1
|
Mitra A, Mandal S, Banerjee K, Ganguly N, Sasmal P, Banerjee D, Gupta S. Cardiac Regeneration in Adult Zebrafish: A Review of Signaling and Metabolic Coordination. Curr Cardiol Rep 2025; 27:15. [PMID: 39792206 DOI: 10.1007/s11886-024-02162-y] [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] [Accepted: 11/20/2024] [Indexed: 01/12/2025]
Abstract
PURPOSE OF REVIEW This review investigates how post-injury cellular signaling and energy metabolism are two pivotal points in zebrafish's cardiomyocyte cell cycle re-entry and proliferation. It seeks to highlight the probable mechanism of action in proliferative cardiomyocytes compared to mammals and identify gaps in the current understanding of metabolic regulation of cardiac regeneration. RECENT FINDINGS Metabolic substrate changes after birth correlate with reduced cardiomyocyte proliferation in mammals. Unlike adult mammalian hearts, zebrafish can regenerate cardiomyocytes by re-entering the cell cycle, characterized by a metabolic switch from oxidative metabolism to increased glycolysis. Zebrafish provide a valuable model for studying metabolic regulation during cell cycle re-entry and cardiac regeneration. Proliferative cardiomyocytes have upregulated Notch, hippo, and Wnt signaling and decreased ROS generation, DNA damage in different zebrafish cardiac regeneration models. Understanding the correlation between metabolic switches during cell cycle re-entry of already differentiated zebrafish cardiomyocytes is being increasingly recognized as a critical factor in heart regeneration. Zebrafish studies provide insights into metabolic adaptations during heart regeneration, emphasizing the importance of a metabolic switch. However, there are mechanistic gaps, and extensive studies are required to aid in formulating therapeutic strategies for cardiac regenerative medicine.
Collapse
Affiliation(s)
- Arkadeep Mitra
- Department of Zoology, City College, 102/1, Raja Rammohan Sarani, Kolkata, 700009, West Bengal, India
| | - Subhadeep Mandal
- Department of Zoology, Trivenidevi Bhalotia College (Affiliated to Kazi Nazrul University), College Para Rd, Raniganj, 713347, West Bengal, India
| | - Kalyan Banerjee
- Department of Zoology, Trivenidevi Bhalotia College (Affiliated to Kazi Nazrul University), College Para Rd, Raniganj, 713347, West Bengal, India
| | - Nilanjan Ganguly
- Department of Zoology, Trivenidevi Bhalotia College (Affiliated to Kazi Nazrul University), College Para Rd, Raniganj, 713347, West Bengal, India
| | - Pramit Sasmal
- Department of Zoology, Trivenidevi Bhalotia College (Affiliated to Kazi Nazrul University), College Para Rd, Raniganj, 713347, West Bengal, India
| | - Durba Banerjee
- Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican St, Seattle, WA, 98109, USA.
| | - Shreyasi Gupta
- Department of Zoology, Trivenidevi Bhalotia College (Affiliated to Kazi Nazrul University), College Para Rd, Raniganj, 713347, West Bengal, India.
| |
Collapse
|
2
|
Cano-Martínez A, Rubio-Ruiz ME, Guarner-Lans V. Homeostasis and evolution in relation to regeneration and repair. J Physiol 2024; 602:2627-2648. [PMID: 38781025 DOI: 10.1113/jp284426] [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: 06/22/2023] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Homeostasis constitutes a key concept in physiology and refers to self-regulating processes that maintain internal stability when adjusting to changing external conditions. It diminishes internal entropy constituting a driving force behind evolution. Natural selection might act on homeostatic regulatory mechanisms and control mechanisms including homeodynamics, allostasis, hormesis and homeorhesis, where different stable stationary states are reached. Regeneration is under homeostatic control through hormesis. Damage to tissues initiates a response to restore the impaired equilibrium caused by mild stress using cell proliferation, cell differentiation and cell death to recover structure and function. Repair is a homeorhetic change leading to a new stable stationary state with decreased functionality and fibrotic scarring without reconstruction of the 3-D pattern. Mechanisms determining entrance of the tissue or organ to regeneration or repair include the balance between innate and adaptive immune cells in relation to cell plasticity and stromal stem cell responses, and redox balance. The regenerative and reparative capacities vary in different species, distinct tissues and organs, and at different stages of development including ageing. Many cell signals and pathways play crucial roles determining regeneration or repair by regulating protein synthesis, cellular growth, inflammation, proliferation, autophagy, lysosomal function, metabolism and metalloproteinase cell signalling. Attempts to favour the entrance of damaged tissues to regeneration in those with low proliferative rates have been made; however, there are evolutionary constraint mechanisms leading to poor proliferation of stem cells in unfavourable environments or tumour development. More research is required to better understand the regulatory processes of these mechanisms.
Collapse
Affiliation(s)
- Agustina Cano-Martínez
- Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, México, México
| | | | - Verónica Guarner-Lans
- Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, México, México
| |
Collapse
|
3
|
Lai ZY, Yang CC, Chen PH, Chen WC, Lai TY, Lu GY, Yang CY, Wang KY, Liu WC, Chen YC, Liu LYM, Chuang YJ. Syndecan-4 is required for early-stage repair responses during zebrafish heart regeneration. Mol Biol Rep 2024; 51:604. [PMID: 38700644 PMCID: PMC11068835 DOI: 10.1007/s11033-024-09531-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 04/08/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND The healing process after a myocardial infarction (MI) in humans involves complex events that replace damaged tissue with a fibrotic scar. The affected cardiac tissue may lose its function permanently. In contrast, zebrafish display a remarkable capacity for scar-free heart regeneration. Previous studies have revealed that syndecan-4 (SDC4) regulates inflammatory response and fibroblast activity following cardiac injury in higher vertebrates. However, whether and how Sdc4 regulates heart regeneration in highly regenerative zebrafish remains unknown. METHODS AND RESULTS This study showed that sdc4 expression was differentially regulated during zebrafish heart regeneration by transcriptional analysis. Specifically, sdc4 expression increased rapidly and transiently in the early regeneration phase upon ventricular cryoinjury. Moreover, the knockdown of sdc4 led to a significant reduction in extracellular matrix protein deposition, immune cell accumulation, and cell proliferation at the lesion site. The expression of tgfb1a and col1a1a, as well as the protein expression of Fibronectin, were all down-regulated under sdc4 knockdown. In addition, we verified that sdc4 expression was required for cardiac repair in zebrafish via in vivo electrocardiogram analysis. Loss of sdc4 expression caused an apparent pathological Q wave and ST elevation, which are signs of human MI patients. CONCLUSIONS Our findings support that Sdc4 is required to mediate pleiotropic repair responses in the early stage of zebrafish heart regeneration.
Collapse
Grants
- MOST 110-2311-B-007-005-MY3 Ministry of Science and Technology, Taiwan
- MOST 110-2311-B-007-005-MY3 Ministry of Science and Technology, Taiwan
- MOST 110-2311-B-007-005-MY3 Ministry of Science and Technology, Taiwan
- MOST 110-2311-B-007-005-MY3 Ministry of Science and Technology, Taiwan
- MOST 110-2311-B-007-005-MY3 Ministry of Science and Technology, Taiwan
- MOST 110-2311-B-007-005-MY3 Ministry of Science and Technology, Taiwan
- MOST 110-2311-B-007-005-MY3 Ministry of Science and Technology, Taiwan
- MOST 110-2311-B-007-005-MY3 Ministry of Science and Technology, Taiwan
- MOST 110-2311-B-007-005-MY3 Ministry of Science and Technology, Taiwan
- MOST 110-2311-B-007-005-MY3 Ministry of Science and Technology, Taiwan
- MOST 110-2311-B-007-005-MY3 Ministry of Science and Technology, Taiwan
- MOST 110-2311-B-007-005-MY3 Ministry of Science and Technology, Taiwan
Collapse
Affiliation(s)
- Zih-Yin Lai
- School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Chung-Chi Yang
- School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
- Division of Cardiovascular Medicine, Taoyuan Armed Forces General Hospital, Taoyuan City, 325208, Taiwan, ROC
- Cardiovascular Division, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114201, Taiwan, ROC
| | - Po-Hsun Chen
- School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Wei-Chen Chen
- School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Ting-Yu Lai
- School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Guan-Yun Lu
- School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Chiao-Yu Yang
- School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Ko-Ying Wang
- School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Wei-Cen Liu
- School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Yu-Chieh Chen
- School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Lawrence Yu-Min Liu
- Department of Internal Medicine, Division of Cardiology, Hsinchu MacKay Memorial Hospital, Hsinchu, 300044, Taiwan, ROC.
- Department of Medicine, MacKay Medical College, New Taipei City, 252005, Taiwan, ROC.
| | - Yung-Jen Chuang
- School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC.
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC.
| |
Collapse
|
4
|
Tan WH, Winkler C. Lineage Tracing of Bone Cells in the Regenerating Fin and During Repair of Bone Lesions. Methods Mol Biol 2024; 2707:99-110. [PMID: 37668907 DOI: 10.1007/978-1-0716-3401-1_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Small teleost fishes such as zebrafish and medaka show remarkable regeneration capabilities upon tissue injury or amputation. To elucidate cellular mechanisms of teleost tissue repair and regeneration processes, the Cre/LoxP recombination system for cell lineage tracing is a widely used technique. In this chapter, we describe protocols used for inducible Cre/LoxP recombination-mediated lineage tracing of osteoblast progenitors during medaka fin regeneration as well as during the repair of osteoporosis-like bone lesions in the medaka vertebral column. Our approach can be adapted for lineage tracing of other cell populations in the regenerating teleost fin or in other tissues undergoing repair.
Collapse
Affiliation(s)
- Wen Hui Tan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
| | - Christoph Winkler
- Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
5
|
Tajer B, Savage AM, Whited JL. The salamander blastema within the broader context of metazoan regeneration. Front Cell Dev Biol 2023; 11:1206157. [PMID: 37635872 PMCID: PMC10450636 DOI: 10.3389/fcell.2023.1206157] [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: 04/15/2023] [Accepted: 07/26/2023] [Indexed: 08/29/2023] Open
Abstract
Throughout the animal kingdom regenerative ability varies greatly from species to species, and even tissue to tissue within the same organism. The sheer diversity of structures and mechanisms renders a thorough comparison of molecular processes truly daunting. Are "blastemas" found in organisms as distantly related as planarians and axolotls derived from the same ancestral process, or did they arise convergently and independently? Is a mouse digit tip blastema orthologous to a salamander limb blastema? In other fields, the thorough characterization of a reference model has greatly facilitated these comparisons. For example, the amphibian Spemann-Mangold organizer has served as an amazingly useful comparative template within the field of developmental biology, allowing researchers to draw analogies between distantly related species, and developmental processes which are superficially quite different. The salamander limb blastema may serve as the best starting point for a comparative analysis of regeneration, as it has been characterized by over 200 years of research and is supported by a growing arsenal of molecular tools. The anatomical and evolutionary closeness of the salamander and human limb also add value from a translational and therapeutic standpoint. Tracing the evolutionary origins of the salamander blastema, and its relatedness to other regenerative processes throughout the animal kingdom, will both enhance our basic biological understanding of regeneration and inform our selection of regenerative model systems.
Collapse
Affiliation(s)
| | | | - Jessica L. Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, United States
| |
Collapse
|
6
|
Mehdipour M, Park S, Huang GN. Unlocking cardiomyocyte renewal potential for myocardial regeneration therapy. J Mol Cell Cardiol 2023; 177:9-20. [PMID: 36801396 PMCID: PMC10699255 DOI: 10.1016/j.yjmcc.2023.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/28/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Cardiovascular disease remains the leading cause of mortality worldwide. Cardiomyocytes are irreversibly lost due to cardiac ischemia secondary to disease. This leads to increased cardiac fibrosis, poor contractility, cardiac hypertrophy, and subsequent life-threatening heart failure. Adult mammalian hearts exhibit notoriously low regenerative potential, further compounding the calamities described above. Neonatal mammalian hearts, on the other hand, display robust regenerative capacities. Lower vertebrates such as zebrafish and salamanders retain the ability to replenish lost cardiomyocytes throughout life. It is critical to understand the varying mechanisms that are responsible for these differences in cardiac regeneration across phylogeny and ontogeny. Adult mammalian cardiomyocyte cell cycle arrest and polyploidization have been proposed as major barriers to heart regeneration. Here we review current models about why adult mammalian cardiac regenerative potential is lost including changes in environmental oxygen levels, acquisition of endothermy, complex immune system development, and possible cancer risk tradeoffs. We also discuss recent progress and highlight conflicting reports pertaining to extrinsic and intrinsic signaling pathways that control cardiomyocyte proliferation and polyploidization in growth and regeneration. Uncovering the physiological brakes of cardiac regeneration could illuminate novel molecular targets and offer promising therapeutic strategies to treat heart failure.
Collapse
Affiliation(s)
- Melod Mehdipour
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94158, USA; Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sangsoon Park
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94158, USA; Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Guo N Huang
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94158, USA; Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
7
|
Sorbini M, Arab S, Soni T, Frisiras A, Mehta S. How can the adult zebrafish and neonatal mice teach us about stimulating cardiac regeneration in the human heart? Regen Med 2023; 18:85-99. [PMID: 36416596 DOI: 10.2217/rme-2022-0161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The proliferative capacity of mammalian cardiomyocytes diminishes shortly after birth. In contrast, adult zebrafish and neonatal mice can regenerate cardiac tissues, highlighting new potential therapeutic avenues. Different factors have been found to promote cardiomyocyte proliferation in zebrafish and neonatal mice; these include maintenance of mononuclear and diploid cardiomyocytes and upregulation of the proto-oncogene c-Myc. The growth factor NRG-1 controls cell proliferation and interacts with the Hippo-Yap pathway to modulate regeneration. Key components of the extracellular matrix such as Agrin are also crucial for cardiac regeneration. Novel therapies explored in this review, include intramyocardial injection of Agrin or zebrafish-ECM and NRG-1 administration. These therapies may induce regeneration in patients and should be further explored.
Collapse
Affiliation(s)
- Michela Sorbini
- Barts and the London School of Medicien and Dentistry, Queen Mary University of London, E1 2AD, London, UK.,Imperial College School of Medicine, SW7 2AZ, London, UK
| | - Sammy Arab
- Imperial College School of Medicine, SW7 2AZ, London, UK
| | - Tara Soni
- Imperial College School of Medicine, SW7 2AZ, London, UK
| | | | - Samay Mehta
- Imperial College School of Medicine, SW7 2AZ, London, UK
| |
Collapse
|
8
|
Hendin N, Gordon T, Shenkar N, Wurtzel O. Molecular characterization of the immediate wound response of the solitary ascidian Polycarpa mytiligera. Dev Dyn 2022; 251:1968-1981. [PMID: 36001356 PMCID: PMC10087333 DOI: 10.1002/dvdy.526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 06/16/2022] [Accepted: 07/05/2022] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Injury response is key to successful regeneration. Yet, transcriptome analyses of injury response were performed only on a handful of regenerative organisms. Here, we studied the injury response of the solitary ascidian Polycarpa mytiligera, an emerging model system, capable of regenerating any body part. We used the siphon as a model for studying transcriptional changes following injury, and identified genes that were activated in the initial 24 hours post amputation (hpa). RESULTS Highly conserved genes, such as bone morphogenetic protein-1 (BMP1), growth hormone secretagogue receptor (GHSR) and IL-17, were upregulated by 12 hpa, yet their expression was sustained only in non-regenerating tissue fragments. We optimized fluorescent in situ hybridization, and found that the majority of BMP1+ cells were localized to the rigid tunic that covers the animal. This highlights the importance of this tissue, particularly during injury response. BMP1 was overexpressed following injuries to other body regions, suggesting that it was a part of a common injury-induced program. CONCLUSION Our study suggests that, initially, specific injury-induced genes were upregulated in P. mytiligera organs, yet, later, a unique transcriptional profile was observed only in regenerating tissues. These findings highlight the importance of studying diverse regenerating and non-regenerating organisms for complete understanding of regeneration.
Collapse
Affiliation(s)
- Noam Hendin
- The School of Neurobiology, Biochemistry & Biophysics, George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
| | - Tal Gordon
- The School of Neurobiology, Biochemistry & Biophysics, George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
| | - Noa Shenkar
- School of Zoology, George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
- The Steinhardt Museum of Natural History, Israel National Center for Biodiversity StudiesTel‐Aviv UniversityTel‐AvivIsrael
| | - Omri Wurtzel
- The School of Neurobiology, Biochemistry & Biophysics, George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
9
|
Qian C, Zhang Z, Zhao R, Wang D, Li H. Effect of acellular nerve scaffold containing human umbilical cord-derived mesenchymal stem cells on nerve repair and regeneration in rats with sciatic nerve defect. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:483. [PMID: 35571424 PMCID: PMC9096419 DOI: 10.21037/atm-22-1578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/20/2022] [Indexed: 11/11/2022]
Abstract
Background The aim of the present study was to investigate the effect of acellular nerve scaffold (ANS) containing human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) on nerve repair and regeneration in rats with sciatic nerve defect. Methods Sciatic nerve trunks were removed from 6 female Sprague-Dawley (SD) rats, and ANS was prepared by lyophilization + enzymatic method and divided into A, B, C, D and E groups according to different treatment times. hUC-MSCs were isolated from the collected umbilical cords and cultured, and then ANS-hUC-MSCs complexes were made. The other 24 adult female SD rats were randomly divided into the control, autograft, ANS, and ANS-hUC-MSCs groups, and a rat model of sciatic nerve defect was established. Hematoxylin-eosin (HE) staining, Luxol fast blue (LFB) staining, Masson staining, and scanning electron microscopy were used to observe the morphology and tissue structure of ANS. The performance of ANS was evaluated by mechanical detection, and hydroxyproline (HYP) content was evaluated using a biochemical kit. Flow cytometry was adopted to detect the levels of hUC-MSCs surface antigens CD29, CD44, and CD34, as well as electrophysiological detection and muscle wet weight recovery rate for measuring rat muscle performance. Results ANS was prepared according to group A method and had good mechanical properties, with less residues of cells and myelin, and higher HYP content, indicating that this scaffold had the best performance. ANS-hUC-MSCs significantly reduced myelin injury in the sciatic nerve, and increased axonal regeneration, effectively improving sciatic nerve injury in rats. In addition, ANS-hUC-MSCs significantly increased compound muscle action potential (CMAP), nerve conduction velocity (NCV), and muscle wet weight, and reduced muscle atrophy. Conclusions ANS containing hUC-MSCs can promote nerve repair and regeneration in rats with sciatic nerve defects.
Collapse
Affiliation(s)
- Chuang Qian
- Department of Orthopaedics, Children’s Hospital of Fudan University and National Children’s Medical Center, Shanghai, China
| | - Zhiqiang Zhang
- Department of Orthopaedics, Children’s Hospital of Fudan University and National Children’s Medical Center, Shanghai, China
| | - Rui Zhao
- Department of Neurosurgery, Children’s Hospital of Fudan University and National Children’s Medical Center, Shanghai, China
| | - Dahui Wang
- Department of Orthopaedics, Children’s Hospital of Fudan University and National Children’s Medical Center, Shanghai, China
| | - Hao Li
- Department of Neurosurgery, Children’s Hospital of Fudan University and National Children’s Medical Center, Shanghai, China
| |
Collapse
|
10
|
Lavecchia AM, Pelekanos K, Mavelli F, Xinaris C. Cell Hypertrophy: A “Biophysical Roadblock” to Reversing Kidney Injury. Front Cell Dev Biol 2022; 10:854998. [PMID: 35309910 PMCID: PMC8927721 DOI: 10.3389/fcell.2022.854998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/10/2022] [Indexed: 11/24/2022] Open
Abstract
In anamniotes cell loss can typically be compensated for through proliferation, but in amniotes, this capacity has been significantly diminished to accommodate tissue complexity. In order to cope with the increased workload that results from cell death, instead of proliferation highly specialised post-mitotic cells undergo polyploidisation and hypertrophy. Although compensatory hypertrophy is the main strategy of repair/regeneration in various parenchymal tissues, the long-term benefits and its capacity to sustain complete recovery of the kidney has not been addressed sufficiently. In this perspective article we integrate basic principles from biophysics and biology to examine whether renal cell hypertrophy is a sustainable adaptation that can efficiently regenerate tissue mass and restore organ function, or a maladaptive detrimental response.
Collapse
Affiliation(s)
- Angelo Michele Lavecchia
- Laboratory of Organ Regeneration, Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy
| | | | - Fabio Mavelli
- Department of Chemistry, University of Bari Aldo Moro, Bari, Italy
| | - Christodoulos Xinaris
- Laboratory of Organ Regeneration, Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy
- *Correspondence: Christodoulos Xinaris,
| |
Collapse
|
11
|
Wen X, Jiao L, Tan H. MAPK/ERK Pathway as a Central Regulator in Vertebrate Organ Regeneration. Int J Mol Sci 2022; 23:ijms23031464. [PMID: 35163418 PMCID: PMC8835994 DOI: 10.3390/ijms23031464] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023] Open
Abstract
Damage to organs by trauma, infection, diseases, congenital defects, aging, and other injuries causes organ malfunction and is life-threatening under serious conditions. Some of the lower order vertebrates such as zebrafish, salamanders, and chicks possess superior organ regenerative capacity over mammals. The extracellular signal-regulated kinases 1 and 2 (ERK1/2), as key members of the mitogen-activated protein kinase (MAPK) family, are serine/threonine protein kinases that are phylogenetically conserved among vertebrate taxa. MAPK/ERK signaling is an irreplaceable player participating in diverse biological activities through phosphorylating a broad variety of substrates in the cytoplasm as well as inside the nucleus. Current evidence supports a central role of the MAPK/ERK pathway during organ regeneration processes. MAPK/ERK signaling is rapidly excited in response to injury stimuli and coordinates essential pro-regenerative cellular events including cell survival, cell fate turnover, migration, proliferation, growth, and transcriptional and translational activities. In this literature review, we recapitulated the multifaceted MAPK/ERK signaling regulations, its dynamic spatio-temporal activities, and the profound roles during multiple organ regeneration, including appendages, heart, liver, eye, and peripheral/central nervous system, illuminating the possibility of MAPK/ERK signaling as a critical mechanism underlying the vastly differential regenerative capacities among vertebrate species, as well as its potential applications in tissue engineering and regenerative medicine.
Collapse
|
12
|
Bijarchian F, Taghiyar L, Azhdari Z, Baghaban Eslaminejad M. M2c Macrophages enhance phalange regeneration of amputated mice digits in an organ co-culture system. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:1602-1612. [PMID: 35317116 PMCID: PMC8917845 DOI: 10.22038/ijbms.2021.57887.12870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 10/26/2021] [Indexed: 11/17/2022]
Abstract
Objectives Delayed anti-inflammatory responses and scar-formation are the main causes for inability of injured body parts such as phalanges to regrow in mammals. Salamanders can regenerate fully scar-free body structures, followed by the appearance of anti-inflammatory responses at the injured site immediately after amputation. This study aimed to evaluate the local regenerative effects of direct amplified anti-inflammatory signals on regeneration of amputated mice digit tips using M2c-macrophages in a co-cultured organ system for the first time. Materials and Methods We used the amputated digits from the paws of 18.5E day old C57BL/6J mice. Monocytes were obtained from peripheral blood and co-cultured with amputated digits, which subsequently enhanced the M2c macrophage phenotype induced by IL-10. We also examined the regenerative effects of IL-10 and transcription growth factor-beta 1 (TGF-β1). Results The regrowth of new tissue occurred 10 days post-amputation in all groups. This regrowth was related to enhanced Msh homeobox-1 (Msx1), Msh homeobox-2 (Msx2), and bone morphogenic protein-4 (Bmp4) genes. Increased expression of fibroblast growth factor-8 (Fgf-8) also increased the proliferation rate. Histological analyses indicated that epidermal-closure occurred at 3-dpa in all groups. We observed full digit tip regeneration in the co-cultured group. Particularly, there was new tissue regrowth observed with 40 µg/ml of IL-10 and 120 µg/ml of TGF-β. In contrast, the control group had no remarkable digit elongation. Conclusion We propose that a direct amplified anti-inflammatory response at the digit injury site can regenerate epithelial and mesenchymal tissues, and might be useful for limb regeneration without scar formation in adult mammals.
Collapse
Affiliation(s)
- Fatemeh Bijarchian
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Leila Taghiyar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Zahra Azhdari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran,Corresponding author: Mohamadreza Baghaban Eslaminejad. Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem Street, Tehran, Iran. Tel: +98-21-23562524; Fax: +98-21-23562507;
| |
Collapse
|
13
|
Womersley F, Hancock J, Perry CT, Rowat D. Wound-healing capabilities of whale sharks ( Rhincodon typus) and implications for conservation management. CONSERVATION PHYSIOLOGY 2021; 9:coaa120. [PMID: 33569175 PMCID: PMC7859907 DOI: 10.1093/conphys/coaa120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 10/08/2020] [Accepted: 12/03/2020] [Indexed: 05/03/2023]
Abstract
Wound healing is important for marine taxa such as elasmobranchs, which can incur a range of natural and anthropogenic wounds throughout their life history. There is evidence that this group shows a high capacity for external wound healing. However, anthropogenic wounds may become more frequent due to increasing commercial and recreational marine activities. Whale sharks are particularly at risk of attaining injuries given their use of surface waters and wildlife tourism interest. There is limited understanding as to how whale sharks recover from injuries, and often insights are confined to singular opportunistic observations. The present study makes use of a unique and valuable photographic data source from two whale shark aggregation sites in the Indian Ocean. Successional injury-healing progression cases were reviewed to investigate the characteristics of injuries and quantify a coarse healing timeframe. Wounds were measured over time using an image standardization method. This work shows that by Day 25 major injury surface area decreased by an average of 56% and the most rapid healing case showed a surface area reduction of 50% in 4 days. All wounds reached a point of 90% surface area closure by Day 35. There were differences in healing rate based on wound type, with lacerations and abrasions taking 50 and 22 days to reach 90% healing, respectively. This study provides baseline information for wound healing in whale sharks and the methods proposed could act as a foundation for future research. Use of a detailed classification system, as presented here, may also assist in ocean scale injury comparisons between research groups and aid reliable descriptive data. Such findings can contribute to discussions regarding appropriate management in aggregation areas with an aim to reduce the likelihood of injuries, such as those resulting from vessel collisions, in these regions or during movements between coastal waters.
Collapse
Affiliation(s)
- Freya Womersley
- Marine Biological Association of the United Kingdom, The Laboratory, Citadel Hill, Plymouth, PL1 2PB, UK
- Ocean and Earth Science, National Oceanography Centre Southampton, University of Southampton, Southampton, SO17 1BJ, UK
- Marine Conservation Society Seychelles, Mahé, PO Box 384, Seychelles
- Corresponding author: Marine Biological Association of the United Kingdom, The Laboratory, Citadel Hill, Plymouth, PL1 2PB, UK.
| | - James Hancock
- Maldives Whale Shark Research Programme, Popeshead Court Offices, Peter Lane, York, Yorkshire, Y01 8SU, UK
| | - Cameron T Perry
- Maldives Whale Shark Research Programme, Popeshead Court Offices, Peter Lane, York, Yorkshire, Y01 8SU, UK
| | - David Rowat
- Marine Conservation Society Seychelles, Mahé, PO Box 384, Seychelles
| |
Collapse
|
14
|
Rodrigues SC, Cardoso RMS, Duarte FV. Mitochondrial microRNAs: A Putative Role in Tissue Regeneration. BIOLOGY 2020; 9:biology9120486. [PMID: 33371511 PMCID: PMC7767490 DOI: 10.3390/biology9120486] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/16/2020] [Accepted: 12/19/2020] [Indexed: 12/12/2022]
Abstract
The most famous role of mitochondria is to generate ATP through oxidative phosphorylation, a metabolic pathway that involves a chain of four protein complexes (the electron transport chain, ETC) that generates a proton-motive force that in turn drives the ATP synthesis by the Complex V (ATP synthase). An impressive number of more than 1000 mitochondrial proteins have been discovered. Since mitochondrial proteins have a dual genetic origin, it is predicted that ~99% of these proteins are nuclear-encoded and are synthesized in the cytoplasmatic compartment, being further imported through mitochondrial membrane transporters. The lasting 1% of mitochondrial proteins are encoded by the mitochondrial genome and synthesized by the mitochondrial ribosome (mitoribosome). As a result, an appropriate regulation of mitochondrial protein synthesis is absolutely required to achieve and maintain normal mitochondrial function. Regarding miRNAs in mitochondria, it is well-recognized nowadays that several cellular mechanisms involving mitochondria are regulated by many genetic players that originate from either nuclear- or mitochondrial-encoded small noncoding RNAs (sncRNAs). Growing evidence collected from whole genome and transcriptome sequencing highlight the role of distinct members of this class, from short interfering RNAs (siRNAs) to miRNAs and long noncoding RNAs (lncRNAs). Some of the mechanisms that have been shown to be modulated are the expression of mitochondrial proteins itself, as well as the more complex coordination of mitochondrial structure and dynamics with its function. We devote particular attention to the role of mitochondrial miRNAs and to their role in the modulation of several molecular processes that could ultimately contribute to tissue regeneration accomplishment.
Collapse
Affiliation(s)
- Sílvia C. Rodrigues
- Exogenus Therapeutics, 3060-197 Cantanhede, Portugal;
- Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-504 Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | | | - Filipe V. Duarte
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Correspondence:
| |
Collapse
|
15
|
The Future of Direct Cardiac Reprogramming: Any GMT Cocktail Variety? Int J Mol Sci 2020; 21:ijms21217950. [PMID: 33114756 PMCID: PMC7663133 DOI: 10.3390/ijms21217950] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/13/2022] Open
Abstract
Direct cardiac reprogramming has emerged as a novel therapeutic approach to treat and regenerate injured hearts through the direct conversion of fibroblasts into cardiac cells. Most studies have focused on the reprogramming of fibroblasts into induced cardiomyocytes (iCMs). The first study in which this technology was described, showed that at least a combination of three transcription factors, GATA4, MEF2C and TBX5 (GMT cocktail), was required for the reprogramming into iCMs in vitro using mouse cells. However, this was later demonstrated to be insufficient for the reprogramming of human cells and additional factors were required. Thereafter, most studies have focused on implementing reprogramming efficiency and obtaining fully reprogrammed and functional iCMs, by the incorporation of other transcription factors, microRNAs or small molecules to the original GMT cocktail. In this respect, great advances have been made in recent years. However, there is still no consensus on which of these GMT-based varieties is best, and robust and highly reproducible protocols are still urgently required, especially in the case of human cells. On the other hand, apart from CMs, other cells such as endothelial and smooth muscle cells to form new blood vessels will be fundamental for the correct reconstruction of damaged cardiac tissue. With this aim, several studies have centered on the direct reprogramming of fibroblasts into induced cardiac progenitor cells (iCPCs) able to give rise to all myocardial cell lineages. Especially interesting are reports in which multipotent and highly expandable mouse iCPCs have been obtained, suggesting that clinically relevant amounts of these cells could be created. However, as of yet, this has not been achieved with human iCPCs, and exactly what stage of maturity is appropriate for a cell therapy product remains an open question. Nonetheless, the major concern in regenerative medicine is the poor retention, survival, and engraftment of transplanted cells in the cardiac tissue. To circumvent this issue, several cell pre-conditioning approaches are currently being explored. As an alternative to cell injection, in vivo reprogramming may face fewer barriers for its translation to the clinic. This approach has achieved better results in terms of efficiency and iCMs maturity in mouse models, indicating that the heart environment can favor this process. In this context, in recent years some studies have focused on the development of safer delivery systems such as Sendai virus, Adenovirus, chemical cocktails or nanoparticles. This article provides an in-depth review of the in vitro and in vivo cardiac reprograming technology used in mouse and human cells to obtain iCMs and iCPCs, and discusses what challenges still lie ahead and what hurdles are to be overcome before results from this field can be transferred to the clinical settings.
Collapse
|
16
|
Cadiz L, Jonz MG. A comparative perspective on lung and gill regeneration. ACTA ACUST UNITED AC 2020; 223:223/19/jeb226076. [PMID: 33037099 DOI: 10.1242/jeb.226076] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The ability to continuously grow and regenerate the gills throughout life is a remarkable property of fish and amphibians. Considering that gill regeneration was first described over one century ago, it is surprising that the underlying mechanisms of cell and tissue replacement in the gills remain poorly understood. By contrast, the mammalian lung is a largely quiescent organ in adults but is capable of facultative regeneration following injury. In the course of the past decade, it has been recognized that lungs contain a population of stem or progenitor cells with an extensive ability to restore tissue; however, despite recent advances in regenerative biology of the lung, the signaling pathways that underlie regeneration are poorly understood. In this Review, we discuss the common evolutionary and embryological origins shared by gills and mammalian lungs. These are evident in homologies in tissue structure, cell populations, cellular function and genetic pathways. An integration of the literature on gill and lung regeneration in vertebrates is presented using a comparative approach in order to outline the challenges that remain in these areas, and to highlight the importance of using aquatic vertebrates as model organisms. The study of gill regeneration in fish and amphibians, which have a high regenerative potential and for which genetic tools are widely available, represents a unique opportunity to uncover common signaling mechanisms that may be important for regeneration of respiratory organs in all vertebrates. This may lead to new advances in tissue repair following lung disease.
Collapse
Affiliation(s)
- Laura Cadiz
- Department of Biology, University of Ottawa, 30 Marie Curie Pvt., Ottawa, ON, Canada, K1N 6N5
| | - Michael G Jonz
- Department of Biology, University of Ottawa, 30 Marie Curie Pvt., Ottawa, ON, Canada, K1N 6N5
| |
Collapse
|
17
|
Alibardi L. Appendage regeneration in anamniotes utilizes genes active during larval-metamorphic stages that have been lost or altered in amniotes: The case for studying lizard tail regeneration. J Morphol 2020; 281:1358-1381. [PMID: 32865265 DOI: 10.1002/jmor.21251] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/20/2020] [Accepted: 07/25/2020] [Indexed: 12/17/2022]
Abstract
This review elaborates the idea that organ regeneration derives from specific evolutionary histories of vertebrates. Regenerative ability depends on genomic regulation of genes specific to the life-cycles that have differentially evolved in anamniotes and amniotes. In aquatic environments, where fish and amphibians live, one or multiple metamorphic transitions occur before the adult stage is reached. Each transition involves the destruction and remodeling of larval organs that are replaced with adult organs. After organ injury or loss in adult anamniotes, regeneration uses similar genes and developmental process than those operating during larval growth and metamorphosis. Therefore, the broad presence of regenerative capability across anamniotes is possible because generating new organs is included in their life history at metamorphic stages. Soft hyaluronate-rich regenerative blastemas grow in submersed or in hydrated environments, that is, essential conditions for regeneration, like during development. In adult anamniotes, the ability to regenerate different organs decreases in comparison to larval stages and becomes limited during aging. Comparisons of genes activated during metamorphosis and regeneration in anamniotes identify key genes unique to these processes, and include thyroid, wnt and non-coding RNAs developmental pathways. In the terrestrial environment, some genes or developmental pathways for metamorphic transitions were lost during amniote evolution, determining loss of regeneration. Among amniotes, the formation of soft and hydrated blastemas only occurs in lizards, a morphogenetic process that evolved favoring their survival through tail autotomy, leading to a massive although imperfect regeneration of the tail. Deciphering genes activity during lizard tail regeneration would address future attempts to recreate in other amniotes regenerative blastemas that grow into variably completed organs.
Collapse
|
18
|
The effects of Piper sarmentosum aqueous extracts on zebrafish (Danio rerio) embryos and caudal fin tissue regeneration. Sci Rep 2020; 10:14165. [PMID: 32843675 PMCID: PMC7447815 DOI: 10.1038/s41598-020-70962-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 08/06/2020] [Indexed: 11/30/2022] Open
Abstract
In Malaysia, Piper sarmentosum or ‘kaduk’ is commonly used in traditional medicines. However, its biological effects including in vivo embryonic toxicity and tissue regenerative properties are relatively unknown. The purpose of this study was to determine zebrafish (Danio rerio) embryo toxicities and caudal fin tissue regeneration in the presence of P. sarmentosum aqueous extracts. The phytochemical components and antioxidant activity of the extract were studied using GC–MS analysis and DPPH assay, respectively. Embryo toxicity tests involving survival, heartbeat, and morphological analyses were conducted to determine P. sarmentosum extract toxicity (0–60 µg/mL); concentrations of 0–400 µg/mL of the extract were used to study tissue regeneration in the zebrafish caudal fin. The extract contained several phytochemicals with antioxidant activity and exhibited DPPH scavenging activity (IC50 = 50.56 mg/mL). Embryo toxicity assays showed that a concentration of 60 μg/mL showed the highest rates of lethality regardless of exposure time. Slower embryogenesis was observed at 40 µg/mL, with non-viable embryos first detected at 50 µg/mL. Extracts showed significant differences (p < 0.01) for tissue regeneration at all concentrations when compared to non-treated samples. In conclusion, Piper sarmentosum extracts accelerated tissue regeneration, and extract concentrations at 60 µg/mL showed the highest toxicity levels for embryo viability.
Collapse
|
19
|
de Wit L, Fang J, Neef K, Xiao J, A. Doevendans P, Schiffelers RM, Lei Z, Sluijter JP. Cellular and Molecular Mechanism of Cardiac Regeneration: A Comparison of Newts, Zebrafish, and Mammals. Biomolecules 2020; 10:biom10091204. [PMID: 32825069 PMCID: PMC7564143 DOI: 10.3390/biom10091204] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/06/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide. Current palliative treatments can slow the progression of heart failure, but ultimately, the only curative treatment for end-stage heart failure is heart transplantation, which is only available for a minority of patients due to lack of donors' hearts. Explorative research has shown the replacement of the damaged and lost myocardium by inducing cardiac regeneration from preexisting myocardial cells. Lower vertebrates, such as the newt and zebrafish, can regenerate lost myocardium through cardiomyocyte proliferation. The preexisting adult cardiomyocytes replace the lost cells through subsequent dedifferentiation, proliferation, migration, and re-differentiation. Similarly, neonatal mice show complete cardiac regeneration post-injury; however, this regenerative capacity is remarkably diminished one week after birth. In contrast, the adult mammalian heart presents a fibrotic rather than a regenerative response and only shows signs of partial pathological cardiomyocyte dedifferentiation after injury. In this review, we explore the cellular and molecular responses to myocardial insults in different adult species to give insights for future interventional directions by which one can promote or activate cardiac regeneration in mammals.
Collapse
Affiliation(s)
- Lousanne de Wit
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
| | - Juntao Fang
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
| | - Klaus Neef
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
- UMC Utrecht RM Center, Circulatory Health Laboratory, 3584CT Utrecht, The Netherlands
| | - Junjie Xiao
- Institute of Cardiovascular Sciences, Shanghai University, Shanghai 200444, China;
| | - Pieter A. Doevendans
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
- Utrecht University, 3584CS Utrecht, The Netherlands
- Netherlands Heart Institute (NHI), Central Military Hospital (CMH), 3511EP Utrecht, The Netherlands
| | | | - Zhiyong Lei
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
- Division LAB, CDL Research, UMC Utrecht, 3584CX Utrecht, The Netherlands;
- Correspondence: (Z.L.); (J.P.G.S.)
| | - Joost P.G. Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
- UMC Utrecht RM Center, Circulatory Health Laboratory, 3584CT Utrecht, The Netherlands
- Utrecht University, 3584CS Utrecht, The Netherlands
- Correspondence: (Z.L.); (J.P.G.S.)
| |
Collapse
|
20
|
Multiple cryoinjuries modulate the efficiency of zebrafish heart regeneration. Sci Rep 2020; 10:11551. [PMID: 32665622 PMCID: PMC7360767 DOI: 10.1038/s41598-020-68200-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/18/2020] [Indexed: 01/18/2023] Open
Abstract
Zebrafish can regenerate their damaged hearts throughout their lifespan. It is, however, unknown, whether regeneration remains effective when challenged with successive cycles of cardiac damage in the same animals. Here, we assessed ventricular restoration after two, three and six cryoinjuries interspaced by recovery periods. Using transgenic cell-lineage tracing analysis, we demonstrated that the second cryoinjury damages the regenerated area from the preceding injury, validating the experimental approach. We identified that after multiple cryoinjuries, all hearts regrow a thickened myocardium, similarly to hearts after one cryoinjury. However, the efficiency of scar resorption decreased with the number of repeated cryoinjuries. After six cryoinjuries, all examined hearts failed to completely resolve the fibrotic tissue, demonstrating reduced myocardial restoration. This phenotype was associated with enhanced recruitment of neutrophils and decreased cardiomyocyte proliferation and dedifferentiation at the early regenerative phase. Furthermore, we found that each repeated cryoinjury increased the accumulation of collagen at the injury site. Our analysis demonstrates that the cardiac regenerative program can be successfully activated many times, despite a persisting scar in the wounded area. This finding provides a new perspective for regenerative therapies, aiming in stimulation of organ regeneration in the presence of fibrotic tissue in mammalian models and humans.
Collapse
|
21
|
Midkine-a functions as a universal regulator of proliferation during epimorphic regeneration in adult zebrafish. PLoS One 2020; 15:e0232308. [PMID: 32530962 PMCID: PMC7292404 DOI: 10.1371/journal.pone.0232308] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022] Open
Abstract
Zebrafish have the ability to regenerate damaged cells and tissues by activating quiescent stem and progenitor cells or reprogramming differentiated cells into regeneration-competent precursors. Proliferation among the cells that will functionally restore injured tissues is a fundamental biological process underlying regeneration. Midkine-a is a cytokine growth factor, whose expression is strongly induced by injury in a variety of tissues across a range of vertebrate classes. Using a zebrafish Midkine-a loss of function mutant, we evaluated regeneration of caudal fin, extraocular muscle and retinal neurons to investigate the function of Midkine-a during epimorphic regeneration. In wildtype zebrafish, injury among these tissues induces robust proliferation and rapid regeneration. In Midkine-a mutants, the initial proliferation in each of these tissues is significantly diminished or absent. Regeneration of the caudal fin and extraocular muscle is delayed; regeneration of the retina is nearly completely absent. These data demonstrate that Midkine-a is universally required in the signaling pathways that convert tissue injury into the initial burst of cell proliferation. Further, these data highlight differences in the molecular mechanisms that regulate epimorphic regeneration in zebrafish.
Collapse
|
22
|
Jaźwińska A, Blanchoud S. Towards deciphering variations of heart regeneration in fish. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2019.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
23
|
Lv L, Liao Z, Luo J, Chen H, Guo H, Yang J, Huang R, Pu Q, Zhao H, Yuan Z, Feng S, Qi X, Cai D. Cardiac telocytes exist in the adult Xenopus tropicalis heart. J Cell Mol Med 2020; 24:2531-2541. [PMID: 31930692 PMCID: PMC7028868 DOI: 10.1111/jcmm.14947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 12/02/2019] [Accepted: 12/05/2019] [Indexed: 12/16/2022] Open
Abstract
Recent research has revealed that cardiac telocytes (CTs) play an important role in cardiac physiopathology and the regeneration of injured myocardium. Recently, we reported that the adult Xenopus tropicalis heart can regenerate perfectly in a nearly scar‐free manner after injury via apical resection. However, whether telocytes exist in the X tropicalis heart and are affected in the regeneration of injured X tropicalis myocardium is still unknown. The present ultrastructural and immunofluorescent double staining results clearly showed that CTs exist in the X tropicalis myocardium. CTs in the X tropicalis myocardium were mainly twined around the surface of cardiomyocyte trabeculae and linked via nanocontacts between the ends of the telopodes, forming a three‐dimensional network. CTs might play a role in the regeneration of injured myocardium.
Collapse
Affiliation(s)
- Luocheng Lv
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China.,Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China.,International Base of Collaboration for Science and Technology (JNU), Ministry of Science and Technology, Guangdong Province, Guangzhou, China.,Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Zhaofu Liao
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China.,Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China.,International Base of Collaboration for Science and Technology (JNU), Ministry of Science and Technology, Guangdong Province, Guangzhou, China.,Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Jiali Luo
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China.,Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China.,International Base of Collaboration for Science and Technology (JNU), Ministry of Science and Technology, Guangdong Province, Guangzhou, China.,Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Hongyi Chen
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China.,Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China.,International Base of Collaboration for Science and Technology (JNU), Ministry of Science and Technology, Guangdong Province, Guangzhou, China.,Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Hongyan Guo
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China.,Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China.,International Base of Collaboration for Science and Technology (JNU), Ministry of Science and Technology, Guangdong Province, Guangzhou, China.,Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Jifeng Yang
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China.,Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China.,International Base of Collaboration for Science and Technology (JNU), Ministry of Science and Technology, Guangdong Province, Guangzhou, China.,Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Ruijin Huang
- Department of Neuroanatomy, Institute of Anatomy, University of Bonn, Bonn, Germany.,Department of Anatomy and Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
| | - Qin Pu
- Department of Neuroanatomy, Institute of Anatomy, University of Bonn, Bonn, Germany
| | - Hui Zhao
- Stem Cell and Regeneration TRP, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Ziqiang Yuan
- Department of Medical Oncology, Cancer Institute of New Jersey, Robert Wood Johnson of Medical School, New Brunswick, NJ, USA
| | - Shanshan Feng
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China.,Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China.,International Base of Collaboration for Science and Technology (JNU), Ministry of Science and Technology, Guangdong Province, Guangzhou, China.,Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Xufeng Qi
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China.,Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China.,International Base of Collaboration for Science and Technology (JNU), Ministry of Science and Technology, Guangdong Province, Guangzhou, China.,Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Dongqing Cai
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China.,Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China.,International Base of Collaboration for Science and Technology (JNU), Ministry of Science and Technology, Guangdong Province, Guangzhou, China.,Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| |
Collapse
|
24
|
Liu L, Tian D, Liu C, Yu K, Bai J. Metformin Enhances Functional Recovery of Peripheral Nerve in Rats with Sciatic Nerve Crush Injury. Med Sci Monit 2019; 25:10067-10076. [PMID: 31882570 PMCID: PMC6946044 DOI: 10.12659/msm.918277] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background The aim of this study was to explore the effect of metformin by inducing autophagy for enhancing functional recovery of peripheral nerve in rats with sciatic nerve crush injury. Material/Method Autophagy was determined by electron microscopy, immunofluorescence, and Western blot analysis. Motor function recovery was studied by the footprint intensity method. Axonal growth and regeneration were detected through Western blot while axonal remyelination was analysed through immunocytochemistry. Sensory and functional recovery were assessed by reflexive motor function analysis. Results The present study deciphered the role of autophagy induction by metformin in motor functions and peripheral nerve regeneration following sciatic nerve crush injury in rats. The process was detected by measuring autophagosomes and the expression of microtubule-associated protein 1A/1B-light chain 3 upon metformin treatment of sciatic nerve crush-injured rats. Neurobehavioral recovery by metformin was tested by CatWalk gait analysis, and we quantified expression of myelin basic protein MBP and neurofilament NF200 at the damage sight by immunoblotting. In metformin-treated injured rats, autophagy was upregulated, by which the number of dead cells was decreased. Motor function was also recovered after metformin treatment, which was accompanied by upregulation of MBP and NF200 through autophagy induction. Surprisingly, the motor regenerative capability was reduced by treatment with 3-methyl adenine (an autophagy inhibitor) in nerve-injured rats. Conclusions Our study revealed that pharmacological induction of autophagy has an important and active role in the regeneration of nerve and motor function regain.
Collapse
Affiliation(s)
- Lei Liu
- Department of Hand Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Dehu Tian
- Department of Hand Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Chunjie Liu
- Department of Hand Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Kunlun Yu
- Department of Hand Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Jiangbo Bai
- Department of Hand Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| |
Collapse
|
25
|
Alibardi L. Microscopic observations on amputated and scarring lizard digits show an intense inflammatory reaction. ZOOLOGY 2019; 139:125737. [PMID: 32062299 DOI: 10.1016/j.zool.2019.125737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/28/2019] [Accepted: 12/09/2019] [Indexed: 01/27/2023]
Abstract
The microscopic details of the failure of digit regeneration in lizards are not known. The present study reports some histological, ultrastructural and 5BrdU-immunohistochemical observations on healing digits after amputation in the lizard Podarcis muralis. At 7-12 days post-amputation, the stump of digits forms a multilayered wound epidermis covering a loose connective tissue that is invaded by granulocytes, macrophages and lymphocytes. In addition to macrophages also electron-pale multinuclear giant cells are seen underneath or penetrating the wound epidermis while osteoclasts are present in the degrading bone of the severed phalanges. Granulocytes and macrophages invading the wound epidermis indicate the formation of an intra-epidermal immune barrier beneath the scab where numerous bacteria remain entrapped. Immunofluorescence for 5BrdU reveals that few proliferating cells are present in the wound epidermis and the underlying connective tissue at 12 and 32 days post-amputation. Outgrowths of less than 1mm stop growing and at 32 days they appear scaling. Most of connective cells give rise to fibrocytes and large irregular collagen bundles, as is typical for scar tissue. In conclusion, like for the amputated limb, the intense inflammatory reaction and scarring here described after digit loss appears associated with immune cells invasion.
Collapse
Affiliation(s)
- Lorenzo Alibardi
- Comparative Histolab Padova and Dipartimento di Biologia, University of Bologna, via Semi 3, 40126 Bologna, Italy.
| |
Collapse
|
26
|
Pfefferli C, Jaźwińska A. Lymphatic vessels help mend broken hearts. eLife 2019; 8:52200. [PMID: 31709981 PMCID: PMC6845218 DOI: 10.7554/elife.52200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 10/22/2019] [Indexed: 12/18/2022] Open
Abstract
Experiments on zebrafish show that the regeneration of the heart after an injury is supported by lymphatic vessels.
Collapse
Affiliation(s)
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
27
|
Garcia-Puig A, Mosquera JL, Jiménez-Delgado S, García-Pastor C, Jorba I, Navajas D, Canals F, Raya A. Proteomics Analysis of Extracellular Matrix Remodeling During Zebrafish Heart Regeneration. Mol Cell Proteomics 2019; 18:1745-1755. [PMID: 31221719 PMCID: PMC6731076 DOI: 10.1074/mcp.ra118.001193] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 06/03/2019] [Indexed: 12/13/2022] Open
Abstract
Adult zebrafish, in contrast to mammals, are able to regenerate their hearts in response to injury or experimental amputation. Our understanding of the cellular and molecular bases that underlie this process, although fragmentary, has increased significantly over the last years. However, the role of the extracellular matrix (ECM) during zebrafish heart regeneration has been comparatively rarely explored. Here, we set out to characterize the ECM protein composition in adult zebrafish hearts, and whether it changed during the regenerative response. For this purpose, we first established a decellularization protocol of adult zebrafish ventricles that significantly enriched the yield of ECM proteins. We then performed proteomic analyses of decellularized control hearts and at different times of regeneration. Our results show a dynamic change in ECM protein composition, most evident at the earliest (7 days postamputation) time point analyzed. Regeneration associated with sharp increases in specific ECM proteins, and with an overall decrease in collagens and cytoskeletal proteins. We finally tested by atomic force microscopy that the changes in ECM composition translated to decreased ECM stiffness. Our cumulative results identify changes in the protein composition and mechanical properties of the zebrafish heart ECM during regeneration.
Collapse
Affiliation(s)
- Anna Garcia-Puig
- ‡Center of Regenerative Medicine in Barcelona (CMRB), 3rd Floor Hospital Duran i Reynals, Avinguda de la Gran Via 199-203, 08908 Hospitalet de Llobregat (Barcelona), Spain; §Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08908 Hospitalet de Llobregat (Barcelona), Spain
| | - Jose Luis Mosquera
- ¶Bioinformatics Unit, Institut d'Investigació Biomèdica de Bellvitge IDIBELL), 3rd Floor Hospital Duran i Reynals, Avinguda de la Gran Via 199-203, 08908 Hospitalet de Llobregat (Barcelona), Spain
| | - Senda Jiménez-Delgado
- ‡Center of Regenerative Medicine in Barcelona (CMRB), 3rd Floor Hospital Duran i Reynals, Avinguda de la Gran Via 199-203, 08908 Hospitalet de Llobregat (Barcelona), Spain
| | - Cristina García-Pastor
- ‡Center of Regenerative Medicine in Barcelona (CMRB), 3rd Floor Hospital Duran i Reynals, Avinguda de la Gran Via 199-203, 08908 Hospitalet de Llobregat (Barcelona), Spain
| | - Ignasi Jorba
- ‖Institute for Bioengineering of Catalonia (IBEC), Barcelona Science Park, Baldiri Reixac 15-21, 08028 Barcelona, Spain; **Unit of Biophysics and Bioengineering, Department of Physiological Sciences I, School of Medicine, University of Barcelona, Casanova 143, 08036 Barcelona, Spain; ‡‡Center for Networked Biomedical Research on Respiratory Diseases (CIBERES), 08036 Barcelona, Spain
| | - Daniel Navajas
- ‖Institute for Bioengineering of Catalonia (IBEC), Barcelona Science Park, Baldiri Reixac 15-21, 08028 Barcelona, Spain; **Unit of Biophysics and Bioengineering, Department of Physiological Sciences I, School of Medicine, University of Barcelona, Casanova 143, 08036 Barcelona, Spain; ‡‡Center for Networked Biomedical Research on Respiratory Diseases (CIBERES), 08036 Barcelona, Spain
| | - Francesc Canals
- §§Proteomics group, Vall d'Hebron Institut of Oncology (VHIO), Cellex center, Natzaret 115-117, 08035 Barcelona, Spain
| | - Angel Raya
- ‡Center of Regenerative Medicine in Barcelona (CMRB), 3rd Floor Hospital Duran i Reynals, Avinguda de la Gran Via 199-203, 08908 Hospitalet de Llobregat (Barcelona), Spain; §Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08908 Hospitalet de Llobregat (Barcelona), Spain; ¶¶Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain.
| |
Collapse
|
28
|
Paredes LC, Olsen Saraiva Camara N, Braga TT. Understanding the Metabolic Profile of Macrophages During the Regenerative Process in Zebrafish. Front Physiol 2019; 10:617. [PMID: 31178754 PMCID: PMC6543010 DOI: 10.3389/fphys.2019.00617] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/01/2019] [Indexed: 12/14/2022] Open
Abstract
In contrast to mammals, lower vertebrates, including zebrafish (Danio rerio), have the ability to regenerate damaged or lost tissues, such as the caudal fin, which makes them an ideal model for tissue and organ regeneration studies. Since several diseases involve the process of transition between fibrosis and tissue regeneration, it is necessary to attain a better understanding of these processes. It is known that the cells of the immune system, especially macrophages, play essential roles in regeneration by participating in the removal of cellular debris, release of pro- and anti-inflammatory factors, remodeling of components of the extracellular matrix and alteration of oxidative patterns during proliferation and angiogenesis. Immune cells undergo phenotypical and functional alterations throughout the healing process due to growth factors and cytokines that are produced in the tissue microenvironment. However, some aspects of the molecular mechanisms through which macrophages orchestrate the formation and regeneration of the blastema remain unclear. In the present review, we outline how macrophages orchestrate the regenerative process in zebrafish and give special attention to the redox balance in the context of tail regeneration.
Collapse
Affiliation(s)
| | - Niels Olsen Saraiva Camara
- Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo, São Paulo, Brazil.,Nephrology Division, Federal University of São Paulo, São Paulo, Brazil.,Renal Pathophysiology Laboratory, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
29
|
König D, Jaźwińska A. Zebrafish fin regeneration involves transient serotonin synthesis. Wound Repair Regen 2019; 27:375-385. [DOI: 10.1111/wrr.12719] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/13/2019] [Accepted: 04/02/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Désirée König
- Department of BiologyUniversity of Fribourg Chemin du Musée 10, 1700, Fribourg Switzerland
| | - Anna Jaźwińska
- Department of BiologyUniversity of Fribourg Chemin du Musée 10, 1700, Fribourg Switzerland
| |
Collapse
|
30
|
Ciliary neurotrophic factor stimulates cardioprotection and the proliferative activity in the adult zebrafish heart. NPJ Regen Med 2019; 4:2. [PMID: 30701084 PMCID: PMC6345746 DOI: 10.1038/s41536-019-0064-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 12/27/2018] [Indexed: 12/29/2022] Open
Abstract
Unlike mammals, adult zebrafish can regenerate their hearts after injury via proliferation of cardiomyocytes. The cell-cycle entry of zebrafish cardiac cells can also be stimulated through preconditioning by thoracotomy, a chest incision without myocardial damage. To identify effector genes of heart preconditioning, we performed transcriptome analysis of ventricles from thoracotomized zebrafish. This intervention led to enrichment of cardioprotective factors, epithelial-to-mesenchymal transition genes, matrix proteins and components of LIFR/gp130 signaling. We identified that inhibition of the downstream signal transducer of the LIFR/gp130 pathway through treatment with Ruxolitinib, a specific JAK1/2 antagonist, suppressed the cellular effects of preconditioning. Activation of LIFR/gp130 signaling by a single injection of the ligand Cilliary Neurotrophic Factor, CNTF, was sufficient to trigger cardiomyocyte proliferation in the intact heart. In addition, CNTF induced other pro-regenerative processes, including expression of cardioprotective genes, activation of the epicardium, enhanced intramyocardial Collagen XII deposition and leucocyte recruitment. These effects were abrogated by the concomitant inhibition of the JAK/STAT activity. Mutation of the cntf gene suppressed the proliferative response of cardiomyocytes after thoracotomy. In the regenerating zebrafish heart, CNTF injection prior to ventricular cryoinjury improved the initiation of regeneration via reduced cell apoptosis and boosted cardiomyocyte proliferation. Our findings reveal the molecular effectors of preconditioning and demonstrate that exogenous CNTF exerts beneficial regenerative effects by rendering the heart more resilient to injury and efficient in activation of the proliferative programs.
Collapse
|
31
|
Suzuki Y, Chou J, Garvey SL, Wang VR, Yanes KO. Evolution and Regulation of Limb Regeneration in Arthropods. Results Probl Cell Differ 2019; 68:419-454. [PMID: 31598866 DOI: 10.1007/978-3-030-23459-1_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Regeneration has fascinated both scientists and non-scientists for centuries. Many organisms can regenerate, and arthropod limbs are no exception although their ability to regenerate is a product shaped by natural and sexual selection. Recent studies have begun to uncover cellular and molecular processes underlying limb regeneration in several arthropod species. Here we argue that an evo-devo approach to the study of arthropod limb regeneration is needed to understand aspects of limb regeneration that are conserved and divergent. In particular, we argue that limbs of different species are comprised of cells at distinct stages of differentiation at the time of limb loss and therefore provide insights into regeneration involving both stem cell-like cells/precursor cells and differentiated cells. In addition, we review recent studies that demonstrate how limb regeneration impacts the development of the whole organism and argue that studies on the link between local tissue damage and the rest of the body should provide insights into the integrative nature of development. Molecular studies on limb regeneration are only beginning to take off, but comparative studies on the mechanisms of limb regeneration across various taxa should not only yield interesting insights into development but also answer how this remarkable ability evolved across arthropods and beyond.
Collapse
Affiliation(s)
- Yuichiro Suzuki
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA.
| | - Jacquelyn Chou
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Sarah L Garvey
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Victoria R Wang
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Katherine O Yanes
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| |
Collapse
|
32
|
Jacyniak K, Vickaryous MK. Constitutive cardiomyocyte proliferation in the leopard gecko (Eublepharis macularius
). J Morphol 2018; 279:1355-1367. [DOI: 10.1002/jmor.20850] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/14/2018] [Accepted: 05/19/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Kathy Jacyniak
- Department of Biomedical Sciences; Ontario Veterinary College, University of Guelph; Guelph Ontario Canada
| | - Matthew K. Vickaryous
- Department of Biomedical Sciences; Ontario Veterinary College, University of Guelph; Guelph Ontario Canada
| |
Collapse
|
33
|
Blanchoud S, Rinkevich B, Wilson MJ. Whole-Body Regeneration in the Colonial Tunicate Botrylloides leachii. Results Probl Cell Differ 2018; 65:337-355. [PMID: 30083927 DOI: 10.1007/978-3-319-92486-1_16] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The colonial marine invertebrate Botrylloides leachii belongs to the Tunicata subphylum, the closest invertebrate relatives to the vertebrate group and the only known class of chordates that can undergo whole-body regeneration (WBR). This dramatic developmental process allows a minute isolated fragment of B. leachii's vascular system, or a colony excised of all adults, to restore a functional animal in as little as 10 days. In addition to this exceptional regenerative capacity, B. leachii can reproduce both sexually, through a tadpole larval stage, and asexually, through palleal budding. Thus, three alternative developmental strategies lead to the establishment of filter-feeding adults. Consequently, B. leachii is particularly well suited for comparative studies on regeneration and should provide novel insights into regenerative processes in chordates.Here, after a short introduction on regeneration, we overview the biology of B. leachii as well as the current state of knowledge on WBR in this species and in related species of tunicates. Finally, we highlight the possible future directions that research might take in the study of WBR, including thoughts on technological approaches that appear most promising in this context. Overall, we provide a synthesis of the current knowledge on WBR in B. leachii to support research in this chordate species.
Collapse
Affiliation(s)
- Simon Blanchoud
- Department of Biology, University of Fribourg, Fribourg, Switzerland.
| | - Buki Rinkevich
- Israel Oceanographic and Limnological Research, National Institute of Oceanography, Haifa, Israel
| | - Megan J Wilson
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
34
|
de Preux Charles AS, Bise T, Baier F, Sallin P, Jaźwińska A. Preconditioning boosts regenerative programmes in the adult zebrafish heart. Open Biol 2017; 6:rsob.160101. [PMID: 27440423 PMCID: PMC4967829 DOI: 10.1098/rsob.160101] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/20/2016] [Indexed: 12/22/2022] Open
Abstract
During preconditioning, exposure to a non-lethal harmful stimulus triggers a body-wide increase of survival and pro-regenerative programmes that enable the organism to better withstand the deleterious effects of subsequent injuries. This phenomenon has first been described in the mammalian heart, where it leads to a reduction of infarct size and limits the dysfunction of the injured organ. Despite its important clinical outcome, the actual mechanisms underlying preconditioning-induced cardioprotection remain unclear. Here, we describe two independent models of cardiac preconditioning in the adult zebrafish. As noxious stimuli, we used either a thoracotomy procedure or an induction of sterile inflammation by intraperitoneal injection of immunogenic particles. Similar to mammalian preconditioning, the zebrafish heart displayed increased expression of cardioprotective genes in response to these stimuli. As zebrafish cardiomyocytes have an endogenous proliferative capacity, preconditioning further elevated the re-entry into the cell cycle in the intact heart. This enhanced cycling activity led to a long-term modification of the myocardium architecture. Importantly, the protected phenotype brought beneficial effects for heart regeneration within one week after cryoinjury, such as a more effective cell-cycle reentry, enhanced reactivation of embryonic gene expression at the injury border, and improved cell survival shortly after injury. This study reveals that exposure to antecedent stimuli induces adaptive responses that render the fish more efficient in the activation of the regenerative programmes following heart damage. Our results open a new field of research by providing the adult zebrafish as a model system to study remote cardiac preconditioning.
Collapse
Affiliation(s)
| | - Thomas Bise
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Felix Baier
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Pauline Sallin
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| |
Collapse
|
35
|
de Preux Charles AS, Bise T, Baier F, Marro J, Jaźwińska A. Distinct effects of inflammation on preconditioning and regeneration of the adult zebrafish heart. Open Biol 2017; 6:rsob.160102. [PMID: 27440424 PMCID: PMC4967830 DOI: 10.1098/rsob.160102] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/17/2016] [Indexed: 12/19/2022] Open
Abstract
The adult heart is able to activate cardioprotective programmes and modifies its architecture in response to physiological or pathological changes. While mammalian cardiac remodelling often involves hypertrophic expansion, the adult zebrafish heart exploits hyperplastic growth. This capacity depends on the responsiveness of zebrafish cardiomyocytes to mitogenic signals throughout their entire life. Here, we have examined the role of inflammation on the stimulation of cell cycle activity in the context of heart preconditioning and regeneration. We used thoracotomy as a cardiac preconditioning model and cryoinjury as a model of cardiac infarction in the adult zebrafish. First, we performed a spatio-temporal characterization of leucocytes and cycling cardiac cells after thoracotomy. This analysis revealed a concomitance between the infiltration of inflammatory cells and the stimulation of the mitotic activity. However, decreasing the immune response using clodronate liposome injection, PLX3397 treatment or anti-inflammatory drugs surprisingly had no effect on the re-entry of cardiac cells into the cell cycle. In contrast, reducing inflammation using the same strategies after cryoinjury strongly impaired cardiac cell mitotic activity and the regenerative process. Taken together, our results show that, while the immune response is not necessary to induce cell-cycle activity in intact preconditioned hearts, inflammation is required for the regeneration of injured hearts in zebrafish.
Collapse
Affiliation(s)
| | - Thomas Bise
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Felix Baier
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Jan Marro
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| |
Collapse
|
36
|
Bryant DM, Sousounis K, Payzin-Dogru D, Bryant S, Sandoval AGW, Martinez Fernandez J, Mariano R, Oshiro R, Wong AY, Leigh ND, Johnson K, Whited JL. Identification of regenerative roadblocks via repeat deployment of limb regeneration in axolotls. NPJ Regen Med 2017; 2:30. [PMID: 29302364 PMCID: PMC5677943 DOI: 10.1038/s41536-017-0034-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 09/22/2017] [Accepted: 09/26/2017] [Indexed: 02/07/2023] Open
Abstract
Axolotl salamanders are powerful models for understanding how regeneration of complex body parts can be achieved, whereas mammals are severely limited in this ability. Factors that promote normal axolotl regeneration can be examined in mammals to determine if they exhibit altered activity in this context. Furthermore, factors prohibiting axolotl regeneration can offer key insight into the mechanisms present in regeneration-incompetent species. We sought to determine if we could experimentally compromise the axolotl's ability to regenerate limbs and, if so, discover the molecular changes that might underlie their inability to regenerate. We found that repeated limb amputation severely compromised axolotls' ability to initiate limb regeneration. Using RNA-seq, we observed that a majority of differentially expressed transcripts were hyperactivated in limbs compromised by repeated amputation, suggesting that mis-regulation of these genes antagonizes regeneration. To confirm our findings, we additionally assayed the role of amphiregulin, an EGF-like ligand, which is aberrantly upregulated in compromised animals. During normal limb regeneration, amphiregulin is expressed by the early wound epidermis, and mis-expressing this factor lead to thickened wound epithelium, delayed initiation of regeneration, and severe regenerative defects. Collectively, our results suggest that repeatedly amputated limbs may undergo a persistent wound healing response, which interferes with their ability to initiate the regenerative program. These findings have important implications for human regenerative medicine.
Collapse
Affiliation(s)
- Donald M Bryant
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Konstantinos Sousounis
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA.,The Allen Discovery Center at Tufts University, 200 Boston Ave., Suite 4600, Medford, MA 02155 USA
| | - Duygu Payzin-Dogru
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Sevara Bryant
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Aaron Gabriel W Sandoval
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Jose Martinez Fernandez
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Rachelle Mariano
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Rachel Oshiro
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Alan Y Wong
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Nicholas D Leigh
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Kimberly Johnson
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Jessica L Whited
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA.,The Allen Discovery Center at Tufts University, 200 Boston Ave., Suite 4600, Medford, MA 02155 USA
| |
Collapse
|
37
|
Meda F, Rampon C, Dupont E, Gauron C, Mourton A, Queguiner I, Thauvin M, Volovitch M, Joliot A, Vriz S. Nerves, H 2O 2 and Shh: Three players in the game of regeneration. Semin Cell Dev Biol 2017; 80:65-73. [PMID: 28797840 DOI: 10.1016/j.semcdb.2017.08.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 08/04/2017] [Indexed: 12/12/2022]
Abstract
The tight control of reactive oxygen species (ROS) levels is required during regeneration. H2O2 in particular assumes clear signalling functions at different steps in this process. Injured nerves induce high levels of H2O2 through the activation of the Hedgehog (Shh) pathway, providing an environment that promotes cell plasticity, progenitor recruitment and blastema formation. In turn, high H2O2 levels contribute to growing axon attraction. Once re-innervation is completed, nerves subsequently downregulate H2O2 levels to their original state. A similar regulatory loop between H2O2 levels and nerves also exists during development. This suggests that redox signalling is a major actor in cell plasticity.
Collapse
Affiliation(s)
- Francesca Meda
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France; PSL Research University, Paris, France.
| | - Christine Rampon
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, France; PSL Research University, Paris, France
| | - Edmond Dupont
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France; PSL Research University, Paris, France
| | - Carole Gauron
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France; PSL Research University, Paris, France
| | - Aurélien Mourton
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France; PSL Research University, Paris, France; UPMC, Paris, France
| | - Isabelle Queguiner
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France; PSL Research University, Paris, France
| | - Marion Thauvin
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France; PSL Research University, Paris, France
| | - Michel Volovitch
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France; École Normale Supérieure, Institute of Biology at the Ecole Normale Supérieure (IBENS), CNRS UMR8197, INSERM U1024, Paris, France; PSL Research University, Paris, France
| | - Alain Joliot
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France; PSL Research University, Paris, France
| | - Sophie Vriz
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, France; PSL Research University, Paris, France.
| |
Collapse
|
38
|
Stocum DL. Mechanisms of urodele limb regeneration. REGENERATION (OXFORD, ENGLAND) 2017; 4:159-200. [PMID: 29299322 PMCID: PMC5743758 DOI: 10.1002/reg2.92] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 10/04/2017] [Indexed: 12/21/2022]
Abstract
This review explores the historical and current state of our knowledge about urodele limb regeneration. Topics discussed are (1) blastema formation by the proteolytic histolysis of limb tissues to release resident stem cells and mononucleate cells that undergo dedifferentiation, cell cycle entry and accumulation under the apical epidermal cap. (2) The origin, phenotypic memory, and positional memory of blastema cells. (3) The role played by macrophages in the early events of regeneration. (4) The role of neural and AEC factors and interaction between blastema cells in mitosis and distalization. (5) Models of pattern formation based on the results of axial reversal experiments, experiments on the regeneration of half and double half limbs, and experiments using retinoic acid to alter positional identity of blastema cells. (6) Possible mechanisms of distalization during normal and intercalary regeneration. (7) Is pattern formation is a self-organizing property of the blastema or dictated by chemical signals from adjacent tissues? (8) What is the future for regenerating a human limb?
Collapse
Affiliation(s)
- David L. Stocum
- Department of BiologyIndiana University−Purdue University Indianapolis723 W. Michigan StIndianapolisIN 46202USA
| |
Collapse
|
39
|
König D, Page L, Chassot B, Jaźwińska A. Dynamics of actinotrichia regeneration in the adult zebrafish fin. Dev Biol 2017; 433:416-432. [PMID: 28760345 DOI: 10.1016/j.ydbio.2017.07.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 01/21/2023]
Abstract
The skeleton of adult zebrafish fins comprises lepidotrichia, which are dermal bones of the rays, and actinotrichia, which are non-mineralized spicules at the distal margin of the appendage. Little is known about the regenerative dynamics of the actinotrichia-specific structural proteins called Actinodins. Here, we used immunofluorescence analysis to determine the contribution of two paralogous Actinodin proteins, And1/2, in regenerating fins. Both proteins were detected in the secretory organelles in the mesenchymal cells of the blastema, but only And1 was detected in the epithelial cells of the wound epithelium. The analysis of whole mount fins throughout the entire regenerative process and longitudinal sections revealed that And1-positive fibers are complementary to the lepidotrichia. The analysis of another longfin fish, a gain-of-function mutation in the potassium channel kcnk5b, revealed that the long-fin phenotype is associated with an extended size of actinotrichia during homeostasis and regeneration. Finally, we investigated the role of several signaling pathways in actinotrichia formation and maintenance. This revealed that the pulse-inhibition of either TGFβ/Activin-βA or FGF are sufficient to impair deposition of Actinodin during regeneration. Thus, the dynamic turnover of Actinodin during fin regeneration is regulated by multiple factors, including the osteoblasts, growth rate in a potassium channel mutant, and instructive signaling networks between the epithelium and the blastema of the regenerating fin.
Collapse
Affiliation(s)
- Désirée König
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Lionel Page
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Bérénice Chassot
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland.
| |
Collapse
|
40
|
De Lombaert MC, Rick EL, Krugner-Higby LA, Wolman MA. Behavioral Characteristics of Adult Zebrafish ( Danio rerio) after MS222 Anesthesia for Fin Excision. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2017; 56:377-381. [PMID: 28724486 PMCID: PMC5517326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/27/2017] [Accepted: 03/29/2017] [Indexed: 06/07/2023]
Abstract
The health of laboratory animals is an ethical responsibility of researchers and a critical determinant of experimental outcome. Therefore, all husbandry procedures should be evaluated for their effects on mortality, behavior, and physiology to maximize animal welfare and minimize experimental variability. For adult zebrafish, the excision of a small portion of the caudal fin (that is, 'fin clipping') under MS222 anesthesia is a common procedure to obtain tissue for genotyping. The potential effect of this procedure on behavioral and physiologic assays of feeding, anxiety, and stress has not previously been assessed. Here, we evaluated feeding behavior, anxiety-associated behaviors, and physiologic indicators of stress at multiple time points within 24 h after performing a standard fin-clip procedure under MS222 anesthesia. Within 1 h of the procedure, fin-clipped fish showed a mild increase in anxiety and exhibited reduced feeding; however, these effects were short-lived, and the fish exhibited baseline levels of anxiety and feeding by 6 and 24 h after fin clipping. Together with the zebrafish's ability to regenerate fin tissue and the low mortality associated with fin clipping, our data support the continued practice of this technique under MS222 anesthesia as a routine husbandry procedure that is unlikely to alter experimental outcomes related to feeding, anxiety, or stress.
Collapse
Affiliation(s)
| | - Elizabeth L Rick
- Department of Zoology, University of Wisconsin, Madison, Wisconsin
| | - Lisa A Krugner-Higby
- Research and Animal Resources Center, University of Wisconsin, Madison, Wisconsin
| | - Marc A Wolman
- Department of Zoology, University of Wisconsin, Madison, Wisconsin;,
| |
Collapse
|
41
|
Blanchoud S, Zondag L, Lamare MD, Wilson MJ. Hematological Analysis of the Ascidian Botrylloides leachii (Savigny, 1816) During Whole-Body Regeneration. THE BIOLOGICAL BULLETIN 2017; 232:143-157. [PMID: 28898595 DOI: 10.1086/692841] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Whole-body regeneration (WBR)-the formation of an entire adult from only a small fragment of its own tissue-is extremely rare among chordates. Exceptionally, in the colonial ascidian Botrylloides leachii (Savigny, 1816) a fully functional adult is formed from their common vascular system after ablation of all adults from the colony in just 10 d, thanks to their high blastogenetic potential. While previous studies have identified key genetic markers and morphological changes, no study has yet focused on the hematological aspects of regeneration despite the major involvement of the remaining vascular system and the contained hemocytes in this process. To dissect this process, we analyzed colony blood flow patterns using time-lapse microscopy to obtain a quantitative description of the velocity, reversal pattern, and average distance traveled by hemocytes. We also observed that flows present during regeneration are powered by temporally and spatially synchronized contractions of the terminal ampullae. In addition, we revised previous studies of B. leachii hematology as well as asexual development using histological sectioning and compared the role played by hemocytes during WBR. We found that regeneration starts with a rapid healing response characterized by hemocyte aggregation and infiltration of immunocytes, followed by increased activity of hemoblasts, recruitment of macrophage-like cells for clearing the tissues of debris, and their subsequent disappearance from the circulation concomitant with the maturation of a single regenerated adult. Overall, we provide a detailed account of the hematological properties of regenerating B. leachii colonies, providing novel lines of inquiry toward the decipherment of regeneration in chordates.
Collapse
|
42
|
Zullo L, Fossati SM, Imperadore P, Nödl MT. Molecular Determinants of Cephalopod Muscles and Their Implication in Muscle Regeneration. Front Cell Dev Biol 2017; 5:53. [PMID: 28555185 PMCID: PMC5430041 DOI: 10.3389/fcell.2017.00053] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/27/2017] [Indexed: 12/11/2022] Open
Abstract
The ability to regenerate whole-body structures has been studied for many decades and is of particular interest for stem cell research due to its therapeutic potential. Several vertebrate and invertebrate species have been used as model systems to study pathways involved in regeneration in the past. Among invertebrates, cephalopods are considered as highly evolved organisms, which exhibit elaborate behavioral characteristics when compared to other mollusks including active predation, extraordinary manipulation, and learning abilities. These are enabled by a complex nervous system and a number of adaptations of their body plan, which were acquired over evolutionary time. Some of these novel features show similarities to structures present in vertebrates and seem to have evolved through a convergent evolutionary process. Octopus vulgaris (the common octopus) is a representative of modern cephalopods and is characterized by a sophisticated motor and sensory system as well as highly developed cognitive capabilities. Due to its phylogenetic position and its high regenerative power the octopus has become of increasing interest for studies on regenerative processes. In this paper we provide an overview over the current knowledge of cephalopod muscle types and structures and present a possible link between these characteristics and their high regenerative potential. This may help identify conserved molecular pathways underlying regeneration in invertebrate and vertebrate animal species as well as discover new leads for targeted tissue treatments in humans.
Collapse
Affiliation(s)
- Letizia Zullo
- Centre for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di TecnologiaGenoa, Italy
| | - Sara M Fossati
- Centre for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di TecnologiaGenoa, Italy
| | | | - Marie-Therese Nödl
- Centre for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di TecnologiaGenoa, Italy
| |
Collapse
|
43
|
Pfefferli C, Jaźwińska A. The careg element reveals a common regulation of regeneration in the zebrafish myocardium and fin. Nat Commun 2017; 8:15151. [PMID: 28466843 PMCID: PMC5418624 DOI: 10.1038/ncomms15151] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 03/03/2017] [Indexed: 12/21/2022] Open
Abstract
The existence of common mechanisms regulating organ regeneration is an intriguing concept. Here we report on a regulatory element that is transiently activated during heart and fin regeneration in zebrafish. This element contains a ctgfa upstream sequence, called careg, which is induced by TGFβ/Activin-β signalling in the peri-injury zone of the myocardium and the fin mesenchyme. In addition, this reporter demarcates a primordial cardiac layer and intraray osteoblasts. Using genetic fate mapping, we show the regenerative competence of careg-expressing cells. The analysis of the heart reveals that the primordial cardiac layer is incompletely restored after cryoinjury, whereas trabecular and cortical cardiomyocytes contribute to myocardial regrowth. In regenerating fins, the activated mesenchyme of the stump gives rise to the blastema. Our findings provide evidence of a common regenerative programme in cardiomyocytes and mesenchyme that opens the possibility to further explore conserved mechanisms of the cellular plasticity in diverse vertebrate organs.
Collapse
Affiliation(s)
- Catherine Pfefferli
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| |
Collapse
|
44
|
Marshall L, Vivien C, Girardot F, Péricard L, Demeneix BA, Coen L, Chai N. Persistent fibrosis, hypertrophy and sarcomere disorganisation after endoscopy-guided heart resection in adult Xenopus. PLoS One 2017; 12:e0173418. [PMID: 28278282 PMCID: PMC5344503 DOI: 10.1371/journal.pone.0173418] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/15/2017] [Indexed: 12/30/2022] Open
Abstract
Models of cardiac repair are needed to understand mechanisms underlying failure to regenerate in human cardiac tissue. Such studies are currently dominated by the use of zebrafish and mice. Remarkably, it is between these two evolutionary separated species that the adult cardiac regenerative capacity is thought to be lost, but causes of this difference remain largely unknown. Amphibians, evolutionary positioned between these two models, are of particular interest to help fill this lack of knowledge. We thus developed an endoscopy-based resection method to explore the consequences of cardiac injury in adult Xenopus laevis. This method allowed in situ live heart observation, standardised tissue amputation size and reproducibility. During the first week following amputation, gene expression of cell proliferation markers remained unchanged, whereas those relating to sarcomere organisation decreased and markers of inflammation, fibrosis and hypertrophy increased. One-month post-amputation, fibrosis and hypertrophy were evident at the injury site, persisting through 11 months. Moreover, cardiomyocyte sarcomere organisation deteriorated early following amputation, and was not completely recovered as far as 11 months later. We conclude that the adult Xenopus heart is unable to regenerate, displaying cellular and molecular marks of scarring. Our work suggests that, contrary to urodeles and teleosts, with the exception of medaka, adult anurans share a cardiac injury outcome similar to adult mammals. This observation is at odds with current hypotheses that link loss of cardiac regenerative capacity with acquisition of homeothermy.
Collapse
Affiliation(s)
- Lindsey Marshall
- Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, UMR CNRS 7221, Muséum National d'Histoire Naturelle, Sorbonne Université, Paris, France
| | - Céline Vivien
- Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, UMR CNRS 7221, Muséum National d'Histoire Naturelle, Sorbonne Université, Paris, France
| | - Fabrice Girardot
- Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, UMR CNRS 7221, Muséum National d'Histoire Naturelle, Sorbonne Université, Paris, France
| | - Louise Péricard
- Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, UMR CNRS 7221, Muséum National d'Histoire Naturelle, Sorbonne Université, Paris, France
| | - Barbara A. Demeneix
- Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, UMR CNRS 7221, Muséum National d'Histoire Naturelle, Sorbonne Université, Paris, France
| | - Laurent Coen
- Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, UMR CNRS 7221, Muséum National d'Histoire Naturelle, Sorbonne Université, Paris, France
| | - Norin Chai
- Ménagerie du Jardin des Plantes, Muséum National d’Histoire Naturelle, Paris, France
| |
Collapse
|
45
|
Smith AM, Maguire-Nguyen KK, Rando TA, Zasloff MA, Strange KB, Yin VP. The protein tyrosine phosphatase 1B inhibitor MSI-1436 stimulates regeneration of heart and multiple other tissues. NPJ Regen Med 2017; 2:4. [PMID: 29302341 PMCID: PMC5677970 DOI: 10.1038/s41536-017-0008-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/13/2017] [Accepted: 01/20/2017] [Indexed: 12/13/2022] Open
Abstract
Regenerative medicine holds substantial promise for repairing or replacing tissues and organs damaged by disease, injury, and degeneration. Much of the field has focused on development of cell-based therapeutics, gene-based therapeutics, and tissue engineering-based therapeutics. In contrast, development of small molecule regenerative medicine therapies is an emerging area. Using the adult zebrafish as a novel screening platform, we identified MSI-1436 as a first-in-class regenerative medicine drug candidate. MSI-1436 is a naturally occurring aminosterol that inhibits protein tyrosine phosphatase 1B. Treatment of adult zebrafish by intraperitoneal injection of MSI-1436 increased the rate of regeneration of the amputated caudal fin, which is comprised of bone, connective, skin, vascular and nervous tissues and also increased the rate of adult zebrafish heart regeneration. Intraperitoneal administration of MSI-1436 to adult mice for 4 weeks after induction of myocardial infarction increased survival, improved heart function, reduced infarct size, reduced ventricular wall thinning and increased cardiomyocyte proliferation. Satellite cell activation in injured mouse skeletal muscle was stimulated by MSI-1436. MSI-1436 was well tolerated by patients in Phase 1 and 1b obesity and type 2 diabetes clinical trials. Doses effective at stimulating regeneration are 5–50-times lower than the maximum well tolerated human dose. The demonstrated safety and well established pharmacological properties of MSI-1436 underscore the potential of this molecule as a novel treatment for heart attack and multiple other degenerative diseases. A naturally occurring small molecule shows promise as a drug for tissue and organ repair and regeneration. Viravuth Yin of the Kathryn W. Davis Center for Regenerative Biology and Medicine with colleagues in the US found that treating zebrafish with an intraperitoneal injection of MSI-1436, which inhibits the enzyme ‘protein tyrosine phosphatase 1B’, increased the rate of regeneration of an amputated caudal fin and of partially removed heart muscle without apparent tissue malformation. Intraperitoneal injection of MSI-1436 in adult mice also reduced the size of an induced heart infarction, improved survivability, triggered new heart muscle formation and stimulated regeneration after skeletal muscle injury. Effective doses for tissue regeneration in both animals were much lower than the maximum tolerated doses found for humans in clinical trials for potential treatment of obesity and diabetes.
Collapse
Affiliation(s)
- Ashley M Smith
- Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, Salisbury Cove, ME 04672 USA
| | - Katie K Maguire-Nguyen
- Department of Neurology, Stanford University Medical Center, Stanford, CA 94305-5235 USA
| | - Thomas A Rando
- Department of Neurology, Stanford University Medical Center, Stanford, CA 94305-5235 USA
| | - Michael A Zasloff
- Novo Biosciences, Bar Harbor, ME 04609 USA.,MedStar Georgetown Transplant Institute, Georgetown University Hospital, Washington DC, 20007 USA
| | - Kevin B Strange
- Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, Salisbury Cove, ME 04672 USA.,Novo Biosciences, Bar Harbor, ME 04609 USA
| | - Viravuth P Yin
- Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, Salisbury Cove, ME 04672 USA.,Novo Biosciences, Bar Harbor, ME 04609 USA
| |
Collapse
|
46
|
Abstract
Psychological stress is one of the factors associated with human cardiovascular disease. Here, we demonstrate that acute perceived stress impairs the natural capacity of heart regeneration in zebrafish. Beside physical and chemical disturbances, intermittent crowding triggered an increase in cortisol secretion and blocked the replacement of fibrotic tissue with new myocardium. Pharmacological simulation of stress by pulse treatment with dexamethasone/adrenaline reproduced the regeneration failure, while inhibition of the stress response with anxiolytic drugs partially rescued the regenerative process. Impaired heart regeneration in stressed animals was associated with a reduced cardiomyocyte proliferation and with the downregulation of several genes, including igfbp1b, a modulator of IGF signalling. Notably, daily stress induced a decrease in Igf1r phosphorylation. As cardiomyocyte proliferation was decreased in response to IGF-1 receptor inhibition, we propose that the stress-induced cardiac regenerative failure is partially caused by the attenuation of IGF signalling. These findings indicate that the natural regenerative ability of the zebrafish heart is vulnerable to the systemic paracrine stress response.
Collapse
Affiliation(s)
- Pauline Sallin
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| |
Collapse
|
47
|
Affiliation(s)
| | - Jeffery D. Molkentin
- Department of Pediatrics, University of Cincinnati, Cincinnati Children’s Hospital Medical Center and Howard Hughes Medical Institute, Cincinnati, Ohio
| |
Collapse
|
48
|
Marro J, Pfefferli C, de Preux Charles AS, Bise T, Jaźwińska A. Collagen XII Contributes to Epicardial and Connective Tissues in the Zebrafish Heart during Ontogenesis and Regeneration. PLoS One 2016; 11:e0165497. [PMID: 27783651 PMCID: PMC5081208 DOI: 10.1371/journal.pone.0165497] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/12/2016] [Indexed: 12/25/2022] Open
Abstract
Zebrafish heart regeneration depends on cardiac cell proliferation, epicardium activation and transient reparative tissue deposition. The contribution and the regulation of specific collagen types during the regenerative process, however, remain poorly characterized. Here, we identified that the non-fibrillar type XII collagen, which serves as a matrix-bridging component, is expressed in the epicardium of the zebrafish heart, and is boosted after cryoinjury-induced ventricular damage. During heart regeneration, an intense deposition of Collagen XII covers the outer epicardial cap and the interstitial reparative tissue. Analysis of the activated epicardium and fibroblast markers revealed a heterogeneous cellular origin of Collagen XII. Interestingly, this matrix-bridging collagen co-localized with fibrillar type I collagen and several glycoproteins in the post-injury zone, suggesting its role in tissue cohesion. Using SB431542, a selective inhibitor of the TGF-β receptor, we showed that while the inhibitor treatment did not affect the expression of collagen 12 and collagen 1a2 in the epicardium, it completely suppressed the induction of both genes in the fibrotic tissue. This suggests that distinct mechanisms might regulate collagen expression in the outer heart layer and the inner injury zone. On the basis of this study, we postulate that the TGF-β signaling pathway induces and coordinates formation of a transient collagenous network that comprises fibril-forming Collagen I and fiber-associated Collagen XII, both of which contribute to the reparative matrix of the regenerating zebrafish heart.
Collapse
Affiliation(s)
- Jan Marro
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Catherine Pfefferli
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | | | - Thomas Bise
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
- * E-mail:
| |
Collapse
|
49
|
Vivien CJ, Hudson JE, Porrello ER. Evolution, comparative biology and ontogeny of vertebrate heart regeneration. NPJ Regen Med 2016; 1:16012. [PMID: 29302337 PMCID: PMC5744704 DOI: 10.1038/npjregenmed.2016.12] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 06/01/2016] [Accepted: 06/15/2016] [Indexed: 12/19/2022] Open
Abstract
There are 64,000 living species of vertebrates on our planet and all of them have a heart. Comparative analyses devoted to understanding the regenerative potential of the myocardium have been performed in a dozen vertebrate species with the aim of developing regenerative therapies for human heart disease. Based on this relatively small selection of animal models, important insights into the evolutionary conservation of regenerative mechanisms have been gained. In this review, we survey cardiac regeneration studies in diverse species to provide an evolutionary context for the lack of regenerative capacity in the adult mammalian heart. Our analyses highlight the importance of cardiac adaptations that have occurred over hundreds of millions of years during the transition from aquatic to terrestrial life, as well as during the transition from the womb to an oxygen-rich environment at birth. We also discuss the evolution and ontogeny of cardiac morphological, physiological and metabolic adaptations in the context of heart regeneration. Taken together, our findings suggest that cardiac regenerative potential correlates with a low-metabolic state, the inability to regulate body temperature, low heart pressure, hypoxia, immature cardiomyocyte structure and an immature immune system. A more complete understanding of the evolutionary context and developmental mechanisms governing cardiac regenerative capacity would provide stronger scientific foundations for the translation of cardiac regeneration therapies into the clinic.
Collapse
Affiliation(s)
- Celine J Vivien
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
- Centre for Cardiac and Vascular Biology, The University of Queensland, Brisbane, QLD, Australia
| | - James E Hudson
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
- Centre for Cardiac and Vascular Biology, The University of Queensland, Brisbane, QLD, Australia
| | - Enzo R Porrello
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
- Centre for Cardiac and Vascular Biology, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
50
|
Farah Z, Fan H, Liu Z, He JQ. A concise review of common animal models for the study of limb regeneration. Organogenesis 2016; 12:109-118. [PMID: 27391218 DOI: 10.1080/15476278.2016.1205775] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Correct selection of an appropriate animal mode to closely mimic human extremity diseases or to exhibit desirable phenotypes of limb regeneration is the first critical step for all scientists in biomedical and regenerative researches. The commonly-used animals in limb regeneration and repairing studies, such as axolotl, mice, and rats, are discussed in the review and other models including cockroaches, dogs, and horses are also mentioned. The review weighs the general advantages, disadvantages, and precedent uses of each model in the context of limb and peripheral injury and subsequent regeneration. We hope that this review can provide the reader an overview of each model, from which to select one for their specific purpose.
Collapse
Affiliation(s)
- Zayd Farah
- a Department of Biomedical Sciences & Pathobiology , Center for Veterinary Regenerative Medicine (CVRM), Virginia-Maryland College of Veterinary Medicine, Virginia Tech , Blacksburg , VA , USA
| | - Huimin Fan
- b Research Institute of Heart Failure , Shanghai East Hospital of Tongji University , Shanghai , China
| | - Zhongmin Liu
- b Research Institute of Heart Failure , Shanghai East Hospital of Tongji University , Shanghai , China
| | - Jia-Qiang He
- a Department of Biomedical Sciences & Pathobiology , Center for Veterinary Regenerative Medicine (CVRM), Virginia-Maryland College of Veterinary Medicine, Virginia Tech , Blacksburg , VA , USA
| |
Collapse
|