1
|
Gunasekaran M, Littel HR, Wells NM, Turner J, Campos G, Venigalla S, Estrella EA, Ghosh PS, Daugherty AL, Stafki SA, Kunkel LM, Foley AR, Donkervoort S, Bönnemann CG, Toledo-Bravo de Laguna L, Nascimento A, Natera-de Benito D, Draper I, Bruels CC, Pacak CA, Kang PB. Effects of HMG CoA reductase (HMGCR) deficiency on skeletal muscle development. FEBS J 2025. [PMID: 39823152 DOI: 10.1111/febs.17406] [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: 05/09/2024] [Revised: 10/18/2024] [Accepted: 12/17/2024] [Indexed: 01/19/2025]
Abstract
Pathogenic variants in HMGCR were recently linked to a limb-girdle muscular dystrophy (LGMD) phenotype. The protein product HMG CoA reductase (HMGCR) catalyzes a key component of the cholesterol synthesis pathway. The two other muscle diseases associated with HMGCR, statin-associated myopathy (SAM) and autoimmune anti-HMGCR myopathy, are not inherited in a Mendelian pattern. Statins inhibit HMGCR activity to generate their cholesterol-lowering effects and are known to cause multiple types of adverse effects on skeletal muscle, while the antibodies associated with anti-HMGCR myopathy specifically target this enzyme. The mechanism linking pathogenic variants in HMGCR with skeletal muscle dysfunction is unclear. We knocked down Hmgcr in mouse skeletal myoblasts, knocked down hmgcr in Drosophila, and expressed three pathogenic HMGCR variants (c.1327C>T, p.Arg443Trp; c.1522_1524delTCT, p.Ser508del; and c.1621G>A, p.Ala541Thr) in Hmgcr knockdown mouse myoblasts. Hmgcr deficiency was associated with decreased proliferation, increased apoptosis, and impaired myotube fusion. Transcriptome sequencing of Hmgcr knockdown versus control myoblasts revealed differential expression involving mitochondrial function, with corresponding differences in cellular oxygen consumption rates. Both ubiquitous and muscle-specific knockdown of hmgcr in Drosophila led to lethality. Overexpression of reference HMGCR cDNA rescued myotube fusion in knockdown cells, whereas overexpression of the pathogenic variants of HMGCR cDNA did not. These results suggest that the three HMGCR-related muscle diseases share disease mechanisms related to skeletal muscle development.
Collapse
Affiliation(s)
- Mekala Gunasekaran
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Hannah R Littel
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Natalya M Wells
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Johnnie Turner
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Gloriana Campos
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Sree Venigalla
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Elicia A Estrella
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Partha S Ghosh
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Audrey L Daugherty
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Seth A Stafki
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Louis M Kunkel
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - A Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | | | - Andres Nascimento
- Neuromuscular Unit, Department of Neurology, Hospital Sant Joan de Déu, Barcelona, Spain
- Applied Research in Neuromuscular Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Daniel Natera-de Benito
- Neuromuscular Unit, Department of Neurology, Hospital Sant Joan de Déu, Barcelona, Spain
- Applied Research in Neuromuscular Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Isabelle Draper
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Christine C Bruels
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Christina A Pacak
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Peter B Kang
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
2
|
Masiero C, Aresi C, Forlino A, Tonelli F. Zebrafish Models for Skeletal and Extraskeletal Osteogenesis Imperfecta Features: Unveiling Pathophysiology and Paving the Way for Drug Discovery. Calcif Tissue Int 2024; 115:931-959. [PMID: 39320469 PMCID: PMC11607041 DOI: 10.1007/s00223-024-01282-5] [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: 07/10/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024]
Abstract
In the last decades, the easy genetic manipulation, the external fertilization, the high percentage of homology with human genes and the reduced husbandry costs compared to rodents, made zebrafish a valid model for studying human diseases and for developing new therapeutical strategies. Since zebrafish shares with mammals the same bone cells and ossification types, it became widely used to dissect mechanisms and possible new therapeutic approaches in the field of common and rare bone diseases, such as osteoporosis and osteogenesis imperfecta (OI), respectively. OI is a heritable skeletal disorder caused by defects in gene encoding collagen I or proteins/enzymes necessary for collagen I synthesis and secretion. Nevertheless, OI patients can be also characterized by extraskeletal manifestations such as dentinogenesis imperfecta, muscle weakness, cardiac valve and pulmonary abnormalities and skin laxity. In this review, we provide an overview of the available zebrafish models for both dominant and recessive forms of OI. An updated description of all the main similarities and differences between zebrafish and mammal skeleton, muscle, heart and skin, will be also discussed. Finally, a list of high- and low-throughput techniques available to exploit both larvae and adult OI zebrafish models as unique tools for the discovery of new therapeutic approaches will be presented.
Collapse
Affiliation(s)
- Cecilia Masiero
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| | - Carla Aresi
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy.
| | - Francesca Tonelli
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| |
Collapse
|
3
|
Gunasekaran M, Littel HR, Wells NM, Turner J, Campos G, Venigalla S, Estrella EA, Ghosh PS, Daugherty AL, Stafki SA, Kunkel LM, Foley AR, Donkervoort S, Bönnemann CG, Toledo-Bravo de Laguna L, Nascimento A, Benito DND, Draper I, Bruels CC, Pacak CA, Kang PB. Effects of HMGCR deficiency on skeletal muscle development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.591934. [PMID: 38903061 PMCID: PMC11188090 DOI: 10.1101/2024.05.06.591934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Pathogenic variants in HMGCR were recently linked to a limb-girdle muscular dystrophy (LGMD) phenotype. The protein product HMG CoA reductase (HMGCR) catalyzes a key component of the cholesterol synthesis pathway. The two other muscle diseases associated with HMGCR, statin-associated myopathy (SAM) and autoimmune anti-HMGCR myopathy, are not inherited in a Mendelian pattern. The mechanism linking pathogenic variants in HMGCR with skeletal muscle dysfunction is unclear. We knocked down Hmgcr in mouse skeletal myoblasts, knocked down hmgcr in Drosophila, and expressed three pathogenic HMGCR variants (c.1327C>T, p.Arg443Trp; c.1522_1524delTCT, p.Ser508del; and c.1621G>A, p.Ala541Thr) in Hmgcr knockdown mouse myoblasts. Hmgcr deficiency was associated with decreased proliferation, increased apoptosis, and impaired myotube fusion. Transcriptome sequencing of Hmgcr knockdown versus control myoblasts revealed differential expression involving mitochondrial function, with corresponding differences in cellular oxygen consumption rates. Both ubiquitous and muscle-specific knockdown of hmgcr in Drosophila led to lethality. Overexpression of reference HMGCR cDNA rescued myotube fusion in knockdown cells, whereas overexpression of the pathogenic variants of HMGCR cDNA did not. These results suggest that the three HMGCR-related muscle diseases share disease mechanisms related to skeletal muscle development.
Collapse
|
4
|
Al-Sabri MH, Behare N, Alsehli AM, Berkins S, Arora A, Antoniou E, Moysiadou EI, Anantha-Krishnan S, Cosmen PD, Vikner J, Moulin TC, Ammar N, Boukhatmi H, Clemensson LE, Rask-Andersen M, Mwinyi J, Williams MJ, Fredriksson R, Schiöth HB. Statins Induce Locomotion and Muscular Phenotypes in Drosophila melanogaster That Are Reminiscent of Human Myopathy: Evidence for the Role of the Chloride Channel Inhibition in the Muscular Phenotypes. Cells 2022; 11:3528. [PMID: 36428957 PMCID: PMC9688544 DOI: 10.3390/cells11223528] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/17/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022] Open
Abstract
The underlying mechanisms for statin-induced myopathy (SIM) are still equivocal. In this study, we employ Drosophila melanogaster to dissect possible underlying mechanisms for SIM. We observe that chronic fluvastatin treatment causes reduced general locomotion activity and climbing ability. In addition, transmission microscopy of dissected skeletal muscles of fluvastatin-treated flies reveals strong myofibrillar damage, including increased sarcomere lengths and Z-line streaming, which are reminiscent of myopathy, along with fragmented mitochondria of larger sizes, most of which are round-like shapes. Furthermore, chronic fluvastatin treatment is associated with impaired lipid metabolism and insulin signalling. Mechanistically, knockdown of the statin-target Hmgcr in the skeletal muscles recapitulates fluvastatin-induced mitochondrial phenotypes and lowered general locomotion activity; however, it was not sufficient to alter sarcomere length or elicit myofibrillar damage compared to controls or fluvastatin treatment. Moreover, we found that fluvastatin treatment was associated with reduced expression of the skeletal muscle chloride channel, ClC-a (Drosophila homolog of CLCN1), while selective knockdown of skeletal muscle ClC-a also recapitulated fluvastatin-induced myofibril damage and increased sarcomere lengths. Surprisingly, exercising fluvastatin-treated flies restored ClC-a expression and normalized sarcomere lengths, suggesting that fluvastatin-induced myofibrillar phenotypes could be linked to lowered ClC-a expression. Taken together, these results may indicate the potential role of ClC-a inhibition in statin-associated muscular phenotypes. This study underlines the importance of Drosophila melanogaster as a powerful model system for elucidating the locomotion and muscular phenotypes, promoting a better understanding of the molecular mechanisms underlying SIM.
Collapse
Affiliation(s)
- Mohamed H. Al-Sabri
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
- Department of Pharmaceutical Biosciences, Uppsala University, 751 24 Uppsala, Sweden
| | - Neha Behare
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Ahmed M. Alsehli
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
- Faculty of Medicine, King Abdulaziz University and Hospital, Al Ehtifalat St., Jeddah 21589, Saudi Arabia
| | - Samuel Berkins
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Aadeya Arora
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Eirini Antoniou
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Eleni I. Moysiadou
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Sowmya Anantha-Krishnan
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Patricia D. Cosmen
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Johanna Vikner
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Thiago C. Moulin
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
- Faculty of Medicine, Department of Experimental Medical Science, Lund University, Sölvegatan 19, BMC F10, 221 84 Lund, Sweden
| | - Nourhene Ammar
- Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, CNRS, UMR6290, 35065 Rennes, France
| | - Hadi Boukhatmi
- Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, CNRS, UMR6290, 35065 Rennes, France
| | - Laura E. Clemensson
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Mathias Rask-Andersen
- Department of Immunology, Genetics and Pathology, Uppsala University, 752 37 Uppsala, Sweden
| | - Jessica Mwinyi
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Michael J. Williams
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Biosciences, Uppsala University, 751 24 Uppsala, Sweden
| | - Helgi B. Schiöth
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| |
Collapse
|
5
|
Kilroy EA, Ignacz AC, Brann KL, Schaffer CE, Varney D, Alrowaished SS, Silknitter KJ, Miner JN, Almaghasilah A, Spellen TL, Lewis AD, Tilbury K, King BL, Kelley JB, Henry CA. Beneficial impacts of neuromuscular electrical stimulation on muscle structure and function in the zebrafish model of Duchenne muscular dystrophy. eLife 2022; 11:62760. [PMID: 35324428 PMCID: PMC8947762 DOI: 10.7554/elife.62760] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/10/2022] [Indexed: 12/20/2022] Open
Abstract
Neuromuscular electrical stimulation (NMES) allows activation of muscle fibers in the absence of voluntary force generation. NMES could have the potential to promote muscle homeostasis in the context of muscle disease, but the impacts of NMES on diseased muscle are not well understood. We used the zebrafish Duchenne muscular dystrophy (dmd) mutant and a longitudinal design to elucidate the consequences of NMES on muscle health. We designed four neuromuscular stimulation paradigms loosely based on weightlifting regimens. Each paradigm differentially affected neuromuscular structure, function, and survival. Only endurance neuromuscular stimulation (eNMES) improved all outcome measures. We found that eNMES improves muscle and neuromuscular junction morphology, swimming, and survival. Heme oxygenase and integrin alpha7 are required for eNMES-mediated improvement. Our data indicate that neuromuscular stimulation can be beneficial, suggesting that the right type of activity may benefit patients with muscle disease.
Collapse
Affiliation(s)
- Elisabeth A Kilroy
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, United States
| | - Amanda C Ignacz
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, United States
| | - Kaylee L Brann
- School of Biology and Ecology, University of Maine, Orono, United States
| | - Claire E Schaffer
- School of Biology and Ecology, University of Maine, Orono, United States
| | - Devon Varney
- School of Biology and Ecology, University of Maine, Orono, United States
| | | | - Kodey J Silknitter
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, United States
| | - Jordan N Miner
- Department of Chemical and Biomedical Engineering, University of Maine, Orono, United States
| | - Ahmed Almaghasilah
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, United States
| | - Tashawna L Spellen
- School of Biology and Ecology, University of Maine, Orono, United States
| | - Alexandra D Lewis
- School of Biology and Ecology, University of Maine, Orono, United States
| | - Karissa Tilbury
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, United States.,Department of Chemical and Biomedical Engineering, University of Maine, Orono, United States
| | - Benjamin L King
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, United States.,Department of Molecular and Biomedical Sciences, University of Maine, Orono, United States
| | - Joshua B Kelley
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, United States.,Department of Molecular and Biomedical Sciences, University of Maine, Orono, United States
| | - Clarissa A Henry
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, United States.,School of Biology and Ecology, University of Maine, Orono, United States
| |
Collapse
|
6
|
Campos LM, Guapyassu L, Gomes C, Midlej V, Benchimol M, Mermelstein C, Costa ML. Simvastatin and Muscle: Zebrafish and Chicken Show that the Benefits are not Worth the Damage. Front Cell Dev Biol 2022; 10:778901. [PMID: 35359432 PMCID: PMC8964290 DOI: 10.3389/fcell.2022.778901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 02/17/2022] [Indexed: 11/22/2022] Open
Abstract
Simvastatin is one of the most common medicines prescribed to treat human hypercholesterolemia. Simvastatin acts through the inhibition of cholesterol synthesis. Unfortunately, simvastatin causes unwanted side effects on muscles, such as soreness, tiredness, or weakness. Therefore, to understand the mechanism of action of simvastatin, it is important to study its physiological and structural impacts on muscle in varied animal models. Here we report on the effects of simvastatin on two biological models: zebrafish embryos and chicken muscle culture. In the last years, our group and others showed that simvastatin treatment in zebrafish embryos reduces fish movements and induces major structural alterations in skeletal muscles. We also showed that simvastatin and membrane cholesterol depletion induce major changes in proliferation and differentiation of muscle cells in chick muscle cultures. Here, we review and discuss these observations considering reported data on the use of simvastatin as a potential therapy for Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Laise M. Campos
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Livia Guapyassu
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cyro Gomes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Victor Midlej
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Marlene Benchimol
- Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Universidade do Grande Rio (UNIGRANRIO), Duque de Caxias, Rio de Janeiro, Brazil
| | - Claudia Mermelstein
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Claudia Mermelstein, r
| | - Manoel Luis Costa
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Widrick JJ, Kawahara G, Alexander MS, Beggs AH, Kunkel LM. Discovery of Novel Therapeutics for Muscular Dystrophies using Zebrafish Phenotypic Screens. J Neuromuscul Dis 2020; 6:271-287. [PMID: 31282429 PMCID: PMC6961982 DOI: 10.3233/jnd-190389] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The recent availability and development of mutant and transgenic zebrafish strains that model human muscular dystrophies has created new research opportunities for therapeutic development. Not only do these models mimic many pathological aspects of human dystrophies, but their small size, large clutch sizes, rapid ex utero development, body transparency, and genetic tractability enable research approaches that would be inconceivable with mammalian model systems. Here we discuss the use of zebrafish models of muscular dystrophy to rapidly screen hundreds to thousands of bioactive compounds in order to identify novel therapeutic candidates that modulate pathologic phenotypes. We review the justification and rationale behind this unbiased approach, including how zebrafish screens have identified FDA-approved drugs that are candidates for treating Duchenne and limb girdle muscular dystrophies. Not only can these drugs be re-purposed for treating dystrophies in a fraction of the time and cost of new drug development, but their identification has revealed novel, unexpected directions for future therapy development. Phenotype-driven zebrafish drug screens are an important compliment to the more established mammalian, target-based approaches for rapidly developing and validating therapeutics for muscular dystrophies.
Collapse
Affiliation(s)
- Jeffrey J Widrick
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Genri Kawahara
- Department of Pathophysiology, Tokyo Medical University, Tokyo, Japan
| | - Matthew S Alexander
- Department of Pediatrics, Division of Neurology at the University of Alabama at Birmingham and Children's of Alabama; University of Alabama at Birmingham Center for Exercise Medicine; University of Alabama at Birmingham Civitan International Research Center; University of Alabama at Birmingham Department of Genetics; Birmingham, Alabama, USA
| | - Alan H Beggs
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Louis M Kunkel
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
8
|
Hoppstädter J, Valbuena Perez JV, Linnenberger R, Dahlem C, Legroux TM, Hecksteden A, Tse WKF, Flamini S, Andreas A, Herrmann J, Herr C, Müller R, Meyer T, Bals R, Riccardi C, Bruscoli S, Kiemer AK. The glucocorticoid-induced leucine zipper mediates statin-induced muscle damage. FASEB J 2020; 34:4684-4701. [PMID: 32030813 DOI: 10.1096/fj.201902557rrr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/21/2020] [Accepted: 01/21/2020] [Indexed: 12/31/2022]
Abstract
Statins, the most prescribed class of drugs for the treatment of hypercholesterolemia, can cause muscle-related adverse effects. It has been shown that the glucocorticoid-induced leucine zipper (GILZ) plays a key role in the anti-myogenic action of dexamethasone. In the present study, we aimed to evaluate the role of GILZ in statin-induced myopathy. Statins induced GILZ expression in C2C12 cells, primary murine myoblasts/myotubes, primary human myoblasts, and in vivo in zebrafish embryos and human quadriceps femoris muscle. Gilz induction was mediated by FOXO3 activation and binding to the Gilz promoter, and could be reversed by the addition of geranylgeranyl, but not farnesyl, pyrophosphate. Atorvastatin decreased Akt phosphorylation and increased cleaved caspase-3 levels in myoblasts. This effect was reversed in myoblasts from GILZ knockout mice. Similarly, myofibers isolated from knockout animals were more resistant toward statin-induced cell death than their wild-type counterparts. Statins also impaired myoblast differentiation, and this effect was accompanied by GILZ induction. The in vivo relevance of our findings was supported by the observation that gilz overexpression in zebrafish embryos led to impaired embryonic muscle development. Taken together, our data point toward GILZ as an essential mediator of the molecular mechanisms leading to statin-induced muscle damage.
Collapse
Affiliation(s)
- Jessica Hoppstädter
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany.,Department of Medicine, Section of Pharmacology, University of Perugia, Perugia, Italy
| | | | - Rebecca Linnenberger
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Charlotte Dahlem
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Thierry M Legroux
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Anne Hecksteden
- Institute of Sports and Preventive Medicine, Saarland University, Saarbrücken, Germany
| | - William K F Tse
- Center for Promotion of International Education and Research, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Sara Flamini
- Department of Medicine, Section of Pharmacology, University of Perugia, Perugia, Italy
| | - Anastasia Andreas
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Jennifer Herrmann
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Christian Herr
- Department of Internal Medicine V-Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Rolf Müller
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Tim Meyer
- Institute of Sports and Preventive Medicine, Saarland University, Saarbrücken, Germany
| | - Robert Bals
- Department of Internal Medicine V-Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Carlo Riccardi
- Department of Medicine, Section of Pharmacology, University of Perugia, Perugia, Italy
| | - Stefano Bruscoli
- Department of Medicine, Section of Pharmacology, University of Perugia, Perugia, Italy
| | - Alexandra K Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| |
Collapse
|
9
|
Clemente Z, Silva GH, de Souza Nunes MC, Martinez DST, Maurer-Morelli CV, Thomaz AA, Castro VLSS. Exploring the mechanisms of graphene oxide behavioral and morphological changes in zebrafish. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:30508-30523. [PMID: 31463743 DOI: 10.1007/s11356-019-05870-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/28/2019] [Indexed: 06/10/2023]
Abstract
The presence of natural organic matter such as humic acid (HA) can influence the behavior of graphene oxide (GO) in the aquatic environment. In this study, zebrafish embryos were analyzed after 5 and 7 days of exposure to GO (100 mg L-1) and HA (20 mg L-1) alone or together. The results indicated that, regardless of the presence of HA, larvae exposed to GO for 5 days showed an increase in locomotor activity, reduction in the yolk sac size, and total length and inhibition of AChE activity, but there was no difference in enzyme expression. The statistical analysis indicated that the reductions in total larval length, yolk sac size, and AChE activity in larvae exposed to GO persisted in relation to the control group, but there was a recovery of these parameters in groups also exposed to HA. Larvae exposed to GO for 7 days did not show significant differences in locomotor activity, but the RT-PCR gene expression analysis evidenced an increase in the AChE expression. Since the embryos exposed to GO showed a reduction in overall length, they were submitted to confocal microscopy and their muscle tissue configuration investigated. No changes were observed in the muscle tissue. The results indicated that HA is associated with the toxicity risk modulation by GO and that some compensatory homeostasis mechanisms may be involved in the developmental effects observed in zebrafish.
Collapse
Affiliation(s)
- Zaira Clemente
- Laboratory of Ecotoxicology and Biosafety, Embrapa Environment, Jaguariúna, São Paulo, 13820-000, Brazil.
- Brazilian National Nanotechnology Laboratory (LNNano), Brazilian Center for Research on Energy and Materials (CNPEM), Campinas, São Paulo, 13083-970, Brazil.
| | - Gabriela Helena Silva
- Laboratory of Ecotoxicology and Biosafety, Embrapa Environment, Jaguariúna, São Paulo, 13820-000, Brazil
- Brazilian National Nanotechnology Laboratory (LNNano), Brazilian Center for Research on Energy and Materials (CNPEM), Campinas, São Paulo, 13083-970, Brazil
- Center for Nuclear Energy in Agriculture (CENA), University of São Paulo (USP), Piracicaba, São Paulo, 13416-000, Brazil
| | - Miriam Celi de Souza Nunes
- Department of Medical Genetics, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, 13087-883, Brazil
| | - Diego Stéfani Teodoro Martinez
- Brazilian National Nanotechnology Laboratory (LNNano), Brazilian Center for Research on Energy and Materials (CNPEM), Campinas, São Paulo, 13083-970, Brazil
- Center for Nuclear Energy in Agriculture (CENA), University of São Paulo (USP), Piracicaba, São Paulo, 13416-000, Brazil
| | - Claudia Vianna Maurer-Morelli
- Department of Medical Genetics, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, 13087-883, Brazil
| | - Andre Alexandre Thomaz
- Department of Quantum Electronics, Institute of Physics "Gleb Wataghin", University of Campinas (UNICAMP), Campinas, São Paulo, 13083-859, Brazil
- National Institute of Photonics Applied to Cell Biology (INFABIC), University of Campinas (UNICAMP), Campinas, São Paulo, 13083-859, Brazil
| | | |
Collapse
|
10
|
Da Silva GH, Clemente Z, Khan LU, Coa F, Neto LLR, Carvalho HWP, Castro VL, Martinez DST, Monteiro RTR. Toxicity assessment of TiO 2-MWCNT nanohybrid material with enhanced photocatalytic activity on Danio rerio (Zebrafish) embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 165:136-143. [PMID: 30195205 DOI: 10.1016/j.ecoenv.2018.08.093] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/22/2018] [Accepted: 08/24/2018] [Indexed: 05/24/2023]
Abstract
The increasing production and use of nanomaterials is causing serious concerns about their safety to human and environmental health. However, the applications of titanium dioxide nanoparticles (TiO2NP) and multiwalled carbon nanotubes (MWCNT) hybrids has grown considerably, due to their enhanced photocatalytic efficiency. To our knowledge, there are no reports available to the scientific community about their toxicity. In this work, we perform a toxicity assessment of TiO2NP and TiO2-MWCNT nanohybrid materials using Zebrafish embryos standardized 96 h early life stage assay, under different exposure conditions (with and without UV light exposure). After exposure the parameters assessed were acute toxicity, hatching rate, growth, yolk sac size, and sarcomere length. In addition, μ-probe X-ray fluorescence spectroscopy (µ-XRF) was employed to observe if nanoparticles were uptaken by zebrafish embryos and consequently accumulated in their organisms. Neither TiO2NP nor TiO2-MWCNT nanohybrids presented acute toxicity to the zebrafish embryos. Moreover, TiO2NP presents sublethal effects for total length (with and without UV light exposure) on the embryos. This work contributes to the understanding of the potential adverse effects of the emerging nanohybrid materials towards safe innovation approaches in nanotechnology.
Collapse
Affiliation(s)
- Gabriela H Da Silva
- Center of Nuclear Energy in Agriculture (CENA), University of São Paulo (USP), Piracicaba, São Paulo, Brazil; Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil; Laboratory of Ecotoxicology and Biosafety, EMBRAPA Environment, Jaguariuna, São Paulo, Brazil.
| | - Zaira Clemente
- Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil; Laboratory of Ecotoxicology and Biosafety, EMBRAPA Environment, Jaguariuna, São Paulo, Brazil
| | - Latif U Khan
- Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
| | - Francine Coa
- Center of Nuclear Energy in Agriculture (CENA), University of São Paulo (USP), Piracicaba, São Paulo, Brazil; Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
| | - Lais L R Neto
- Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil; School of Technology, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Hudson W P Carvalho
- Center of Nuclear Energy in Agriculture (CENA), University of São Paulo (USP), Piracicaba, São Paulo, Brazil
| | - Vera L Castro
- Laboratory of Ecotoxicology and Biosafety, EMBRAPA Environment, Jaguariuna, São Paulo, Brazil
| | - Diego Stéfani T Martinez
- Center of Nuclear Energy in Agriculture (CENA), University of São Paulo (USP), Piracicaba, São Paulo, Brazil; Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil; School of Technology, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.
| | - Regina T R Monteiro
- Center of Nuclear Energy in Agriculture (CENA), University of São Paulo (USP), Piracicaba, São Paulo, Brazil.
| |
Collapse
|
11
|
de Wergifosse M, Botek E, De Meulenaere E, Clays K, Champagne B. ONIOM Investigation of the Second-Order Nonlinear Optical Responses of Fluorescent Proteins. J Phys Chem B 2018; 122:4993-5005. [DOI: 10.1021/acs.jpcb.8b01430] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Marc de Wergifosse
- Laboratory of Theoretical Chemistry, Unit of Theoretical and Structural Physical Chemistry, Namur Institute of Structured Matter, University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Edith Botek
- Laboratory of Theoretical Chemistry, Unit of Theoretical and Structural Physical Chemistry, Namur Institute of Structured Matter, University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Evelien De Meulenaere
- Centre of Microbial and Plant Genetics, KU Leuven, Kasteelpark Arenberg 20, B-3001 Leuven, Belgium
- Laboratory for Molecular Electronics and Photonics, Department of Chemistry, KU Leuven, Celestijnenlaan 200D, B-3001 Leuven, Belgium
| | - Koen Clays
- Laboratory for Molecular Electronics and Photonics, Department of Chemistry, KU Leuven, Celestijnenlaan 200D, B-3001 Leuven, Belgium
| | - Benoît Champagne
- Laboratory of Theoretical Chemistry, Unit of Theoretical and Structural Physical Chemistry, Namur Institute of Structured Matter, University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| |
Collapse
|
12
|
Boltaña S, Sanhueza N, Aguilar A, Gallardo-Escarate C, Arriagada G, Valdes JA, Soto D, Quiñones RA. Influences of thermal environment on fish growth. Ecol Evol 2017; 7:6814-6825. [PMID: 28904762 PMCID: PMC5587470 DOI: 10.1002/ece3.3239] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/19/2017] [Accepted: 06/20/2017] [Indexed: 01/17/2023] Open
Abstract
Thermoregulation in ectothermic animals is influenced by the ability to effectively respond to thermal variations. While it is known that ectotherms are affected by thermal changes, it remains unknown whether physiological and/or metabolic traits are impacted by modifications to the thermal environment. Our research provides key evidence that fish ectotherms are highly influenced by thermal variability during development, which leads to important modifications at several metabolic levels (e.g., growth trajectories, microstructural alterations, muscle injuries, and molecular mechanisms). In Atlantic salmon (Salmo salar), a wide thermal range (ΔT 6.4°C) during development (posthatch larvae to juveniles) was associated with increases in key thermal performance measures for survival and growth trajectory. Other metabolic traits were also significantly influenced, such as size, muscle cellularity, and molecular growth regulators possibly affected by adaptive processes. In contrast, a restricted thermal range (ΔT 1.4°C) was detrimental to growth, survival, and cellular microstructure as muscle growth could not keep pace with increased metabolic demands. These findings provide a possible basic explanation for the effects of thermal environment during growth. In conclusion, our results highlight the key role of thermal range amplitude on survival and on interactions with major metabolism‐regulating processes that have positive adaptive effects for organisms.
Collapse
Affiliation(s)
- Sebastián Boltaña
- Interdisciplinary Center for Aquaculture Research (INCAR) Department of Oceanography Biotechnology Center University of Concepción Concepción Chile
| | - Nataly Sanhueza
- Interdisciplinary Center for Aquaculture Research (INCAR) Department of Oceanography Biotechnology Center University of Concepción Concepción Chile
| | - Andrea Aguilar
- Interdisciplinary Center for Aquaculture Research (INCAR) Department of Oceanography Biotechnology Center University of Concepción Concepción Chile
| | - Cristian Gallardo-Escarate
- Interdisciplinary Center for Aquaculture Research (INCAR) Department of Oceanography Biotechnology Center University of Concepción Concepción Chile
| | - Gabriel Arriagada
- Interdisciplinary Center for Aquaculture Research (INCAR) Department of Oceanography Biotechnology Center University of Concepción Concepción Chile
| | | | - Doris Soto
- Interdisciplinary Center for Aquaculture Research (INCAR) Department of Oceanography Biotechnology Center University of Concepción Concepción Chile
| | - Renato A Quiñones
- Interdisciplinary Center for Aquaculture Research (INCAR) Department of Oceanography Biotechnology Center University of Concepción Concepción Chile
| |
Collapse
|
13
|
Miller MA, Weissleder R. Imaging the pharmacology of nanomaterials by intravital microscopy: Toward understanding their biological behavior. Adv Drug Deliv Rev 2017; 113:61-86. [PMID: 27266447 PMCID: PMC5136524 DOI: 10.1016/j.addr.2016.05.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 05/25/2016] [Indexed: 12/15/2022]
Abstract
Therapeutic nanoparticles (NPs) can deliver cytotoxic chemotherapeutics and other drugs more safely and efficiently to patients; furthermore, selective delivery to target tissues can theoretically be accomplished actively through coating NPs with molecular ligands, and passively through exploiting physiological "enhanced permeability and retention" features. However, clinical trial results have been mixed in showing improved efficacy with drug nanoencapsulation, largely due to heterogeneous NP accumulation at target sites across patients. Thus, a clear need exists to better understand why many NP strategies fail in vivo and not result in significantly improved tumor uptake or therapeutic response. Multicolor in vivo confocal fluorescence imaging (intravital microscopy; IVM) enables integrated pharmacokinetic and pharmacodynamic (PK/PD) measurement at the single-cell level, and has helped answer key questions regarding the biological mechanisms of in vivo NP behavior. This review summarizes progress to date and also describes useful technical strategies for successful IVM experimentation.
Collapse
Affiliation(s)
- Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, Boston, MA 02114, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA.
| |
Collapse
|
14
|
Lee IJ, Yang YC, Hsu JW, Chang WT, Chuang YJ, Liau I. Zebrafish model of photochemical thrombosis for translational research and thrombolytic screening in vivo. JOURNAL OF BIOPHOTONICS 2017; 10:494-502. [PMID: 27174426 DOI: 10.1002/jbio.201500287] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 04/22/2016] [Accepted: 04/22/2016] [Indexed: 06/05/2023]
Abstract
Acute thromboembolic diseases remain the major global cause of death or disability. Although an array of thrombolytic and antithrombotic drugs has been approved to treat or prevent thromboembolic diseases, many more drugs that target specific clotting mechanisms are under development. Here a novel zebrafish model of photochemical thrombosis is reported and its prospective application for the screening and preclinical testing of thrombolytic agents in vivo is demonstrated. Through photochemical excitation, a thrombus was induced to form at a selected section of the dorsal aorta of larval zebrafish, which had been injected with photosensitizers. Such photochemical thrombosis can be consistently controlled to occlude partially or completely the targeted blood vessel. Detailed mechanistic tests indicate that the zebrafish model of photochemical thrombosis exhibits essential features of classical coagulation and a thrombolytic pathway. For demonstration, tissue plasminogen activator (tPA), a clinically feasible thrombolytic agent, was shown to effectively dissolve photochemically induced blood clots. In light of the numerous unique advantages of zebrafish as a model organism, our approach is expected to benefit not only the development of novel thrombolytic and antithrombotic strategies but also the fundamental or translational research targeting hereditary thrombotic or coagulation disorders.
Collapse
Affiliation(s)
- I-Ju Lee
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Yi-Cyun Yang
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Jia-Wen Hsu
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Wei-Tien Chang
- Department of Emergency Medicine and Cardiovascular Center, National Taiwan University Hospital and College of Medicine, Taipei, 100, Taiwan
| | - Yung-Jen Chuang
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Ian Liau
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, 300, Taiwan
| |
Collapse
|
15
|
|
16
|
Chen X, Delp SL. Human soleus sarcomere lengths measured using in vivo microendoscopy at two ankle flexion angles. J Biomech 2016; 49:4164-4167. [PMID: 27866676 PMCID: PMC6050010 DOI: 10.1016/j.jbiomech.2016.11.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 11/08/2016] [Accepted: 11/08/2016] [Indexed: 11/19/2022]
Abstract
The forces generated by the soleus muscle play an important role in standing and locomotion. The lengths of the sarcomeres of the soleus affect its force-generating capacity, yet it is unknown how sarcomere lengths in the soleus change as a function of ankle flexion angle. In this study, we used microendoscopy to measure resting sarcomere lengths at 10° plantarflexion and 20° dorsiflexion in 7 healthy individuals. Mean sarcomere lengths at 10° plantarflexion were 2.84±0.09µm (mean±S.E.M.), near the optimal length for sarcomere force generation. Sarcomere lengths were 3.43±0.09µm at 20° dorsiflexion, indicating that they were longer than optimal length when the ankle was in dorsiflexion and the muscle was inactive. Our results indicate a smaller sarcomere length difference between two ankle flexion angles compared to estimates from musculoskeletal models and suggest why these models frequently underestimate the force-generating capacity of the soleus.
Collapse
Affiliation(s)
- Xuefeng Chen
- Department of Mechanical Engineering, Stanford University, United States
| | - Scott L Delp
- Department of Mechanical Engineering, Stanford University, United States; Department of Bioengineering, Stanford University, United States.
| |
Collapse
|
17
|
Dubińska-Magiera M, Daczewska M, Lewicka A, Migocka-Patrzałek M, Niedbalska-Tarnowska J, Jagla K. Zebrafish: A Model for the Study of Toxicants Affecting Muscle Development and Function. Int J Mol Sci 2016; 17:E1941. [PMID: 27869769 PMCID: PMC5133936 DOI: 10.3390/ijms17111941] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 11/10/2016] [Accepted: 11/14/2016] [Indexed: 01/08/2023] Open
Abstract
The rapid progress in medicine, agriculture, and allied sciences has enabled the development of a large amount of potentially useful bioactive compounds, such as drugs and pesticides. However, there is another side of this phenomenon, which includes side effects and environmental pollution. To avoid or minimize the uncontrollable consequences of using the newly developed compounds, researchers seek a quick and effective means of their evaluation. In achieving this goal, the zebrafish (Danio rerio) has proven to be a highly useful tool, mostly because of its fast growth and development, as well as the ability to absorb the molecules diluted in water through its skin and gills. In this review, we focus on the reports concerning the application of zebrafish as a model for assessing the impact of toxicants on skeletal muscles, which share many structural and functional similarities among vertebrates, including zebrafish and humans.
Collapse
Affiliation(s)
- Magda Dubińska-Magiera
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, 21 Sienkiewicza Street, 50-335 Wroclaw, Poland.
| | - Małgorzata Daczewska
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, 21 Sienkiewicza Street, 50-335 Wroclaw, Poland.
| | - Anna Lewicka
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, 21 Sienkiewicza Street, 50-335 Wroclaw, Poland.
| | - Marta Migocka-Patrzałek
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, 21 Sienkiewicza Street, 50-335 Wroclaw, Poland.
| | - Joanna Niedbalska-Tarnowska
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, 21 Sienkiewicza Street, 50-335 Wroclaw, Poland.
| | - Krzysztof Jagla
- GReD-Genetics, Reproduction and Development Laboratory, INSERM U1103, CNRS UMR6293, University of Clermont-Auvergne, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France.
| |
Collapse
|
18
|
Exposure to gemfibrozil and atorvastatin affects cholesterol metabolism and steroid production in zebrafish (Danio rerio). Comp Biochem Physiol B Biochem Mol Biol 2016; 199:87-96. [DOI: 10.1016/j.cbpb.2015.11.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 11/19/2015] [Accepted: 11/23/2015] [Indexed: 01/09/2023]
|
19
|
Chen X, Sanchez GN, Schnitzer MJ, Delp SL. Changes in sarcomere lengths of the human vastus lateralis muscle with knee flexion measured using in vivo microendoscopy. J Biomech 2016; 49:2989-2994. [PMID: 27481293 DOI: 10.1016/j.jbiomech.2016.07.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 06/05/2016] [Accepted: 07/16/2016] [Indexed: 11/30/2022]
Abstract
Sarcomeres are the basic contractile units of muscle, and their lengths influence muscle force-generating capacity. Despite their importance, in vivo sarcomere lengths remain unknown for many human muscles. Second harmonic generation (SHG) microendoscopy is a minimally invasive technique for imaging sarcomeres in vivo and measuring their lengths. In this study, we used SHG microendoscopy to visualize sarcomeres of the human vastus lateralis, a large knee extensor muscle important for mobility, to examine how sarcomere lengths change with knee flexion and thus affect the muscle׳s force-generating capacity. We acquired in vivo sarcomere images of several muscle fibers of the resting vastus lateralis in six healthy individuals. Mean sarcomere lengths increased (p=0.031) from 2.84±0.16μm at 50° of knee flexion to 3.17±0.13μm at 110° of knee flexion. The standard deviation of sarcomere lengths among different fibers within a muscle was 0.21±0.09μm. Our results suggest that the sarcomeres of the resting vastus lateralis at 50° of knee flexion are near optimal length. At a knee flexion angle of 110° the resting sarcomeres of vastus lateralis are longer than optimal length. These results show a smaller sarcomere length change and greater conservation of force-generating capacity with knee flexion than estimated in previous studies.
Collapse
Affiliation(s)
- Xuefeng Chen
- Department of Mechanical Engineering, Stanford University, United States
| | - Gabriel N Sanchez
- Department of Mechanical Engineering, Stanford University, United States; Department of Bioengineering, Stanford University, United States
| | - Mark J Schnitzer
- Department of Biology, Stanford University, United States; Department of Applied Physics, Stanford University, United States; Howard Hughes Medical Institute, United States
| | - Scott L Delp
- Department of Mechanical Engineering, Stanford University, United States; Department of Bioengineering, Stanford University, United States.
| |
Collapse
|
20
|
Abstract
Modern optical imaging has progressed rapidly with the ability to noninvasively image cellular and subcellular phenomena with high spatial and temporal resolution. In particular, emerging techniques such as second harmonic generation (SHG) microscopy can allow for the monitoring of intrinsic contrast, such as that from collagen, in live and fixed samples. When coupled with multiphoton fluorescence microscopy, SHG can be used to image interactions between cells and the surrounding extracellular environment. There is recent interest in using these approaches to study inflammation and wound healing in zebrafish, an important model for studying these processes. In this chapter we present the practical aspects of using second harmonic generation to image interactions between leukocytes and collagen during wound healing in zebrafish.
Collapse
|
21
|
Lin HJ, Hong ZY, Li YK, Liau I. Fluorescent tracer of dopamine enables selective labelling and interrogation of dopaminergic amacrine cells in the retina of living zebrafish. RSC Adv 2016. [DOI: 10.1039/c6ra13073a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
A ‘fluorescent dopamine’ that enables selective labeling and interrogation of retinal dopaminergic amacrine cells in living zebrafish was demonstrated.
Collapse
Affiliation(s)
- Hui-Jen Lin
- Department of Applied Chemistry
- Institute of Molecular Science
- National Chiao Tung University
- Hsinchu 300
- Taiwan
| | - Zhen-Yi Hong
- Department of Applied Chemistry
- Institute of Molecular Science
- National Chiao Tung University
- Hsinchu 300
- Taiwan
| | - Yaw-Kuen Li
- Department of Applied Chemistry
- Institute of Molecular Science
- National Chiao Tung University
- Hsinchu 300
- Taiwan
| | - Ian Liau
- Department of Applied Chemistry
- Institute of Molecular Science
- National Chiao Tung University
- Hsinchu 300
- Taiwan
| |
Collapse
|
22
|
Rengo JL, Callahan DM, Savage PD, Ades PA, Toth MJ. Skeletal muscle ultrastructure and function in statin-tolerant individuals. Muscle Nerve 2015; 53:242-51. [PMID: 26059690 DOI: 10.1002/mus.24722] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2015] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Statins have well-known benefits on cardiovascular mortality, though up to 15% of patients experience side effects. With guidelines from the American Heart Association, American College of Cardiology, and American Diabetes Association expected to double the number of statin users, the overall incidence of myalgia and myopathy will increase. METHODS We evaluated skeletal muscle structure and contractile function at the molecular, cellular, and whole tissue levels in 12 statin tolerant and 12 control subjects. RESULTS Myosin isoform expression, fiber type distributions, single fiber maximal Ca(2+) -activated tension, and whole muscle contractile force were similar between groups. No differences were observed in myosin-actin cross-bridge kinetics in myosin heavy chain I or IIA fibers. CONCLUSIONS We found no evidence for statin-induced changes in muscle morphology at the molecular, cellular, or whole tissue levels. Collectively, our data show that chronic statin therapy in healthy asymptomatic individuals does not promote deleterious myofilament structural or functional adaptations.
Collapse
Affiliation(s)
- Jason L Rengo
- Department of Cardiology, University of Vermont Medical Center, Burlington, Vermont, USA
| | - Damien M Callahan
- Department of Medicine, University of Vermont, College of Medicine, Burlington, Vermont, USA.,Department of Molecular Physiology and Biophysics, University of Vermont, College of Medicine, Burlington, Vermont, USA
| | - Patrick D Savage
- Department of Cardiology, University of Vermont Medical Center, Burlington, Vermont, USA
| | - Philip A Ades
- Department of Cardiology, University of Vermont Medical Center, Burlington, Vermont, USA.,Department of Medicine, University of Vermont, College of Medicine, Burlington, Vermont, USA
| | - Michael J Toth
- Department of Medicine, University of Vermont, College of Medicine, Burlington, Vermont, USA.,Department of Molecular Physiology and Biophysics, University of Vermont, College of Medicine, Burlington, Vermont, USA
| |
Collapse
|
23
|
Awasthi S, Izu LT, Mao Z, Jian Z, Landas T, Lerner A, Shimkunas R, Woldeyesus R, Bossuyt J, Wood BM, Chen YJ, Matthews DL, Lieu DK, Chiamvimonvat N, Lam KS, Chen-Izu Y, Chan JW. Multimodal SHG-2PF Imaging of Microdomain Ca2+-Contraction Coupling in Live Cardiac Myocytes. Circ Res 2015; 118:e19-28. [PMID: 26643875 DOI: 10.1161/circresaha.115.307919] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 12/07/2015] [Indexed: 01/24/2023]
Abstract
RATIONALE Cardiac myocyte contraction is caused by Ca(2+) binding to troponin C, which triggers the cross-bridge power stroke and myofilament sliding in sarcomeres. Synchronized Ca(2+) release causes whole cell contraction and is readily observable with current microscopy techniques. However, it is unknown whether localized Ca(2+) release, such as Ca(2+) sparks and waves, can cause local sarcomere contraction. Contemporary imaging methods fall short of measuring microdomain Ca(2+)-contraction coupling in live cardiac myocytes. OBJECTIVE To develop a method for imaging sarcomere level Ca(2+)-contraction coupling in healthy and disease model cardiac myocytes. METHODS AND RESULTS Freshly isolated cardiac myocytes were loaded with the Ca(2+)-indicator fluo-4. A confocal microscope equipped with a femtosecond-pulsed near-infrared laser was used to simultaneously excite second harmonic generation from A-bands of myofibrils and 2-photon fluorescence from fluo-4. Ca(2+) signals and sarcomere strain correlated in space and time with short delays. Furthermore, Ca(2+) sparks and waves caused contractions in subcellular microdomains, revealing a previously underappreciated role for these events in generating subcellular strain during diastole. Ca(2+) activity and sarcomere strain were also imaged in paced cardiac myocytes under mechanical load, revealing spontaneous Ca(2+) waves and correlated local contraction in pressure-overload-induced cardiomyopathy. CONCLUSIONS Multimodal second harmonic generation 2-photon fluorescence microscopy enables the simultaneous observation of Ca(2+) release and mechanical strain at the subsarcomere level in living cardiac myocytes. The method benefits from the label-free nature of second harmonic generation, which allows A-bands to be imaged independently of T-tubule morphology and simultaneously with Ca(2+) indicators. Second harmonic generation 2-photon fluorescence imaging is widely applicable to the study of Ca(2+)-contraction coupling and mechanochemotransduction in both health and disease.
Collapse
Affiliation(s)
- Samir Awasthi
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis
| | - Leighton T Izu
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis
| | - Ziliang Mao
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis
| | - Zhong Jian
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis
| | - Trevor Landas
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis
| | - Aaron Lerner
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis
| | - Rafael Shimkunas
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis
| | - Rahwa Woldeyesus
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis
| | - Julie Bossuyt
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis
| | - Brent M Wood
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis
| | - Yi-Je Chen
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis
| | - Dennis L Matthews
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis
| | - Deborah K Lieu
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis
| | - Nipavan Chiamvimonvat
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis
| | - Kit S Lam
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis
| | - Ye Chen-Izu
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis.
| | - James W Chan
- From the Center for Biophotonics (S.A., Z.M., A.L., D.L.M., J.W.C.), Division of Cardiology (D.K.L., N.C., Y.C.-I.), Division of Hematology/Oncology, Department of Internal Medicine (K.S.L.), and Department of Pathology and Laboratory Medicine (J.W.C.), UC Davis School of Medicine, University of California, Davis, Sacramento; and Departments of Pharmacology (L.T.I., Z.J., T.L., J.B., B.W., Y.-J.C., Y.C.-I.), Biomedical Engineering (S.A., R.S., R.W., Y.C.-I.), Biochemistry and Molecular Medicine (K.S.L.), and Microsurgery Core (Y.-J.C.), University of California, Davis.
| |
Collapse
|
24
|
Vågberg W, Larsson DH, Li M, Arner A, Hertz HM. X-ray phase-contrast tomography for high-spatial-resolution zebrafish muscle imaging. Sci Rep 2015; 5:16625. [PMID: 26564785 PMCID: PMC4643221 DOI: 10.1038/srep16625] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 10/16/2015] [Indexed: 12/11/2022] Open
Abstract
Imaging of muscular structure with cellular or subcellular detail in whole-body animal models is of key importance for understanding muscular disease and assessing interventions. Classical histological methods for high-resolution imaging methods require excision, fixation and staining. Here we show that the three-dimensional muscular structure of unstained whole zebrafish can be imaged with sub-5 μm detail with X-ray phase-contrast tomography. Our method relies on a laboratory propagation-based phase-contrast system tailored for detection of low-contrast 4-6 μm subcellular myofibrils. The method is demonstrated on 20 days post fertilization zebrafish larvae and comparative histology confirms that we resolve individual myofibrils in the whole-body animal. X-ray imaging of healthy zebrafish show the expected structured muscle pattern while specimen with a dystrophin deficiency (sapje) displays an unstructured pattern, typical of Duchenne muscular dystrophy. The method opens up for whole-body imaging with sub-cellular detail also of other types of soft tissue and in different animal models.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Dystrophin/deficiency
- Dystrophin/genetics
- Imaging, Three-Dimensional/methods
- Larva/genetics
- Larva/metabolism
- Microscopy, Confocal
- Microscopy, Phase-Contrast
- Muscles/diagnostic imaging
- Muscular Dystrophy, Animal/diagnostic imaging
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Duchenne/diagnostic imaging
- Muscular Dystrophy, Duchenne/genetics
- Myofibrils/diagnostic imaging
- Radiographic Image Enhancement/instrumentation
- Radiographic Image Enhancement/methods
- Reproducibility of Results
- Tomography, X-Ray Computed/instrumentation
- Tomography, X-Ray Computed/methods
- Zebrafish
Collapse
Affiliation(s)
- William Vågberg
- Department of Applied Physics, KTH Royal Institute of Technology/Albanova, Stockholm, Sweden
| | - Daniel H. Larsson
- Department of Applied Physics, KTH Royal Institute of Technology/Albanova, Stockholm, Sweden
| | - Mei Li
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Anders Arner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Hans M. Hertz
- Department of Applied Physics, KTH Royal Institute of Technology/Albanova, Stockholm, Sweden
| |
Collapse
|
25
|
Combining microscopy with mesoscopy using optical and optoacoustic label-free modes. Sci Rep 2015; 5:12902. [PMID: 26306396 PMCID: PMC4549672 DOI: 10.1038/srep12902] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/08/2015] [Indexed: 01/05/2023] Open
Abstract
Biology requires observations at multiple geometrical scales, a feature that is not typically offered by a single imaging modality. We developed a hybrid optical system that not only provides different contrast modes but also offers imaging at different geometrical scales, achieving uniquely broad resolution and a 1000-fold volume sampling increase compared to volumes scanned by optical microscopy. The system combines optoacoustic mesoscopy, optoacoustic microscopy and two-photon microscopy, the latter integrating second and third harmonic generation modes. Label-free imaging of a mouse ear and zebrafish larva ex-vivo demonstrates the contrast and scale complementarity provided by the hybrid system. We showcase the superior anatomical orientation offered by the label-free capacity and hybrid operation, over fluorescence microscopy, and the dynamic selection between field of view and resolution achieved, leading to new possibilities in biological visualization.
Collapse
|
26
|
Nance ME, Whitfield JT, Zhu Y, Gibson AK, Hanft LM, Campbell KS, Meininger GA, McDonald KS, Segal SS, Domeier TL. Attenuated sarcomere lengthening of the aged murine left ventricle observed using two-photon fluorescence microscopy. Am J Physiol Heart Circ Physiol 2015. [PMID: 26209054 DOI: 10.1152/ajpheart.00315.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The Frank-Starling mechanism, whereby increased diastolic filling leads to increased cardiac output, depends on increasing the sarcomere length (Ls) of cardiomyocytes. Ventricular stiffness increases with advancing age, yet it remains unclear how such changes in compliance impact sarcomere dynamics in the intact heart. We developed an isolated murine heart preparation to monitor Ls as a function of left ventricular pressure and tested the hypothesis that sarcomere lengthening in response to ventricular filling is impaired with advanced age. Mouse hearts isolated from young (3-6 mo) and aged (24-28 mo) C57BL/6 mice were perfused via the aorta under Ca(2+)-free conditions with the left ventricle cannulated to control filling pressure. Two-photon imaging of 4-{2-[6-(dioctylamino)-2-naphthalenyl]ethenyl}1-(3-sulfopropyl)-pyridinium fluorescence was used to monitor t-tubule striations and obtain passive Ls between pressures of 0 and 40 mmHg. Ls values (in μm, aged vs. young, respectively) were 2.02 ± 0.04 versus 2.01 ± 0.02 at 0 mmHg, 2.13 ± 0.04 versus 2.23 ± 0.02 at 5 mmHg, 2.21 ± 0.03 versus 2.27 ± 0.03 at 10 mmHg, and 2.28 ± 0.02 versus 2.36 ± 0.01 at 40 mmHg, indicative of impaired sarcomere lengthening in aged hearts. Atomic force microscopy nanoindentation revealed that intact cardiomyocytes enzymatically isolated from aged hearts had increased stiffness compared with those of young hearts (elastic modulus: aged, 41.9 ± 5.8 kPa vs. young, 18.6 ± 3.3 kPa; P = 0.006). Impaired sarcomere lengthening during left ventricular filling may contribute to cardiac dysfunction with advancing age by attenuating the Frank-Starling mechanism and reducing stroke volume.
Collapse
Affiliation(s)
- Michael E Nance
- Molecular Pathogenesis and Therapeutics Graduate Program, University of Missouri School of Medicine, Columbia, Missouri
| | - Justin T Whitfield
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Yi Zhu
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Anne K Gibson
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Laurin M Hanft
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Kenneth S Campbell
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Gerald A Meininger
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri; and
| | - Kerry S McDonald
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri; and
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri;
| |
Collapse
|
27
|
Lai YC, Chang WT, Lin KY, Liau I. Optical assessment of the cardiac rhythm of contracting cardiomyocytes in vitro and a pulsating heart in vivo for pharmacological screening. BIOMEDICAL OPTICS EXPRESS 2014; 5:1616-1625. [PMID: 24877019 PMCID: PMC4026895 DOI: 10.1364/boe.5.001616] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/15/2014] [Accepted: 04/15/2014] [Indexed: 06/03/2023]
Abstract
Our quest in the pathogenesis and therapies targeting human heart diseases requires assessment of the contractile dynamics of cardiac models of varied complexity, such as isolated cardiomyocytes and the heart of a model animal. It is hence beneficial to have an integral means that can interrogate both cardiomyocytes in vitro and a heart in vivo. Herein we report an application of dual-beam optical reflectometry to determine noninvasively the rhythm of two representative cardiac models-chick embryonic cardiomyocytes and the heart of zebrafish. We probed self-beating cardiomyocytes and revealed the temporally varying contractile frequency with a short-time Fourier transform. Our unique dual-beam setup uniquely records the atrial and ventricular pulsations of zebrafish simultaneously. To minimize the cross talk between signals associated with atrial and ventricular chambers, we particularly modulated the two probe beams at distinct frequencies and extracted the signals specific to individual cardiac chambers with phase-sensitive detection. With this setup, we determined the atrio-ventricular interval, a parameter that is manifested by the electrical conduction from the atrium to the ventricle. To demonstrate pharmacological applications, we characterized zebrafish treated with various cardioactive and cardiotoxic drugs, and identified abnormal cardiac rhythms and atrioventricular (AV) blocks of varied degree. In light of its potential capability to assess cardiac models both in vitro and in vivo and to screen drugs with cardioactivity or toxicity, we expect this approach to have broad applications ranging from cardiopharmacology to developmental biology.
Collapse
Affiliation(s)
- Yu-Cheng Lai
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu 300, Taiwan
- Equal contribution
| | - Wei-Tien Chang
- National Taiwan University Hospital and College of Medicine, Taipei 100, Taiwan
- Equal contribution
| | - Kuen-You Lin
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Ian Liau
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu 300, Taiwan
| |
Collapse
|
28
|
Yang YC, Chang WT, Huang SK, Liau I. Characterization of the Pharmaceutical Effect of Drugs on Atherosclerotic Lesions in Vivo Using Integrated Fluorescence Imaging and Raman Spectral Measurements. Anal Chem 2014; 86:3863-8. [DOI: 10.1021/ac404051f] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yi-Cyun Yang
- Department
of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Wei-Tien Chang
- National Taiwan University Hospital and College of Medicine, Taipei 100, Taiwan
| | - Shao-Kang Huang
- Department
of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Ian Liau
- Department
of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu 300, Taiwan
| |
Collapse
|
29
|
van Spreeuwel ACC, Bax NAM, Bastiaens AJ, Foolen J, Loerakker S, Borochin M, van der Schaft DWJ, Chen CS, Baaijens FPT, Bouten CVC. The influence of matrix (an)isotropy on cardiomyocyte contraction in engineered cardiac microtissues. Integr Biol (Camb) 2014; 6:422-9. [PMID: 24549279 DOI: 10.1039/c3ib40219c] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In the cardiac microenvironment, cardiomyocytes (CMs) are embedded in an aligned and structured extracellular matrix (ECM) to maintain the coordinated contractile function of the heart. The cardiac fibroblast (cFB) is the main cell type responsible for producing and remodeling this matrix. In cardiac diseases, however, adverse remodeling and CM death may lead to deterioration of the aligned myocardial structure. Here, we present an in vitro cardiac model system with uniaxial and biaxial constraints to induce (an)isotropy in 3D microtissues, thereby mimicking 'healthy' aligned and 'diseased' disorganized cardiac matrices. A mixture of neonatal mouse CMs and cFBs was resuspended in a collagen-matrigel hydrogel and seeded to form microtissues to recapitulate the in vivo cellular composition. Matrix disarray led to a stellate cell shape and a disorganized sarcomere organization, while CMs in aligned matrices were more elongated and had aligned sarcomeres. Although matrix disarray has no detrimental effect on the force generated by the CMs, it did have a negative effect on the homogeneity of contraction force distribution. Furthermore, proliferation of the cFBs affected microtissue contraction as indicated by the negative correlation between the percentage of cFBs in the microtissues and their beating frequency. These results suggest that in regeneration of the diseased heart, reorganization of the disorganized matrix will contribute to recover the coordinated contraction but restoring the ratio in cellular composition (CMs and cFBs) is also a prerequisite to completely regain tissue function.
Collapse
Affiliation(s)
- A C C van Spreeuwel
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Lin KY, Chang WT, Lai YC, Liau I. Toward functional screening of cardioactive and cardiotoxic drugs with zebrafish in vivo using pseudodynamic three-dimensional imaging. Anal Chem 2014; 86:2213-20. [PMID: 24456565 DOI: 10.1021/ac403877h] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Given the high mortality in patients with cardiovascular diseases and the life-threatening consequences of drugs with unforeseen adverse effects on hearts, a critical evaluation of the pharmacological response of cardiovascular function on model animals is important especially in the early stages of drug development. We report a proof-of-principle study to demonstrate the utility of zebrafish as an analytical platform to predict the cardiac response of new drugs or chemicals on human beings. With pseudodynamic 3D imaging, we derive individual parameters that are central to the cardiac function of zebrafish, including the ventricular stroke volume, ejection fraction, cardiac output, heart rate, diastolic filling function, and ventricular mass. We evaluate both inotropic and chronotropic responses of the heart of zebrafish treated with drugs that are commonly prescribed and possess varied known cardiac activities. We reveal deranged cardiac function of a zebrafish model of cardiomyopathy induced with a cardiotoxic drug. The cardiac function of zebrafish exhibits a pharmacological response similar to that of human beings. We compare also cardiac parameters obtained in this work with those derived with conventional 2D approximation and show that the latter tends to overestimate the cardiac parameters and produces results of greater variation. In view of the growing interest of using zebrafish in both fundamental and translational biomedical research, we envisage that our approach should benefit not only contemporary pharmaceutical development but also exploratory research such as gene, stem cell, or regenerative therapies targeting congenital or acquired heart diseases.
Collapse
Affiliation(s)
- Kuen-You Lin
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University , Hsinchu 300, Taiwan
| | | | | | | |
Collapse
|
31
|
Kabir MM, Inavalli VVGK, Lau TY, Toussaint KC. Application of quantitative second-harmonic generation microscopy to dynamic conditions. BIOMEDICAL OPTICS EXPRESS 2013; 4:2546-2554. [PMID: 24298415 PMCID: PMC3829549 DOI: 10.1364/boe.4.002546] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 08/22/2013] [Accepted: 10/14/2013] [Indexed: 06/02/2023]
Abstract
We present a quantitative second-harmonic generation (SHG) imaging technique that quantifies the 2D spatial organization of collagen fiber samples under dynamic conditions, as an image is acquired. The technique is demonstrated for both a well-aligned tendon sample and a randomly aligned, sparsely distributed collagen scaffold sample. For a fixed signal-to-noise ratio, we confirm the applicability of this method for various window sizes (pixel areas) as well as with using a gridded overlay map that allows for correlations of fiber orientations within a given image. This work has direct impact to in vivo biological studies by incorporating simultaneous SHG image acquisition and analysis.
Collapse
Affiliation(s)
- Mohammad M. Kabir
- Photonics Research of Bio/nano Environments (PROBE) Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, 1206 W Green St, Urbana, IL 61801, USA
| | - V. V. G. Krishna Inavalli
- Photonics Research of Bio/nano Environments (PROBE) Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, 1206 W Green St, Urbana, IL 61801, USA
| | - Tung-Yuen Lau
- Photonics Research of Bio/nano Environments (PROBE) Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, 1206 W Green St, Urbana, IL 61801, USA
| | - Kimani C. Toussaint
- Photonics Research of Bio/nano Environments (PROBE) Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, 1206 W Green St, Urbana, IL 61801, USA
- Affiliate in the departments of Electrical and Computer Engineering, and Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|