniversityRiyadhKingdom of Saudi Arabia
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCataniaItaly
| | - Naomi Osakabe
- Systems Engineering and Science, Graduate School of Engineering and ScienceShibaura Institute of TechnologySaitamaJapan
- Department of Bioscience and Engineering, Faculty of System Science and EngineeringShibaura Institute of TechnologySaitamaJapan
| |
Collapse
|
2
|
Ono D, Kawai H, Kuwahara H, Yokota T. Refining Muscle Morphometry Through Machine Learning and Spatial Analysis. Neuropathol Appl Neurobiol 2025; 51:e70012. [PMID: 40125691 DOI: 10.1111/nan.70012] [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: 08/17/2024] [Revised: 02/18/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
AIMS Muscle morphology provides important information in differentiating disease aetiology, but its measurement remains challenging because of the lack of an efficient and objective method. This study aimed to quantitatively refine the morphological features of muscle fibres in neuromuscular diseases using machine learning. METHODS In this retrospective study, we analysed muscle biopsy specimens on haematoxylin and eosin-staining. Machine learning-based software was developed to segment muscle fibre contours and perform automated muscle morphometry and subsequent graph theory-based spatial analysis of atrophied fibre grouping. A decision tree-based framework, LightGBM, was trained to predict underlying aetiologies based on morphometric and spatial variables. RESULTS The study included 100 muscle samples, including 20 normal muscles, 49 myopathies and 19 neuropathies. The fine-tuned segmentation model, YOLOv8, achieved a mask average precision of 0.819. The muscle morphometry revealed the significance of fibre circularity. The mean circularity was higher in the myopathy group, and the SD of circularity was elevated in the neuropathy group. Although most cases were consistent with textbook findings, atypical presentations, such as dermatomyositis with angular atrophy and amyotrophic lateral sclerosis with round atrophy, were objectively documented. Spatial analysis quantified grouped atrophy, showing the potential to feature specific atrophy patterns. The LightGBM model successfully predicted the final clinical diagnosis of the myopathies and neuropathies with an accuracy of 0.852, which exceeded that of 0.808 by human annotation. CONCLUSION Automated muscle morphometry and spatial analysis provide quantification of muscle morphology and patterns of atrophy, which will facilitate objective and efficient investigation of neuromuscular diseases.
Collapse
Affiliation(s)
- Daisuke Ono
- Department of Neurology and Neurological Science, Institute of Science Tokyo, Bunkyo-Ku, Tokyo, Japan
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Honami Kawai
- Department of Neurology and Neurological Science, Institute of Science Tokyo, Bunkyo-Ku, Tokyo, Japan
| | - Hiroya Kuwahara
- Department of Neurology and Neurological Science, Institute of Science Tokyo, Bunkyo-Ku, Tokyo, Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Institute of Science Tokyo, Bunkyo-Ku, Tokyo, Japan
| |
Collapse
|
3
|
Mill L, Aust O, Ackermann JA, Burger P, Pascual M, Palumbo-Zerr K, Krönke G, Uderhardt S, Schett G, Clemen CS, Holtzhausen C, Jabari S, Schröder R, Maier A, Grüneboom A. Deep learning-based image analysis in muscle histopathology using photo-realistic synthetic data. COMMUNICATIONS MEDICINE 2025; 5:64. [PMID: 40050400 PMCID: PMC11885816 DOI: 10.1038/s43856-025-00777-y] [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: 01/30/2024] [Accepted: 02/20/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Artificial intelligence (AI), specifically Deep learning (DL), has revolutionized biomedical image analysis, but its efficacy is limited by the need for representative, high-quality large datasets with manual annotations. While latest research on synthetic data using AI-based generative models has shown promising results to tackle this problem, several challenges such as lack of interpretability and need for vast amounts of real data remain. This study aims to introduce a new approach-SYNTA-for the generation of photo-realistic synthetic biomedical image data to address the challenges associated with state-of-the art generative models and DL-based image analysis. METHODS The SYNTA method employs a fully parametric approach to create photo-realistic synthetic training datasets tailored to specific biomedical tasks. Its applicability is tested in the context of muscle histopathology and skeletal muscle analysis. This new approach is evaluated for two real-world datasets to validate its applicability to solve complex image analysis tasks on real data. RESULTS Here we show that SYNTA enables expert-level segmentation of unseen real-world biomedical data using only synthetic training data. By addressing the lack of representative and high-quality real-world training data, SYNTA achieves robust performance in muscle histopathology image analysis, offering a scalable, controllable and interpretable alternative to generative models such as Generative Adversarial Networks (GANs) or Diffusion Models. CONCLUSIONS SYNTA demonstrates great potential to accelerate and improve biomedical image analysis. Its ability to generate high-quality photo-realistic synthetic data reduces reliance on extensive collection of data and manual annotations, paving the way for advancements in histopathology and medical research.
Collapse
Affiliation(s)
- Leonid Mill
- MIRA Vision Microscopy GmbH, 73037, Göppingen, Germany.
- Pattern Recognition Lab, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91058, Erlangen, Germany.
| | - Oliver Aust
- Department of Medicine 3 - Rheumatology and Immunology & Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Jochen A Ackermann
- Department of Medicine 3 - Rheumatology and Immunology & Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Philipp Burger
- Department of Medicine 3 - Rheumatology and Immunology & Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Monica Pascual
- Department of Medicine 3 - Rheumatology and Immunology & Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Katrin Palumbo-Zerr
- Department of Medicine 3 - Rheumatology and Immunology & Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Gerhard Krönke
- Department of Medicine 3 - Rheumatology and Immunology & Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Stefan Uderhardt
- Department of Medicine 3 - Rheumatology and Immunology & Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Georg Schett
- Department of Medicine 3 - Rheumatology and Immunology & Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Christoph S Clemen
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Institute of Vegetative Physiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Christian Holtzhausen
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Samir Jabari
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Klinikum Nuremberg, Institute of Pathology, Paracelsus Medical University, 90419, Nuremberg, Germany
| | - Rolf Schröder
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Andreas Maier
- Pattern Recognition Lab, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91058, Erlangen, Germany
| | - Anika Grüneboom
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V, 44139, Dortmund, Germany.
| |
Collapse
|
4
|
Sancerni T, Montel V, Dereumetz J, Cochon L, Coq JO, Bastide B, Canu MH. Enduring effects of acute prenatal ischemia in rat soleus muscle, and protective role of erythropoietin. J Muscle Res Cell Motil 2025; 46:23-34. [PMID: 39549147 DOI: 10.1007/s10974-024-09684-6] [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: 09/30/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024]
Abstract
Motor disorders are considered to originate mainly from brain lesions. Placental dysfunction or maternal exposure to a persistently hypoxic environment is a major cause of further motor disorders such as cerebral palsy. Our main goal was to determine the long-term effects of mild intrauterine acute ischemic stress on rat soleus myofibres and whether erythropoietin treatment could prevent these changes. Rat embryos were subjected to ischemic stress at embryonic day E17. They then received an intraperitoneal erythropoietin injection at postnatal days 1-5. Soleus muscles were collected at postnatal day 28. Prenatal ischemic stress durably affected muscle structure, as indicated by the greater fiber cross-sectional area (+ 18%) and the greater number of mature vessels (i.e. vessels with mature endothelial cells) per myofibres (+ 43%), and muscle biochemistry, as shown by changes in signaling pathways involved in protein synthesis/degradation balance (-81% for 4EBP1; -58% for AKT) and Hif1α expression levels (+ 95%). Erythropoietin injection in ischemic pups had a weak protective effect: it increased muscle mass (+ 25% with respect to ischemic pups) and partially prevented the increase in muscle degradation pathways and mature vascularization, whereas it exacerbated the decrease in synthesis pathways. Hence, erythropoietin treatment after acute ischemic stress contributes to muscle adaptation to ischemic conditions.
Collapse
Affiliation(s)
- Tiphaine Sancerni
- Univ. Lille, Univ Artois, Univ Littoral Côte d'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, ULR7369, Lille, F-59000, France
| | - Valérie Montel
- Univ. Lille, Univ Artois, Univ Littoral Côte d'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, ULR7369, Lille, F-59000, France
| | - Julie Dereumetz
- Univ. Lille, Univ Artois, Univ Littoral Côte d'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, ULR7369, Lille, F-59000, France
| | - Laetitia Cochon
- Univ. Lille, Univ Artois, Univ Littoral Côte d'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, ULR7369, Lille, F-59000, France
| | - Jacques-Olivier Coq
- Institut des Sciences du Mouvement (ISM), Team 'Plasticité des Systèmes Nerveux et Musculaires', UMR 7287 CNRS, Aix-Marseille Université Faculté des Sports, Marseille Cedex 09, F-13288, France
| | - Bruno Bastide
- Univ. Lille, Univ Artois, Univ Littoral Côte d'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, ULR7369, Lille, F-59000, France
| | - Marie-Hélène Canu
- Univ. Lille, Univ Artois, Univ Littoral Côte d'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, ULR7369, Lille, F-59000, France.
| |
Collapse
|
5
|
Kahn RE, Zhu P, Roy I, Peek C, Hawley JA, Dayanidhi S. Ablation of satellite cell-specific clock gene, Bmal1, alters force production, muscle damage, and repair following contractile-induced injury. FASEB J 2025; 39:e70325. [PMID: 39812604 PMCID: PMC11734708 DOI: 10.1096/fj.202402145rr] [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: 09/09/2024] [Revised: 12/24/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
Following injury, skeletal muscle undergoes repair via satellite cell (SC)-mediated myogenic progression. In SCs, the circadian molecular clock gene, Bmal1, is necessary for appropriate myogenic progression and repair with evidence that muscle molecular clocks can also affect force production. Utilizing a mouse model allowing for inducible depletion of Bmal1 within SCs, we determined contractile function, SC myogenic progression and muscle damage and repair following eccentric contractile-induced injury. At baseline, SC-Bmal1iKO animals exhibited a ~20-25% reduction in normalized force production (ex vivo and in vivo) versus control SC-Bmal1Cntrl and SC-Bmal1iKO untreated littermates (p < .05). Following contractile injury, SC-Bmal1iKO animals displayed reduced muscle damage and subsequent repair post-injury (Dystrophinnegative fibers 24 h: SC-Bmal1Cntrl 199 ± 41; SC-Bmal1iKO 36 ± 13, p < .05) (eMHC+ fibers 7 day: SC-Bmal1Cntrl 217.8 ± 115.5; SC-Bmal1iKO 27.8 ± 17.3; Centralized nuclei 7 day: SC-Bmal1Cntrl 160.7 ± 70.5; SC-Bmal1iKO 46.2 ± 15.7). SC-Bmal1iKO animals also showed reduced neutrophil infiltration, consistent with less injury (Neutrophil content 24 h: SC-Bmal1Cntrl 2.4 ± 0.4; SC-Bmal1iKO 0.4 ± 0.2, % area fraction, p < .05). SC-Bmal1iKO animals had greater SC activation/proliferation at an earlier timepoint (p < .05) and an unexplained increase in activation 7 days post injury. Collectively, these data suggest SC-Bmal1 plays a regulatory role in force production, influencing the magnitude of muscle damage/repair, with an altered SC myogenic progression following contractile-induced muscle injury.
Collapse
Affiliation(s)
- Ryan E. Kahn
- Exercise and Nutrition Research Program, The Mary MacKillop Institute for Health ResearchAustralian Catholic UniversityMelbourneAustralia
- Shirley Ryan AbilityLabChicagoIllinoisUSA
| | - Pei Zhu
- Department of Biochemistry and Molecular GeneticsNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Ishan Roy
- Shirley Ryan AbilityLabChicagoIllinoisUSA
- Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Clara Peek
- Department of Biochemistry and Molecular GeneticsNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - John A. Hawley
- Exercise and Nutrition Research Program, The Mary MacKillop Institute for Health ResearchAustralian Catholic UniversityMelbourneAustralia
- Department of Sport and Exercise SciencesManchester Metropolitan University Institute of SportManchester
| | - Sudarshan Dayanidhi
- Shirley Ryan AbilityLabChicagoIllinoisUSA
- Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| |
Collapse
|
6
|
Munoz AZ, Soni K, Li A, Lakkundi V, Iyer A, Adler A, Kirkendall K, Petrigliano F, Benayoun BA, Lozito TP, Almada AE. Ilastik: a machine learning image analysis platform to interrogate stem cell fate decisions across multiple vertebrate species. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.21.629913. [PMID: 40236229 PMCID: PMC11996584 DOI: 10.1101/2024.12.21.629913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Stem cells are the key cellular source for regenerating tissues and organs in vertebrate species. Historically, the investigation of stem cell fate decisions in vivo has been assessed in tissue sections using immunohistochemistry (IHC), where a trained user quantifies fluorescent signal in multiple randomly selected images using manual counting-which is prone to inaccuracies, bias, and is very labor intensive. Here, we highlight the performance of a recently developed machine-learning (ML)-based image analysis program called Ilastik using skeletal muscle as a model system. Interestingly, we demonstrate that Ilastik accurately quantifies Paired Box Protein 7 (PAX7)-positive muscle stem cells (MuSCs) before and during the regenerative process in whole muscle sections from mice, humans, axolotl salamanders, and short-lived African turquoise killifish, to a precision that exceeds human capabilities and in a fraction of the time. Overall, Ilastik is a free user-friendly ML-based program that will expedite the analysis of stained tissue sections in vertebrate animals.
Collapse
|
7
|
Cho S, Servián-Morilla E, Garrido VN, Rodriguez-Gonzalez B, Yuan Y, Cano R, Rambhiya AA, Darabi R, Haltiwanger RS, Paradas C, Jafar-Nejad H. The glycosyltransferase POGLUT1 regulates muscle stem cell development and maintenance in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625261. [PMID: 39651163 PMCID: PMC11623641 DOI: 10.1101/2024.11.25.625261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Mutations in protein O -glucosyltransferase 1 ( POGLUT1 ) cause a recessive form of limb-girdle muscular dystrophy (LGMD-R21) associated with reduced satellite cell number and NOTCH1 signaling in adult patient muscles and impaired myogenic capacity of patient-derived muscle progenitors. However, the in vivo roles of POGLUT1 in the development, function, and maintenance of satellite cells are not well understood. Here, we show that conditional deletion of mouse Poglut1 in myogenic progenitors leads to early lethality, postnatal muscle growth defects, reduced Pax7 expression, abnormality in muscle extracellular matrix, and impaired muscle repair. Poglut1 -deficient muscle progenitors exhibit reduced proliferation, enhanced differentiation, and accelerated fusion into myofibers. Inducible loss of Poglut1 in adult satellite cells leads to their precocious differentiation and impairs muscle repair upon serial injury. Cell-based signaling assays and mass spectrometric analysis indicate that POGLUT1 is required for the activation of NOTCH1, NOTCH2, and NOTCH3 in myoblasts and that NOTCH3 is a target of POGLUT1 like NOTCH1 and NOTCH2. These observations provide insight into the roles of POGLUT1 in muscle development and repair and the pathophysiology of LGMD-R21.
Collapse
|
8
|
Yamaoka Y, Chan WI, Seno S, Iwamori K, Fukada SI, Matsuda H. Quantifying the recovery process of skeletal muscle on hematoxylin and eosin stained images via learning from label proportion. Sci Rep 2024; 14:27044. [PMID: 39511433 PMCID: PMC11544229 DOI: 10.1038/s41598-024-78433-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
Visual observing muscle tissue regeneration is used to measure experimental effect size in biological research to discover the mechanism of muscle strength decline due to illness or aging. Quantitative computer imaging analysis for support evaluating the recovery phase has not been established because of the localized nature of recovery and the difficulty in selecting image features for cells in regeneration. We constructed MyoRegenTrack for segmenting cells and classifying their regeneration phase in hematoxylin-eosin (HE) stained images. A straightforward approach to classification is supervised learning. However, obtaining detailed annotations for each fiber in a whole slide image is impractical in terms of cost and accuracy. Thus, we propose to learn individual recovery phase classification utilizing the proportions of cell class depending on the days after muscle injection to induce regeneration. We extract implicit multidimensional features from the HE-stained tissue images and train a classifier using weakly supervised learning, guided by their class proportion for elapsed time on recovery. We confirmed the effectiveness of MyoRegenTrack by comparing its results with expert annotations. A comparative study of the recovery relation between two different muscle injections shows that the analysis result using MyoRegenTrack is consistent with findings from previous studies.
Collapse
Affiliation(s)
- Yu Yamaoka
- Graduate School of Information Science and Technology, Osaka University, Osaka, 565-0871, Japan
| | - Weng Ian Chan
- Graduate School of Information Science and Technology, Osaka University, Osaka, 565-0871, Japan
| | - Shigeto Seno
- Graduate School of Information Science and Technology, Osaka University, Osaka, 565-0871, Japan.
| | - Kanako Iwamori
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - So-Ichiro Fukada
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Hideo Matsuda
- Graduate School of Information Science and Technology, Osaka University, Osaka, 565-0871, Japan
| |
Collapse
|
9
|
Van de Casteele F, Van Thienen R, Horwath O, Apró W, Van der Stede T, Moberg M, Lievens E, Derave W. Does one biopsy cut it? Revisiting human muscle fiber type composition variability using repeated biopsies in the vastus lateralis and gastrocnemius medialis. J Appl Physiol (1985) 2024; 137:1341-1353. [PMID: 39359186 DOI: 10.1152/japplphysiol.00394.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/06/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024] Open
Abstract
Human skeletal muscle fiber type composition varies greatly along the muscle, so one biopsy may not accurately represent the whole muscle. Recommendations on the number of biopsies and fiber counts using immunohistochemistry and whether these findings can be extrapolated to other muscles are lacking. We assessed fiber type composition in the vastus lateralis and gastrocnemius medialis muscles of 40 individuals. Per muscle, we took four biopsy samples from one incision, collecting two samples each from a proximally and distally directed needle. Based on another dataset involving 10 vastus lateralis biopsies per participant (n = 7), we calculated 95% limits of agreement for subsets of biopsies and fiber counts compared with the 10-biopsy average. Average absolute differences in type I fiber proportions between proximal and distal, and between within-needle samples were 6.9 and 4.5 percentage points in the vastus lateralis, and 5.5 and 4.4 percentage points in the gastrocnemius medialis, respectively. The 95% limits of agreement narrowed to ±10 percentage points when 200 fibers from at least three biopsies were analyzed, with minimal improvements with greater fiber counts. Type I fiber proportions in the vastus lateralis and gastrocnemius medialis showed a moderate positive association (r2 = 0.22; P = 0.006; at least 200 fibers in each of three to four samples per muscle). In conclusion, three biopsies with a minimum of 200 counted fibers are required to estimate the vastus lateralis fiber type composition within ±10 percentage points. Even when using these standards, researchers should be cautious when extrapolating muscle fiber type proportions from one muscle to another.NEW & NOTEWORTHY Fiber type composition is equally variable in muscle biopsy samples taken from one incision as from multiple incisions. Hence, we propose two biopsies from a single incision-needles directed proximally and distally, and each rotated 180° for cutting a second sample-as a more feasible, less invasive alternative to three biopsies from as many incisions. In addition, we identified the gastrocnemius medialis as a slow-twitch muscle with an average of 64.7% slow fibers.
Collapse
Affiliation(s)
| | - Ruud Van Thienen
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
| | - Oscar Horwath
- Department of Physiology, Nutrition and Biomechanics, Åstrand Laboratory, Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - William Apró
- Department of Physiology, Nutrition and Biomechanics, Åstrand Laboratory, Swedish School of Sport and Health Sciences, Stockholm, Sweden
- Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| | - Thibaux Van der Stede
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Marcus Moberg
- Department of Physiology, Nutrition and Biomechanics, Åstrand Laboratory, Swedish School of Sport and Health Sciences, Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Eline Lievens
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
| | - Wim Derave
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
10
|
Fefeu M, Blatzer M, Kneppers A, Briand D, Rocheteau P, Haroche A, Hardy D, Juchet-Martin M, Danckaert A, Coudoré F, Tutakhail A, Huchet C, Lafoux A, Mounier R, Mir O, Gaillard R, Chrétien F. Serotonin reuptake inhibitors improve muscle stem cell function and muscle regeneration in male mice. Nat Commun 2024; 15:6457. [PMID: 39085209 PMCID: PMC11291725 DOI: 10.1038/s41467-024-50220-4] [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: 06/16/2021] [Accepted: 07/03/2024] [Indexed: 08/02/2024] Open
Abstract
Serotonin reuptake inhibitor antidepressants such as fluoxetine are widely used to treat mood disorders. The mechanisms of action include an increase in extracellular level of serotonin, neurogenesis, and growth of vessels in the brain. We investigated whether fluoxetine could have broader peripheral regenerative properties. Following prolonged administration of fluoxetine in male mice, we showed that fluoxetine increases the number of muscle stem cells and muscle angiogenesis, associated with positive changes in skeletal muscle function. Fluoxetine also improved skeletal muscle regeneration after single and multiples injuries with an increased muscle stem cells pool and vessel density associated with reduced fibrotic lesions and inflammation. Mice devoid of peripheral serotonin treated with fluoxetine did not exhibit beneficial effects during muscle regeneration. Specifically, pharmacological, and genetic inactivation of the 5-HT1B subtype serotonin receptor also abolished the enhanced regenerative process induced by fluoxetine. We highlight here a regenerative property of serotonin on skeletal muscle.
Collapse
Affiliation(s)
- Mylène Fefeu
- GHU Paris Psychiatrie & Neurosciences, site Sainte Anne, Service Hospitalo-Universitaire de psychiatrie, Paris, France
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
- Université de Paris Cité, Paris, France
| | - Michael Blatzer
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
| | - Anita Kneppers
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - David Briand
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
| | - Pierre Rocheteau
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
| | - Alexandre Haroche
- GHU Paris Psychiatrie & Neurosciences, site Sainte Anne, Service Hospitalo-Universitaire de psychiatrie, Paris, France
| | - David Hardy
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
| | - Mélanie Juchet-Martin
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
| | | | - François Coudoré
- CESP, MOODS Team, Inserm, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France
| | - Abdulkarim Tutakhail
- CESP, MOODS Team, Inserm, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France
| | - Corinne Huchet
- TaRGeT, INSERM UMR 1089, Nantes Université, CHU Nantes, Nantes, France
| | - Aude Lafoux
- Therassay Platform, Capacités, Université de Nantes, IRS 2 Nantes Biotech, Nantes, France
| | - Rémi Mounier
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Olivier Mir
- Sarcoma Group, Gustave Roussy, Villejuif, France
| | - Raphaël Gaillard
- GHU Paris Psychiatrie & Neurosciences, site Sainte Anne, Service Hospitalo-Universitaire de psychiatrie, Paris, France.
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France.
- Université de Paris Cité, Paris, France.
| | - Fabrice Chrétien
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France.
- Université de Paris Cité, Paris, France.
- GHU Paris Psychiatrie & Neurosciences, site Sainte Anne, Service Hospitalo-Universitaire de neuropathologie, Paris, France.
| |
Collapse
|
11
|
Lu-Nguyen N, Snowden S, Popplewell L, Malerba A. Systemic Pharmacotherapeutic Treatment of the ACTA1-MCM/FLExDUX4 Preclinical Mouse Model of FSHD. Int J Mol Sci 2024; 25:6994. [PMID: 39000102 PMCID: PMC11241187 DOI: 10.3390/ijms25136994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/18/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Aberrant expression of the double homeobox 4 (DUX4) gene in skeletal muscle predominantly drives the pathogenesis of facioscapulohumeral muscular dystrophy (FSHD). We recently demonstrated that berberine, an herbal extract known for its ability to stabilize guanine-quadruplex structures, effectively downregulates DUX4 expression in FSHD patient-derived myoblasts and in mice overexpressing exogenous DUX4 after viral vector-based treatment. Here, we sought to confirm berberine's inhibitory efficacy on DUX4 in the widely used FSHD-like transgenic mouse model, ACTA1-MCM/FLExDUX4, where DUX4 is induced at pathogenic levels using tamoxifen. Animals repeatedly treated with berberine via intraperitoneal injections for 4 weeks exhibited significant reductions in both mRNA and protein levels of DUX4, and in mRNA expression of murine DUX4-related genes. This inhibition translated into improved forelimb muscle strength and positive alterations in important FSHD-relevant cellular pathways, although its impact on muscle mass and histopathology was less pronounced. Collectively, our data confirm the efficacy of berberine in downregulating DUX4 expression in the most relevant FSHD mouse model. However, further optimization of dosing regimens and new studies to enhance the bioavailability of berberine in skeletal muscle are warranted to fully leverage its therapeutic potential for FSHD treatment.
Collapse
Affiliation(s)
- Ngoc Lu-Nguyen
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK; (N.L.-N.); (S.S.)
| | - Stuart Snowden
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK; (N.L.-N.); (S.S.)
| | - Linda Popplewell
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK; (N.L.-N.); (S.S.)
- National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
| | - Alberto Malerba
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK; (N.L.-N.); (S.S.)
| |
Collapse
|
12
|
Longo L, Bartikoski BJ, de Souza VEG, Salvati F, Uribe‐Cruz C, Lenz G, Xavier RM, Álvares‐da‐Silva MR, Filippi‐Chiela EC. Muscle fibre morphometric analysis (MusMA) correlates with muscle function and cardiovascular risk prognosis. Int J Exp Pathol 2024; 105:100-113. [PMID: 38722178 PMCID: PMC11129960 DOI: 10.1111/iep.12504] [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: 11/20/2023] [Revised: 03/14/2024] [Accepted: 03/22/2024] [Indexed: 05/29/2024] Open
Abstract
Morphometry of striated muscle fibres is critical for monitoring muscle health and function. Here, we evaluated functional parameters of skeletal and cardiac striated muscle in two experimental models using the Morphometric Analysis of Muscle Fibre tool (MusMA). The collagen-induced arthritis model was used to evaluate the function of skeletal striated muscle and the non-alcoholic fatty liver disease model was used for cardiac striated muscle analysis. After euthanasia, we used haeamatoxylin and eosin stained sections of skeletal and cardiac muscle to perform muscle fibre segmentation and morphometric analysis. Morphometric analysis classified muscle fibres into six subpopulations: normal, regular hypertrophic, irregular hypertrophic, irregular, irregular atrophic and regular atrophic. The percentage of atrophic fibres was associated with lower walking speed (p = 0.009) and lower body weight (p = 0.026), respectively. Fibres categorized as normal were associated with maximum grip strength (p < 0.001) and higher march speed (p < 0.001). In the evaluation of cardiac striated muscle fibres, the percentage of normal cardiomyocytes negatively correlated with cardiovascular risk markers such as the presence of abdominal adipose tissue (p = .003), miR-33a expression (p = .001) and the expression of miR-126 (p = .042) Furthermore, the percentage of atrophic cardiomyocytes correlated significantly with the Castelli risk index II (p = .014). MusMA is a simple and objective tool that allows the screening of striated muscle fibre morphometry, which can complement the diagnosis of muscle diseases while providing functional and prognostic information in basic and clinical research.
Collapse
Affiliation(s)
- Larisse Longo
- Graduate Program in Gastroenterology and HepatologyUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Experimental Hepatology and Gastroenterology Laboratory, Center for Experimental ResearchHospital de Clínicas de Porto AlegrePorto AlegreBrazil
| | - Bárbara Jonson Bartikoski
- Autoimmune Diseases Laboratory, Rheumatology ServiceHospital de Clínicas de Porto AlegrePorto AlegreBrazil
| | - Valessa Emanoele Gabriel de Souza
- Experimental Hepatology and Gastroenterology Laboratory, Center for Experimental ResearchHospital de Clínicas de Porto AlegrePorto AlegreBrazil
| | - Fernando Salvati
- Experimental Hepatology and Gastroenterology Laboratory, Center for Experimental ResearchHospital de Clínicas de Porto AlegrePorto AlegreBrazil
| | - Carolina Uribe‐Cruz
- Graduate Program in Gastroenterology and HepatologyUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Experimental Hepatology and Gastroenterology Laboratory, Center for Experimental ResearchHospital de Clínicas de Porto AlegrePorto AlegreBrazil
- Universidad Católica de las MisionesPosadasArgentina
| | - Guido Lenz
- Department of Biophysics and Biotechnology CenterUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Ricardo Machado Xavier
- Graduate Program in Gastroenterology and HepatologyUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Graduate Program in Medical SciencesUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Mário Reis Álvares‐da‐Silva
- Graduate Program in Gastroenterology and HepatologyUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Experimental Hepatology and Gastroenterology Laboratory, Center for Experimental ResearchHospital de Clínicas de Porto AlegrePorto AlegreBrazil
- Division of GastroenterologyHospital de Clínicas de Porto AlegrePorto AlegreBrazil
| | - Eduardo Cremonese Filippi‐Chiela
- Graduate Program in Gastroenterology and HepatologyUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Department of Morphological SciencesUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Experimental Research ServiceHospital de Clínicas de Porto AlegrePorto AlegreBrazil
| |
Collapse
|
13
|
Oliver T, Nguyen NY, Tully CB, McCormack NM, Sun CM, Fiorillo AA, Heier CR. The glucocorticoid receptor acts locally to protect dystrophic muscle and heart during disease. Dis Model Mech 2024; 17:dmm050397. [PMID: 38770680 PMCID: PMC11139035 DOI: 10.1242/dmm.050397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 03/28/2024] [Indexed: 05/22/2024] Open
Abstract
Absence of dystrophin results in muscular weakness, chronic inflammation and cardiomyopathy in Duchenne muscular dystrophy (DMD). Pharmacological corticosteroids are the DMD standard of care; however, they have harsh side effects and unclear molecular benefits. It is uncertain whether signaling by physiological corticosteroids and their receptors plays a modifying role in the natural etiology of DMD. Here, we knocked out the glucocorticoid receptor (GR, encoded by Nr3c1) specifically in myofibers and cardiomyocytes within wild-type and mdx52 mice to dissect its role in muscular dystrophy. Double-knockout mice showed significantly worse phenotypes than mdx52 littermate controls in measures of grip strength, hang time, inflammatory pathology and gene expression. In the heart, GR deletion acted additively with dystrophin loss to exacerbate cardiomyopathy, resulting in enlarged hearts, pathological gene expression and systolic dysfunction, consistent with imbalanced mineralocorticoid signaling. The results show that physiological GR functions provide a protective role during muscular dystrophy, directly contrasting its degenerative role in other disease states. These data provide new insights into corticosteroids in disease pathophysiology and establish a new model to investigate cell-autonomous roles of nuclear receptors and mechanisms of pharmacological corticosteroids.
Collapse
MESH Headings
- Animals
- Mice
- Cardiomyopathies/pathology
- Cardiomyopathies/metabolism
- Dystrophin/metabolism
- Dystrophin/genetics
- Dystrophin/deficiency
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Mice, Knockout
- Muscle, Skeletal/pathology
- Muscle, Skeletal/metabolism
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Animal/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/metabolism
- Myocardium/pathology
- Myocardium/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/drug effects
- Phenotype
- Receptors, Glucocorticoid/metabolism
Collapse
Affiliation(s)
- Trinitee Oliver
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
- Department of Biology, Howard University, Washington, DC 20059, USA
- Graduate School of Biomedical Sciences, Cedars-Sinai Medical Center, West Hollywood, CA 90048, USA
| | - Nhu Y. Nguyen
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
| | - Christopher B. Tully
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
| | - Nikki M. McCormack
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
| | - Christina M. Sun
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
| | - Alyson A. Fiorillo
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
- Department of Genomics and Precision Medicine, The George Washington University, Washington, DC 20037, USA
- Center for Inherited Muscle Research, Department of Neurology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Christopher R. Heier
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
- Department of Genomics and Precision Medicine, The George Washington University, Washington, DC 20037, USA
- Center for Inherited Muscle Research, Department of Neurology, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
14
|
Thome T, Vugman NA, Stone LE, Wimberly K, Scali ST, Ryan TE. A tryptophan-derived uremic metabolite/Ahr/Pdk4 axis governs skeletal muscle mitochondrial energetics in chronic kidney disease. JCI Insight 2024; 9:e178372. [PMID: 38652558 PMCID: PMC11141944 DOI: 10.1172/jci.insight.178372] [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: 12/08/2023] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
Chronic kidney disease (CKD) causes accumulation of uremic metabolites that negatively affect skeletal muscle. Tryptophan-derived uremic metabolites are agonists of the aryl hydrocarbon receptor (AHR), which has been shown to be activated in CKD. This study investigated the role of the AHR in skeletal muscle pathology of CKD. Compared with controls with normal kidney function, AHR-dependent gene expression (CYP1A1 and CYP1B1) was significantly upregulated in skeletal muscle of patients with CKD, and the magnitude of AHR activation was inversely correlated with mitochondrial respiration. In mice with CKD, muscle mitochondrial oxidative phosphorylation (OXPHOS) was markedly impaired and strongly correlated with the serum level of tryptophan-derived uremic metabolites and AHR activation. Muscle-specific deletion of the AHR substantially improved mitochondrial OXPHOS in male mice with the greatest uremic toxicity (CKD + probenecid) and abolished the relationship between uremic metabolites and OXPHOS. The uremic metabolite/AHR/mitochondrial axis in skeletal muscle was verified using muscle-specific AHR knockdown in C57BL/6J mice harboring a high-affinity AHR allele, as well as ectopic viral expression of constitutively active mutant AHR in mice with normal renal function. Notably, OXPHOS changes in AHRmKO mice were present only when mitochondria were fueled by carbohydrates. Further analyses revealed that AHR activation in mice led to significantly increased pyruvate dehydrogenase kinase 4 (Pdk4) expression and phosphorylation of pyruvate dehydrogenase enzyme. These findings establish a uremic metabolite/AHR/Pdk4 axis in skeletal muscle that governs mitochondrial deficits in carbohydrate oxidation during CKD.
Collapse
Affiliation(s)
- Trace Thome
- Department of Applied Physiology and Kinesiology and
| | | | | | - Keon Wimberly
- Department of Applied Physiology and Kinesiology and
| | - Salvatore T. Scali
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, Florida, USA
- Malcom Randall VA Medical Center, Gainesville, Florida, USA
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology and
- Center for Exercise Science and
- Myology Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
15
|
Racine JJ, Bachman JF, Zhang JG, Misherghi A, Khadour R, Kaisar S, Bedard O, Jenkins C, Abbott A, Forte E, Rainer P, Rosenthal N, Sattler S, Serreze DV. Murine MHC-Deficient Nonobese Diabetic Mice Carrying Human HLA-DQ8 Develop Severe Myocarditis and Myositis in Response to Anti-PD-1 Immune Checkpoint Inhibitor Cancer Therapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1287-1306. [PMID: 38426910 PMCID: PMC10984778 DOI: 10.4049/jimmunol.2300841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
Myocarditis has emerged as an immune-related adverse event of immune checkpoint inhibitor (ICI) cancer therapy associated with significant mortality. To ensure patients continue to safely benefit from life-saving cancer therapy, an understanding of fundamental immunological phenomena underlying ICI myocarditis is essential. We recently developed the NOD-cMHCI/II-/-.DQ8 mouse model that spontaneously develops myocarditis with lower mortality than observed in previous HLA-DQ8 NOD mouse strains. Our strain was rendered murine MHC class I and II deficient using CRISPR/Cas9 technology, making it a genetically clean platform for dissecting CD4+ T cell-mediated myocarditis in the absence of classically selected CD8+ T cells. These mice are highly susceptible to myocarditis and acute heart failure following anti-PD-1 ICI-induced treatment. Additionally, anti-PD-1 administration accelerates skeletal muscle myositis. Using histology, flow cytometry, adoptive transfers, and RNA sequencing analyses, we performed a thorough characterization of cardiac and skeletal muscle T cells, identifying shared and unique characteristics of both populations. Taken together, this report details a mouse model with features of a rare, but highly lethal clinical presentation of overlapping myocarditis and myositis following ICI therapy. This study sheds light on underlying immunological mechanisms in ICI myocarditis and provides the basis for further detailed analyses of diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Adel Misherghi
- The Jackson Laboratory, Bar Harbor, Maine, USA
- College of the Atlantic, Bar Harbor, Maine, USA
| | - Raheem Khadour
- The Jackson Laboratory, Bar Harbor, Maine, USA
- College of the Atlantic, Bar Harbor, Maine, USA
| | | | | | | | | | | | - Peter Rainer
- Medical University of Graz, Graz, 8053 Austria
- BioTechMed Graz, Graz, Austria
- BKH St. Johann in Tirol, 6380 St. Johann in Tirol, Austria
| | - Nadia Rosenthal
- The Jackson Laboratory, Bar Harbor, Maine, USA
- Imperial College London, London SW7 2AZ, UK
| | - Susanne Sattler
- Imperial College London, London SW7 2AZ, UK
- Medical University of Graz, Graz, 8053 Austria
| | | |
Collapse
|
16
|
Kim K, Fazzone B, Cort TA, Kunz EM, Alvarez S, Moerschel J, Palzkill VR, Dong G, Anderson EM, O'Malley KA, Berceli SA, Ryan TE, Scali ST. Mitochondrial targeted catalase improves muscle strength following arteriovenous fistula creation in mice with chronic kidney disease. Sci Rep 2024; 14:8288. [PMID: 38594299 PMCID: PMC11004135 DOI: 10.1038/s41598-024-58805-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/03/2024] [Indexed: 04/11/2024] Open
Abstract
Hand dysfunction is a common observation after arteriovenous fistula (AVF) creation for hemodialysis access and has a variable clinical phenotype; however, the underlying mechanism responsible is unclear. Grip strength changes are a common metric used to assess AVF-associated hand disability but has previously been found to poorly correlate with the hemodynamic perturbations post-AVF placement implicating other tissue-level factors as drivers of hand outcomes. In this study, we sought to test if expression of a mitochondrial targeted catalase (mCAT) in skeletal muscle could reduce AVF-related limb dysfunction in mice with chronic kidney disease (CKD). Male and female C57BL/6J mice were fed an adenine-supplemented diet to induce CKD prior to placement of an AVF in the iliac vascular bundle. Adeno-associated virus was used to drive expression of either a green fluorescent protein (control) or mCAT using the muscle-specific human skeletal actin (HSA) gene promoter prior to AVF creation. As expected, the muscle-specific AAV-HSA-mCAT treatment did not impact blood urea nitrogen levels (P = 0.72), body weight (P = 0.84), or central hemodynamics including infrarenal aorta and inferior vena cava diameters (P > 0.18) or velocities (P > 0.38). Hindlimb perfusion recovery and muscle capillary densities were also unaffected by AAV-HSA-mCAT treatment. In contrast to muscle mass and myofiber size which were not different between groups, both absolute and specific muscle contractile forces measured via a nerve-mediated in-situ preparation were significantly greater in AAV-HSA-mCAT treated mice (P = 0.0012 and P = 0.0002). Morphological analysis of the post-synaptic neuromuscular junction uncovered greater acetylcholine receptor cluster areas (P = 0.0094) and lower fragmentation (P = 0.0010) in AAV-HSA-mCAT treated mice. Muscle mitochondrial oxidative phosphorylation was not different between groups, but AAV-HSA-mCAT treated mice had lower succinate-fueled mitochondrial hydrogen peroxide emission compared to AAV-HSA-GFP mice (P < 0.001). In summary, muscle-specific scavenging of mitochondrial hydrogen peroxide significantly improves neuromotor function in mice with CKD following AVF creation.
Collapse
Affiliation(s)
- Kyoungrae Kim
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA
| | - Brian Fazzone
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, P.O. Box 100128, Gainesville, FL, 32610, USA
- Malcom Randall Veteran Affairs Medical Center, Gainesville, FL, USA
| | - Tomas A Cort
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA
| | - Eric M Kunz
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA
| | - Samuel Alvarez
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA
| | - Jack Moerschel
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA
| | - Victoria R Palzkill
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA
| | - Gengfu Dong
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA
| | - Erik M Anderson
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, P.O. Box 100128, Gainesville, FL, 32610, USA
- Malcom Randall Veteran Affairs Medical Center, Gainesville, FL, USA
| | - Kerri A O'Malley
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, P.O. Box 100128, Gainesville, FL, 32610, USA
- Malcom Randall Veteran Affairs Medical Center, Gainesville, FL, USA
| | - Scott A Berceli
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, P.O. Box 100128, Gainesville, FL, 32610, USA
- Malcom Randall Veteran Affairs Medical Center, Gainesville, FL, USA
| | - Terence E Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA.
- Center for Exercise Science, University of Florida, Gainesville, FL, USA.
| | - Salvatore T Scali
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, P.O. Box 100128, Gainesville, FL, 32610, USA.
- Malcom Randall Veteran Affairs Medical Center, Gainesville, FL, USA.
| |
Collapse
|
17
|
Yao Z, Wo J, Zheng E, Yang J, Li H, Li X, Li J, Luo Y, Wang T, Fan Z, Zhan Y, Yang Y, Wu Z, Yin L, Meng F. A deep learning-based approach for fully automated segmentation and quantitative analysis of muscle fibers in pig skeletal muscle. Meat Sci 2024; 213:109506. [PMID: 38603965 DOI: 10.1016/j.meatsci.2024.109506] [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: 10/05/2023] [Revised: 02/06/2024] [Accepted: 04/01/2024] [Indexed: 04/13/2024]
Abstract
Muscle fiber properties exert a significant influence on pork quality, with cross-sectional area (CSA) being a crucial parameter closely associated with various meat quality indicators, such as shear force. Effectively identifying and segmenting muscle fibers in a robust manner constitutes a vital initial step in determining CSA. This step is highly intricate and time-consuming, necessitating an accurate and automated analytical approach. One limitation of existing methods is their tendency to perform well on high signal-to-noise ratio images of intact, healthy muscle fibers but their lack of validation on more complex image datasets featuring significant morphological changes, such as the presence of ice crystals. In this study, we undertake the fully automatic segmentation of muscle fiber microscopic images stained with myosin adenosine triphosphate (mATPase) activity using a deep learning architecture known as SOLOv2. Our objective is to efficiently derive accurate measurements of muscle fiber size and distribution. Tests conducted on actual images demonstrate that our method adeptly handles the intricate task of muscle fiber segmentation, yielding quantitative results amenable to statistical analysis and displaying reliability comparable to manual analysis.
Collapse
Affiliation(s)
- Zekai Yao
- State Key Laboratory of Swine and Poultry Breeding Industry/ Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, PR China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, PR China
| | - Jingjie Wo
- College of Mathematics and Informatics, South China Agricultural University, Guangzhou 510642, PR China
| | - Enqin Zheng
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, PR China
| | - Jie Yang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, PR China
| | - Hao Li
- State Key Laboratory of Swine and Poultry Breeding Industry/ Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, PR China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, PR China
| | - Xinxin Li
- State Key Laboratory of Swine and Poultry Breeding Industry/ Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, PR China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, PR China
| | - Jianhao Li
- State Key Laboratory of Swine and Poultry Breeding Industry/ Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, PR China
| | - Yizhi Luo
- State Key Laboratory of Swine and Poultry Breeding Industry/ Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, PR China; Institute of Facility Agriculture, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, PR China
| | - Ting Wang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, PR China
| | - Zhenfei Fan
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, PR China
| | - Yuexin Zhan
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, PR China
| | - Yingshan Yang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, PR China
| | - Zhenfang Wu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, PR China; Yunfu Subcenter of Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu 527400, PR China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, PR China.
| | - Ling Yin
- College of Mathematics and Informatics, South China Agricultural University, Guangzhou 510642, PR China.
| | - Fanming Meng
- State Key Laboratory of Swine and Poultry Breeding Industry/ Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, PR China.
| |
Collapse
|
18
|
Milani M, Della Valle I, Rossi S, Fabbrizio P, Margotta C, Nardo G, Cozzolino M, D'Ambrosi N, Apolloni S. Neuroprotective effects of niclosamide on disease progression via inflammatory pathways modulation in SOD1-G93A and FUS-associated amyotrophic lateral sclerosis models. Neurotherapeutics 2024; 21:e00346. [PMID: 38493058 PMCID: PMC11070272 DOI: 10.1016/j.neurot.2024.e00346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/28/2024] [Accepted: 03/06/2024] [Indexed: 03/18/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a complex neurodegenerative disease influenced by genetic, epigenetic, and environmental factors, resulting in dysfunction in cellular and molecular pathways. The limited efficacy of current treatments highlights the need for combination therapies targeting multiple aspects of the disease. Niclosamide, an anthelminthic drug listed as an essential medicine, has been repurposed in clinical trials for different diseases due to its anti-inflammatory and anti-fibrotic properties. Niclosamide can inhibit various molecular pathways (e.g., STAT3, mTOR) that are dysregulated in ALS, suggesting its potential to disrupt these altered mechanisms associated with the pathology. We administered niclosamide intraperitoneally to two transgenic murine models, SOD1-G93A and FUS mice, mimicking key pathological processes of ALS. The treatment was initiated at the onset of symptoms, and we assessed disease progression by neurological scores, rotarod and wire tests, and monitored survival. Furthermore, we investigated cellular and molecular mechanisms affected by niclosamide in the spinal cord and muscle of ALS mice. In both models, the administration of niclosamide resulted in a slowdown of disease progression, an increase in survival rates, and an improvement in tissue pathology. This was characterised by reduced gliosis, motor neuron loss, muscle atrophy, and inflammatory pathways. Based on these results, our findings demonstrate that niclosamide can impact multiple pathways involved in ALS. This multi-targeted approach leads to a slowdown in the progression of the disease, positioning niclosamide as a promising candidate for repurposing in the treatment of ALS.
Collapse
Affiliation(s)
- Martina Milani
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; Program in Cellular and Molecular Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ilaria Della Valle
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; Program in Cellular and Molecular Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Simona Rossi
- Institute of Translational Pharmacology, CNR, 00133 Rome, Italy
| | - Paola Fabbrizio
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Cassandra Margotta
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Giovanni Nardo
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Mauro Cozzolino
- Institute of Translational Pharmacology, CNR, 00133 Rome, Italy
| | - Nadia D'Ambrosi
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Savina Apolloni
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy.
| |
Collapse
|
19
|
Hung M, Lo HF, Beckmann AG, Demircioglu D, Damle G, Hasson D, Radice GL, Krauss RS. Cadherin-dependent adhesion is required for muscle stem cell niche anchorage and maintenance. Development 2024; 151:dev202387. [PMID: 38456551 PMCID: PMC11057819 DOI: 10.1242/dev.202387] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/25/2024] [Indexed: 03/09/2024]
Abstract
Adhesion between stem cells and their niche provides stable anchorage and signaling cues to sustain properties such as quiescence. Skeletal muscle stem cells (MuSCs) adhere to an adjacent myofiber via cadherin-catenin complexes. Previous studies on N- and M-cadherin in MuSCs revealed that although N-cadherin is required for quiescence, they are collectively dispensable for MuSC niche localization and regenerative activity. Although additional cadherins are expressed at low levels, these findings raise the possibility that cadherins are unnecessary for MuSC anchorage to the niche. To address this question, we conditionally removed from MuSCs β- and γ-catenin, and, separately, αE- and αT-catenin, factors that are essential for cadherin-dependent adhesion. Catenin-deficient MuSCs break quiescence similarly to N-/M-cadherin-deficient MuSCs, but exit the niche and are depleted. Combined in vivo, ex vivo and single cell RNA-sequencing approaches reveal that MuSC attrition occurs via precocious differentiation, re-entry to the niche and fusion to myofibers. These findings indicate that cadherin-catenin-dependent adhesion is required for anchorage of MuSCs to their niche and for preservation of the stem cell compartment. Furthermore, separable cadherin-regulated functions govern niche localization, quiescence and MuSC maintenance.
Collapse
Affiliation(s)
- Margaret Hung
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hsiao-Fan Lo
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Aviva G. Beckmann
- Pathos AI, 600 West Chicago Avenue, Suite 510, Chicago, IL 60654, USA
| | - Deniz Demircioglu
- Bioinformatics for Next Generation Sequencing Core, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Gargi Damle
- Bioinformatics for Next Generation Sequencing Core, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dan Hasson
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Bioinformatics for Next Generation Sequencing Core, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Glenn L. Radice
- Cardiovascular Research Center, Department of Medicine, Division of Cardiology, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Robert S. Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Bioinformatics for Next Generation Sequencing Core, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
20
|
Fitzgerald LF, Lackey J, Moussa A, Shah SV, Castellanos AM, Khan S, Schonk M, Thome T, Salyers ZR, Jakkidi N, Kim K, Yang Q, Hepple RT, Ryan TE. Chronic aryl hydrocarbon receptor activity impairs muscle mitochondrial function with tobacco smoking. J Cachexia Sarcopenia Muscle 2024; 15:646-659. [PMID: 38333944 PMCID: PMC10995249 DOI: 10.1002/jcsm.13439] [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: 06/06/2023] [Revised: 11/21/2023] [Accepted: 01/14/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Accumulating evidence has demonstrated that chronic tobacco smoking directly contributes to skeletal muscle dysfunction independent of its pathological impact to the cardiorespiratory systems. The mechanisms underlying tobacco smoke toxicity in skeletal muscle are not fully resolved. In this study, the role of the aryl hydrocarbon receptor (AHR), a transcription factor known to be activated with tobacco smoke, was investigated. METHODS AHR related gene (mRNA) expression was quantified in skeletal muscle from adult controls and patients with chronic obstructive pulmonary disease (COPD), as well as mice with and without cigarette smoke exposure. Utilizing both skeletal muscle-specific AHR knockout mice exposed to chronic repeated (5 days per week for 16 weeks) cigarette smoke and skeletal muscle-specific expression of a constitutively active mutant AHR in healthy mice, a battery of assessments interrogating muscle size, contractile function, mitochondrial energetics, and RNA sequencing were employed. RESULTS Skeletal muscle from COPD patients (N = 79, age = 67.0 ± 8.4 years) had higher levels of AHR (P = 0.0451) and CYP1B1 (P < 0.0001) compared to healthy adult controls (N = 16, age = 66.5 ± 6.5 years). Mice exposed to cigarette smoke displayed higher expression of Ahr (P = 0.008), Cyp1b1 (P < 0.0001), and Cyp1a1 (P < 0.0001) in skeletal muscle compared to air controls. Cigarette smoke exposure was found to impair skeletal muscle mitochondrial oxidative phosphorylation by ~50% in littermate controls (Treatment effect, P < 0.001), which was attenuated by deletion of the AHR in muscle in male (P = 0.001), but not female, mice (P = 0.37), indicating there are sex-dependent pathological effects of smoking-induced AHR activation in skeletal muscle. Viral mediated expression of a constitutively active mutant AHR in the muscle of healthy mice recapitulated the effects of cigarette smoking by decreasing muscle mitochondrial oxidative phosphorylation by ~40% (P = 0.003). CONCLUSIONS These findings provide evidence linking chronic AHR activation secondary to cigarette smoke exposure to skeletal muscle bioenergetic deficits in male, but not female, mice. AHR activation is a likely contributor to the decline in muscle oxidative capacity observed in smokers and AHR antagonism may provide a therapeutic avenue aimed to improve muscle function in COPD.
Collapse
Affiliation(s)
| | - Jacob Lackey
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFLUSA
| | - Ahmad Moussa
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFLUSA
| | - Sohan V. Shah
- Department of Physical TherapyUniversity of FloridaGainesvilleFLUSA
| | - Ana Maria Castellanos
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFLUSA
| | - Shawn Khan
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFLUSA
| | - Martin Schonk
- Department of Physical TherapyUniversity of FloridaGainesvilleFLUSA
| | - Trace Thome
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFLUSA
| | - Zachary R. Salyers
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFLUSA
| | - Nishka Jakkidi
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFLUSA
| | - Kyoungrae Kim
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFLUSA
| | - Qingping Yang
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFLUSA
| | - Russell T. Hepple
- Department of Physical TherapyUniversity of FloridaGainesvilleFLUSA
- Myology InstituteUniversity of FloridaGainesvilleFLUSA
| | - Terence E. Ryan
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFLUSA
- Myology InstituteUniversity of FloridaGainesvilleFLUSA
- Center for Exercise Science, University of FloridaGainesvilleFLUSA
| |
Collapse
|
21
|
Brun A, Mougeot G, Denis P, Collin ML, Pouchin P, Montaurier C, Walrand S, Capel F, Gueugneau M. A new bio imagery user-friendly tool for automatic morphometry measurement on muscle cell cultures and histological sections. Sci Rep 2024; 14:3108. [PMID: 38326394 PMCID: PMC11269594 DOI: 10.1038/s41598-024-53658-0] [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: 06/17/2023] [Accepted: 02/03/2024] [Indexed: 02/09/2024] Open
Abstract
TRUEFAD (TRUE Fiber Atrophy Distinction) is a bioimagery user-friendly tool developed to allow consistent and automatic measurement of myotube diameter in vitro, muscle fiber size and type using rodents and human muscle biopsies. This TRUEFAD package was set up to standardize and dynamize muscle research via easy-to-obtain images run on an open-source plugin for FIJI. We showed here both the robustness and the performance of our pipelines to correctly segment muscle cells and fibers. We evaluated our pipeline on real experiment image sets and showed consistent reliability across images and conditions. TRUEFAD development makes possible systematical and rapid screening of substances impacting muscle morphology for helping scientists focus on their hypothesis rather than image analysis.
Collapse
Affiliation(s)
- Aurélien Brun
- UMR1019 Unité de Nutrition Humaine (UNH), INRAE, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Guillaume Mougeot
- iGReD CNRS, INSERM Université Clermont Auvergne, Clermont-Ferrand, France
| | - Philippe Denis
- UMR1019 Unité de Nutrition Humaine (UNH), INRAE, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Marie Laure Collin
- UMR1019 Unité de Nutrition Humaine (UNH), INRAE, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Pierre Pouchin
- iGReD CNRS, INSERM Université Clermont Auvergne, Clermont-Ferrand, France
| | - Christophe Montaurier
- UMR1019 Unité de Nutrition Humaine (UNH), INRAE, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Stéphane Walrand
- UMR1019 Unité de Nutrition Humaine (UNH), INRAE, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Frédéric Capel
- UMR1019 Unité de Nutrition Humaine (UNH), INRAE, Université Clermont Auvergne, Clermont-Ferrand, France.
| | - Marine Gueugneau
- UMR1019 Unité de Nutrition Humaine (UNH), INRAE, Université Clermont Auvergne, Clermont-Ferrand, France
| |
Collapse
|
22
|
Englund DA, Jolliffe AM, Hanson GJ, Aversa Z, Zhang X, Jiang X, White TA, Zhang L, Monroe DG, Robbins PD, Niedernhofer LJ, Kamenecka TM, Khosla S, LeBrasseur NK. Senotherapeutic drug treatment ameliorates chemotherapy-induced cachexia. JCI Insight 2024; 9:e169512. [PMID: 38051584 PMCID: PMC10906225 DOI: 10.1172/jci.insight.169512] [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: 02/15/2023] [Accepted: 12/01/2023] [Indexed: 12/07/2023] Open
Abstract
Cachexia is a debilitating skeletal muscle wasting condition for which we currently lack effective treatments. In the context of cancer, certain chemotherapeutics cause DNA damage and cellular senescence. Senescent cells exhibit chronic activation of the transcription factor NF-κB, a known mediator of the proinflammatory senescence-associated secretory phenotype (SASP) and skeletal muscle atrophy. Thus, targeting NF-κB represents a logical therapeutic strategy to alleviate unintended consequences of genotoxic drugs. Herein, we show that treatment with the IKK/NF-κB inhibitor SR12343 during a course of chemotherapy reduces markers of cellular senescence and the SASP in liver, skeletal muscle, and circulation and, correspondingly, attenuates features of skeletal muscle pathology. Lastly, we demonstrate that SR12343 mitigates chemotherapy-induced reductions in body weight, lean mass, fat mass, and muscle strength. These findings support senescent cells as a promising druggable target to counteract the SASP and skeletal muscle wasting in the context of chemotherapy.
Collapse
Affiliation(s)
- Davis A. Englund
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Alyssa M. Jolliffe
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Gabriel J. Hanson
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Zaira Aversa
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Xu Zhang
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Xinyi Jiang
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Thomas A. White
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Lei Zhang
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - David G. Monroe
- Robert and Arlene Kogod Center on Aging, and
- Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging, and
- Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Nathan K. LeBrasseur
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
- Paul F. Glenn Center for the Biology of Aging at Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
23
|
Gu S, Wen C, Xiao Z, Huang Q, Jiang Z, Liu H, Gao J, Li J, Sun C, Yang N. MyoV: a deep learning-based tool for the automated quantification of muscle fibers. Brief Bioinform 2024; 25:bbad528. [PMID: 38271484 PMCID: PMC10810329 DOI: 10.1093/bib/bbad528] [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: 09/25/2023] [Revised: 12/06/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Accurate approaches for quantifying muscle fibers are essential in biomedical research and meat production. In this study, we address the limitations of existing approaches for hematoxylin and eosin-stained muscle fibers by manually and semiautomatically labeling over 660 000 muscle fibers to create a large dataset. Subsequently, an automated image segmentation and quantification tool named MyoV is designed using mask regions with convolutional neural networks and a residual network and feature pyramid network as the backbone network. This design enables the tool to allow muscle fiber processing with different sizes and ages. MyoV, which achieves impressive detection rates of 0.93-0.96 and precision levels of 0.91-0.97, exhibits a superior performance in quantification, surpassing both manual methods and commonly employed algorithms and software, particularly for whole slide images (WSIs). Moreover, MyoV is proven as a powerful and suitable tool for various species with different muscle development, including mice, which are a crucial model for muscle disease diagnosis, and agricultural animals, which are a significant meat source for humans. Finally, we integrate this tool into visualization software with functions, such as segmentation, area determination and automatic labeling, allowing seamless processing for over 400 000 muscle fibers within a WSI, eliminating the model adjustment and providing researchers with an easy-to-use visual interface to browse functional options and realize muscle fiber quantification from WSIs.
Collapse
Affiliation(s)
- Shuang Gu
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Chaoliang Wen
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Sanya Institute of China Agricultural University, Hainan 572025, China
| | - Zhen Xiao
- School of Computer and Information, Hefei University of Technology, Anhui 230009, China
| | - Qiang Huang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Zheyi Jiang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Honghong Liu
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jia Gao
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Junying Li
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Sanya Institute of China Agricultural University, Hainan 572025, China
| | - Congjiao Sun
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Sanya Institute of China Agricultural University, Hainan 572025, China
| | - Ning Yang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Sanya Institute of China Agricultural University, Hainan 572025, China
| |
Collapse
|
24
|
Rønning SB, Carlsen H, Rocha SDC, Rud I, Solberg N, Høst V, Veiseth-Kent E, Arnesen H, Bergum S, Kirkhus B, Böcker U, Abedali N, Rundblad A, Bålsrud P, Måge I, Holven KB, Ulven SM, Pedersen ME. Dietary intake of micronized avian eggshell membrane in aged mice reduces circulating inflammatory markers, increases microbiota diversity, and attenuates skeletal muscle aging. Front Nutr 2024; 10:1336477. [PMID: 38288061 PMCID: PMC10822908 DOI: 10.3389/fnut.2023.1336477] [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: 11/10/2023] [Accepted: 12/27/2023] [Indexed: 01/31/2024] Open
Abstract
Introduction Avian eggshell membrane (ESM) is a complex extracellular matrix comprising collagens, glycoproteins, proteoglycans, and hyaluronic acid. We have previously demonstrated that ESM possesses anti-inflammatory properties in vitro and regulates wound healing processes in vivo. The present study aimed to investigate if oral intake of micronized ESM could attenuate skeletal muscle aging associated with beneficial alterations in gut microbiota profile and reduced inflammation. Methods Elderly male C57BL/6 mice were fed an AIN93G diet supplemented with 0, 0.1, 1, or 8% ESM. Young mice were used as reference. The digestibility of ESM was investigated using the static in vitro digestion model INFOGEST for older people and adults, and the gut microbiota profile was analyzed in mice. In addition, we performed a small-scale pre-clinical human study with healthy home-dwelling elderly (>70 years) who received capsules with a placebo or 500 mg ESM every day for 4 weeks and studied the effect on circulating inflammatory markers. Results and discussion Intake of ESM in elderly mice impacted and attenuated several well-known hallmarks of aging, such as a reduction in the number of skeletal muscle fibers, the appearance of centronucleated fibers, a decrease in type IIa/IIx fiber type proportion, reduced gene expression of satellite cell markers Sdc3 and Pax7 and increased gene expression of the muscle atrophy marker Fbxo32. Similarly, a transition toward the phenotypic characteristics of young mice was observed for several proteins involved in cellular processes and metabolism. The digestibility of ESM was poor, especially for the elderly condition. Furthermore, our experiments showed that mice fed with 8% ESM had increased gut microbiota diversity and altered microbiota composition compared with the other groups. ESM in the diet also lowered the expression of the inflammation marker TNFA in mice and in vitro in THP-1 macrophages. In the human study, intake of ESM capsules significantly reduced the inflammatory marker CRP. Altogether, our results suggest that ESM, a natural extracellular biomaterial, may be attractive as a nutraceutical candidate with a possible effect on skeletal muscle aging possibly through its immunomodulating effect or gut microbiota.
Collapse
Affiliation(s)
| | - Harald Carlsen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | | | - Ida Rud
- Nofima AS, Food Division, Ås, Norway
| | | | | | | | - Henriette Arnesen
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | | | | | | | - Nada Abedali
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Amanda Rundblad
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Pia Bålsrud
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | - Kirsten Bjørklund Holven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Stine Marie Ulven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | |
Collapse
|
25
|
Du G, Zhang P, Guo J, Pang X, Kan G, Zeng B, Chen X, Liang J, Zhan Y. MF-Net: Automated Muscle Fiber Segmentation From Immunofluorescence Images Using a Local-Global Feature Fusion Network. J Digit Imaging 2023; 36:2411-2426. [PMID: 37714969 PMCID: PMC10584774 DOI: 10.1007/s10278-023-00890-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 09/17/2023] Open
Abstract
Histological assessment of skeletal muscle slices is very important for the accurate evaluation of weightless muscle atrophy. The accurate identification and segmentation of muscle fiber boundary is an important prerequisite for the evaluation of skeletal muscle fiber atrophy. However, there are many challenges to segment muscle fiber from immunofluorescence images, including the presence of low contrast in fiber boundaries in immunofluorescence images and the influence of background noise. Due to the limitations of traditional convolutional neural network-based segmentation methods in capturing global information, they cannot achieve ideal segmentation results. In this paper, we propose a muscle fiber segmentation network (MF-Net) method for effective segmentation of macaque muscle fibers in immunofluorescence images. The network adopts a dual encoder branch composed of convolutional neural networks and transformer to effectively capture local and global feature information in the immunofluorescence image, highlight foreground features, and suppress irrelevant background noise. In addition, a low-level feature decoder module is proposed to capture more global context information by combining different image scales to supplement the missing detail pixels. In this study, a comprehensive experiment was carried out on the immunofluorescence datasets of six macaques' weightlessness models and compared with the state-of-the-art deep learning model. It is proved from five segmentation indices that the proposed automatic segmentation method can be accurately and effectively applied to muscle fiber segmentation in shank immunofluorescence images.
Collapse
Affiliation(s)
| | - Peng Zhang
- China Astronaut Research and Training Center, Beijing, 100094, People's Republic of China
| | - Jianzhong Guo
- Institute of Applied Acoustics, School of Physics and Information Technology, Shaanxi Normal University, Xi'an, 710062, China
| | - Xiangsheng Pang
- China Astronaut Research and Training Center, Beijing, 100094, People's Republic of China
| | - Guanghan Kan
- China Astronaut Research and Training Center, Beijing, 100094, People's Republic of China
| | - Bin Zeng
- China Astronaut Research and Training Center, Beijing, 100094, People's Republic of China
| | - Xiaoping Chen
- China Astronaut Research and Training Center, Beijing, 100094, People's Republic of China.
| | - Jimin Liang
- School of Electronic Engineering, Xidian University, Xi'an, Shaanxi, 710071, China.
| | - Yonghua Zhan
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi, 710126, China.
| |
Collapse
|
26
|
Zeng Q, Du ZQ. Advances in the discovery of genetic elements underlying longissimus dorsi muscle growth and development in the pig. Anim Genet 2023; 54:709-720. [PMID: 37796678 DOI: 10.1111/age.13365] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 07/08/2023] [Accepted: 07/08/2023] [Indexed: 10/07/2023]
Abstract
As a major source of protein in human diets, pig meat plays a crucial role in ensuring global food security. Key determinants of meat production refer to the chemical and physical compositions or characteristics of muscle fibers, such as the number, hypertrophy potential, fiber-type conversion and intramuscular fat deposition. However, the growth and formation of muscle fibers comprises a complex process under spatio-temporal regulation, that is, the intermingled and concomitant proliferation, differentiation, migration and fusion of myoblasts. Recently, with the fast and continuous development of next-generation sequencing technology, the integration of quantitative trait loci mapping with genome-wide association studies (GWAS) has greatly helped animal geneticists to discover and explore thousands of functional or causal genetic elements underlying muscle growth and development. However, owing to the underlying complex molecular mechanisms, challenges to in-depth understanding and utilization remain, and the cost of large-scale sequencing, which requires integrated analyses of high-throughput omics data, is high. In this review, we mainly elaborate on research advances in integrative analyses (e.g. GWAS, omics) for identifying functional genes or genomic elements for longissimus dorsi muscle growth and development for different pig breeds, describing several successful transcriptome analyses and functional genomics cases, in an attempt to provide some perspective on the future functional annotation of genetic elements for muscle growth and development in pigs.
Collapse
Affiliation(s)
- Qingjie Zeng
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Zhi-Qiang Du
- College of Animal Science, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
27
|
Noviello C, Kobon K, Randrianarison-Huetz V, Maire P, Pietri-Rouxel F, Falcone S, Sotiropoulos A. RhoA Is a Crucial Regulator of Myoblast Fusion. Cells 2023; 12:2673. [PMID: 38067102 PMCID: PMC10705320 DOI: 10.3390/cells12232673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 12/18/2023] Open
Abstract
Satellite cells (SCs) are adult muscle stem cells that are mobilized when muscle homeostasis is perturbed. Here we show that RhoA in SCs is indispensable to have correct muscle regeneration and hypertrophy. In particular, the absence of RhoA in SCs prevents a correct SC fusion both to other RhoA-deleted SCs (regeneration context) and to growing control myofibers (hypertrophy context). We demonstrated that RhoA is dispensable for SCs proliferation and differentiation; however, RhoA-deleted SCs have an inefficient movement even if their cytoskeleton assembly is not altered. Proliferative myoblast and differentiated myotubes without RhoA display a decreased expression of Chordin, suggesting a crosstalk between these genes for myoblast fusion regulation. These findings demonstrate the importance of RhoA in SC fusion regulation and its requirement to achieve an efficient skeletal muscle homeostasis restoration.
Collapse
Affiliation(s)
- Chiara Noviello
- Institut Cochin, Université de Paris, INSERM U1016, CNRS, F-75014 Paris, France (P.M.); (A.S.)
- Centre de Recherche en Myologie, Sorbonne Université, INSERM UMRS 974, Institut de Myologie, F-75013 Paris, France;
| | - Kassandra Kobon
- Institut Cochin, Université de Paris, INSERM U1016, CNRS, F-75014 Paris, France (P.M.); (A.S.)
| | | | - Pascal Maire
- Institut Cochin, Université de Paris, INSERM U1016, CNRS, F-75014 Paris, France (P.M.); (A.S.)
| | - France Pietri-Rouxel
- Centre de Recherche en Myologie, Sorbonne Université, INSERM UMRS 974, Institut de Myologie, F-75013 Paris, France;
| | - Sestina Falcone
- Centre de Recherche en Myologie, Sorbonne Université, INSERM UMRS 974, Institut de Myologie, F-75013 Paris, France;
| | - Athanassia Sotiropoulos
- Institut Cochin, Université de Paris, INSERM U1016, CNRS, F-75014 Paris, France (P.M.); (A.S.)
| |
Collapse
|
28
|
Baltrusch S. Automated in-depth fiber and nuclei typing in cross-sectional muscle images can pave the way to a better understanding of skeletal muscle diseases. Acta Physiol (Oxf) 2023; 239:e14031. [PMID: 37551418 DOI: 10.1111/apha.14031] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/27/2023] [Accepted: 07/30/2023] [Indexed: 08/09/2023]
Affiliation(s)
- Simone Baltrusch
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Rostock and Department Life, Light & Matter, University of Rostock, Rostock, Germany
| |
Collapse
|
29
|
Lundquist A, Lázár E, Han NS, Emanuelsson EB, Reitzner SM, Chapman MA, Shirokova V, Alkass K, Druid H, Petri S, Sundberg CJ, Bergmann O. FiNuTyper: Design and validation of an automated deep learning-based platform for simultaneous fiber and nucleus type analysis in human skeletal muscle. Acta Physiol (Oxf) 2023; 239:e13982. [PMID: 37097015 DOI: 10.1111/apha.13982] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/30/2023] [Accepted: 04/21/2023] [Indexed: 04/26/2023]
Abstract
AIM While manual quantification is still considered the gold standard for skeletal muscle histological analysis, it is time-consuming and prone to investigator bias. To address this challenge, we assembled an automated image analysis pipeline, FiNuTyper (Fiber and Nucleus Typer). METHODS We integrated recently developed deep learning-based image segmentation methods, optimized for unbiased evaluation of fresh and postmortem human skeletal muscle, and utilized SERCA1 and SERCA2 as type-specific myonucleus and myofiber markers after validating them against the traditional use of MyHC isoforms. RESULTS Parameters including cross-sectional area, myonuclei per fiber, myonuclear domain, central myonuclei per fiber, and grouped myofiber ratio were determined in a fiber-type-specific manner, revealing that a large degree of sex- and muscle-related heterogeneity could be detected using the pipeline. Our platform was also tested on pathological muscle tissue (ALS and IBM) and adapted for the detection of other resident cell types (leucocytes, satellite cells, capillary endothelium). CONCLUSION In summary, we present an automated image analysis tool for the simultaneous quantification of myofiber and myonuclear types, to characterize the composition and structure of healthy and diseased human skeletal muscle.
Collapse
Affiliation(s)
- August Lundquist
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Enikő Lázár
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Nan S Han
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Eric B Emanuelsson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Stefan M Reitzner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department for Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Mark A Chapman
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Integrated Engineering, University of San Diego, San Diego, USA
| | - Vera Shirokova
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Kanar Alkass
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Druid
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Petri
- Department of Neurology, Hanover Medical School, Hanover, Germany
| | - Carl J Sundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Learning, Informatics, Management, and Ethics, Karolinska Institutet, Stockholm, Sweden
| | - Olaf Bergmann
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- Pharmacology and Toxicology, University Medical Center Göttingen (UMG), Göttingen, Germany
| |
Collapse
|
30
|
Kim K, Cort TA, Kunz EM, Moerschel J, Palzkill VR, Dong G, Moparthy CN, Anderson EM, Fazzone B, O'Malley KA, Robinson ST, Berceli SA, Ryan TE, Scali ST. N-acetylcysteine treatment attenuates hemodialysis access-related limb pathophysiology in mice with chronic kidney disease. Am J Physiol Renal Physiol 2023; 325:F271-F282. [PMID: 37439200 PMCID: PMC10511162 DOI: 10.1152/ajprenal.00083.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/20/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023] Open
Abstract
The objective of the present study was to determine if treatment with N-acetylcysteine (NAC) could reduce access-related limb dysfunction in mice. Male and female C57BL6J mice were fed an adenine-supplemented diet to induce chronic kidney disease (CKD) prior to the surgical creation of an arteriovenous fistula (AVF) in the iliac vascular bundle. AVF creation significantly increased peak aortic and infrarenal vena cava blood flow velocities, but NAC treatment had no significant impact, indicating that fistula maturation was not impacted by NAC treatment. Hindlimb muscle and paw perfusion recovery and muscle capillary density in the AVF limb were unaffected by NAC treatment. However, NAC treatment significantly increased the mass of the tibialis anterior (P = 0.0120) and soleus (P = 0.0452) muscles post-AVF. There was a significant main effect of NAC treatment on hindlimb grip strength at postoperative day 12 (POD 12) (P = 0.0003), driven by significantly higher grip strength in both male (P = 0.0273) and female (P = 0.0031) mice treated with NAC. There was also a significant main effect of NAC treatment on the walking speed at postoperative day 12 (P = 0.0447), and post hoc testing revealed an improvement in NAC-treated male mice (P = 0.0091). The area of postsynaptic acetylcholine receptors (P = 0.0263) and motor endplates (P = 0.0240) was also increased by NAC treatment. Interestingly, hindlimb skeletal muscle mitochondrial oxidative phosphorylation trended higher in NAC-treated female mice but was not statistically significant (P = 0.0973). Muscle glutathione levels and redox status were not significantly impacted by NAC treatment in either sex. In summary, NAC treatment attenuated some aspects of neuromotor pathology in mice with chronic kidney disease following AVF creation.NEW & NOTEWORTHY Hemodialysis via autogenous arteriovenous fistula (AVF) is the preferred first-line modality for renal replacement therapy in patients with end-stage kidney disease. However, patients undergoing AVF surgery frequently experience a spectrum of hand disability symptoms postsurgery including weakness and neuromotor dysfunction. Unfortunately, no treatment is currently available to prevent or mitigate these symptoms. Here, we provide evidence that daily N-acetylcysteine supplementation can attenuate some aspects of limb neuromotor function in a preclinical mouse model of AVF.
Collapse
Affiliation(s)
- Kyoungrae Kim
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, United States
| | - Tomas A Cort
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, United States
| | - Eric M Kunz
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, United States
| | - Jack Moerschel
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, United States
| | - Victoria R Palzkill
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, United States
| | - Gengfu Dong
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, United States
| | - Chatick N Moparthy
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, United States
| | - Erik M Anderson
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, Florida, United States
- Malcom Randall Veteran Affairs Medical Center, University of Florida, Gainesville, Florida, United States
| | - Brian Fazzone
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, Florida, United States
- Malcom Randall Veteran Affairs Medical Center, University of Florida, Gainesville, Florida, United States
| | - Kerri A O'Malley
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, Florida, United States
- Malcom Randall Veteran Affairs Medical Center, University of Florida, Gainesville, Florida, United States
| | - Scott T Robinson
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, Florida, United States
- Malcom Randall Veteran Affairs Medical Center, University of Florida, Gainesville, Florida, United States
| | - Scott A Berceli
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, Florida, United States
- Malcom Randall Veteran Affairs Medical Center, University of Florida, Gainesville, Florida, United States
| | - Terence E Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, United States
- Center for Exercise Science, University of Florida, Gainesville, Florida, United States
| | - Salvatore T Scali
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, Florida, United States
- Malcom Randall Veteran Affairs Medical Center, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
31
|
Danckaert A, Trignol A, Le Loher G, Loubens S, Staels B, Duez H, Shorte SL, Mayeuf-Louchart A. MuscleJ2: a rebuilding of MuscleJ with new features for high-content analysis of skeletal muscle immunofluorescence slides. Skelet Muscle 2023; 13:14. [PMID: 37612778 PMCID: PMC10463807 DOI: 10.1186/s13395-023-00323-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 07/25/2023] [Indexed: 08/25/2023] Open
Abstract
Histological analysis of skeletal muscle is of major interest for understanding its behavior in different pathophysiological conditions, such as the response to different environments or myopathies. In this context, many software programs have been developed to perform automated high-content analysis. We created MuscleJ, a macro that runs in ImageJ/Fiji on batches of images. MuscleJ is a multianalysis tool that initially allows the analysis of muscle fibers, capillaries, and satellite cells. Since its creation, it has been used in many studies, and we have further developed the software and added new features, which are presented in this article. We converted the macro into a Java-language plugin with an improved user interface. MuscleJ2 provides quantitative analysis of fibrosis, vascularization, and cell phenotype in whole muscle sections. It also performs analysis of the peri-myonuclei, the individual capillaries, and any staining in the muscle fibers, providing accurate quantification within regional sublocalizations of the fiber. A multicartography option allows users to visualize multiple results simultaneously. The plugin is freely available to the muscle science community.
Collapse
Affiliation(s)
- Anne Danckaert
- UTechS Photonic BioImaging/C2RT, Institut Pasteur, Université Paris Cité, 75015, Paris, France.
| | - Aurélie Trignol
- French Armed Forces Biomedical Research Institute - IRBA, Brétigny-sur-Orge, France
| | - Guillaume Le Loher
- UTechS Photonic BioImaging/C2RT, Institut Pasteur, Université Paris Cité, 75015, Paris, France
- École Centrale d'Electronique (ECE), Paris, France
| | - Sébastien Loubens
- CHU Lille, INSERM, Institut Pasteur de Lille, Univ. Lille, U1011-EGID, Lille, 59000, France
- Service Neuropédiatrie, CHU Lille, 59000, Lille, France
| | - Bart Staels
- CHU Lille, INSERM, Institut Pasteur de Lille, Univ. Lille, U1011-EGID, Lille, 59000, France
| | - Hélène Duez
- CHU Lille, INSERM, Institut Pasteur de Lille, Univ. Lille, U1011-EGID, Lille, 59000, France
| | - Spencer L Shorte
- UTechS Photonic BioImaging/C2RT, Institut Pasteur, Université Paris Cité, 75015, Paris, France
| | - Alicia Mayeuf-Louchart
- CHU Lille, INSERM, Institut Pasteur de Lille, Univ. Lille, U1011-EGID, Lille, 59000, France.
| |
Collapse
|
32
|
Dubuisson N, Versele R, Davis-López de Carrizosa MA, Selvais CM, Noel L, Planchon C, Van den Bergh PYK, Brichard SM, Abou-Samra M. The Adiponectin Receptor Agonist, ALY688: A Promising Therapeutic for Fibrosis in the Dystrophic Muscle. Cells 2023; 12:2101. [PMID: 37626911 PMCID: PMC10453606 DOI: 10.3390/cells12162101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is one of the most devastating myopathies, where severe inflammation exacerbates disease progression. Previously, we demonstrated that adiponectin (ApN), a hormone with powerful pleiotropic effects, can efficiently improve the dystrophic phenotype. However, its practical therapeutic application is limited. In this study, we investigated ALY688, a small peptide ApN receptor agonist, as a potential novel treatment for DMD. Four-week-old mdx mice were subcutaneously treated for two months with ALY688 and then compared to untreated mdx and wild-type mice. In vivo and ex vivo tests were performed to assess muscle function and pathophysiology. Additionally, in vitro tests were conducted on human DMD myotubes. Our results showed that ALY688 significantly improved the physical performance of mice and exerted potent anti-inflammatory, anti-oxidative and anti-fibrotic actions on the dystrophic muscle. Additionally, ALY688 hampered myonecrosis, partly mediated by necroptosis, and enhanced the myogenic program. Some of these effects were also recapitulated in human DMD myotubes. ALY688's protective and beneficial properties were mainly mediated by the AMPK-PGC-1α axis, which led to suppression of NF-κβ and TGF-β. Our results demonstrate that an ApN mimic may be a promising and effective therapeutic prospect for a better management of DMD.
Collapse
Affiliation(s)
- Nicolas Dubuisson
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
- Neuromuscular Reference Center, Department of Neurology, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium;
| | - Romain Versele
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
| | - Maria A. Davis-López de Carrizosa
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Seville, Spain
| | - Camille M. Selvais
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
| | - Laurence Noel
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
| | - Chloé Planchon
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
| | - Peter Y. K. Van den Bergh
- Neuromuscular Reference Center, Department of Neurology, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium;
| | - Sonia M. Brichard
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
| | - Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
| |
Collapse
|
33
|
McCormack NM, Nguyen NY, Tully CB, Oliver T, Fiorillo AA, Heier CR. Vamorolone improves Becker muscular dystrophy and increases dystrophin protein in bmx model mice. iScience 2023; 26:107161. [PMID: 37534133 PMCID: PMC10391915 DOI: 10.1016/j.isci.2023.107161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/15/2023] [Accepted: 06/13/2023] [Indexed: 08/04/2023] Open
Abstract
There is no approved therapy for Becker muscular dystrophy (BMD), a genetic muscle disease caused by in-frame dystrophin deletions. We previously developed the dissociative corticosteroid vamorolone for treatment of the allelic, dystrophin-null disease Duchenne muscular dystrophy. We hypothesize vamorolone can treat BMD by safely reducing inflammatory signaling in muscle and through a novel mechanism of increasing dystrophin protein via suppression of dystrophin-targeting miRNAs. Here, we test this in the bmx mouse model of BMD. Daily oral treatment with vamorolone or prednisolone improves bmx grip strength and hang time phenotypes. Both drugs reduce myofiber size and decrease the percentage of centrally nucleated fibers. Vamorolone shows improved safety versus prednisolone by avoiding or reducing key side effects to behavior and growth. Intriguingly, vamorolone increases dystrophin protein in both heart and skeletal muscle. These data indicate that vamorolone, nearing approval for Duchenne, shows efficacy in bmx mice and therefore warrants clinical investigation in BMD.
Collapse
Affiliation(s)
- Nikki M. McCormack
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Nhu Y. Nguyen
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Christopher B. Tully
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Trinitee Oliver
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
- Department of Biology, Howard University, Washington, DC, USA
| | - Alyson A. Fiorillo
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
- Department of Genomics and Precision Medicine, The George Washington University, Washington, DC, USA
| | - Christopher R. Heier
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
- Department of Genomics and Precision Medicine, The George Washington University, Washington, DC, USA
| |
Collapse
|
34
|
Dong G, Moparthy C, Thome T, Kim K, Yue F, Ryan TE. IGF-1 Therapy Improves Muscle Size and Function in Experimental Peripheral Arterial Disease. JACC Basic Transl Sci 2023; 8:702-719. [PMID: 37426532 PMCID: PMC10322901 DOI: 10.1016/j.jacbts.2022.12.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 03/11/2023]
Abstract
Lower-extremity peripheral arterial disease (PAD) has increased in prevalence, yet therapeutic development has remained stagnant. Skeletal muscle health and function has been strongly linked to quality of life and medical outcomes in patients with PAD. Using a rodent model of PAD, this study demonstrates that treatment of the ischemic limb with insulin-like growth factor (IGF)-1 significantly increases muscle size and strength without improving limb hemodynamics. Interestingly, the effect size of IGF1 therapy was larger in female mice than in male mice, highlighting the need to carefully examine sex-dependent effects in experimental PAD therapies.
Collapse
Affiliation(s)
- Gengfu Dong
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Chatick Moparthy
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Trace Thome
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Kyoungrae Kim
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Feng Yue
- Department of Animal Sciences, University of Florida, Gainesville, Florida, USA
- Myology Institute, University of Florida, Gainesville, Florida, USA
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
- Center for Exercise Science, University of Florida, Gainesville, Florida, USA
- Myology Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
35
|
Paul TA, Macpherson PC, Janetzke TL, Davis CS, Jackson MJ, McArdle A, Brooks SV. Older mice show decreased regeneration of neuromuscular junctions following lengthening contraction-induced injury. GeroScience 2023; 45:1899-1912. [PMID: 36952126 PMCID: PMC10400502 DOI: 10.1007/s11357-023-00774-w] [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: 11/11/2022] [Accepted: 03/13/2023] [Indexed: 03/24/2023] Open
Abstract
Progressive muscle atrophy and loss of muscle strength associated with old age have been well documented. Although age-associated impairments in skeletal muscle regeneration following injury have been demonstrated, less is known about whether aging impacts the regenerative response of neuromuscular junctions (NMJ) following contraction-induced injury. Reduced ability of NMJs to regenerate could lead to increased numbers of denervated muscle fibers and therefore play a contributing role to age-related sarcopenia. To investigate the relationship between age and NMJ regeneration following injury, extensor digitorum longus (EDL) muscles of middle-aged (18-19 months) and old mice (27-28 months) were subjected to a protocol of lengthening contractions (LC) that resulted in an acute force deficit of ~55% as well as functional and histological evidence of a similar magnitude of injury 3 days post LCs that was not different between age groups. After 28 days, the architecture and innervation of the NMJs were evaluated. The numbers of fragmented endplates increased and of fully innervated NMJs decreased post-injury for the muscle of both middle-aged and old mice and for contralateral uninjured muscles of old compared with uninjured muscles of middle-aged controls. Thus, the diminished ability of the skeletal muscle of old mice to recover following injury may be due in part to an age-related decrease in the ability to regenerate NMJs in injured muscles. The impaired ability to regenerate NMJs may be a triggering factor for degenerative changes at the NMJ contributing to muscle fiber weakness and loss in old age.
Collapse
Affiliation(s)
- Thomas A. Paul
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI USA
- Department of Biomedical Engineering, University of Michigan, 2029 Biomedical Sciences Building, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200 USA
| | - Peter C. Macpherson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI USA
| | - Tara L. Janetzke
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI USA
| | - Carol S. Davis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI USA
| | - Malcolm J. Jackson
- MRC-Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Institute of Life Course and Ageing Science, University of Liverpool, Liverpool, UK
| | - Anne McArdle
- MRC-Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Institute of Life Course and Ageing Science, University of Liverpool, Liverpool, UK
| | - Susan V. Brooks
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI USA
- Department of Biomedical Engineering, University of Michigan, 2029 Biomedical Sciences Building, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200 USA
| |
Collapse
|
36
|
Samakkarnthai P, Saul D, Zhang L, Aversa Z, Doolittle ML, Sfeir JG, Kaur J, Atkinson EJ, Edwards JR, Russell GG, Pignolo RJ, Kirkland JL, Tchkonia T, Niedernhofer LJ, Monroe DG, Lebrasseur NK, Farr JN, Robbins PD, Khosla S. In vitro and in vivo effects of zoledronic acid on senescence and senescence-associated secretory phenotype markers. Aging (Albany NY) 2023; 15:3331-3355. [PMID: 37154858 PMCID: PMC10449299 DOI: 10.18632/aging.204701] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/18/2023] [Indexed: 05/10/2023]
Abstract
In addition to reducing fracture risk, zoledronic acid has been found in some studies to decrease mortality in humans and extend lifespan and healthspan in animals. Because senescent cells accumulate with aging and contribute to multiple co-morbidities, the non-skeletal actions of zoledronic acid could be due to senolytic (killing of senescent cells) or senomorphic (inhibition of the secretion of the senescence-associated secretory phenotype [SASP]) actions. To test this, we first performed in vitro senescence assays using human lung fibroblasts and DNA repair-deficient mouse embryonic fibroblasts, which demonstrated that zoledronic acid killed senescent cells with minimal effects on non-senescent cells. Next, in aged mice treated with zoledronic acid or vehicle for 8 weeks, zoledronic acid significantly reduced circulating SASP factors, including CCL7, IL-1β, TNFRSF1A, and TGFβ1 and improved grip strength. Analysis of publicly available RNAseq data from CD115+ (CSF1R/c-fms+) pre-osteoclastic cells isolated from mice treated with zoledronic acid demonstrated a significant downregulation of senescence/SASP genes (SenMayo). To establish that these cells are potential senolytic/senomorphic targets of zoledronic acid, we used single cell proteomic analysis (cytometry by time of flight [CyTOF]) and demonstrated that zoledronic acid significantly reduced the number of pre-osteoclastic (CD115+/CD3e-/Ly6G-/CD45R-) cells and decreased protein levels of p16, p21, and SASP markers in these cells without affecting other immune cell populations. Collectively, our findings demonstrate that zoledronic acid has senolytic effects in vitro and modulates senescence/SASP biomarkers in vivo. These data point to the need for additional studies testing zoledronic acid and/or other bisphosphonate derivatives for senotherapeutic efficacy.
Collapse
Affiliation(s)
- Parinya Samakkarnthai
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Division of Endocrinology, Phramongkutklao Hospital and College of Medicine, Bangkok 10400, Thailand
| | - Dominik Saul
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Trauma Center Tübingen, Tübingen 72076, Germany
| | - Lei Zhang
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zaira Aversa
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN 55905, USA
| | - Madison L. Doolittle
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Jad G. Sfeir
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Japneet Kaur
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | | | - James R. Edwards
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7FY, UK
| | - Graham G. Russell
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7FY, UK
- Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, S10 2RX, UK
| | - Robert J. Pignolo
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Division of Hospital Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - James L. Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - David G. Monroe
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Nathan K. Lebrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN 55905, USA
| | - Joshua N. Farr
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sundeep Khosla
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
37
|
Heier CR, McCormack NM, Tully CB, Novak JS, Newell‐Stamper BL, Russell AJ, Fiorillo AA. The X-linked Becker muscular dystrophy (bmx) mouse models Becker muscular dystrophy via deletion of murine dystrophin exons 45-47. J Cachexia Sarcopenia Muscle 2023; 14:940-954. [PMID: 36628607 PMCID: PMC10067474 DOI: 10.1002/jcsm.13171] [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: 07/20/2022] [Revised: 11/22/2022] [Accepted: 12/04/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Becker muscular dystrophy (BMD) is a genetic neuromuscular disease of growing importance caused by in-frame, partial loss-of-function mutations in the dystrophin (DMD) gene. BMD presents with reduced severity compared with Duchenne muscular dystrophy (DMD), the allelic disorder of complete dystrophin deficiency. Significant therapeutic advancements have been made in DMD, including four FDA-approved drugs. BMD, however, is understudied and underserved-there are no drugs and few clinical trials. Discordance in therapeutic efforts is due in part to lack of a BMD mouse model which would enable greater understanding of disease and de-risk potential therapeutics before first-in-human trials. Importantly, a BMD mouse model is becoming increasingly critical as emerging DMD dystrophin restoration therapies aim to convert a DMD genotype into a BMD phenotype. METHODS We use CRISPR/Cas9 technology to generate bmx (Becker muscular dystrophy, X-linked) mice, which express an in-frame ~40 000 bp deletion of exons 45-47 in the murine Dmd gene, reproducing the most common BMD patient mutation. Here, we characterize muscle pathogenesis using molecular and histological techniques and then test skeletal muscle and cardiac function using muscle function assays and echocardiography. RESULTS Overall, bmx mice present with significant muscle weakness and heart dysfunction versus wild-type (WT) mice, despite a substantial improvement in pathology over dystrophin-null mdx52 mice. bmx mice show impaired motor function in grip strength (-39%, P < 0.0001), wire hang (P = 0.0025), and in vivo as well as ex vivo force assays. In aged bmx, echocardiography reveals decreased heart function through reduced fractional shortening (-25%, P = 0.0036). Additionally, muscle-specific serum CK is increased >60-fold (P < 0.0001), indicating increased muscle damage. Histologically, bmx muscles display increased myofibre size variability (minimal Feret's diameter: P = 0.0017) and centrally located nuclei indicating degeneration/regeneration (P < 0.0001). bmx muscles also display dystrophic pathology; however, levels of the following parameters are moderate in comparison with mdx52: inflammatory/necrotic foci (P < 0.0001), collagen deposition (+1.4-fold, P = 0.0217), and sarcolemmal damage measured by intracellular IgM (P = 0.0878). Like BMD patients, bmx muscles show reduced dystrophin protein levels (~20-50% of WT), whereas Dmd transcript levels are unchanged. At the molecular level, bmx muscles express increased levels of inflammatory genes, inflammatory miRNAs and fibrosis genes. CONCLUSIONS The bmx mouse recapitulates BMD disease phenotypes with histological, molecular and functional deficits. Importantly, it can inform both BMD pathology and DMD dystrophin restoration therapies. This novel model will enable further characterization of BMD disease progression, identification of biomarkers, identification of therapeutic targets and new preclinical drug studies aimed at developing therapies for BMD patients.
Collapse
Affiliation(s)
- Christopher R. Heier
- Center for Genetic Medicine ResearchChildren's National HospitalWashingtonDCUSA
- Department of Genomics and Precision MedicineGeorge Washington University School of Medicine and Health SciencesWashingtonDCUSA
| | - Nikki M. McCormack
- Center for Genetic Medicine ResearchChildren's National HospitalWashingtonDCUSA
| | | | - James S. Novak
- Center for Genetic Medicine ResearchChildren's National HospitalWashingtonDCUSA
- Department of Genomics and Precision MedicineGeorge Washington University School of Medicine and Health SciencesWashingtonDCUSA
| | | | - Alan J. Russell
- Edgewise Therapeutics, BioFrontiers InstituteUniversity of ColoradoBoulderCO80303USA
| | - Alyson A. Fiorillo
- Center for Genetic Medicine ResearchChildren's National HospitalWashingtonDCUSA
- Department of Genomics and Precision MedicineGeorge Washington University School of Medicine and Health SciencesWashingtonDCUSA
| |
Collapse
|
38
|
Pierre A, Bourel C, Favory R, Brassart B, Wallet F, Daussin FN, Normandin S, Howsam M, Romien R, Lemaire J, Grolaux G, Durand A, Frimat M, Bastide B, Amouyel P, Boulanger E, Preau S, Lancel S. Sepsis-like Energy Deficit Is Not Sufficient to Induce Early Muscle Fiber Atrophy and Mitochondrial Dysfunction in a Murine Sepsis Model. BIOLOGY 2023; 12:529. [PMID: 37106730 PMCID: PMC10136327 DOI: 10.3390/biology12040529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/17/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
Sepsis-induced myopathy is characterized by muscle fiber atrophy, mitochondrial dysfunction, and worsened outcomes. Whether whole-body energy deficit participates in the early alteration of skeletal muscle metabolism has never been investigated. Three groups were studied: "Sepsis" mice, fed ad libitum with a spontaneous decrease in caloric intake (n = 17), and "Sham" mice fed ad libitum (Sham fed (SF), n = 13) or subjected to pair-feeding (Sham pair fed (SPF), n = 12). Sepsis was induced by the intraperitoneal injection of cecal slurry in resuscitated C57BL6/J mice. The feeding of the SPF mice was restricted according to the food intake of the Sepsis mice. Energy balance was evaluated by indirect calorimetry over 24 h. The tibialis anterior cross-sectional area (TA CSA), mitochondrial function (high-resolution respirometry), and mitochondrial quality control pathways (RTqPCR and Western blot) were assessed 24 h after sepsis induction. The energy balance was positive in the SF group and negative in both the SPF and Sepsis groups. The TA CSA did not differ between the SF and SPF groups, but was reduced by 17% in the Sepsis group compared with the SPF group (p < 0.05). The complex-I-linked respiration in permeabilized soleus fibers was higher in the SPF group than the SF group (p < 0.05) and lower in the Sepsis group than the SPF group (p < 0.01). Pgc1α protein expression increased 3.9-fold in the SPF mice compared with the SF mice (p < 0.05) and remained unchanged in the Sepsis mice compared with the SPF mice; the Pgc1α mRNA expression decreased in the Sepsis compared with the SPF mice (p < 0.05). Thus, the sepsis-like energy deficit did not explain the early sepsis-induced muscle fiber atrophy and mitochondrial dysfunction, but led to specific metabolic adaptations not observed in sepsis.
Collapse
Affiliation(s)
- Alexandre Pierre
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
- Division of Intensive Care, Hôpital Roger Salengro, CHU de Lille, F-59000 Lille, France
| | - Claire Bourel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
- Division of Intensive Care, Hôpital Roger Salengro, CHU de Lille, F-59000 Lille, France
| | - Raphael Favory
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
- Division of Intensive Care, Hôpital Roger Salengro, CHU de Lille, F-59000 Lille, France
| | - Benoit Brassart
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
- Division of Intensive Care, Hôpital Roger Salengro, CHU de Lille, F-59000 Lille, France
| | - Frederic Wallet
- Division of Bacteriology, Biology Pathology Institute of Lille, CHU de Lille, F-59000 Lille, France
| | - Frederic N. Daussin
- Univ. Lille, Univ. Artois, Univ. Littoral Côte d’Opale, ULR 7369-URePSSS-Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France
| | - Sylvain Normandin
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
- Division of Intensive Care, Hôpital Roger Salengro, CHU de Lille, F-59000 Lille, France
| | - Michael Howsam
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| | - Raphael Romien
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| | - Jeremy Lemaire
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| | - Gaelle Grolaux
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| | - Arthur Durand
- Division of Intensive Care, Hôpital Roger Salengro, CHU de Lille, F-59000 Lille, France
| | - Marie Frimat
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
- Division of Nephrology, CHU de Lille, Université de Lille, F-59000 Lille, France
| | - Bruno Bastide
- Univ. Lille, Univ. Artois, Univ. Littoral Côte d’Opale, ULR 7369-URePSSS-Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France
| | - Philippe Amouyel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| | - Eric Boulanger
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| | - Sebastien Preau
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
- Division of Intensive Care, Hôpital Roger Salengro, CHU de Lille, F-59000 Lille, France
| | - Steve Lancel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| |
Collapse
|
39
|
Margotta C, Fabbrizio P, Ceccanti M, Cambieri C, Ruffolo G, D'Agostino J, Trolese MC, Cifelli P, Alfano V, Laurini C, Scaricamazza S, Ferri A, Sorarù G, Palma E, Inghilleri M, Bendotti C, Nardo G. Immune-mediated myogenesis and acetylcholine receptor clustering promote a slow disease progression in ALS mouse models. Inflamm Regen 2023; 43:19. [PMID: 36895050 PMCID: PMC9996869 DOI: 10.1186/s41232-023-00270-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/25/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a heterogeneous disease in terms of onset and progression rate. This may account for therapeutic clinical trial failure. Transgenic SOD1G93A mice on C57 or 129Sv background have a slow and fast disease progression rate, mimicking the variability observed in patients. Based on evidence inferring the active influence of skeletal muscle on ALS pathogenesis, we explored whether dysregulation in hindlimb skeletal muscle reflects the phenotypic difference between the two mouse models. METHODS Ex vivo immunohistochemical, biochemical, and biomolecular methodologies, together with in vivo electrophysiology and in vitro approaches on primary cells, were used to afford a comparative and longitudinal analysis of gastrocnemius medialis between fast- and slow-progressing ALS mice. RESULTS We reported that slow-progressing mice counteracted muscle denervation atrophy by increasing acetylcholine receptor clustering, enhancing evoked currents, and preserving compound muscle action potential. This matched with prompt and sustained myogenesis, likely triggered by an early inflammatory response switching the infiltrated macrophages towards a M2 pro-regenerative phenotype. Conversely, upon denervation, fast-progressing mice failed to promptly activate a compensatory muscle response, exhibiting a rapidly progressive deterioration of muscle force. CONCLUSIONS Our findings further pinpoint the pivotal role of skeletal muscle in ALS, providing new insights into underestimated disease mechanisms occurring at the periphery and providing useful (diagnostic, prognostic, and mechanistic) information to facilitate the translation of cost-effective therapeutic strategies from the laboratory to the clinic.
Collapse
Affiliation(s)
- Cassandra Margotta
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Paola Fabbrizio
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Marco Ceccanti
- Department of Human Neurosciences, Rare Neuromuscular Diseases Centre, Sapienza University of Rome, 00185, Rome, Italy
| | - Chiara Cambieri
- Department of Human Neurosciences, Rare Neuromuscular Diseases Centre, Sapienza University of Rome, 00185, Rome, Italy
| | - Gabriele Ruffolo
- Laboratory Affiliated to Istituto Pasteur Italia, Department of Physiology and Pharmacology, Sapienza University of Rome, 00185, Rome, Italy.,IRCCS San Raffaele Roma, 00163, Rome, Italy
| | - Jessica D'Agostino
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Maria Chiara Trolese
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Pierangelo Cifelli
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | | | - Christian Laurini
- Department of Human Neurosciences, Rare Neuromuscular Diseases Centre, Sapienza University of Rome, 00185, Rome, Italy
| | | | - Alberto Ferri
- IRCCS Fondazione Santa Lucia, Rome, Italy.,Institute of Translational Pharmacology (IFT-CNR), Rome, Italy
| | - Gianni Sorarù
- Department of Neuroscience, Azienda Ospedaliera di Padova, Via Giustiniani 2, 35128, Padua, Italy
| | - Eleonora Palma
- Laboratory Affiliated to Istituto Pasteur Italia, Department of Physiology and Pharmacology, Sapienza University of Rome, 00185, Rome, Italy.,IRCCS San Raffaele Roma, 00163, Rome, Italy
| | - Maurizio Inghilleri
- Department of Human Neurosciences, Rare Neuromuscular Diseases Centre, Sapienza University of Rome, 00185, Rome, Italy
| | - Caterina Bendotti
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy.
| | - Giovanni Nardo
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| |
Collapse
|
40
|
Jurberg AD, Gomes G, Seixas MR, Mermelstein C, Costa ML. Improving quantification of myotube width and nuclear/cytoplasmic ratio in myogenesis research. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 230:107354. [PMID: 36682109 DOI: 10.1016/j.cmpb.2023.107354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/05/2023] [Accepted: 01/12/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND AND OBJECTIVE The culture of skeletal muscle cells is particularly relevant to basic biomedical research and translational medicine. The incubation of dissociated cells under controlled conditions has helped to dissect several molecular mechanisms associated with muscle cell differentiation, in addition to contributing for the evaluation of drug effects and prospective cell therapies for patients with degenerative muscle pathologies. The formation of mature multinucleated myotubes is a stepwise process involving well defined events of cell proliferation, commitment, migration, and fusion easily identified through optical microscopy methods including immunofluorescence and live cell imaging. The characterization of each step is usually based on muscle cell morphology and nuclei number, as well as the presence and intracellular location of specific cell markers. However, manual quantification of these parameters in large datasets of images is work-intensive and prone to researcher's subjectivity, mostly because of the extremely elongated cell shape of large myotubes and because myotubes are multinucleated. METHODS Here we provide two semi-automated ImageJ macros aimed to measure the width of myotubes and the nuclear/cytoplasmic localization of molecules in fluorescence images. The width measuring macro automatically determines the best angle, perpendicular to most cells, to draw a profile plot and identify and measure individual myotubes. The nuclear/cytoplasmic ratio macro compares the intensity values along lines, drawn by the user, over cytoplasm and nucleus. RESULTS We show that the macro measurements are more consistent than manual measurements by comparing with our own results and with the literature. CONCLUSIONS By relying on semi-automated muscle specific ImageJ macros, we seek to improve measurement accuracy and to alleviate the laborious routine of counting and measuring muscle cell features.
Collapse
Affiliation(s)
- Arnon Dias Jurberg
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), RJ, Brazil; Instituto de Educação Médica (IDOMED), Campus Vista Carioca, Universidade Estácio de Sá (UNESA), RJ, Brazil
| | - Geyse Gomes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), RJ, Brazil
| | - Marianna Reis Seixas
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), RJ, Brazil
| | - Claudia Mermelstein
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), RJ, Brazil
| | - Manoel Luis Costa
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), RJ, Brazil.
| |
Collapse
|
41
|
Kahn RE, Krater T, Larson JE, Encarnacion M, Karakostas T, Patel NM, Swaroop VT, Dayanidhi S. Resident muscle stem cell myogenic characteristics in postnatal muscle growth impairments in children with cerebral palsy. Am J Physiol Cell Physiol 2023; 324:C614-C631. [PMID: 36622072 PMCID: PMC9942895 DOI: 10.1152/ajpcell.00499.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/13/2022] [Accepted: 12/28/2022] [Indexed: 01/10/2023]
Abstract
Children with cerebral palsy (CP), a perinatal brain alteration, have impaired postnatal muscle growth, with some muscles developing contractures. Functionally, children are either able to walk or primarily use wheelchairs. Satellite cells are muscle stem cells (MuSCs) required for postnatal development and source of myonuclei. Only MuSC abundance has been previously reported in contractured muscles, with myogenic characteristics assessed only in vitro. We investigated whether MuSC myogenic, myonuclear, and myofiber characteristics in situ differ between contractured and noncontractured muscles, across functional levels, and compared with typically developing (TD) children with musculoskeletal injury. Open muscle biopsies were obtained from 36 children (30 CP, 6 TD) during surgery; contracture correction for adductors or gastrocnemius, or from vastus lateralis [bony surgery in CP, anterior cruciate ligament (ACL) repair in TD]. Muscle cross sections were immunohistochemically labeled for MuSC abundance, activation, proliferation, nuclei, myofiber borders, type-1 fibers, and collagen content in serial sections. Although MuSC abundance was greater in contractured muscles, primarily in type-1 fibers, their myogenic characteristics (activation, proliferation) were lower compared with noncontractured muscles. Overall, MuSC abundance, activation, and proliferation appear to be associated with collagen content. Myonuclear number was similar between all muscles, but only in contractured muscles were there associations between myonuclear number, MuSC abundance, and fiber cross-sectional area. Puzzlingly, MuSC characteristics were similar between ambulatory and nonambulatory children. Noncontractured muscles in children with CP had a lower MuSC abundance compared with TD-ACL injured children, but similar myogenic characteristics. Contractured muscles may have an intrinsic deficiency in developmental progression for postnatal MuSC pool establishment, needed for lifelong efficient growth and repair.
Collapse
Affiliation(s)
| | | | - Jill E Larson
- Shirley Ryan AbilityLab, Chicago, Illinois
- Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | | | - Tasos Karakostas
- Shirley Ryan AbilityLab, Chicago, Illinois
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Neeraj M Patel
- Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Vineeta T Swaroop
- Shirley Ryan AbilityLab, Chicago, Illinois
- Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Sudarshan Dayanidhi
- Shirley Ryan AbilityLab, Chicago, Illinois
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
42
|
Samakkarnthai P, Saul D, Zhang L, Aversa Z, Doolittle ML, Sfeir JG, Kaur J, Atkinson EJ, Edwards JR, Russell RGG, Pignolo RJ, Kirkland JL, Tchkonia T, Niedernhofer LJ, Monroe DG, LeBrasseur NK, Farr JN, Robbins PD, Khosla S. In vitro and in vivo effects of zoledronate on senescence and senescence-associated secretory phenotype markers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.23.529777. [PMID: 36865244 PMCID: PMC9980119 DOI: 10.1101/2023.02.23.529777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
In addition to reducing fracture risk, zoledronate has been found in some studies to decrease mortality in humans and extend lifespan and healthspan in animals. Because senescent cells accumulate with aging and contribute to multiple co-morbidities, the non-skeletal actions of zoledronate could be due to senolytic (killing of senescent cells) or senomorphic (inhibition of the secretion of the senescence-associated secretory phenotype [SASP]) actions. To test this, we first performed in vitro senescence assays using human lung fibroblasts and DNA repair-deficient mouse embryonic fibroblasts, which demonstrated that zoledronate killed senescent cells with minimal effects on non-senescent cells. Next, in aged mice treated with zoledronate or vehicle for 8 weeks, zoledronate significantly reduced circulating SASP factors, including CCL7, IL-1β, TNFRSF1A, and TGFβ1 and improved grip strength. Analysis of publicly available RNAseq data from CD115+ (CSF1R/c-fms+) pre-osteoclastic cells isolated from mice treated with zoledronate demonstrated a significant downregulation of senescence/SASP genes (SenMayo). To establish that these cells are potential senolytic/senomorphic targets of zoledronate, we used single cell proteomic analysis (cytometry by time of flight [CyTOF]) and demonstrated that zoledronate significantly reduced the number of pre-osteoclastic (CD115+/CD3e-/Ly6G-/CD45R-) cells and decreased protein levels of p16, p21, and SASP markers in these cells without affecting other immune cell populations. Collectively, our findings demonstrate that zoledronate has senolytic effects in vitro and modulates senescence/SASP biomarkers in vivo . These data point to the need for additional studies testing zoledronate and/or other bisphosphonate derivatives for senotherapeutic efficacy.
Collapse
|
43
|
Monteyne AJ, Coelho MOC, Murton AJ, Abdelrahman DR, Blackwell JR, Koscien CP, Knapp KM, Fulford J, Finnigan TJA, Dirks ML, Stephens FB, Wall BT. Vegan and Omnivorous High Protein Diets Support Comparable Daily Myofibrillar Protein Synthesis Rates and Skeletal Muscle Hypertrophy in Young Adults. J Nutr 2023:S0022-3166(23)12680-0. [PMID: 36822394 DOI: 10.1016/j.tjnut.2023.02.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/30/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND It remains unclear whether non-animal-derived dietary protein sources (and therefore vegan diets) can support resistance training-induced skeletal muscle remodeling to the same extent as animal-derived protein sources. METHODS In Phase 1, 16 healthy young adults (m = 8, f = 8; age: 23 ± 1 y; BMI: 23 ± 1 kg/m2) completed a 3-d dietary intervention (high protein, 1.8 g·kg bm-1·d-1) where protein was derived from omnivorous (OMNI1; n = 8) or exclusively non-animal (VEG1; n = 8) sources, alongside daily unilateral leg resistance exercise. Resting and exercised daily myofibrillar protein synthesis (MyoPS) rates were assessed using deuterium oxide. In Phase 2, 22 healthy young adults (m = 11, f = 11; age: 24 ± 1 y; BMI: 23 ± 0 kg/m2) completed a 10 wk, high-volume (5 d/wk), progressive resistance exercise program while consuming an omnivorous (OMNI2; n = 12) or non-animal-derived (VEG2; n = 10) high-protein diet (∼2 g·kg bm-1·d-1). Muscle fiber cross-sectional area (CSA), whole-body lean mass (via DXA), thigh muscle volume (via MRI), muscle strength, and muscle function were determined pre, after 2 and 5 wk, and postintervention. OBJECTIVES To investigate whether a high-protein, mycoprotein-rich, non-animal-derived diet can support resistance training-induced skeletal muscle remodeling to the same extent as an isonitrogenous omnivorous diet. RESULTS Daily MyoPS rates were ∼12% higher in the exercised than in the rested leg (2.46 ± 0.27%·d-1 compared with 2.20 ± 0.33%·d-1 and 2.62 ± 0.56%·d-1 compared with 2.36 ± 0.53%·d-1 in OMNI1 and VEG1, respectively; P < 0.001) and not different between groups (P > 0.05). Resistance training increased lean mass in both groups by a similar magnitude (OMNI2 2.6 ± 1.1 kg, VEG2 3.1 ± 2.5 kg; P > 0.05). Likewise, training comparably increased thigh muscle volume (OMNI2 8.3 ± 3.6%, VEG2 8.3 ± 4.1%; P > 0.05), and muscle fiber CSA (OMNI2 33 ± 24%, VEG2 32 ± 48%; P > 0.05). Both groups increased strength (1 repetition maximum) of multiple muscle groups, to comparable degrees. CONCLUSIONS Omnivorous and vegan diets can support comparable rested and exercised daily MyoPS rates in healthy young adults consuming a high-protein diet. This translates to similar skeletal muscle adaptive responses during prolonged high-volume resistance training, irrespective of dietary protein provenance. This trial was registered at clinicaltrials.gov as NCT03572127.
Collapse
Affiliation(s)
- Alistair J Monteyne
- Department of Public Health and Sports Sciences, Nutritional Physiology Research Group, University of Exeter, Exeter, United Kingdom
| | - Mariana O C Coelho
- Department of Public Health and Sports Sciences, Nutritional Physiology Research Group, University of Exeter, Exeter, United Kingdom
| | - Andrew J Murton
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, United States; Sealy Center of Aging, University of Texas Medical Branch, Galveston, Texas, United States
| | - Doaa R Abdelrahman
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, United States; Sealy Center of Aging, University of Texas Medical Branch, Galveston, Texas, United States
| | - Jamie R Blackwell
- Department of Public Health and Sports Sciences, Nutritional Physiology Research Group, University of Exeter, Exeter, United Kingdom
| | - Christopher P Koscien
- Department of Public Health and Sports Sciences, Nutritional Physiology Research Group, University of Exeter, Exeter, United Kingdom
| | - Karen M Knapp
- College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Jonathan Fulford
- College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | | | - Marlou L Dirks
- Department of Public Health and Sports Sciences, Nutritional Physiology Research Group, University of Exeter, Exeter, United Kingdom
| | - Francis B Stephens
- Department of Public Health and Sports Sciences, Nutritional Physiology Research Group, University of Exeter, Exeter, United Kingdom
| | - Benjamin T Wall
- Department of Public Health and Sports Sciences, Nutritional Physiology Research Group, University of Exeter, Exeter, United Kingdom.
| |
Collapse
|
44
|
Abbassi-Daloii T, el Abdellaoui S, Voortman LM, Veeger TTJ, Cats D, Mei H, Meuffels DE, van Arkel E, 't Hoen PAC, Kan HE, Raz V. A transcriptome atlas of leg muscles from healthy human volunteers reveals molecular and cellular signatures associated with muscle location. eLife 2023; 12:e80500. [PMID: 36744868 PMCID: PMC9988256 DOI: 10.7554/elife.80500] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 02/03/2023] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscles support the stability and mobility of the skeleton but differ in biomechanical properties and physiological functions. The intrinsic factors that regulate muscle-specific characteristics are poorly understood. To study these, we constructed a large atlas of RNA-seq profiles from six leg muscles and two locations from one muscle, using biopsies from 20 healthy young males. We identified differential expression patterns and cellular composition across the seven tissues using three bioinformatics approaches confirmed by large-scale newly developed quantitative immune-histology procedures. With all three procedures, the muscle samples clustered into three groups congruent with their anatomical location. Concomitant with genes marking oxidative metabolism, genes marking fast- or slow-twitch myofibers differed between the three groups. The groups of muscles with higher expression of slow-twitch genes were enriched in endothelial cells and showed higher capillary content. In addition, expression profiles of Homeobox (HOX) transcription factors differed between the three groups and were confirmed by spatial RNA hybridization. We created an open-source graphical interface to explore and visualize the leg muscle atlas (https://tabbassidaloii.shinyapps.io/muscleAtlasShinyApp/). Our study reveals the molecular specialization of human leg muscles, and provides a novel resource to study muscle-specific molecular features, which could be linked with (patho)physiological processes.
Collapse
Affiliation(s)
| | - Salma el Abdellaoui
- Department of Human Genetics, Leiden University Medical CenterLeidenNetherlands
| | - Lenard M Voortman
- Division of Cell and Chemical Biology, Leiden University Medical CenterLeidenNetherlands
| | - Thom TJ Veeger
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical CenterLeidenNetherlands
| | - Davy Cats
- Sequencing Analysis Support Core, Leiden University Medical CenterLeidenNetherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical CenterLeidenNetherlands
| | - Duncan E Meuffels
- Orthopedic and Sport Medicine Department, Erasmus MC, University Medical Center RotterdamRotterdamNetherlands
| | | | - Peter AC 't Hoen
- Department of Human Genetics, Leiden University Medical CenterLeidenNetherlands
- Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical CenterRadboudNetherlands
| | - Hermien E Kan
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical CenterLeidenNetherlands
- Duchenne Center NetherlandsLeidenNetherlands
| | - Vered Raz
- Department of Human Genetics, Leiden University Medical CenterLeidenNetherlands
| |
Collapse
|
45
|
Dubuisson N, Davis-López de Carrizosa MA, Versele R, Selvais CM, Noel L, Van den Bergh PYD, Brichard SM, Abou-Samra M. Inhibiting the inflammasome with MCC950 counteracts muscle pyroptosis and improves Duchenne muscular dystrophy. Front Immunol 2022; 13:1049076. [PMID: 36569900 PMCID: PMC9770793 DOI: 10.3389/fimmu.2022.1049076] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is the most common inherited human myopathy. Typically, the secondary process involving severe inflammation and necrosis exacerbate disease progression. Previously, we reported that the NLRP3 inflammasome complex plays a crucial role in this disorder. Moreover, pyroptosis, a form of programmed necrotic cell death, is triggered by NLRP3 via gasdermin D (GSDMD). So far, pyroptosis has never been described either in healthy muscle or in dystrophic muscle. The aim of this study was to unravel the role of NLRP3 inflammasome in DMD and explore a potentially promising treatment with MCC950 that selectively inhibits NLRP3. Methods Four-week-old mdx mice (n=6 per group) were orally treated for 2 months with MCC950 (mdx-T), a highly potent, specific, small-molecule inhibitor of NLRP3, and compared with untreated (mdx) and wild-type (WT) mice. In vivo functional tests were carried out to measure the global force and endurance of mice. Ex vivo biochemical and molecular analyses were performed to evaluate the pathophysiology of the skeletal muscle. Finally, in vitro tests were conducted on primary cultures of DMD human myotubes. Results After MCC950 treatment, mdx mice exhibited a significant reduction of inflammation, macrophage infiltration and oxidative stress (-20 to -65%, P<0.05 vs untreated mdx). Mdx-T mice displayed considerably less myonecrosis (-54%, P<0.05 vs mdx) and fibrosis (-75%, P<0.01 vs mdx). Moreover, a more mature myofibre phenotype, characterized by larger-sized fibres and higher expression of mature myosin heavy chains 1 and 7 was observed. Mdx-T also exhibited enhanced force and resistance to fatigue (+20 to 60%, P<0.05 or less). These beneficial effects resulted from MCC950 inhibition of both active caspase-1 (-46%, P=0.075) and cleaved gasdermin D (N-GSDMD) (-42% in medium-sized-fibres, P<0.001). Finally, the anti-inflammatory action and the anti-pyroptotic effect of MCC950 were also recapitulated in DMD human myotubes. Conclusion Specific inhibition of the NLRP3 inflammasome can significantly attenuate the dystrophic phenotype. A novel finding of this study is the overactivation of GSDMD, which is hampered by MCC950. This ultimately leads to less inflammation and pyroptosis and to a better muscle maturation and function. Targeting NLRP3 might lead to an effective therapeutic approach for a better management of DMD.
Collapse
Affiliation(s)
- Nicolas Dubuisson
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Brussels, Belgium,Neuromuscular Reference Center, Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium,*Correspondence: Nicolas Dubuisson,
| | - María A. Davis-López de Carrizosa
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Brussels, Belgium,Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Romain Versele
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Camille M. Selvais
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Laurence Noel
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - P. Y. D. Van den Bergh
- Neuromuscular Reference Center, Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Sonia M. Brichard
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
46
|
Mueller M, Thompson R, Osman KL, Andel E, Dejonge C, Kington S, Stephenson Z, Hamad A, Bunyak F, Nichols NL, Lever TE. Impact of Limb Phenotype on Tongue Denervation Atrophy, Dysphagia Penetrance, and Survival Time in a Mouse Model of ALS. Dysphagia 2022; 37:1777-1795. [PMID: 35426522 PMCID: PMC9568622 DOI: 10.1007/s00455-022-10442-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 03/28/2022] [Indexed: 12/16/2022]
Abstract
Current treatments for dysphagia in ALS do not target the underlying tongue weakness and denervation atrophy that is prevalent in spinal and bulbar ALS cases. To address this clinical gap, we studied the low copy number SOD1-G93A (LCN-SOD1) mouse model of ALS to quantify the impact of limb phenotype on tongue denervation atrophy, dysphagia penetrance, and survival time in preparation for future treatment-based studies. Two male LCN-SOD1 breeders and 125 offspring were followed for limb phenotype inheritance, of which 52 (30 LCN-SOD1 and 22 wild-type/WT, both sexes) underwent characterization of dysphagia penetrance (via videofluoroscopic swallow study; VFSS) and survival time at disease end-stage (15-20% body weight loss). From these, 16 mice (8/genotype) underwent postmortem histological analysis of the genioglossus for evidence of denervation atrophy. Results revealed that both breeders displayed a mixed (hindlimb and forelimb) ALS phenotype and sired equal proportions of hindlimb vs. mixed phenotype offspring. Dysphagia penetrance was complete for mixed (100%) versus incomplete for hindlimb (64%) phenotype mice; yet survival times were similar. Regardless of limb phenotype, LCN-SOD1 mice had significantly smaller genioglossus myofibers and more centralized myonuclei compared to WT mice (p < 0.05). These biomarkers of denervation atrophy were significantly correlated with VFSS metrics (lick and swallow rates, p < 0.05) but not survival time. In conclusion, both LCN-SOD1 phenotypes had significant tongue denervation atrophy, even hindlimb phenotype mice without dysphagia. This finding recapitulates human ALS, providing robust rationale for using this preclinical model to explore targeted treatments for tongue denervation atrophy and ensuing dysphagia.
Collapse
Affiliation(s)
| | | | - Kate L. Osman
- Department of Otolaryngology – Head and Neck Surgery, University of Missouri School of Medicine, Columbia, MO, USA
| | - Ellyn Andel
- University of Missouri Students, Columbia, MO, USA
| | | | | | | | - Ali Hamad
- University of Missouri Students, Columbia, MO, USA
| | - Filiz Bunyak
- Department of Electrical Engineering and Computer Science, University of Missouri College of Engineering, Columbia, MO, USA
| | - Nicole L. Nichols
- Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, MO, USA
| | - Teresa E. Lever
- Department of Otolaryngology – Head and Neck Surgery, University of Missouri School of Medicine, Columbia, MO, USA,Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, MO, USA
| |
Collapse
|
47
|
Reinbigler M, Cosette J, Guesmia Z, Jimenez S, Fetita C, Brunet E, Stockholm D. Artificial intelligence workflow quantifying muscle features on Hematoxylin-Eosin stained sections reveals dystrophic phenotype amelioration upon treatment. Sci Rep 2022; 12:19913. [PMID: 36402802 PMCID: PMC9675753 DOI: 10.1038/s41598-022-24139-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 11/10/2022] [Indexed: 11/21/2022] Open
Abstract
Cell segmentation is a key step for a wide variety of biological investigations, especially in the context of muscle science. Currently, automated methods still struggle to perform skeletal muscle fiber quantification on Hematoxylin-Eosin (HE) stained histopathological whole slide images due to low contrast. On the other hand, the Deep Learning algorithm Cellpose offers new perspectives considering its increasing adoption for segmentation of a wide range of cells. Combining two open-source tools, Cellpose and QuPath, we developed MyoSOTHES, an automated Myofibers Segmentation wOrkflow Tuned for HE Staining. MyoSOTHES enables solving segmentation inconsistencies encountered by default Cellpose model in presence of large range size cells and provides information related to muscle Feret's diameter distribution and Centrally Nucleated Fibers, thus depicting muscle health and treatment effects. MyoSOTHES achieves high quality segmentation compared to baseline workflow with a detection F1-score increasing from 0.801 to 0.919 and a Root Mean Square Error (RMSE) on diameter improved by 31%. MyoSOTHES was validated on an animal study featuring gene transfer in [Formula: see text]-Sarcoglycanopathy, for which dose-response effect is visible and conclusions drawn are consistent with those previously published. MyoSOTHES thus paves the way for wide quantification of HE stained muscle sections and retrospective analysis of HE labeled slices used in laboratories for decades.
Collapse
Affiliation(s)
- Marie Reinbigler
- grid.508893.fTélécom SudParis, Institut Polytechnique de Paris, 91120 Palaiseau, France
| | - Jérémie Cosette
- grid.419946.70000 0004 0641 2700Généthon, 91000 Evry, France
| | - Zoheir Guesmia
- grid.419946.70000 0004 0641 2700Généthon, 91000 Evry, France ,grid.418250.a0000 0001 0308 8843Centre de Recherche en Myologie, UMR-S 974, Institut de Myologie, 75000 Paris, France
| | - Simon Jimenez
- grid.419946.70000 0004 0641 2700Généthon, 91000 Evry, France
| | - Catalin Fetita
- grid.508893.fTélécom SudParis, Institut Polytechnique de Paris, 91120 Palaiseau, France
| | - Elisabeth Brunet
- grid.508893.fTélécom SudParis, Institut Polytechnique de Paris, 91120 Palaiseau, France
| | - Daniel Stockholm
- grid.419946.70000 0004 0641 2700Généthon, 91000 Evry, France ,grid.424469.90000 0001 2195 5365École Pratique des Hautes Études, PSL University, 75000 Paris, France
| |
Collapse
|
48
|
Monceau A, Moutachi D, Lemaitre M, Garcia L, Trollet C, Furling D, Klein A, Ferry A. Dystrophin Restoration after Adeno-Associated Virus U7-Mediated Dmd Exon Skipping Is Modulated by Muscular Exercise in the Severe D2-Mdx Duchenne Muscular Dystrophy Murine Model. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1604-1618. [PMID: 36113555 DOI: 10.1016/j.ajpath.2022.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/18/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a severe neuromuscular disease caused by Dmd mutations, resulting in the absence of dystrophin in skeletal muscle, and a greater susceptibility to damage during contraction (exercise). The current study evaluated whether voluntary exercise impacts a Dmd exon skipping and muscle physiology in a severe DMD murine model. D2-mdx mice were intramuscularly injected with an adeno-associated virus (AAV) U7 snRNA to correct Dmd reading frame, and allowed to voluntary run on a wheel for 1 month. Voluntary running did not induce muscle fiber regeneration, as indicated by the percentage of centronucleated fibers, Myh3 and Myh4 expression, and maximal force production, and thus possibly did not compromise the gene therapy approach. Voluntary running did not impact the number of viral genomes and the expression of U7 and Dmd 1 month after injection of AAV-U7 injected just before exercise initiation, but reduced the amount of dystrophin in dystrophin-expressing fibers from 80% to 65% of the muscle cross-sectional area. In conclusion, voluntary running did not induce muscle damage and had no drastic detrimental effect on the AAV gene therapy exon skipping approach in a severe murine DMD model. Moreover, these results suggest considering exercise as an additional element in the design and conception of future therapeutic approaches for DMD.
Collapse
Affiliation(s)
- Alexandra Monceau
- UMRS974 INSERM, Association of Myology Institute, Myology Center of Research, UMRS974, Sorbonne Université, Paris, France
| | - Dylan Moutachi
- UMRS974 INSERM, Association of Myology Institute, Myology Center of Research, UMRS974, Sorbonne Université, Paris, France
| | | | - Luis Garcia
- U1179 INSERM, Université de Versailles Saint-Quentin-en-Yvelines, Montigny le Bretonneux, Paris, France
| | - Capucine Trollet
- UMRS974 INSERM, Association of Myology Institute, Myology Center of Research, UMRS974, Sorbonne Université, Paris, France
| | - Denis Furling
- UMRS974 INSERM, Association of Myology Institute, Myology Center of Research, UMRS974, Sorbonne Université, Paris, France
| | - Arnaud Klein
- UMRS974 INSERM, Association of Myology Institute, Myology Center of Research, UMRS974, Sorbonne Université, Paris, France
| | - Arnaud Ferry
- UMRS974 INSERM, Association of Myology Institute, Myology Center of Research, UMRS974, Sorbonne Université, Paris, France; Faculty of Science Sport, Université Paris Cité, Paris, France.
| |
Collapse
|
49
|
Khattri RB, Kim K, Anderson EM, Fazzone B, Harland KC, Hu Q, Palzkill VR, Cort TA, O'Malley KA, Berceli SA, Scali ST, Ryan TE. Metabolomic profiling reveals muscle metabolic changes following iliac arteriovenous fistula creation in mice. Am J Physiol Renal Physiol 2022; 323:F577-F589. [PMID: 36007889 PMCID: PMC9602894 DOI: 10.1152/ajprenal.00156.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 12/31/2022] Open
Abstract
End-stage kidney disease, the most advanced stage of chronic kidney disease (CKD), requires renal replacement therapy or kidney transplant to sustain life. To accomplish durable dialysis access, the creation of an arteriovenous fistula (AVF) has emerged as a preferred approach. Unfortunately, a significant proportion of patients that receive an AVF experience some form of hand dysfunction; however, the mechanisms underlying these side effects are not understood. In this study, we used nuclear magnetic resonance spectroscopy to investigate the muscle metabolome following iliac AVF placement in mice with CKD. To induce CKD, C57BL6J mice were fed an adenine-supplemented diet for 3 wk and then randomized to receive AVF or sham surgery. Two weeks following surgery, the quadriceps muscles were rapidly dissected and snap frozen for metabolite extraction and subsequent nuclear magnetic resonance analysis. Principal component analysis demonstrated clear separation between groups, confirming a unique metabolome in mice that received an AVF. AVF creation resulted in reduced levels of creatine, ATP, and AMP as well as increased levels of IMP and several tricarboxylic acid cycle metabolites suggesting profound energetic stress. Pearson correlation and multiple linear regression analyses identified several metabolites that were strongly linked to measures of limb function (grip strength, gait speed, and mitochondrial respiration). In summary, AVF creation generates a unique metabolome profile in the distal skeletal muscle indicative of an energetic crisis and myosteatosis.NEW & NOTEWORTHY Creation of an arteriovenous fistula (AVF) is the preferred approach for dialysis access, but some patients experience hand dysfunction after AVF creation. In this study, we provide a detailed metabolomic analysis of the limb muscle in a murine model of AVF. AVF creation resulted in metabolite changes associated with an energetic crisis and myosteatosis that associated with limb function.
Collapse
Affiliation(s)
- Ram B Khattri
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Kyoungrae Kim
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Erik M Anderson
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, Florida
- Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida
| | - Brian Fazzone
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, Florida
- Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida
| | - Kenneth C Harland
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, Florida
- Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida
| | - Qiongyao Hu
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, Florida
- Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida
| | - Victoria R Palzkill
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Tomas A Cort
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Kerri A O'Malley
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, Florida
- Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida
| | - Scott A Berceli
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, Florida
- Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida
| | - Salvatore T Scali
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, Florida
- Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida
| | - Terence E Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
- Center for Exercise Science, University of Florida, Gainesville, Florida
| |
Collapse
|
50
|
Anderson EM, Kim K, Fazzone BJ, Harland KC, Hu Q, Salyers Z, Palzkill VR, Cort TA, Kunz EM, Martin AJ, Neal D, O’Malley KA, Berceli SA, Ryan TE, Scali ST. Influences of renal insufficiency and ischemia on mitochondrial bioenergetics and limb dysfunction in a novel murine iliac arteriovenous fistula model. JVS Vasc Sci 2022; 3:345-362. [PMID: 36439698 PMCID: PMC9692039 DOI: 10.1016/j.jvssci.2022.10.001] [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: 01/24/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022] Open
Abstract
Objective Hand disability after hemodialysis access surgery has been common yet has remained poorly understood. Arteriovenous fistula (AVF) hemodynamic perturbations have not reliably correlated with the observed measures of hand function. Chronic kidney disease (CKD) is known to precipitate myopathy; however, the interactive influences of renal insufficiency and ischemia on limb outcomes have remained unknown. We hypothesized that CKD would contribute to access-related hand dysfunction via altered mitochondrial bioenergetics. Using a novel murine AVF model, we sought to characterize the skeletal muscle outcomes in mice with and without renal insufficiency. Methods Male, 8-week-old C57BL/6J mice were fed either an adenine-supplemented diet to induce renal insufficiency (CKD) or a casein-based control chow (CON). After 2 weeks of dietary intervention, the mice were randomly assigned to undergo iliac AVF surgery (n = 12/group) or a sham operation (n = 5/group). Measurements of aortoiliac hemodynamics, hindlimb perfusion, and hindlimb motor function were collected for 2 weeks. The mice were sacrificed on postoperative day 14 to assess skeletal muscle histopathologic features and mitochondrial function. To assess the late outcome trends, 20 additional mice had undergone CKD induction and sham (n = 5) or AVF (n = 15) surgery and followed up for 6 weeks postoperatively before sacrifice. Results The adenine-fed mice had had a significantly reduced glomerular filtration rate and elevated blood urea nitrogen, confirming the presence of CKD. The sham mice had a 100% survival rate and AVF cohorts an 82.1% survival rate with an 84.4% AVF patency rate. The aorta and inferior vena cava velocity measurements and the vessel diameter had increased after AVF creation (P < .0001 vs sham). The AVF groups had had a 78.4% deficit in paw perfusion compared with the contralateral limb after surgery (P < .0001 vs sham). Mitochondrial function was influenced by the presence of CKD. The respiratory capacity of the CKD-sham mice (8443 ± 1509 pmol/s/mg at maximal energy demand) was impaired compared with that of the CON-sham mice (12,870 ± 1203 pmol/s/mg; P = .0001). However, this difference was muted after AVF creation (CKD-AVF, 4478 ± 3685 pmol/s/mg; CON-AVF, 5407 ± 3582 pmol/s/mg; P = .198). The AVF cohorts had had impairments in grip strength (vs sham; P < .0001) and gait (vs sham; P = .012). However, the presence of CKD did not significantly alter the measurements of gross muscle function. The paw perfusion deficits had persisted 6 weeks postoperatively for the AVF mice (P < .0001 vs sham); however, the myopathy had resolved (grip strength, P = .092 vs sham; mitochondrial respiration, P = .108 vs sham). Conclusions CKD and AVF-induced distal limb ischemia both impaired skeletal muscle mitochondrial function. Renal insufficiency was associated with a baseline myopathy that was exacerbated by the acute ischemic injury resulting from AVF creation. However, ischemia was the primary driver of the observed phenotype of gross motor impairment. This model reliably reproduced the local and systemic influences that contribute to access-related hand dysfunction and provides a platform for further mechanistic and therapeutic investigation.
Collapse
Affiliation(s)
- Erik M. Anderson
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| | - Kyoungrae Kim
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL
| | - Brian J. Fazzone
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| | - Kenneth C. Harland
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| | - Qiongyao Hu
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| | - Zach Salyers
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL
| | - Victoria R. Palzkill
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL
| | - Tomas A. Cort
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL
| | - Eric M. Kunz
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL
| | - Andrew J. Martin
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| | - Dan Neal
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
| | - Kerri A. O’Malley
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| | - Scott A. Berceli
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL
| | - Salvatore T. Scali
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL
- Malcolm Randall Veterans Affairs Medical Center, Gainesville, FL
| |
Collapse
|