1
|
Cheung CV, Atube KJ, Colonna NA, Carter GJ, Marchena T, McCarthy S, Krusen KE, McCain RS, Frizzell N, Gower RM. A microparticle delivery system for extended release of all-trans retinoic acid and its impact on macrophage insulin-like growth factor 1 release and myotube formation. Int J Pharm 2024; 666:124821. [PMID: 39396656 DOI: 10.1016/j.ijpharm.2024.124821] [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: 06/24/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/15/2024]
Abstract
Muscle atrophy secondary to disuse, aging, or illness increases the risk of injury, prolonged recovery, and permanent disability. The recovery process involves macrophages and their secretions, such as insulin-like growth factor 1 (IGF-1), which direct muscle to regenerate and grow. Retinoic acid receptor (RAR) activation in macrophages increases IGF-1 expression and can be achieved with all-trans retinoic acid (ATRA). However, poor bioavailability limits its clinical application. Thus, we encapsulated ATRA into poly(lactide-co-glycolide) microparticles (ATRA-PLG) to maintain bioactivity and achieve extended release. ATRA-PLG induces IGF-1 release by RAW 264.7 macrophages, and conditioned media from these cells enhances C2C12 myotube formation through IGF-1. Additionally, ATRA released from ATRA-PLG enhances myotube formation in the absence of macrophages. Toward clinical translation, we envision that ATRA-PLG will be injected in the vicinity of debilitated muscle where it can be taken up by macrophages and induce IGF-1 release over a predetermined therapeutic window. Along these lines, we demonstrate that ATRA-PLG microparticles are readily taken up by bone marrow-derived macrophages and reside within the cytosol for at least 12 days with no toxicity. Interestingly, ATRA-PLG induced IGF-1 secretion by thioglycolate-elicited macrophages, but not bone marrow derived macrophages. We found that the RAR isoforms present in lysate differed between the macrophages studied, which could explain the different IGF-1 responses to ATRA. Given that ATRA-PLG enhances myotube formation directly (through ATRA) and indirectly (through macrophage IGF-1) this study supports the further testing of this promising pharmaceutical using rodent models of muscle regeneration and growth.
Collapse
Affiliation(s)
- Candice V Cheung
- Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208, USA
| | - Kidochukwu J Atube
- Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA
| | - Nicholas A Colonna
- Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208, USA
| | - Griffin J Carter
- Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208, USA
| | - Tristan Marchena
- Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208, USA
| | - Samantha McCarthy
- Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208, USA
| | - Kelsey E Krusen
- Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208, USA
| | - Richard S McCain
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Norma Frizzell
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - R Michael Gower
- Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208, USA; Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA; Veterans Affairs Medical Center, Columbia, SC 29209, USA.
| |
Collapse
|
2
|
Alonso-Puyo J, Izagirre-Fernandez O, Crende O, Valdivia A, García-Gallastegui P, Sanz B. Experimental models as a tool for research on sarcopenia: A narrative review. Ageing Res Rev 2024; 101:102534. [PMID: 39369798 DOI: 10.1016/j.arr.2024.102534] [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: 02/27/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Sarcopenia is a musculoskeletal disorder related to muscle mass and function; as the worldwide population ages, its growing prevalence means a decline in quality of life and an increased burden for public health systems. As sarcopenia is a reversible condition, its early diagnosis is of utmost importance. Consensus definitions and diagnosis protocols for sarcopenia have been evolving for a long time, and the identification of molecular pathways subjacent to sarcopenia is a growing research area. The use of liquid biopsies to identify circulating molecules does not provide information about specific regulatory pathways or biomarkers in relevant tissue, and the use of skeletal muscle biopsies from older people has many limitations. Complementary tools are therefore necessary to advance the knowledge of relevant molecular aspects. The development of experimental models, such as animal, cellular, or bioengineered tissue, together with knock-in or knock-out strategies, could therefore be of great interest. This narrative review will explore experimental models of healthy muscle and aged muscle cells as a tool for research on sarcopenia. We will summarize the literature and present relevant experimental models in terms of their advantages and disadvantages. All of the presented approaches could potentially contribute to the accurate and early diagnosis, follow-up, and possible treatment of sarcopenia.
Collapse
Affiliation(s)
- Janire Alonso-Puyo
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain
| | - Oihane Izagirre-Fernandez
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain
| | - Olatz Crende
- Cell Biology and Histology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain
| | - Asier Valdivia
- Cell Biology and Histology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain
| | - Patricia García-Gallastegui
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain.
| | - Begoña Sanz
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain; Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia 48903, Spain.
| |
Collapse
|
3
|
Xu YY, Chen T, Ding H, Chen Q, Fan QL. Melatonin inhibits circadian gene DEC1 and TLR2/MyD88/NF-κB signaling pathway to alleviate renal injury in type 2 diabetic mice. Acta Diabetol 2024; 61:1455-1474. [PMID: 38896283 DOI: 10.1007/s00592-024-02312-2] [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: 02/07/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Diabetic Kidney Disease (DKD) is a complex disease associated with circadian rhythm and biological clock regulation disorders. Melatonin (MT) is considered a hormone with renal protective effects, but its mechanism of action in DKD is unclear. METHODS We used the GSE151325 dataset from the GEO database for differential gene analysis and further explored related genes and pathways through GO and KEGG analysis and PPI network analysis. Additionally, this study used a type 2 diabetes db/db mouse model and investigated the role of melatonin in DKD and its relationship with clock genes through immunohistochemistry, Western blot, real-time PCR, ELISA, chromatin immunoprecipitation (ChIP), dual-luciferase reporter technology, and liposome transfection technology to study DEC1 siRNA. RESULTS Bioinformatics analysis revealed the central position of clock genes such as CLOCK, DEC1, Bhlhe41, CRY1, and RORB in DKD. Their interaction with key inflammatory regulators may reveal melatonin's potential mechanism in treating diabetic kidney disease. Further experimental results showed that melatonin significantly improved the renal pathological changes in db/db mice, reduced body weight and blood sugar, regulated clock genes in renal tissue, and downregulated the TLR2/MyD88/NF-κB signaling pathway. We found that the transcription factor DEC1 can bind to the TLR2 promoter and activate its transcription, while CLOCK's effect is unclear. Liposome transfection experiments further confirmed the effect of DEC1 on the TLR2/MyD88/NF-κB signaling pathway. CONCLUSION Melatonin shows significant renal protective effects by regulating clock genes and downregulating the TLR2/MyD88/NF-κB signaling pathway. The transcription factor DEC1 may become a key regulatory factor for renal inflammation and fibrosis by activating TLR2 promoter transcription. These findings provide new perspectives and directions for the potential application of melatonin in DKD treatment.
Collapse
Affiliation(s)
- Yan-Yan Xu
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Tong Chen
- Department of Nephrology, Shenyang Seventh People's Hospital, Shenyang, China
| | - Hong Ding
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Qiong Chen
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200082, China.
| | - Qiu-Ling Fan
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200082, China.
| |
Collapse
|
4
|
Boeno FP, Roesch LFW, Efron PA, Laitano O. Proteomic Profiling of Hindlimb Skeletal Muscle Disuse in a Murine Model of Sepsis. Crit Care Explor 2024; 6:e1144. [PMID: 39162648 PMCID: PMC11338252 DOI: 10.1097/cce.0000000000001144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024] Open
Abstract
CONTEXT Sepsis leads to multiple organ dysfunction and negatively impacts patient outcomes. Skeletal muscle disuse is a significant comorbidity in septic patients during their ICU stay due to prolonged immobilization. HYPOTHESIS Combination of sepsis and muscle disuse will promote a unique proteomic signature in skeletal muscle in comparison to disuse and sepsis separately. METHODS AND MODELS Following cecal ligation and puncture (CLP) or Sham surgeries, mice were subjected to hindlimb suspension (HLS) or maintained normal ambulation (NA). Tibialis anterior muscles from 24 C57BL6/J male mice were harvested for proteomic analysis. Proteomic profiles were assessed using nano-liquid chromatography with tandem mass spectrometry, followed by data analysis including Partial Least Squares Discriminant Analysis (PLS-DA), to compare the differential protein expression across groups. RESULTS A total of 2876 differentially expressed proteins were identified, with marked differences between groups. In mice subjected to CLP and HLS combined, there was a distinctive proteomic signature characterized by a significant decrease in the expression of proteins involved in mitochondrial function and muscle metabolism, alongside a marked increase in proteins related to muscle degradation pathways. The PLS-DA demonstrated a clear separation among experimental groups, highlighting the unique profile of the CLP/HLS group. This suggests an important interaction between sepsis-induced inflammation and disuse atrophy mechanisms in sepsis-induced myopathy. INTERPRETATIONS AND CONCLUSIONS Our findings reveal a complex proteomic landscape in skeletal muscle exposed to sepsis and disuse, consistent with an exacerbation of muscle protein degradation under these combined stressors. The identified proteins and their roles in cellular stress responses and muscle pathology provide potential targets for intervention to mitigate muscle dysfunction in septic conditions, highlighting the importance of addressing both sepsis and disuse concurrently in clinical and experimental settings.
Collapse
Affiliation(s)
- Franccesco P. Boeno
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL
| | - Luiz Fernando W. Roesch
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL
| | - Philip A. Efron
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL
| | - Orlando Laitano
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL
| |
Collapse
|
5
|
Karimi R, Yanovich A, Elbarbry F, Cleven A. Adaptive Effects of Endocrine Hormones on Metabolism of Macronutrients during Fasting and Starvation: A Scoping Review. Metabolites 2024; 14:336. [PMID: 38921471 PMCID: PMC11205672 DOI: 10.3390/metabo14060336] [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: 03/29/2024] [Revised: 06/08/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Food deprivation can occur for different reasons. Fasting (<24 h duration) occurs to meet religious or well-being goals. Starvation (>1-day duration) occurs when there is intentional (hunger strike or treatment of a medical condition) or unintentional (anorexia nervosa, drought, epidemic famine, war, or natural disaster) food deprivation. A scoping review was undertaken using the PubMed database to explore 1805 abstracts and review 88 eligible full-text articles to explore the adaptive relationships that emerge between cortisol, insulin, glucagon, and thyroid hormones on the metabolic pathways of macronutrients in humans during fasting and starvation. The collected data indicate that fasting and starvation prime the human body to increase cortisol levels and decrease the insulin/glucagon ratio and triiodothyronine (T3) levels. During fasting, increased levels of cortisol and a decreased insulin/glucagon ratio enhance glycogenolysis and reduce the peripheral uptake of glucose and glycogenesis, whereas decreased T3 levels potentially reduce glycogenolysis. During starvation, increased levels of cortisol and a decreased insulin/glucagon ratio enhance lipolysis, proteolysis, fatty acid and amino acid oxidation, ketogenesis, and ureagenesis, and decreased T3 levels reduce thermogenesis. We present a potential crosstalk between T3 and the above hormones, including between T3 and leptin, to extend their adaptive roles in the metabolism of endogenous macronutrients during food deprivation.
Collapse
Affiliation(s)
- Reza Karimi
- Pacific University School of Pharmacy, 222 SE 8th Avenue, HPC-Ste 451, Hillsboro, OR 97123, USA; (A.Y.); (F.E.); (A.C.)
| | | | | | | |
Collapse
|
6
|
Jara TC, Park K, Vahmani P, Van Eenennaam AL, Smith LR, Denicol AC. Stem cell-based strategies and challenges for production of cultivated meat. NATURE FOOD 2023; 4:841-853. [PMID: 37845547 DOI: 10.1038/s43016-023-00857-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 09/05/2023] [Indexed: 10/18/2023]
Abstract
Cultivated meat scale-up and industrial production will require multiple stable cell lines from different species to recreate the organoleptic and nutritional properties of meat from livestock. In this Review, we explore the potential of stem cells to create the major cellular components of cultivated meat. By using developments in the fields of tissue engineering and biomedicine, we explore the advantages and disadvantages of strategies involving primary adult and pluripotent stem cells for generating cell sources that can be grown at scale. These myogenic, adipogenic or extracellular matrix-producing adult stem cells as well as embryonic or inducible pluripotent stem cells are discussed for their proliferative and differentiation capacity, necessary for cultivated meat. We examine the challenges for industrial scale-up, including differentiation and culture protocols, as well as genetic modification options for stem cell immortalization and controlled differentiation. Finally, we discuss stem cell-related safety and regulatory challenges for bringing cultivated meat to the marketplace.
Collapse
Affiliation(s)
- T C Jara
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - K Park
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - P Vahmani
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - A L Van Eenennaam
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - L R Smith
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA, USA.
| | - A C Denicol
- Department of Animal Science, University of California Davis, Davis, CA, USA
| |
Collapse
|
7
|
Roberts MD, McCarthy JJ, Hornberger TA, Phillips SM, Mackey AL, Nader GA, Boppart MD, Kavazis AN, Reidy PT, Ogasawara R, Libardi CA, Ugrinowitsch C, Booth FW, Esser KA. Mechanisms of mechanical overload-induced skeletal muscle hypertrophy: current understanding and future directions. Physiol Rev 2023; 103:2679-2757. [PMID: 37382939 PMCID: PMC10625844 DOI: 10.1152/physrev.00039.2022] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
Mechanisms underlying mechanical overload-induced skeletal muscle hypertrophy have been extensively researched since the landmark report by Morpurgo (1897) of "work-induced hypertrophy" in dogs that were treadmill trained. Much of the preclinical rodent and human resistance training research to date supports that involved mechanisms include enhanced mammalian/mechanistic target of rapamycin complex 1 (mTORC1) signaling, an expansion in translational capacity through ribosome biogenesis, increased satellite cell abundance and myonuclear accretion, and postexercise elevations in muscle protein synthesis rates. However, several lines of past and emerging evidence suggest that additional mechanisms that feed into or are independent of these processes are also involved. This review first provides a historical account of how mechanistic research into skeletal muscle hypertrophy has progressed. A comprehensive list of mechanisms associated with skeletal muscle hypertrophy is then outlined, and areas of disagreement involving these mechanisms are presented. Finally, future research directions involving many of the discussed mechanisms are proposed.
Collapse
Affiliation(s)
- Michael D Roberts
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Troy A Hornberger
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital-Bispebjerg and Frederiksberg, and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gustavo A Nader
- Department of Kinesiology and Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States
| | - Marni D Boppart
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Andreas N Kavazis
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Paul T Reidy
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, United States
| | - Riki Ogasawara
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Cleiton A Libardi
- MUSCULAB-Laboratory of Neuromuscular Adaptations to Resistance Training, Department of Physical Education, Federal University of São Carlos, São Carlos, Brazil
| | - Carlos Ugrinowitsch
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Karyn A Esser
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
8
|
Lang CH. IMPORTANCE OF THE INNATE IMMUNE RESPONSE IN SKELETAL MUSCLE TO SEPSIS-INDUCED ALTERATIONS IN PROTEIN BALANCE. Shock 2023; 59:214-223. [PMID: 36730901 PMCID: PMC9957944 DOI: 10.1097/shk.0000000000002029] [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] [Indexed: 02/04/2023]
Abstract
ABSTRACT There is growing appreciation that skeletal muscle is a fully functional component of the body's innate immune system with the potential to actively participate in the host response to invading bacteria as opposed to being a passive target. In this regard, skeletal muscle in general and myocytes specifically possess an afferent limb that recognizes a wide variety of host pathogens via their interaction with multiple classes of cell membrane-bound and intracellular receptors, including toll-like receptors, cytokine receptors, NOD-like receptors, and the NLRP inflammasome. The efferent limb of the innate immune system in muscle is equally robust and with an increased synthesis and secretion of a variety of myocyte-derived cytokines (i.e., myokines), including TNF-α, IL-1, IL-6, and NO as well as multiple chemokines in response to appropriate stimulation. Herein, the current narrative review focuses primarily on the immune response of myocytes per se as opposed to other cell types within whole muscle. Moreover, because there are important differences, this review focuses specifically on systemic infection and inflammation as opposed to the response of muscle to direct injury and various types of muscular dystrophies. To date, however, there are few definitive muscle-specific studies that are necessary to directly address the relative importance of muscle-derived immune activation as a contributor to either the systemic immune response or the local immune microenvironment within muscle during sepsis and the resultant downstream metabolic disturbances.
Collapse
Affiliation(s)
- Charles H Lang
- Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
9
|
Jengelley DHA, Wang M, Narasimhan A, Rupert JE, Young AR, Zhong X, Horan DJ, Robling AG, Koniaris LG, Zimmers TA. Exogenous Oncostatin M induces Cardiac Dysfunction, Musculoskeletal Atrophy, and Fibrosis. Cytokine 2022; 159:155972. [PMID: 36054964 PMCID: PMC10468097 DOI: 10.1016/j.cyto.2022.155972] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 01/21/2023]
Abstract
Musculoskeletal diseases such as muscular dystrophy, cachexia, osteoarthritis, and rheumatoid arthritis impair overall physical health and reduce survival. Patients suffer from pain, dysfunction, and dysmobility due to inflammation and fibrosis in bones, muscles, and joints, both locally and systemically. The Interleukin-6 (IL-6) family of cytokines, most notably IL-6, is implicated in musculoskeletal disorders and cachexia. Here we show elevated circulating levels of OSM in murine pancreatic cancer cachexia and evaluate the effects of the IL-6 family member, Oncostatin M (OSM), on muscle and bone using adeno-associated virus (AAV) mediated over-expression of murine OSM in wildtype and IL-6 deficient mice. Initial studies with high titer AAV-OSM injection yielded high circulating OSM and IL-6, thrombocytosis, inflammation, and 60% mortality without muscle loss within 4 days. Subsequently, to mimic OSM levels in cachexia, a lower titer of AAV-OSM was used in wildtype and Il6 null mice, observing effects out to 4 weeks and 12 weeks. AAV-OSM caused muscle atrophy and fibrosis in the gastrocnemius, tibialis anterior, and quadriceps of the injected limb, but these effects were not observed on the non-injected side. In contrast, OSM induced both local and distant trabecular bone loss as shown by reduced bone volume, trabecular number, and thickness, and increased trabecular separation. OSM caused cardiac dysfunction including reduced ejection fraction and reduced fractional shortening. RNA-sequencing of cardiac muscle revealed upregulation of genes related to inflammation and fibrosis. None of these effects were different in IL-6 knockout mice. Thus, OSM induces local muscle atrophy, systemic bone loss, tissue fibrosis, and cardiac dysfunction independently of IL-6, suggesting a role for OSM in musculoskeletal conditions with these characteristics, including cancer cachexia.
Collapse
Affiliation(s)
- Daenique H A Jengelley
- Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA
| | - Meijing Wang
- Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA
| | - Ashok Narasimhan
- Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA
| | - Joseph E Rupert
- Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA
| | - Andrew R Young
- Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiaoling Zhong
- Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA; Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA
| | - Daniel J Horan
- Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA
| | - Alexander G Robling
- Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA
| | - Leonidas G Koniaris
- Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA; Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA
| | - Teresa A Zimmers
- Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Otolaryngology, Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA; Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA.
| |
Collapse
|
10
|
Choi Y, Morlino G, Toboso-Navasa A, Hopf R, Pramotton FM, Bigot A, Taddei A, Cesarovic N, Falk V, Mazza E, Giampietro C. A novel bistable device to study mechanosensitive cell responses to instantaneous stretch. BIOMATERIALS ADVANCES 2022; 141:213134. [PMID: 36191540 DOI: 10.1016/j.bioadv.2022.213134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/17/2022] [Accepted: 09/25/2022] [Indexed: 06/16/2023]
Abstract
The behavior of cells and tissues in vivo is determined by the integration of multiple biochemical and mechanical signals. Of the mechanical signals, stretch has been studied for decades and shown to contribute to pathophysiological processes. Several different stretch devices have been developed for in vitro investigations of cell stretch. In this work, we describe a new 3D-printed uniaxial stretching device for studying cell response to rapid deformation. The device is a bistable compliant mechanism holding two equilibrium states-an unstretched and stretched configuration-without the need of an external actuator. Furthermore, it allows multiple simultaneous measurements of different levels of stretch on a single substrate and is compatible with standard immunofluorescence imaging of fixed cells as well as live-cell imaging. To demonstrate the effectiveness of the device to stretch cells, a test case using aligned myotubes is presented. Leveraging material area changes associated with deformation of the substrate, changes in nuclei density provided evidence of affine deformation between cells and substrate. Furthermore, intranuclear deformations were also assessed and shown to deform non-affinely. As a proof-of-principle of the use of the device for mechanobiological studies, we uniaxially stretched aligned healthy and dystrophic myotubes that displayed different passive mechanical responses, consistent with previous literature in the field. We also identified a new feature in the mechanoresponse of dystrophic myotubes, which is of potential interest for identifying the diseased cells based on a quick mechanical readout. While some applications of the device for elucidating passive mechanical responses are demonstrated, the simplicity of the device allows it to be potentially used for other modes of deformation with little modifications.
Collapse
Affiliation(s)
- Young Choi
- Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland
| | | | | | - Raoul Hopf
- Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland; Swiss Federal Laboratories for Materials Science and Technology (EMPA), Dübendorf 8600, Switzerland; Senecell AG, Zurich 8057, Switzerland
| | - Francesca Michela Pramotton
- Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland; Swiss Federal Laboratories for Materials Science and Technology (EMPA), Dübendorf 8600, Switzerland
| | - Anne Bigot
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | | | - Nikola Cesarovic
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland; Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Volkmar Falk
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland; Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Edoardo Mazza
- Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland; Swiss Federal Laboratories for Materials Science and Technology (EMPA), Dübendorf 8600, Switzerland.
| | - Costanza Giampietro
- Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland; Swiss Federal Laboratories for Materials Science and Technology (EMPA), Dübendorf 8600, Switzerland; Senecell AG, Zurich 8057, Switzerland.
| |
Collapse
|
11
|
Ohlendieck K, Swandulla D. Complexity of skeletal muscle degeneration: multi-systems pathophysiology and organ crosstalk in dystrophinopathy. Pflugers Arch 2021; 473:1813-1839. [PMID: 34553265 PMCID: PMC8599371 DOI: 10.1007/s00424-021-02623-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy is a highly progressive muscle wasting disorder due to primary abnormalities in one of the largest genes in the human genome, the DMD gene, which encodes various tissue-specific isoforms of the protein dystrophin. Although dystrophinopathies are classified as primary neuromuscular disorders, the body-wide abnormalities that are associated with this disorder and the occurrence of organ crosstalk suggest that a multi-systems pathophysiological view should be taken for a better overall understanding of the complex aetiology of X-linked muscular dystrophy. This article reviews the molecular and cellular effects of deficiency in dystrophin isoforms in relation to voluntary striated muscles, the cardio-respiratory system, the kidney, the liver, the gastrointestinal tract, the nervous system and the immune system. Based on the establishment of comprehensive biomarker signatures of X-linked muscular dystrophy using large-scale screening of both patient specimens and genetic animal models, this article also discusses the potential usefulness of novel disease markers for more inclusive approaches to differential diagnosis, prognosis and therapy monitoring that also take into account multi-systems aspects of dystrophinopathy. Current therapeutic approaches to combat muscular dystrophy are summarised.
Collapse
Affiliation(s)
- Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Co. Kildare, Maynooth, W23F2H6, Ireland.
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Co. Kildare, Maynooth, W23F2H6, Ireland.
| | - Dieter Swandulla
- Institute of Physiology, University of Bonn, 53115, Bonn, Germany.
| |
Collapse
|
12
|
What's New in Shock, May 2020? Shock 2020; 53:525-527. [PMID: 32287153 DOI: 10.1097/shk.0000000000001520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|