1
|
Butt FA, De Simone A, Di Talia S, Poss KD. In toto live imaging of Erk signaling dynamics in developing zebrafish hepatocytes. Dev Biol 2025; 523:43-50. [PMID: 40228782 PMCID: PMC12068954 DOI: 10.1016/j.ydbio.2025.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Regional and tissue-wide regulation of signaling pathways orchestrates cellular proliferation and differentiation during organ development. In this study, we established an imaging platform for longitudinal analysis of liver development in live developing zebrafish. We generated hepatocyte-specific transgenic lines for kinase translocation reporters of extracellular signal-regulated kinase (Erk) and c-Jun N-terminal kinase (Jnk) signaling, and with these we captured signaling dynamics that govern rapid expansion of hepatocytes toward creation of the functioning liver at single-cell resolution. Our findings reveal Erk signaling fluctuations as the liver develops and introduce methodology for investigating cell-type specific signaling dynamics during organ morphogenesis.
Collapse
Affiliation(s)
- Faraz Ahmed Butt
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Alessandro De Simone
- Department of Genetics and Evolution, University of Geneva, 1211, Geneva, Switzerland
| | - Stefano Di Talia
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC, 27710, USA; Duke Center for Quantitative Living Systems, Duke University Medical Center, Durham, NC, 27710, USA
| | - Kenneth D Poss
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC, 27710, USA; Morgridge Institute for Research, Madison, WI, 53715, USA; Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, 53705, USA.
| |
Collapse
|
2
|
Raz AA, Yamashita YM. Rewinding the clock: mechanisms of dedifferentiation. Curr Opin Genet Dev 2025; 93:102353. [PMID: 40311173 DOI: 10.1016/j.gde.2025.102353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 05/03/2025]
Abstract
Adult stem cells maintain tissue homeostasis through the production of differentiating cells. Considerable recent work has identified that stem cells themselves are replaceable through the process of dedifferentiation. The capacity and mechanisms of dedifferentiation vary widely among species and organ contexts. However, some core features are commonly present. In this review, we summarize 'hallmarks' of dedifferentiation, including mechanisms for maintenance of potency, sensation of loss, and migration, and review the current understanding of dedifferentiation as a true replacement mechanism.
Collapse
Affiliation(s)
- Amelie A Raz
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
| | - Yukiko M Yamashita
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Howard Hughes Medical Institute, Cambridge, MA, USA; Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
3
|
Barnes M, Burton D, Marsden K, Kullman S. Early disruptions in vitamin D receptor signaling induces persistent developmental behavior deficits in zebrafish larvae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645997. [PMID: 40235984 PMCID: PMC11996324 DOI: 10.1101/2025.03.28.645997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
A critical function of the nervous system is to rapidly process sensory information and initiate appropriate behavioral responses. Defects in sensory processing and behavior selection are commonly observed in neuro-psychiatric conditions including anxiety, autism (ASD), and schizophrenia. The etiology of sensory processing disorders remains equivocal; however, it is hypothesized that extrinsic environmental factors can play fundamental roles. In this study we examine the importance of vitamin D (1α, 25-dihydroxyvitamin D3) receptor signaling during early life stage development on sensory processing and neurobehavioral health outcomes. While vitamin D has traditionally been associated with mineral ion homeostasis, accumulating evidence suggests non-calcemic roles for vitamin D including early neurodevelopment. Here we demonstrate that systemic disruption of vitamin D receptor (VDR) signaling with a conditional dominant negative (dnVDR) transgenic zebrafish line results in specific visual and acoustic sensorimotor behavior defects. Induction of dnVDR between 24-72 hours post fertilization (hpf) results in modulation of visual motor response with demonstrate attenuation in acute activity and hypolocomotion across multiple swimming metrics when assayed at 6- and 28-days post fertilization (dpf). Disruption in VDR signaling additionally resulted in a strong and specific attenuation of the Long-Latency C-bends (LLC) within the acoustic startle response at 6 dpf while Short-Latency C-bends (SLC) were moderately impacted. Pre-pulse inhibition (PPI) was not impacted in young larvae however exhibited a significantly attenuated response at 28 dpf suggesting an inability to properly modulate their startle responses later in development and persistent effects of VDR modulation during early development. Overall, our data demonstrate that modulation of vitamin D signaling during critical windows of development irreversibly disrupts the development of neuronal circuitry associated with sensory processing behaviors which may have significant implications to neurobehavioral health outcomes.
Collapse
|
4
|
Liang J, Jiang P, Yan S, Cheng T, Chen S, Xian K, Xu P, Xiong JW, He A, Li J, Han P. Genetically encoded tension heterogeneity sculpts cardiac trabeculation. SCIENCE ADVANCES 2025; 11:eads2998. [PMID: 40053597 PMCID: PMC11887796 DOI: 10.1126/sciadv.ads2998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025]
Abstract
The myocardial wall arises from a single layer of cardiomyocytes, some delaminate to create trabeculae while others remain in the compact layer. However, the mechanisms governing cardiomyocyte fate decisions remain unclear. Using single-cell RNA sequencing, genetically encoded biosensors, and in toto live imaging, we observe intrinsic variations in erbb2 expression and its association with trabecular fate. Specifically, erbb2 promotes PI3K activity and recruits the Arp2/3 complex, inducing a polarized accumulation of the actomyosin network to drive cell delamination. Subsequently, the lineage-committed nascent trabeculae trigger Notch activity in neighboring cardiomyocytes to suppress erbb2 expression and reduce cell tension, thereby confining them to the compact layer. Overall, this genetic and cellular interplay governs compact and trabecular cell fate determination to orchestrate myocardial pattern formation.
Collapse
Affiliation(s)
- Jinxiu Liang
- Department of Cardiology, Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
- International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Peijun Jiang
- Department of Cardiology, Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
- International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shuaifang Yan
- Institute of Molecular Medicine, National Biomedical Imaging Center, College of Future Technology, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Tao Cheng
- Department of Cardiology, Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
- International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shuo Chen
- Department of Cardiology, Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
- International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Kexin Xian
- Institute of Molecular Medicine, National Biomedical Imaging Center, College of Future Technology, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Pengfei Xu
- Department of Cardiology, Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
- International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jing-Wei Xiong
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | - Aibin He
- Institute of Molecular Medicine, National Biomedical Imaging Center, College of Future Technology, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Jia Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Peidong Han
- Department of Cardiology, Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
- International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Vasudevarao MD, Posadas Pena D, Ihle M, Bongiovanni C, Maity P, Geissler D, Mohammadi HF, Rall-Scharpf M, Niemann J, Mommersteeg MTM, Redaelli S, Happ K, Wu CC, Beisaw A, Scharffetter-Kochanek K, D'Uva G, Malek Mohammadi M, Wiesmüller L, Geiger H, Weidinger G. BMP signaling promotes zebrafish heart regeneration via alleviation of replication stress. Nat Commun 2025; 16:1708. [PMID: 39962064 PMCID: PMC11832743 DOI: 10.1038/s41467-025-56993-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
In contrast to mammals, adult zebrafish achieve complete heart regeneration via proliferation of cardiomyocytes. Surprisingly, we found that regenerating cardiomyocytes experience DNA replication stress, which represents one reason for declining tissue regeneration during aging in mammals. Pharmacological inhibition of ATM and ATR kinases revealed that DNA damage response signaling is essential for zebrafish heart regeneration. Manipulation of Bone Morphogenetic Protein (BMP)-Smad signaling using transgenics and mutants showed that BMP signaling alleviates cardiomyocyte replication stress. BMP signaling also rescues neonatal mouse cardiomyocytes, human fibroblasts and human hematopoietic stem and progenitor cells (HSPCs) from replication stress. DNA fiber spreading assays indicate that BMP signaling facilitates re-start of replication forks after replication stress-induced stalling. Our results identify the ability to overcome replication stress as key factor for the elevated zebrafish heart regeneration capacity and reveal a conserved role for BMP signaling in promotion of stress-free DNA replication.
Collapse
Affiliation(s)
| | - Denise Posadas Pena
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Michaela Ihle
- Department of Obstetrics and Gynecology, Ulm University, Prittwitzstraße 43, 89075, Ulm, Germany
| | - Chiara Bongiovanni
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, via Massarenti 9, 40138, Bologna, Italy
| | - Pallab Maity
- Department of Dermatology and Allergic Diseases, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Dominik Geissler
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Hossein Falah Mohammadi
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Melanie Rall-Scharpf
- Department of Obstetrics and Gynecology, Ulm University, Prittwitzstraße 43, 89075, Ulm, Germany
| | - Julian Niemann
- Institute of Molecular Medicine, Ulm University, Meyerhofstrasse N27, 89081, Ulm, Germany
| | - Mathilda T M Mommersteeg
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3PT, United Kingdom
| | - Simone Redaelli
- Institute of Biomedical Ethics and History of Medicine, University of Zurich, Winterthurerstrasse 30, 8006, Zurich, Switzerland
| | - Kathrin Happ
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Chi-Chung Wu
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Arica Beisaw
- Institute of Experimental Cardiology, Heidelberg University, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
| | - Karin Scharffetter-Kochanek
- Department of Dermatology and Allergic Diseases, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Gabriele D'Uva
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, via Massarenti 9, 40138, Bologna, Italy
| | - Mona Malek Mohammadi
- Institute of Physiology I, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Lisa Wiesmüller
- Department of Obstetrics and Gynecology, Ulm University, Prittwitzstraße 43, 89075, Ulm, Germany
| | - Hartmut Geiger
- Institute of Molecular Medicine, Ulm University, Meyerhofstrasse N27, 89081, Ulm, Germany
| | - Gilbert Weidinger
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
6
|
Rich A, Lu Z, Simone AD, Garcia L, Janssen J, Ando K, Ou J, Vergassola M, Poss KD, Talia SD. Decaying and expanding Erk gradients process memory of skeletal size during zebrafish fin regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634576. [PMID: 39896678 PMCID: PMC11785216 DOI: 10.1101/2025.01.23.634576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Regeneration of an amputated salamander limb or fish fin restores pre-injury size and structure, illustrating the phenomenon of positional memory. Although appreciated for centuries, the identity of position-dependent cues and how they control tissue growth are not resolved. Here, we quantify Erk signaling events in whole populations of osteoblasts during zebrafish fin regeneration. We find that osteoblast Erk activity is dependent on Fgf receptor signaling and organized into millimeter-long gradients that extend from the distal tip to the amputation site. Erk activity scales with the amount of tissue amputated, predicts the likelihood of osteoblast cycling, and predicts the size of regenerated skeletal structures. Mathematical modeling suggests gradients are established by the transient deposition of long-lived ligands that are transported by tissue growth. This concept is supported by the observed scaling of expression of the essential epidermal ligand fgf20a with extents of amputation. Our work provides evidence that localized, scaled expression of pro-regenerative ligands instructs long-range signaling and cycling to control skeletal size in regenerating appendages.
Collapse
Affiliation(s)
- Ashley Rich
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Center for Quantitative Living Systems, Duke University Medical Center, Durham, NC, USA
| | - Ziqi Lu
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Center for Quantitative Living Systems, Duke University Medical Center, Durham, NC, USA
| | - Alessandro De Simone
- Department of Genetics and Evolution, University of Geneva, 1211 Geneva, Switzerland
| | - Lucas Garcia
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | | | - Kazunori Ando
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Regeneration Center, Duke University Medical Center, Durham, NC, USA
- Morgridge Institute for Research, Madison WI USA
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, USA
| | - Jianhong Ou
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Regeneration Center, Duke University Medical Center, Durham, NC, USA
- Morgridge Institute for Research, Madison WI USA
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, USA
| | - Massimo Vergassola
- Department of Physics, École Normale Supérieure, Paris 75005, France
- Department of Physics, University of California, San Diego, CA, USA
| | - Kenneth D. Poss
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Regeneration Center, Duke University Medical Center, Durham, NC, USA
- Morgridge Institute for Research, Madison WI USA
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, USA
| | - Stefano Di Talia
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Center for Quantitative Living Systems, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
7
|
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
|
8
|
Masiero C, Aresi C, Forlino A, Tonelli F. Zebrafish Models for Skeletal and Extraskeletal Osteogenesis Imperfecta Features: Unveiling Pathophysiology and Paving the Way for Drug Discovery. Calcif Tissue Int 2024; 115:931-959. [PMID: 39320469 PMCID: PMC11607041 DOI: 10.1007/s00223-024-01282-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024]
Abstract
In the last decades, the easy genetic manipulation, the external fertilization, the high percentage of homology with human genes and the reduced husbandry costs compared to rodents, made zebrafish a valid model for studying human diseases and for developing new therapeutical strategies. Since zebrafish shares with mammals the same bone cells and ossification types, it became widely used to dissect mechanisms and possible new therapeutic approaches in the field of common and rare bone diseases, such as osteoporosis and osteogenesis imperfecta (OI), respectively. OI is a heritable skeletal disorder caused by defects in gene encoding collagen I or proteins/enzymes necessary for collagen I synthesis and secretion. Nevertheless, OI patients can be also characterized by extraskeletal manifestations such as dentinogenesis imperfecta, muscle weakness, cardiac valve and pulmonary abnormalities and skin laxity. In this review, we provide an overview of the available zebrafish models for both dominant and recessive forms of OI. An updated description of all the main similarities and differences between zebrafish and mammal skeleton, muscle, heart and skin, will be also discussed. Finally, a list of high- and low-throughput techniques available to exploit both larvae and adult OI zebrafish models as unique tools for the discovery of new therapeutic approaches will be presented.
Collapse
Affiliation(s)
- Cecilia Masiero
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| | - Carla Aresi
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy.
| | - Francesca Tonelli
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| |
Collapse
|
9
|
Chen X, Zhong X, Huang GN. Heart regeneration from the whole-organism perspective to single-cell resolution. NPJ Regen Med 2024; 9:34. [PMID: 39548113 PMCID: PMC11568173 DOI: 10.1038/s41536-024-00378-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Cardiac regenerative potential in the animal kingdom displays striking divergence across ontogeny and phylogeny. Here we discuss several fundamental questions in heart regeneration and provide both a holistic view of heart regeneration in the organism as a whole, as well as a single-cell perspective on intercellular communication among diverse cardiac cell populations. We hope to provide valuable insights that advance our understanding of organ regeneration and future therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoxin Chen
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Xiaochen Zhong
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Guo N Huang
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA, USA.
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
- Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
10
|
Cheng X, Ju J, Huang W, Duan Z, Han Y. cpt1b Regulates Cardiomyocyte Proliferation Through Modulation of Glutamine Synthetase in Zebrafish. J Cardiovasc Dev Dis 2024; 11:344. [PMID: 39590187 PMCID: PMC11594654 DOI: 10.3390/jcdd11110344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 11/28/2024] Open
Abstract
Carnitine palmitoyltransferase 1b (Cpt1b) is a crucial rate-limiting enzyme in fatty acid metabolism, but its role and mechanism in early cardiac development remains unclear. Here, we show that cpt1b regulates cardiomyocyte proliferation during zebrafish development. Knocking out entire cpt1b coding sequences leads to impaired cardiomyocyte proliferation, while cardiomyocyte-specific overexpression of cpt1b promotes cardiomyocyte proliferation. RNA sequencing analysis and pharmacological studies identified glutamine synthetase as a key downstream effector of cpt1b in regulating cardiomyocyte proliferation. Our study elucidates a novel mechanism whereby cpt1b promotes zebrafish cardiomyocyte proliferation through glutamine synthetase, which provides new perspectives on the significance of fatty acid metabolism in heart development and the interplay between fatty acid and amino acid metabolic pathways.
Collapse
Affiliation(s)
| | | | | | | | - Yanchao Han
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou 215000, China
| |
Collapse
|
11
|
Kim MJ, Cho SH, Seo Y, Kim SD, Park HC, Kim BJ. Neuro-Restorative Effect of Nimodipine and Calcitriol in 1-Methyl 4-Phenyl 1,2,3,6 Tetrahydropyridine-Induced Zebrafish Parkinson's Disease Model. J Korean Neurosurg Soc 2024; 67:510-520. [PMID: 38130142 PMCID: PMC11375070 DOI: 10.3340/jkns.2023.0189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/20/2023] [Accepted: 12/16/2023] [Indexed: 12/23/2023] Open
Abstract
OBJECTIVE Parkinson's disease (PD) is one of the most prevalent neurodegenerative diseases, characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta. The treatment of PD aims to alleviate motor symptoms by replacing the reduced endogenous dopamine. Currently, there are no disease-modifying agents for the treatment of PD. Zebrafish (Danio rerio) have emerged as an effective tool for new drug discovery and screening in the age of translational research. The neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is known to cause a similar loss of dopaminergic neurons in the human midbrain, with corresponding Parkinsonian symptoms. L-type calcium channels (LTCCs) have been implicated in the generation of mitochondrial oxidative stress, which underlies the pathogenesis of PD. Therefore, we investigated the neuro-restorative effect of LTCC inhibition in an MPTP-induced zebrafish PD model and suggested a possible drug candidate that might modify the progression of PD. METHODS All experiments were conducted using a line of transgenic zebrafish, Tg(dat:EGFP), in which green fluorescent protein (GFP) is expressed in dopaminergic neurons. The experimental groups were exposed to 500 μmol MPTP from 1 to 3 days post fertilization (dpf). The drug candidates : levodopa 1 mmol, nifedipine 10 μmol, nimodipine 3.5 μmol, diethylstilbestrol 0.3 μmol, luteolin 100 μmol, and calcitriol 0.25 μmol were exposed from 3 to 5 dpf. Locomotor activity was assessed by automated tracking and dopaminergic neurons were visualized in vivo by confocal microscopy. RESULTS Levodopa, nimodipine, diethylstilbestrol, and calcitriol had significant positive effects on the restoration of motor behavior, which was damaged by MPTP. Nimodipine and calcitriol have significant positive effects on the restoration of dopaminergic neurons, which were reduced by MPTP. Through locomotor analysis and dopaminergic neuron quantification, we identified the neuro-restorative effects of nimodipine and calcitriol in zebrafish MPTP-induced PD model. CONCLUSION The present study identified the neuro-restorative effects of nimodipine and calcitriol in an MPTP-induced zebrafish model of PD. They restored dopaminergic neurons which were damaged due to the effects of MPTP and normalized the locomotor activity. LTCCs have potential pathological roles in neurodevelopmental and neurodegenerative disorders. Zebrafish are highly amenable to high-throughput drug screening and might, therefore, be a useful tool to work towards the identification of diseasemodifying treatment for PD. Further studies including zebrafish genetic models to elucidate the mechanism of action of the diseasemodifying candidate by investigating Ca2+ influx and mitochondrial function in dopaminergic neurons, are needed to reveal the pathogenesis of PD and develop disease-modifying treatments for PD.
Collapse
Affiliation(s)
- Myung Ji Kim
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Seoul, Korea
| | - Su Hee Cho
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Seoul, Korea
| | - Yongbo Seo
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | - Sang-Dae Kim
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Seoul, Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | - Bum-Joon Kim
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
12
|
Freeburg SH, Shwartz A, Kemény LV, Smith CJ, Weeks O, Miller BM, PenkoffLidbeck N, Fisher DE, Evason KJ, Goessling W. Hepatocyte vitamin D receptor functions as a nutrient sensor that regulates energy storage and tissue growth in zebrafish. Cell Rep 2024; 43:114393. [PMID: 38944835 PMCID: PMC11708751 DOI: 10.1016/j.celrep.2024.114393] [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: 11/07/2023] [Revised: 03/20/2024] [Accepted: 06/07/2024] [Indexed: 07/02/2024] Open
Abstract
Vitamin D receptor (VDR) has been implicated in fatty liver pathogenesis, but its role in the regulation of organismal energy usage remains unclear. Here, we illuminate the evolutionary function of VDR by demonstrating that zebrafish Vdr coordinates hepatic and organismal energy homeostasis through antagonistic regulation of nutrient storage and tissue growth. Hepatocyte-specific Vdr impairment increases hepatic lipid storage, partially through acsl4a induction, while simultaneously diminishing fatty acid oxidation and liver growth. Importantly, Vdr impairment exacerbates the starvation-induced hepatic storage of systemic fatty acids, indicating that loss of Vdr signaling elicits hepatocellular energy deficiency. Strikingly, hepatocyte Vdr impairment diminishes diet-induced systemic growth while increasing hepatic and visceral fat in adult fish, revealing that hepatic Vdr signaling is required for complete adaptation to food availability. These data establish hepatocyte Vdr as a regulator of organismal energy expenditure and define an evolutionary function for VDR as a transcriptional effector of environmental nutrient supply.
Collapse
Affiliation(s)
- Scott H Freeburg
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Arkadi Shwartz
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lajos V Kemény
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; HCEMM-SU Translational Dermatology Research Group, Department of Physiology, Semmelweis University, 1085 Budapest Hungary; Department of Dermatology, Venereology, and Dermatooncology, Semmelweis University, 1085 Budapest Hungary
| | - Colton J Smith
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Olivia Weeks
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Bess M Miller
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nadia PenkoffLidbeck
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David E Fisher
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Kimberley J Evason
- Huntsman Cancer Institute and Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Wolfram Goessling
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard-MIT Division of Health Sciences and Technology, Boston, MA 02115, USA; Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
13
|
Abe K, Ino H, Niwa T, Semmy D, Takaochi A, Nishimura T, Mogi C, Uenaka M, Ishii M, Tanaka K, Ohkawa Y, Ishitani T. Sex-dependent regulation of vertebrate somatic growth and aging by germ cells. SCIENCE ADVANCES 2024; 10:eadi1621. [PMID: 38865462 PMCID: PMC11168456 DOI: 10.1126/sciadv.adi1621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/23/2024] [Indexed: 06/14/2024]
Abstract
The function of germ cells in somatic growth and aging has been demonstrated in invertebrate models but remains unclear in vertebrates. We demonstrated sex-dependent somatic regulation by germ cells in the short-lived vertebrate model Nothobranchius furzeri. In females, germ cell removal shortened life span, decreased estrogen, and increased insulin-like growth factor 1 (IGF-1) signaling. In contrast, germ cell removal in males improved their health with increased vitamin D signaling. Body size increased in both sexes but was caused by different signaling pathways, i.e., IGF-1 and vitamin D in females and males, respectively. Thus, vertebrate germ cells regulate somatic growth and aging through different pathways of the endocrine system, depending on the sex, which may underlie the sexual difference in reproductive strategies.
Collapse
Affiliation(s)
- Kota Abe
- Department of Homeostatic Regulation, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Hikaru Ino
- Department of Homeostatic Regulation, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Tomomi Niwa
- Department of Homeostatic Regulation, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Daniel Semmy
- Department of Homeostatic Regulation, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Ayami Takaochi
- Department of Homeostatic Regulation, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Takashi Nishimura
- Metabolic Regulation and Genetics, Department of Molecular and Cellular Biology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Chihiro Mogi
- Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Maki Uenaka
- Department of Immunology and Cell Biology, Graduate School of Medicine / Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
- Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine / Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
- Immunology Frontier Research Center, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
| | - Kaori Tanaka
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Tohru Ishitani
- Department of Homeostatic Regulation, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
| |
Collapse
|
14
|
Xu HW, Fang XY, Chen H, Zhang SB, Yi YY, Ge XY, Liu XW, Wang SJ. Vitamin D delays intervertebral disc degeneration and improves bone quality in ovariectomized rats. J Orthop Res 2024; 42:1314-1325. [PMID: 38225869 DOI: 10.1002/jor.25778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/01/2023] [Accepted: 12/18/2023] [Indexed: 01/17/2024]
Abstract
Known to be involved in bone-cartilage metabolism, Vitamin D (VD) may play a role in human's disc pathophysiology. Given that postmenopausal women are prone to suffer VD deficiency and intervertebral disc degeneration (IDD), this study is intended to investigate whether VD can delay IDD in ovariectomized rats by improving bone microstructure and antioxidant stress. Female Sprague-Dawley rats were randomly allocated into four groups: sham, oophorectomy (OVX)+VD deficiency (VDD), OVX, and OVX+VD supplementation (VDS). In vivo, after a 6-month intervention, imaging and pathology slice examinations showed that IDD induced by OVX was significantly alleviated in VDS and deteriorated by VDD. The expressions of aggrecan and Collagen II in intervertebral disc were reduced by OVX and VDD, and elevated by VDS. Compared with the OVX+VDD and OVX group vertebrae, OVX+VDS group vertebrae showed significantly improved endplate porosity and lumbar bone mineral density with increased percent bone volume and trabecular thickness. Furthermore, 1α,25(OH)2D3 restored the redox balance (total antioxidant capacity, ratio of oxidized glutathione/glutathione) in the disc. The cocultivation of 1α,25(OH)2D3 and nucleus pulposus cells (NPCs) was conducted to observe its potential ability to resist excessive oxidative stress damage induced by H2O2. In vitro experiments revealed that 1α,25(OH)2D3 reduced the senescence, apoptosis, and extracellular matrix degradation induced by H2O2 in NPCs. In conclusion, VDS exhibits protective effects in OVX-induced IDD, partly by regulating the redox balance and preserving the microstructure of endplate. This finding provides a new idea for the prevention and treatment of IDD.
Collapse
Affiliation(s)
- Hao-Wei Xu
- Department of Spinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin-Yue Fang
- Department of Spinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Chen
- Department of Spinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shu-Bao Zhang
- Department of Spinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yu-Yang Yi
- Department of Spinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiao-Yong Ge
- Department of Spinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiao-Wei Liu
- Department of Spinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shan-Jin Wang
- Department of Spinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Spinal Diseases, Jinggangshan University, Jiangxi, China
- Department of orthopedic, Shanghai East Hospital, Ji'An Hospital, Jiangxi, China
| |
Collapse
|
15
|
Apolínová K, Pérez FA, Dyballa S, Coppe B, Mercader Huber N, Terriente J, Di Donato V. ZebraReg-a novel platform for discovering regulators of cardiac regeneration using zebrafish. Front Cell Dev Biol 2024; 12:1384423. [PMID: 38799508 PMCID: PMC11116629 DOI: 10.3389/fcell.2024.1384423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide with myocardial infarction being the most prevalent. Currently, no cure is available to either prevent or revert the massive death of cardiomyocytes that occurs after a myocardial infarction. Adult mammalian hearts display a limited regeneration capacity, but it is insufficient to allow complete myocardial recovery. In contrast, the injured zebrafish heart muscle regenerates efficiently through robust proliferation of pre-existing myocardial cells. Thus, zebrafish allows its exploitation for studying the genetic programs behind cardiac regeneration, which may be present, albeit dormant, in the adult human heart. To this end, we have established ZebraReg, a novel and versatile automated platform for studying heart regeneration kinetics after the specific ablation of cardiomyocytes in zebrafish larvae. In combination with automated heart imaging, the platform can be integrated with genetic or pharmacological approaches and used for medium-throughput screening of presumed modulators of heart regeneration. We demonstrate the versatility of the platform by identifying both anti- and pro-regenerative effects of genes and drugs. In conclusion, we present a tool which may be utilised to streamline the process of target validation of novel gene regulators of regeneration, and the discovery of new drug therapies to regenerate the heart after myocardial infarction.
Collapse
Affiliation(s)
- Kateřina Apolínová
- ZeClinics SL, Barcelona, Spain
- Biomedicine, Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Pompeu Fabra University, Barcelona, Spain
| | | | | | - Benedetta Coppe
- Developmental Biology and Regeneration, Institute of Anatomy, University of Bern, Bern, Switzerland
- Department for Biomedical Research DBMR, University of Bern, Bern, Switzerland
| | - Nadia Mercader Huber
- Developmental Biology and Regeneration, Institute of Anatomy, University of Bern, Bern, Switzerland
- Department for Biomedical Research DBMR, University of Bern, Bern, Switzerland
- Centro Nacional de Investigaciones Cardiovasculares CNIC, Madrid, Spain
| | | | | |
Collapse
|
16
|
Kwon HJ. Knockdown of vitamin D receptor affects early stages of pectoral fin development in zebrafish. Anat Histol Embryol 2024; 53:e13044. [PMID: 38695121 DOI: 10.1111/ahe.13044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/30/2024] [Accepted: 04/18/2024] [Indexed: 05/29/2024]
Abstract
The vitamin D receptor (VDR) signalling has been implicated in vertebrate limb or fin formation. However, the involvement of VDR signalling in the early stages of limb/fin development remains to be elucidated. In this study, the role of VDR signalling in pectoral fin development was investigated in zebrafish embryos. Knockdown of vdr induced the severe impairment of pectoral fin development. The zebrafish larvae lacking vdr exhibited reduced pectoral fins with no skeletal elements. In situ hybridization revealed depletion of vdr downregulated fibroblast growth factor 24 (fgf24), a marker of early pectoral fin bud mesenchyme, in the presumptive fin field even before fin buds were visible. Moreover, a perturbed expression pattern of bone morphogenetic protein 4 (bmp4), a marker of the pectoral fin fold, was observed in the developing fin buds of zebrafish embryos that lost the vdr function. These findings suggest that VDR signalling is crucial in the early stages of fin development, potentially influencing the process by regulating other signalling molecules such as Fgf24 and Bmp4.
Collapse
Affiliation(s)
- Hye-Joo Kwon
- University of Utah Asia Campus, Incheon, South Korea
| |
Collapse
|
17
|
Xiong Y, Liu X, Jiang L, Hao T, Wang Y, Li T. Inhibition of ferroptosis reverses heart failure with preserved ejection fraction in mice. J Transl Med 2024; 22:199. [PMID: 38402404 PMCID: PMC10894491 DOI: 10.1186/s12967-023-04734-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] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 11/13/2023] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) accounts for approximately 50% of heart failure cases. The molecular mechanisms by which HFpEF leads to impaired diastolic function of the heart have not been clarified, nor have the drugs that target the clinical symptoms of HFpEF patients. METHODS HFpEF chip data (GSE180065) was downloaded from the National Center for Biotechnology Information (NCBI) database. Differentially expressed genes (DEGs) were filtered by the limma package in R and processed for GO and KEGG pathway analyses. Then, ferroptosis-related genes in HFpEF were identified by taking the intersection between DEGs and ferroptosis-related genes. CytoHubba and MCODE were used to screen ferroptosis-related hub DEGs in the protein-protein interaction (PPI) network. Establishment of a mouse HFpEF model to validate the transcript levels of ferroptosis-related hub DEGs and ferroptosis-related phenotypes. Transcript levels of ferroptosis-related hub DEGs and HFpEF phenotypic changes in the hearts of HFpEF mice were further examined after the use of ferroptosis inhibitors. RESULTS GO and KEGG enrichment analyses suggested that the DEGs in HFpEF were significantly enriched in ferroptosis-related pathways. A total of 24 ferroptosis-related DEGs were identified between the ferroptosis gene dataset and the DEGs. The established PPI network was further analyzed by CytoHubba and MCODE modules, and 11 ferroptosis-related hub DEGs in HFpEF were obtained. In animal experiments, HFpEF mice showed significant abnormal activation of ferroptosis. The expression trends of the 11 hub DEGs associated with ferroptosis, except for Cdh1, were consistent with the results of the bioinformatics analysis. Inhibition of ferroptosis alters the transcript levels of 11 ferroptosis-related hub DEGs and ameliorates HFpEF phenotypes. CONCLUSIONS The present study contributes to a deeper understanding of the specific mechanisms by which ferroptosis is involved in the development of HFpEF and suggests that inhibition of ferroptosis may mitigate the progression of HFpEF. In addition, eleven hub genes were recognized as potential drug binding targets.
Collapse
Affiliation(s)
- Yixiao Xiong
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, No 37 Wainan Guoxue Road, Sichuan, 610041, China
- Laboratory of Mitochondria and Metabolism, West China Hospital, Sichuan University, Sichuan, 610041, China
| | - Xin Liu
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, No 37 Wainan Guoxue Road, Sichuan, 610041, China
- Laboratory of Mitochondria and Metabolism, West China Hospital, Sichuan University, Sichuan, 610041, China
| | - Ling Jiang
- Department of Anesthesiology, West China Second Hospital of Sichuan University, Chengdu, 610041, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Tao Hao
- Department of Gastroenterology, Chengdu Fifth People's Hospital, No. 33 Mashi Street, Chengdu, 611130, Sichuan, China
| | - Yanyan Wang
- Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, No 37 Wainan Guoxue Road, Chengdu, 610041, Sichuan, China.
| | - Tao Li
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, No 37 Wainan Guoxue Road, Sichuan, 610041, China.
- Laboratory of Mitochondria and Metabolism, West China Hospital, Sichuan University, Sichuan, 610041, China.
| |
Collapse
|
18
|
Becker CJ, Cigliola V, Gillotay P, Rich A, De Simone A, Han Y, Di Talia S, Poss KD. In toto imaging of glial JNK signaling during larval zebrafish spinal cord regeneration. Development 2023; 150:dev202076. [PMID: 37997694 PMCID: PMC10753585 DOI: 10.1242/dev.202076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023]
Abstract
Identification of signaling events that contribute to innate spinal cord regeneration in zebrafish can uncover new targets for modulating injury responses of the mammalian central nervous system. Using a chemical screen, we identify JNK signaling as a necessary regulator of glial cell cycling and tissue bridging during spinal cord regeneration in larval zebrafish. With a kinase translocation reporter, we visualize and quantify JNK signaling dynamics at single-cell resolution in glial cell populations in developing larvae and during injury-induced regeneration. Glial JNK signaling is patterned in time and space during development and regeneration, decreasing globally as the tissue matures and increasing in the rostral cord stump upon transection injury. Thus, dynamic and regional regulation of JNK signaling help to direct glial cell behaviors during innate spinal cord regeneration.
Collapse
Affiliation(s)
- Clayton J. Becker
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Valentina Cigliola
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose, 06100 Nice, France
| | - Pierre Gillotay
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ashley Rich
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Alessandro De Simone
- Department of Genetics and Evolution, University of Geneva, 1211 Geneva, Switzerland
| | - Yanchao Han
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou, 215006 Jiangsu, China
| | - Stefano Di Talia
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Kenneth D. Poss
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
19
|
Li Y, Teng M, Zhao L, Sun J, Yan J, Zhu W, Wu F. Vitamin D modulates disordered lipid metabolism in zebrafish (Danio rerio) liver caused by exposure to polystyrene nanoplastics. ENVIRONMENT INTERNATIONAL 2023; 182:108328. [PMID: 37979534 DOI: 10.1016/j.envint.2023.108328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 11/20/2023]
Abstract
In this study, zebrafish (Danio rerio) were exposed to polystyrene nanoplastics (PS-NPs, 80 nm) at 0, 15, or 150 μg/L for 21 days and supplied with a low or high vitamin D (VD) diet (280 or 2800 IU/kg, respectively, indicated by - or +) to determine whether and how VD can regulate lipid metabolism disorder induced by PS-NPs. Six groups were created according to the PS-NP concentration and VD diet status: 0-, 0+, 15-, 15+,150-, and 150 +. Transmission electron microscopy showed that PS-NPs accumulated in the livers of zebrafish, which led to large numbers of vacuoles and lipid droplets in liver cell matrices; this accumulation was most prominent in the 150- group, wherein the number of lipid droplets increased significantly by 136.36%. However, the number of lipid droplets decreased significantly by 76.92% in the 150+ group compared with the 150- group. An examination of additional biochemical indicators showed that the high VD diet partially reversed the increases in the triglyceride and total cholesterol contents induced by PS-NPs (e.g., triglycerides decreased by 58.52% in the 150+ group, and total cholesterol decreased by 44.64% in the 15+ group), and regulated lipid metabolism disorder mainly by inhibiting lipid biosynthesis. Untargeted lipidomics analysis showed that exposure to PS-NPs was associated mainly with changes in the lipid molecular content related to cell membrane function and lipid biosynthesis and that the high VD diet reduced the content of lipid molecules related to lipid biosynthesis, effectively alleviating cell membrane damage and lipid accumulation. These findings highlight the potential of VD to alleviate lipid metabolism disorder caused by PS-NP exposure, thereby providing new insights into how the toxic effects of NPs on aquatic organisms could be reduced.
Collapse
Affiliation(s)
- Yunxia Li
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China; School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Miaomiao Teng
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China.
| | - Lihui Zhao
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Jiaqi Sun
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Jin Yan
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Wentao Zhu
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Fengchang Wu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China.
| |
Collapse
|
20
|
Abstract
Tissue regeneration is not simply a local repair event occurring in isolation from the distant, uninjured parts of the body. Rather, evidence indicates that regeneration is a whole-animal process involving coordinated interactions between different organ systems. Here, we review recent studies that reveal how remote uninjured tissues and organ systems respond to and engage in regeneration. We also discuss the need for toolkits and technological advancements to uncover and dissect organ communication during regeneration.
Collapse
Affiliation(s)
- Fei Sun
- Duke Regeneration Center, Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kenneth D. Poss
- Duke Regeneration Center, Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
21
|
Teng M, Li Y, Zhao X, White JC, Zhao L, Sun J, Zhu W, Wu F. Vitamin D modulation of brain-gut-virome disorder caused by polystyrene nanoplastics exposure in zebrafish (Danio rerio). MICROBIOME 2023; 11:266. [PMID: 38008755 PMCID: PMC10680193 DOI: 10.1186/s40168-023-01680-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 09/27/2023] [Indexed: 11/28/2023]
Abstract
BACKGROUND Many studies have investigated how nanoplastics (NPs) exposure mediates nerve and intestinal toxicity through a dysregulated brain-gut axis interaction, but there are few studies aimed at alleviating those effects. To determine whether and how vitamin D can impact that toxicity, fish were supplemented with a vitamin D-low diet and vitamin D-high diet. RESULTS Transmission electron microscopy (TEM) showed that polystyrene nanoplastics (PS-NPs) accumulated in zebrafish brain and intestine, resulting in brain blood-brain barrier basement membrane damage and the vacuolization of intestinal goblet cells and mitochondria. A high concentration of vitamin D reduced the accumulation of PS-NPs in zebrafish brain tissues by 20% and intestinal tissues by 58.8% and 52.2%, respectively, and alleviated the pathological damage induced by PS-NPs. Adequate vitamin D significantly increased the content of serotonin (5-HT) and reduced the anxiety-like behavior of zebrafish caused by PS-NPs exposure. Virus metagenome showed that PS-NPs exposure affected the composition and abundance of zebrafish intestinal viruses. Differentially expressed viruses in the vitamin D-low and vitamin D-high group affected the secretion of brain neurotransmitters in zebrafish. Virus AF191073 was negatively correlated with neurotransmitter 5-HT, whereas KT319643 was positively correlated with malondialdehyde (MDA) content and the expression of cytochrome 1a1 (cyp1a1) and cytochrome 1b1 (cyp1b1) in the intestine. This suggests that AF191073 and KT319643 may be key viruses that mediate the vitamin D reduction in neurotoxicity and immunotoxicity induced by PS-NPs. CONCLUSION Vitamin D can alleviate neurotoxicity and immunotoxicity induced by PS-NPs exposure by directionally altering the gut virome. These findings highlight the potential of vitamin D to alleviate the brain-gut-virome disorder caused by PS-NPs exposure and suggest potential therapeutic strategies to reduce the risk of NPs toxicity in aquaculture, that is, adding adequate vitamin D to diet. Video Abstract.
Collapse
Affiliation(s)
- Miaomiao Teng
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Yunxia Li
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Xiaoli Zhao
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China.
| | - Jason C White
- The Connecticut Agricultural Experiment Station, New Haven, CT, 06511, USA
| | - Lihui Zhao
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Jiaqi Sun
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Wentao Zhu
- Department of Applied Chemistry, Innovation Center of Pesticide Research, College of Science, China Agricultural University, Beijing, 100193, China
| | - Fengchang Wu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China.
| |
Collapse
|
22
|
Ernst A, Piragyte I, Mp AM, Le ND, Grandgirard D, Leib SL, Oates A, Mercader N. Identification of side effects of COVID-19 drug candidates on embryogenesis using an integrated zebrafish screening platform. Sci Rep 2023; 13:17037. [PMID: 37813860 PMCID: PMC10562458 DOI: 10.1038/s41598-023-43911-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/29/2023] [Indexed: 10/11/2023] Open
Abstract
Drug repurposing is an important strategy in COVID-19 treatment, but many clinically approved compounds have not been extensively studied in the context of embryogenesis, thus limiting their administration during pregnancy. Here we used the zebrafish embryo model organism to test the effects of 162 marketed drugs on cardiovascular development. Among the compounds used in the clinic for COVD-19 treatment, we found that Remdesivir led to reduced body size and heart functionality at clinically relevant doses. Ritonavir and Baricitinib showed reduced heart functionality and Molnupiravir and Baricitinib showed effects on embryo activity. Sabizabulin was highly toxic at concentrations only 5 times higher than Cmax and led to a mean mortality of 20% at Cmax. Furthermore, we tested if zebrafish could be used as a model to study inflammatory response in response to spike protein treatment and found that Remdesivir, Ritonavir, Molnupiravir, Baricitinib as well as Sabizabulin counteracted the inflammatory response related gene expression upon SARS-CoV-2 spike protein treatment. Our results show that the zebrafish allows to study immune-modulating properties of COVID-19 compounds and highlights the need to rule out secondary defects of compound treatment on embryogenesis. All results are available on a user friendly web-interface https://share.streamlit.io/alernst/covasc_dataapp/main/CoVasc_DataApp.py that provides a comprehensive overview of all observed phenotypic effects and allows personalized search on specific compounds or group of compounds. Furthermore, the presented platform can be expanded for rapid detection of developmental side effects of new compounds for treatment of COVID-19 and further viral infectious diseases.
Collapse
Affiliation(s)
| | - Indre Piragyte
- Institute of Anatomy, University of Bern, Bern, Switzerland
- Department for Biomedical Research DBMR, University of Bern, Bern, Switzerland
| | - Ayisha Marwa Mp
- Institute of Anatomy, University of Bern, Bern, Switzerland
- Department for Biomedical Research DBMR, University of Bern, Bern, Switzerland
| | - Ngoc Dung Le
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Denis Grandgirard
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Stephen L Leib
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Andrew Oates
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Nadia Mercader
- Institute of Anatomy, University of Bern, Bern, Switzerland.
- Department for Biomedical Research DBMR, University of Bern, Bern, Switzerland.
- Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain.
| |
Collapse
|
23
|
Zhang M, Wang M, Jiang J, Liu W, Zhou S, Wang D, Wang M, Zhao Z, Xu Z, Wu W, Lin X, Zhang J, Xu W, Tang Q, Zhan R, Liu W, Yang L, Zhou X, Zhou W, Lei M. COX2-ATP Synthase Regulates Spine Follicle Size in Hedgehogs. Int J Biol Sci 2023; 19:4763-4777. [PMID: 37781513 PMCID: PMC10539703 DOI: 10.7150/ijbs.83387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 08/26/2023] [Indexed: 10/03/2023] Open
Abstract
Skin evolves essential appendages with adaptive patterns that synergistically insulate the body from environmental insults. How similar appendages in different animals generate diversely-sized appendages remain elusive. Here we used hedgehog spine follicles and mouse hair follicles as models to investigate how similar follicles form in different sizes postnatally. Histology and immunostaining show that the spine follicles have a significantly greater size than the hair follicles. By RNA-sequencing analysis, we found that ATP synthases are highly expressed in hedgehog skin compared to mouse skin. Inhibition of ATP synthase resulted in smaller spine follicle formation during regeneration. We also identified that the mitochondrial gene COX2 functions upstream of ATP synthase that influences energy metabolism and cell proliferation to control the size of the spine follicles. Our study identified molecules that function differently in forming diversely-sized skin appendages across different animals, allowing them to adapt to the living environment and benefit from self-protection.
Collapse
Affiliation(s)
- Man Zhang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Mengyue Wang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jingwei Jiang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Weiwei Liu
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Siyi Zhou
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Dehuan Wang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Miaomiao Wang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Zixian Zhao
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Zhiling Xu
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Wang Wu
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Three Gorges Hospital, Chongqing University, Chongqing 404000, China
| | - Xia Lin
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Three Gorges Hospital, Chongqing University, Chongqing 404000, China
| | - Jinwei Zhang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Department of Dermatology and Cosmetology, The First Affiliated Hospital of Chongqing College of Traditional Chinese Medicine, Chongqing 400021, China
| | - Wei Xu
- Department of Dermatology and Cosmetology, The First Affiliated Hospital of Chongqing College of Traditional Chinese Medicine, Chongqing 400021, China
| | - Qu Tang
- Three Gorges Hospital, Chongqing University, Chongqing 404000, China
| | - Rixing Zhan
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Wanqian Liu
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Li Yang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Xun Zhou
- Department of Dermatology and Cosmetology, The First Affiliated Hospital of Chongqing College of Traditional Chinese Medicine, Chongqing 400021, China
| | - Wei Zhou
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Mingxing Lei
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| |
Collapse
|
24
|
Kasamoto M, Funakoshi S, Hatani T, Okubo C, Nishi Y, Tsujisaka Y, Nishikawa M, Narita M, Ohta A, Kimura T, Yoshida Y. Am80, a retinoic acid receptor agonist, activates the cardiomyocyte cell cycle and enhances engraftment in the heart. Stem Cell Reports 2023; 18:1672-1685. [PMID: 37451261 PMCID: PMC10444569 DOI: 10.1016/j.stemcr.2023.06.006] [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: 03/10/2022] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
Human induced pluripotent stem cell-derived (hiPSC) cardiomyocytes are a promising source for regenerative therapy. To realize this therapy, however, their engraftment potential after their injection into the host heart should be improved. Here, we established an efficient method to analyze the cell cycle activity of hiPSC cardiomyocytes using a fluorescence ubiquitination-based cell cycle indicator (FUCCI) system. In vitro high-throughput screening using FUCCI identified a retinoic acid receptor (RAR) agonist, Am80, as an effective cell cycle activator in hiPSC cardiomyocytes. The transplantation of hiPSC cardiomyocytes treated with Am80 before the injection significantly enhanced the engraftment in damaged mouse heart for 6 months. Finally, we revealed that the activation of endogenous Wnt pathways through both RARA and RARB underlies the Am80-mediated cell cycle activation. Collectively, this study highlights an efficient method to activate cell cycle in hiPSC cardiomyocytes by Am80 as a means to increase the graft size after cell transplantation into a damaged heart.
Collapse
Affiliation(s)
- Manabu Kasamoto
- Centre for iPS Cell Research and Application, Kyoto University, Kyoto, Japan; Department of Cardiovascular Medicine, Kyoto University Hospital, Kyoto, Japan
| | - Shunsuke Funakoshi
- Centre for iPS Cell Research and Application, Kyoto University, Kyoto, Japan; Takeda-CiRA Joint program (T-CiRA), Fujisawa, Japan.
| | - Takeshi Hatani
- Centre for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Chikako Okubo
- Centre for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Yohei Nishi
- Centre for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Yuta Tsujisaka
- Centre for iPS Cell Research and Application, Kyoto University, Kyoto, Japan; Department of Cardiovascular Medicine, Kyoto University Hospital, Kyoto, Japan
| | - Misato Nishikawa
- Centre for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Megumi Narita
- Centre for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Akira Ohta
- Centre for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Kyoto University Hospital, Kyoto, Japan
| | - Yoshinori Yoshida
- Centre for iPS Cell Research and Application, Kyoto University, Kyoto, Japan; Takeda-CiRA Joint program (T-CiRA), Fujisawa, Japan.
| |
Collapse
|
25
|
Nguyen PD, Gooijers I, Campostrini G, Verkerk AO, Honkoop H, Bouwman M, de Bakker DEM, Koopmans T, Vink A, Lamers GEM, Shakked A, Mars J, Mulder AA, Chocron S, Bartscherer K, Tzahor E, Mummery CL, de Boer TP, Bellin M, Bakkers J. Interplay between calcium and sarcomeres directs cardiomyocyte maturation during regeneration. Science 2023; 380:758-764. [PMID: 37200435 DOI: 10.1126/science.abo6718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/20/2023] [Indexed: 05/20/2023]
Abstract
Zebrafish hearts can regenerate by replacing damaged tissue with new cardiomyocytes. Although the steps leading up to the proliferation of surviving cardiomyocytes have been extensively studied, little is known about the mechanisms that control proliferation and redifferentiation to a mature state. We found that the cardiac dyad, a structure that regulates calcium handling and excitation-contraction coupling, played a key role in the redifferentiation process. A component of the cardiac dyad called leucine-rich repeat-containing 10 (Lrrc10) acted as a negative regulator of proliferation, prevented cardiomegaly, and induced redifferentiation. We found that its function was conserved in mammalian cardiomyocytes. This study highlights the importance of the underlying mechanisms required for heart regeneration and their application to the generation of fully functional cardiomyocytes.
Collapse
Affiliation(s)
- Phong D Nguyen
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Iris Gooijers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Giulia Campostrini
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam University Medical Center, Amsterdam, Netherlands
- Department of Experimental Cardiology, University of Amsterdam, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Hessel Honkoop
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Mara Bouwman
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Dennis E M de Bakker
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Tim Koopmans
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Department of Animal Physiology, Osnabrueck University, Osnabrück, Germany
| | - Aryan Vink
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Gerda E M Lamers
- Core Facility Microscopy, Institute of Biology, Leiden University, Leiden, Netherlands
| | - Avraham Shakked
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jonas Mars
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Aat A Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Sonja Chocron
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Kerstin Bartscherer
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Department of Animal Physiology, Osnabrueck University, Osnabrück, Germany
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Teun P de Boer
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
- Department of Biology, University of Padua, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Department of Pediatric Cardiology, Division of Pediatrics, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
26
|
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
|
27
|
Zuppo DA, Missinato MA, Santana-Santos L, Li G, Benos PV, Tsang M. Foxm1 regulates cardiomyocyte proliferation in adult zebrafish after cardiac injury. Development 2023; 150:dev201163. [PMID: 36846912 PMCID: PMC10108034 DOI: 10.1242/dev.201163] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023]
Abstract
The regenerative capacity of the mammalian heart is poor, with one potential reason being that adult cardiomyocytes cannot proliferate at sufficient levels to replace lost tissue. During development and neonatal stages, cardiomyocytes can successfully divide under injury conditions; however, as these cells mature their ability to proliferate is lost. Therefore, understanding the regulatory programs that can induce post-mitotic cardiomyocytes into a proliferative state is essential to enhance cardiac regeneration. Here, we report that the forkhead transcription factor Foxm1 is required for cardiomyocyte proliferation after injury through transcriptional regulation of cell cycle genes. Transcriptomic analysis of injured zebrafish hearts revealed that foxm1 expression is increased in border zone cardiomyocytes. Decreased cardiomyocyte proliferation and expression of cell cycle genes in foxm1 mutant hearts was observed, suggesting it is required for cell cycle checkpoints. Subsequent analysis of a candidate Foxm1 target gene, cenpf, revealed that this microtubule and kinetochore binding protein is also required for cardiac regeneration. Moreover, cenpf mutants show increased cardiomyocyte binucleation. Thus, foxm1 and cenpf are required for cardiomyocytes to complete mitosis during zebrafish cardiac regeneration.
Collapse
Affiliation(s)
- Daniel A. Zuppo
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Maria A. Missinato
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
- Avidity Biosciences, 10578 Science Center Dr. Suite 125, San Diego, CA 92121, USA
| | - Lucas Santana-Santos
- Department of Computational and Systems Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Guang Li
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Panayiotis V. Benos
- Department of Computational and Systems Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Michael Tsang
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
28
|
Sørhus E, Donald CE, Nakken CL, Perrichon P, Durif CMF, Shema S, Browman HI, Skiftesvik AB, Lie KK, Rasinger JD, Müller MHB, Meier S. Co-exposure to UV radiation and crude oil increases acute embryotoxicity and sublethal malformations in the early life stages of Atlantic haddock (Melanogrammus aeglefinus). THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 859:160080. [PMID: 36375555 DOI: 10.1016/j.scitotenv.2022.160080] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/28/2022] [Accepted: 11/05/2022] [Indexed: 06/16/2023]
Abstract
Crude oil causes severe abnormalities in developing fish. Photomodification of constituents in crude oil increases its toxicity several fold. We report on the effect of crude oil, in combination with ultraviolet (UV) radiation, on Atlantic haddock (Melanogrammus aeglefinus) embryos. Accumulation of crude oil on the eggshell makes haddock embryos particularly susceptible to exposure. At high latitudes, they can be exposed to UV radiation many hours a day. Haddock embryos were exposed to crude oil (5-300 μg oil/L nominal loading concentrations) for three days in the presence and absence of UV radiation (290-400 nm). UV radiation partly degraded the eggs' outer membrane resulting in less accumulation of oil droplets in the treatment with highest oil concentration (300 μg oil/L). The co-exposure treatments resulted in acute toxicity, manifested by massive tissue necrosis and subsequent mortality, reducing LC50 at hatching stage by 60 % to 0.24 μg totPAH/L compared to 0.62 μg totPAH/L in crude oil only. In the treatment with nominal low oil concentrations (5-30 μg oil/L), only co-exposure to UV led to sublethal morphological heart defects. Including phototoxicity as a parameter in risk assessments of accidental oil spills is recommended.
Collapse
Affiliation(s)
- Elin Sørhus
- Institute of Marine Research, Marine Toxicology Group, Nordnesgaten 50, 5005 Bergen, Norway.
| | - Carey E Donald
- Institute of Marine Research, Marine Toxicology Group, Nordnesgaten 50, 5005 Bergen, Norway
| | - Charlotte L Nakken
- University of Bergen, Department of Chemistry, Allégaten 41, 5020 Bergen, Norway
| | - Prescilla Perrichon
- Institute of Marine Research, Reproduction and Developmental Biology, Austevoll Research Station, Sauganeset 16, 5392 Storebø, Norway
| | - Caroline M F Durif
- Institute of Marine Research, Ecosystem Acoustics Group, Austevoll Research Station, Sauganeset 16, 5392 Storebø, Norway
| | - Steven Shema
- Grótti ehf, Melabraut 22, 220 Hafnarfirði, Iceland
| | - Howard I Browman
- Institute of Marine Research, Ecosystem Acoustics Group, Austevoll Research Station, Sauganeset 16, 5392 Storebø, Norway
| | - Anne Berit Skiftesvik
- Institute of Marine Research, Ecosystem Acoustics Group, Austevoll Research Station, Sauganeset 16, 5392 Storebø, Norway
| | - Kai K Lie
- Institute of Marine Research, Marine Toxicology Group, Nordnesgaten 50, 5005 Bergen, Norway
| | - Josef D Rasinger
- Institute of Marine Research, Marine Toxicology Group, Nordnesgaten 50, 5005 Bergen, Norway
| | - Mette H B Müller
- Norwegian University of Life Sciences, Section for Experimental Biomedicine, Universitetstunet 3, 1433 Ås, Norway
| | - Sonnich Meier
- Institute of Marine Research, Marine Toxicology Group, Nordnesgaten 50, 5005 Bergen, Norway
| |
Collapse
|
29
|
Liu R, Lu Y, Peng X, Jia J, Ruan Y, Shi S, Shu T, Li T, Jin X, Zhai G, He J, Lou Q, Yin Z. Enhanced insulin activity achieved in VDRa/b ablation zebrafish. Front Endocrinol (Lausanne) 2023; 14:1054665. [PMID: 36864841 PMCID: PMC9972578 DOI: 10.3389/fendo.2023.1054665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
INTRODUCTION 1α,25-dihydroxyvitamin D3 (1α,25[OH]2VD3) is a hormone known for its key roles in calcium absorption and nutrient metabolism. In teleost fishes, 1α,25(OH)2VD3 insufficiency causes impaired glucose metabolism and lipid oxidation. However, the cascade and mechanisms of 1α,25(OH)2VD3 and the vitamin d receptor (VDR) signaling are unclear. RESULTS In this study, two genes (vdra and vdrb) encoding paralogs of VDRs were genetically knocked out in zebrafish. Growth retardation and accumulated visceral adipose tissue have been observed in vdra -/-;vdrb -/- deficient line. In the liver elevated accumulation of triglycerides and suppressed lipid oxidation were detected. Morover significantly elevated 1α,25(OH)2VD3 levels were detected in vdra-/-;vdrb-/- zebrafish due to cyp24a1 transcription repression. Furthermore VDRs ablation Enhanced insulin signaling including elevated insulin/insra trancriptional levels, glycolysis, lipogenesis and promoted AKT/mTOR activity. DISCUSSION In conclusion, our present studies provides a zebrafish model with an elevated 1α,25(OH)2VD3 levels in vivo. The 1α,25(OH)2VD3/VDRs signaling promote lipid oxidation activity. However 1α,25(OH)2VD3 activity of regulation of glucose homeostasis through Insulin/Insr was independent of nuclear VDRs in teleosts.
Collapse
Affiliation(s)
- Ruolan Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yao Lu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Xuyan Peng
- The Laboratory of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zheng Zhou, China
| | - Jingyi Jia
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yonglin Ruan
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shengchi Shi
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Tingting Shu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Tianhui Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Xia Jin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Gang Zhai
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Jiangyan He
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Qiyong Lou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- *Correspondence: Qiyong Lou,
| | - Zhan Yin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
- The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
30
|
Kübler IC, Kretzschmar J, Brankatschk M, Sandoval-Guzmán T. Local problems need global solutions: The metabolic needs of regenerating organisms. Wound Repair Regen 2022; 30:652-664. [PMID: 35596643 PMCID: PMC7613859 DOI: 10.1111/wrr.13029] [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: 01/31/2022] [Revised: 05/03/2022] [Accepted: 05/19/2022] [Indexed: 12/01/2022]
Abstract
The vast majority of species that belong to the plant or animal kingdom evolved with two main strategies to counter tissue damage-scar formation and regeneration. Whereas scar formation provides a fast and cost-effective repair to exit life-threatening conditions, complete tissue regeneration is time-consuming and requires vast resources to reinstall functionality of affected organs or structures. Local environments in wound healing are widely studied and findings have provided important biomedical applications. Less well understood are organismic physiological parameters and signalling circuits essential to maintain effective tissue repair. Here, we review accumulated evidence that positions the interplay of local and systemic changes in metabolism as essential variables modulating the injury response. We particularly emphasise the role of lipids and lipid-like molecules as significant components long overlooked.
Collapse
Affiliation(s)
- Ines C. Kübler
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Jenny Kretzschmar
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Marko Brankatschk
- Department of Molecular, Cell and Developmental Biology, Technische Universität Dresden, Dresden, Germany
| | - Tatiana Sandoval-Guzmán
- Department of Internal Medicine III, Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Centre Munich, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
31
|
Zhou S, Liu Z, Kawakami A. A PI3Kγ signal regulates macrophage recruitment to injured tissue for regenerative cell survival. Dev Growth Differ 2022; 64:433-445. [PMID: 36101496 PMCID: PMC9826243 DOI: 10.1111/dgd.12809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/20/2022] [Accepted: 08/03/2022] [Indexed: 01/11/2023]
Abstract
The interaction between immune cells and injured tissues is crucial for regeneration. Previous studies have shown that macrophages attenuate inflammation caused by injuries to support the survival of primed regenerative cells. Macrophage loss in zebrafish mutants like cloche (clo) causes extensive apoptosis in the regenerative cells of the amputated larval fin fold. However, the mechanism of interaction between macrophage and injured tissue is poorly understood. Here, we show that a phosphoinositide 3-kinase gamma (PI3Kγ)-mediated signal is essential for recruiting macrophages to the injured tissue. PI3Kγ inhibition by the PI3Kγ-specific inhibitor, 5-quinoxalin-6-ylmethylene-thiazolidine-2,4-dione (AS605240 or AS), displayed a similar apoptosis phenotype with that observed in clo mutants. We further show that PI3Kγ function during the early regenerative stage is necessary for macrophage recruitment to the injured site. Additionally, protein kinase B (Akt) overexpression in the AS-treated larvae suggested that Akt is not the direct downstream mediator of PI3Kγ for macrophage recruitment, while it independently plays a role for the survival of regenerative cells. Together, our study reveals that PI3Kγ plays a role for recruiting macrophages in response to regeneration.
Collapse
Affiliation(s)
- Siyu Zhou
- School of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
| | - Zhengcheng Liu
- School of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
| | - Atsushi Kawakami
- School of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
| |
Collapse
|
32
|
Hu B, Lelek S, Spanjaard B, El-Sammak H, Simões MG, Mintcheva J, Aliee H, Schäfer R, Meyer AM, Theis F, Stainier DYR, Panáková D, Junker JP. Origin and function of activated fibroblast states during zebrafish heart regeneration. Nat Genet 2022; 54:1227-1237. [PMID: 35864193 PMCID: PMC7613248 DOI: 10.1038/s41588-022-01129-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 06/09/2022] [Indexed: 12/12/2022]
Abstract
The adult zebrafish heart has a high capacity for regeneration following injury. However, the composition of the regenerative niche has remained largely elusive. Here, we dissected the diversity of activated cell states in the regenerating zebrafish heart based on single-cell transcriptomics and spatiotemporal analysis. We observed the emergence of several transient cell states with fibroblast characteristics following injury, and we outlined the proregenerative function of collagen-12-expressing fibroblasts. To understand the cascade of events leading to heart regeneration, we determined the origin of these cell states by high-throughput lineage tracing. We found that activated fibroblasts were derived from two separate sources: the epicardium and the endocardium. Mechanistically, we determined Wnt signalling as a regulator of the endocardial fibroblast response. In summary, our work identifies specialized activated fibroblast cell states that contribute to heart regeneration, thereby opening up possible approaches to modulating the regenerative capacity of the vertebrate heart. Single-cell RNA sequencing and spatiotemporal analysis of the regenerating zebrafish heart identify transient proregenerative fibroblast-like cells that are derived from the epicardium and the endocardium. Wnt signalling regulates the endocardial fibroblast response.
Collapse
Affiliation(s)
- Bo Hu
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Sara Lelek
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research) partner site, Berlin, Germany
| | - Bastiaan Spanjaard
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Hadil El-Sammak
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,DZHK (German Centre for Cardiovascular Research) partner site Rhine/Main, Frankfurt, Germany
| | - Mariana Guedes Simões
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Janita Mintcheva
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Hananeh Aliee
- Helmholtz Center Munich - German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Munich, Germany
| | - Ronny Schäfer
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Alexander M Meyer
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Fabian Theis
- Helmholtz Center Munich - German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Munich, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,DZHK (German Centre for Cardiovascular Research) partner site Rhine/Main, Frankfurt, Germany
| | - Daniela Panáková
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany. .,DZHK (German Centre for Cardiovascular Research) partner site, Berlin, Germany.
| | - Jan Philipp Junker
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology, Berlin, Germany. .,DZHK (German Centre for Cardiovascular Research) partner site, Berlin, Germany.
| |
Collapse
|
33
|
Abstract
Heart disease is the leading cause of death worldwide. Despite decades of research, most heart pathologies have limited treatments, and often the only curative approach is heart transplantation. Thus, there is an urgent need to develop new therapeutic approaches for treating cardiac diseases. Animal models that reproduce the human pathophysiology are essential to uncovering the biology of diseases and discovering therapies. Traditionally, mammals have been used as models of cardiac disease, but the cost of generating and maintaining new models is exorbitant, and the studies have very low throughput. In the last decade, the zebrafish has emerged as a tractable model for cardiac diseases, owing to several characteristics that made this animal popular among developmental biologists. Zebrafish fertilization and development are external; embryos can be obtained in high numbers, are cheap and easy to maintain, and can be manipulated to create new genetic models. Moreover, zebrafish exhibit an exceptional ability to regenerate their heart after injury. This review summarizes 25 years of research using the zebrafish to study the heart, from the classical forward screenings to the contemporary methods to model mutations found in patients with cardiac disease. We discuss the advantages and limitations of this model organism and introduce the experimental approaches exploited in zebrafish, including forward and reverse genetics and chemical screenings. Last, we review the models used to induce cardiac injury and essential ideas derived from studying natural regeneration. Studies using zebrafish have the potential to accelerate the discovery of new strategies to treat cardiac diseases.
Collapse
Affiliation(s)
- Juan Manuel González-Rosa
- Cardiovascular Research Center, Massachusetts General Hospital Research Institute, Harvard Medical School, Charlestown, MA
| |
Collapse
|
34
|
Peters MC, Di Martino S, Boelens T, Qin J, van Mil A, Doevendans PA, Chamuleau SAJ, Sluijter JPG, Neef K. Follistatin-like 1 promotes proliferation of matured human hypoxic iPSC-cardiomyocytes and is secreted by cardiac fibroblasts. Mol Ther Methods Clin Dev 2022; 25:3-16. [PMID: 35317048 PMCID: PMC8917270 DOI: 10.1016/j.omtm.2022.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 02/19/2022] [Indexed: 12/12/2022]
Abstract
The human heart has limited regenerative capacity. Therefore, patients often progress to heart failure after ischemic injury, despite advances in reperfusion therapies generally decreasing mortality. Depending on its glycosylation state, Follistatin-like 1 (FSTL1) has been shown to increase cardiomyocyte (CM) proliferation, decrease CM apoptosis, and prevent cardiac rupture in animal models of ischemic heart disease. To explore its therapeutic potential, we used a human in vitro model of cardiac ischemic injury with human induced pluripotent stem cell-derived CMs (iPSC-CMs) and assessed regenerative effects of two differently glycosylated variants of human FSTL1. Furthermore, we investigated the FSTL1-mediated interplay between human cardiac fibroblasts (cFBs) and iPSC-CMs in hypoxia. Both FSTL1 variants increased viability, while only hypo-glycosylated FSTL1 increased CM proliferation post-hypoxia. Human fetal cardiac fibroblasts (fcFBs) expressed and secreted FSTL1 under normoxic conditions, while FSTL1 secretion increased by iPSC-cFBs upon hypoxia but decreased in iPSC-CMs. Co-culture of iPSC-CMs and cFBs increased FSTL1 secretion compared with cFB mono-culture. Taken together, we confirm that FSTL1 induces iPSC-CM proliferation in a human cardiac in vitro hypoxia damage model. Furthermore, we show hypoxia-related FSTL1 secretion by human cFBs and indications for FSTL1-mediated intercellular communication between cardiac cell types in response to hypoxic conditions.
Collapse
Affiliation(s)
- Marijn C Peters
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Sofia Di Martino
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Thomas Boelens
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Jiabin Qin
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Alain van Mil
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Pieter A Doevendans
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Steven A J Chamuleau
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands.,Department of Cardiology, Amsterdam Medical Centre, 1105 AZ Amsterdam, the Netherlands
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Klaus Neef
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| |
Collapse
|
35
|
Das RN, Tevet Y, Safriel S, Han Y, Moshe N, Lambiase G, Bassi I, Nicenboim J, Brückner M, Hirsch D, Eilam-Altstadter R, Herzog W, Avraham R, Poss KD, Yaniv K. Generation of specialized blood vessels via lymphatic transdifferentiation. Nature 2022; 606:570-575. [PMID: 35614218 PMCID: PMC9875863 DOI: 10.1038/s41586-022-04766-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 04/14/2022] [Indexed: 01/27/2023]
Abstract
The lineage and developmental trajectory of a cell are key determinants of cellular identity. In the vascular system, endothelial cells (ECs) of blood and lymphatic vessels differentiate and specialize to cater to the unique physiological demands of each organ1,2. Although lymphatic vessels were shown to derive from multiple cellular origins, lymphatic ECs (LECs) are not known to generate other cell types3,4. Here we use recurrent imaging and lineage-tracing of ECs in zebrafish anal fins, from early development to adulthood, to uncover a mechanism of specialized blood vessel formation through the transdifferentiation of LECs. Moreover, we demonstrate that deriving anal-fin vessels from lymphatic versus blood ECs results in functional differences in the adult organism, uncovering a link between cell ontogeny and functionality. We further use single-cell RNA-sequencing analysis to characterize the different cellular populations and transition states involved in the transdifferentiation process. Finally, we show that, similar to normal development, the vasculature is rederived from lymphatics during anal-fin regeneration, demonstrating that LECs in adult fish retain both potency and plasticity for generating blood ECs. Overall, our research highlights an innate mechanism of blood vessel formation through LEC transdifferentiation, and provides in vivo evidence for a link between cell ontogeny and functionality in ECs.
Collapse
Affiliation(s)
- Rudra N. Das
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel, Corresponding Authors Karina Yaniv Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100, Israel, , Rudra N. Das Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100, Israel,
| | - Yaara Tevet
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Stav Safriel
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Yanchao Han
- Duke Regeneration Center, Department of Cell Biology, Duke University School of Medicine, Durham, United States, Institute for Cardiovascular Science, Medical College, Soochow University, Suzhou, China
| | - Noga Moshe
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Giuseppina Lambiase
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Ivan Bassi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Julian Nicenboim
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Matthias Brückner
- University of Muenster and Max Plank Institute for Molecular Biomedicine, Muenster, Germany
| | - Dana Hirsch
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | | | - Wiebke Herzog
- University of Muenster and Max Plank Institute for Molecular Biomedicine, Muenster, Germany
| | - Roi Avraham
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Kenneth D. Poss
- Duke Regeneration Center, Department of Cell Biology, Duke University School of Medicine, Durham, United States
| | - Karina Yaniv
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel, Corresponding Authors Karina Yaniv Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100, Israel, , Rudra N. Das Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100, Israel,
| |
Collapse
|
36
|
Kriventsov MA, Yermola YA, Davydova AA, Beketov AA, Makalish TP, Zyablitskaya EY, Geraschenko AV, Kubyshkin AV, Galyshevskaya AA, Zausalina AI. Immunohistochemical Expression of VDR in Myocardium: Postmortem Evaluation of COVID-19 Patients. J Histochem Cytochem 2022; 70:391-399. [PMID: 35357255 PMCID: PMC9058373 DOI: 10.1369/00221554221089916] [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: 11/22/2022] Open
Abstract
Morphological data on heart damage and its mechanisms due to extremely severe course of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection are limited, as well as data on the correlation of damage and expression of vitamin D receptors (VDRs). In this study, we analyzed a series of myocardial samples obtained during postmortem autopsy of 48 critically ill patients with COVID-19 who died with SARS-CoV-2-associated pneumonia. The purpose of the study was to evaluate immunohistochemical VDR expression in the myocardium. The results showed the only minimal or no immunohistochemical expression of VDR in the nuclei of cardiomyocytes in most cases, along with the persisted strong expression in lymphoid cells. To the best of our knowledge, it is the first study and data provided were regarding myocardial VDR expression in COVID-19 patients. The results are of interest in terms of further study of the effects of ligand-associated VDR activation on the cardiovascular system.
Collapse
Affiliation(s)
- Maxim A Kriventsov
- Department of Pathomorphology, Medical Academy named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University, Simferopol, Russia
| | - Yulianna A Yermola
- Department of Pathomorphology, Medical Academy named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University, Simferopol, Russia
| | - Alexandra A Davydova
- Department of Pathomorphology, Medical Academy named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University, Simferopol, Russia
| | - Alexey A Beketov
- Department of Pathomorphology, Medical Academy named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University, Simferopol, Russia
| | - Tatyana P Makalish
- Central Research Laboratory, Medical Academy named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University, Simferopol, Russia
| | - Evgeniya Yu Zyablitskaya
- Central Research Laboratory, Medical Academy named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University, Simferopol, Russia
| | - Alina V Geraschenko
- Central Research Laboratory, Medical Academy named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University, Simferopol, Russia
| | - Anatoly V Kubyshkin
- Department of Pathophysiology, Medical Academy named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University, Simferopol, Russia
| | - Anna A Galyshevskaya
- Department of Pathomorphology, Medical Academy named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University, Simferopol, Russia
| | - Anastasia I Zausalina
- Department of Pathomorphology, Medical Academy named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University, Simferopol, Russia
| |
Collapse
|
37
|
Reconstruction of regulatory network predicts transcription factors driving the dynamics of zebrafish heart regeneration. Gene X 2022; 819:146242. [PMID: 35114280 DOI: 10.1016/j.gene.2022.146242] [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: 09/09/2021] [Revised: 12/21/2021] [Accepted: 01/18/2022] [Indexed: 11/24/2022] Open
Abstract
The limited regenerative capacity in mammals has serious implications for cardiac tissue damage. Meanwhile, zebrafish has a high regenerative capacity, but the regulation of the heart healing process has yet to be elucidated. The dynamic nature of cardiac regeneration requires consideration of the inherent temporal dimension of this process. Here, we conducted a systematic review to find genes that define the regenerative cell state of the zebrafish heart. We then performed an in silico temporal gene regulatory network analysis using transcriptomic data from the zebrafish heart regenerative process obtained from databases. In this analysis, the genes found in the systematic review were used to represent the final cell state of the transition process from a non-regenerative cell state to a regenerative state. We found 135 transcription factors driving the cellular state transition process during zebrafish cardiac regeneration, including Hand2, Nkx2.5, Tbx20, Fosl1, Fosb, Junb, Vdr, Wt1, and Tcf21 previously reported for playing a key role in tissue regeneration. Furthermore, we demonstrate that most regulators are activated in the first days post-injury, indicating that the transition from a non-regenerative to a regenerative state occurs promptly.
Collapse
|
38
|
Zhou L, Yang R, Tian H, Qin X, Ye C, Shi X, Xia C, Cai T, Xie Y, Jia Y, Hu G. Sexual dimorphism in Odontobutis sinensis brain-pituitary-gonad axis and liver highlighted by histological and transcriptomic approach. Gene 2022; 819:146264. [PMID: 35114283 DOI: 10.1016/j.gene.2022.146264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/18/2022] [Accepted: 01/27/2022] [Indexed: 11/24/2022]
Abstract
In this study, sexual dimorphism in Chinese dark sleeper (Odontobutis sinensis) brain-pituitary-gonad axis and liver was highlighted by histological and transcriptomic approach. The results showed that there were two significant differences between males and females. Firstly, males grew larger and faster than females. Transcriptomic analysis and qPCR validation indicated that two key growth genes, insulin-like growth factor (igf) and 25-hydroxyvitamin D-1 alpha hydroxylase (cyp27b), were more highly detected in male liver than that in female liver. Secondly, histological analysis displayed that the liver in males showed an obvious ivory fatty phenomenon with more fat vacuoles and lipid droplet aggregation compared to that in females. Transcriptomic analysis indicated that the transcript level of vitellogenin (vtg) in male liver were significantly lower than that in female liver. After 17β-estradiol (E2) treatment of primary cultured hepatocytes, the vtg mRNA expression was induced significantly, while dihydrotestosterone (DHT) treatment had little effect on it. Generally, this study will provide some ideas for further exploring the mechanism of sexual dimorphism in Odontobutis sinensis.
Collapse
Affiliation(s)
- Lingling Zhou
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Ruibin Yang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Hua Tian
- Institute of Hydroecology, Ministry of Water Resources & Chinese Academy of Sciences, Hubei Engineering Research Center of Hydroecology Protection and Restoration, Wuhan 430079, China
| | - Xiangfeng Qin
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Cheng Ye
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Xuetao Shi
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Chuanhui Xia
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Tianyi Cai
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Yunyi Xie
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Yongyi Jia
- Agriculture Ministry Key Laboratory of Healthy Freshwater Aquaculture, Key Laboratory of Fish Health and Nutrition of Zhejiang Province, Zhejiang Institute of Freshwater Fisheries, Huzhou 313001, China.
| | - Guangfu Hu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
39
|
Göethel G, Augsten LV, das Neves GM, Gonçalves IL, de Souza JPS, Garcia SC, Eifler-Lima VL. The role of alternative toxicological trials in drug discovery programs: The case of Caenorhabditis elegans and other methods. Curr Med Chem 2022; 29:5270-5288. [PMID: 35352642 DOI: 10.2174/0929867329666220329190825] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 11/22/2022]
Abstract
The discovery of a new drug requires over a billion dollars and around 12 years of research efforts, and toxicity is the leading reason for failure to approve candidate drugs. Many alternative methods have been validated to detect toxicity as early as possible to diminish the waste of resources and efforts in medicinal chemistry research, and in vivo alternative methods are especially valuable for the amount of information they can give at little cost and in a short time. In this work, we present a review of the literature published between the years 2000 and 2021 of in vivo alternative methods of toxicity screening employed in medicinal chemistry, which we believe will be useful because, in addition to shortening research times, these studies provide much additional information aside from the toxicity of drug candidate compounds. These in vivo models include zebrafish, Artemia salina, Galleria mellonella, Drosophila melanogaster, planarians, and Caenorhabditis elegans as highlights. The most published ones in the last decade were zebrafish, D. melanogaster and C. elegans due to their reliability, ease and cost-effectiveness of implementation and flexibility. Special attention is given to C. elegans because of its rising popularity, a wide range of uses including toxicity screening, and active effects measurement, from antioxidant effects to anthelmintic and antimicrobial activities, and its fast and reliable results. Over time, C. elegans also became a viable high-throughput (HTS) automated drug screening option. Additionally, this manuscript lists briefly the other screening methods used for the initial toxicological analyses and the role of alternative in vivo methods in these scenarios, classifying them as in silico, in vitro and alternative in vivo models, the latter of which have been receiving a growing increase in interest in recent years.
Collapse
Affiliation(s)
- Gabriela Göethel
- Laboratório de Toxicologia (LATOX). Pharmaceutical Sciences Graduate Program, College of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga, 2752, Porto Alegre - RS, Brazil
| | - Lucas Volnei Augsten
- Laboratório de Síntese Orgânica Medicinal (LaSOM), Pharmaceutical Sciences Graduate Program, College of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga, 2752, Porto Alegre - RS, Brazil
| | - Gustavo Machado das Neves
- Laboratório de Síntese Orgânica Medicinal (LaSOM), Pharmaceutical Sciences Graduate Program, College of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga, 2752, Porto Alegre - RS, Brazil
| | - Itamar Luís Gonçalves
- Laboratório de Síntese Orgânica Medicinal (LaSOM), Pharmaceutical Sciences Graduate Program, College of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga, 2752, Porto Alegre - RS, Brazil
| | - João Pedro Silveira de Souza
- Laboratório de Síntese Orgânica Medicinal (LaSOM), Pharmaceutical Sciences Graduate Program, College of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga, 2752, Porto Alegre - RS, Brazil
| | - Solange Cristina Garcia
- Laboratório de Toxicologia (LATOX). Pharmaceutical Sciences Graduate Program, College of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga, 2752, Porto Alegre - RS, Brazil
| | - Vera Lucia Eifler-Lima
- Laboratório de Síntese Orgânica Medicinal (LaSOM), Pharmaceutical Sciences Graduate Program, College of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga, 2752, Porto Alegre - RS, Brazil
| |
Collapse
|
40
|
Miklas JW, Levy S, Hofsteen P, Mex DI, Clark E, Muster J, Robitaille AM, Sivaram G, Abell L, Goodson JM, Pranoto I, Madan A, Chin MT, Tian R, Murry CE, Moon RT, Wang Y, Ruohola-Baker H. Amino acid primed mTOR activity is essential for heart regeneration. iScience 2022; 25:103574. [PMID: 34988408 PMCID: PMC8704488 DOI: 10.1016/j.isci.2021.103574] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 06/17/2021] [Accepted: 12/03/2021] [Indexed: 01/01/2023] Open
Abstract
Heart disease is the leading cause of death with no method to repair damaged myocardium due to the limited proliferative capacity of adult cardiomyocytes. Curiously, mouse neonates and zebrafish can regenerate their hearts via cardiomyocyte de-differentiation and proliferation. However, a molecular mechanism of why these cardiomyocytes can re-enter cell cycle is poorly understood. Here, we identify a unique metabolic state that primes adult zebrafish and neonatal mouse ventricular cardiomyocytes to proliferate. Zebrafish and neonatal mouse hearts display elevated glutamine levels, predisposing them to amino-acid-driven activation of TOR, and that TOR activation is required for zebrafish cardiomyocyte regeneration in vivo. Through a multi-omics approach with cellular validation we identify metabolic and mitochondrial changes during the first week of regeneration. These data suggest that regeneration of zebrafish myocardium is driven by metabolic remodeling and reveals a unique metabolic regulator, TOR-primed state, in which zebrafish and mammalian cardiomyocytes are regeneration competent.
Collapse
Affiliation(s)
- Jason W. Miklas
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Shiri Levy
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA 98109, USA
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA 98195, USA
| | - Peter Hofsteen
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA 98109, USA
- Department of Pathology, University of Washington, Seattle, WA 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
| | - Diego Ic Mex
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA 98109, USA
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA 98195, USA
| | - Elisa Clark
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Jeanot Muster
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Department of Pharmacology, University of Washington, Seattle, WA 98109, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Aaron M. Robitaille
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA 98109, USA
- Department of Pharmacology, University of Washington, Seattle, WA 98109, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Gargi Sivaram
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA 98109, USA
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA 98195, USA
| | - Lauren Abell
- Department of Pathology, University of Washington, Seattle, WA 98109, USA
- Mitochondria and Metabolism Center, University of Washington, Seattle, WA 98109, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98109, USA
| | - Jamie M. Goodson
- Department of Pathology, University of Washington, Seattle, WA 98109, USA
| | - Inez Pranoto
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA 98109, USA
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA 98195, USA
| | - Anup Madan
- Covance Genomics Laboratory, Redmond, WA 98052, USA
| | - Michael T. Chin
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA 98109, USA
- Department of Pathology, University of Washington, Seattle, WA 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA 98109, USA
| | - Rong Tian
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA 98195, USA
- Mitochondria and Metabolism Center, University of Washington, Seattle, WA 98109, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98109, USA
| | - Charles E. Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Department of Pathology, University of Washington, Seattle, WA 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA 98109, USA
| | - Randall T. Moon
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA 98109, USA
- Department of Pharmacology, University of Washington, Seattle, WA 98109, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Yuliang Wang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA 98109, USA
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA 98195, USA
| | - Hannele Ruohola-Baker
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
41
|
Abstract
Heart regeneration is a remarkable process whereby regrowth of damaged cardiac tissue rehabilitates organ anatomy and function. Unfortunately, the human heart is highly resistant to regeneration, which creates a shortage of cardiomyocytes in the wake of ischemic injury, and explains, in part, why coronary artery disease remains a leading cause of death worldwide. Luckily, a detailed blueprint for achieving therapeutic heart regeneration already exists in nature because several lower vertebrate species successfully regenerate amputated or damaged heart muscle through robust cardiomyocyte proliferation. A growing number of species are being interrogated for cardiac regenerative potential, and several commonalities have emerged between those animals showing high or low innate capabilities. In this review, we provide a historical perspective on the field, discuss how regenerative potential is influenced by cardiomyocyte properties, mitogenic signals, and chromatin accessibility, and highlight unanswered questions under active investigation. Ultimately, delineating why heart regeneration occurs preferentially in some organisms, but not in others, will uncover novel therapeutic inroads for achieving cardiac restoration in humans.
Collapse
Affiliation(s)
- Hui-Min Yin
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
| | - C Geoffrey Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Caroline E Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
| |
Collapse
|
42
|
Bongiovanni C, Sacchi F, Da Pra S, Pantano E, Miano C, Morelli MB, D'Uva G. Reawakening the Intrinsic Cardiac Regenerative Potential: Molecular Strategies to Boost Dedifferentiation and Proliferation of Endogenous Cardiomyocytes. Front Cardiovasc Med 2021; 8:750604. [PMID: 34692797 PMCID: PMC8531484 DOI: 10.3389/fcvm.2021.750604] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/13/2021] [Indexed: 12/27/2022] Open
Abstract
Despite considerable efforts carried out to develop stem/progenitor cell-based technologies aiming at replacing and restoring the cardiac tissue following severe damages, thus far no strategies based on adult stem cell transplantation have been demonstrated to efficiently generate new cardiac muscle cells. Intriguingly, dedifferentiation, and proliferation of pre-existing cardiomyocytes and not stem cell differentiation represent the preponderant cellular mechanism by which lower vertebrates spontaneously regenerate the injured heart. Mammals can also regenerate their heart up to the early neonatal period, even in this case by activating the proliferation of endogenous cardiomyocytes. However, the mammalian cardiac regenerative potential is dramatically reduced soon after birth, when most cardiomyocytes exit from the cell cycle, undergo further maturation, and continue to grow in size. Although a slow rate of cardiomyocyte turnover has also been documented in adult mammals, both in mice and humans, this is not enough to sustain a robust regenerative process. Nevertheless, these remarkable findings opened the door to a branch of novel regenerative approaches aiming at reactivating the endogenous cardiac regenerative potential by triggering a partial dedifferentiation process and cell cycle re-entry in endogenous cardiomyocytes. Several adaptations from intrauterine to extrauterine life starting at birth and continuing in the immediate neonatal period concur to the loss of the mammalian cardiac regenerative ability. A wide range of systemic and microenvironmental factors or cell-intrinsic molecular players proved to regulate cardiomyocyte proliferation and their manipulation has been explored as a therapeutic strategy to boost cardiac function after injuries. We here review the scientific knowledge gained thus far in this novel and flourishing field of research, elucidating the key biological and molecular mechanisms whose modulation may represent a viable approach for regenerating the human damaged myocardium.
Collapse
Affiliation(s)
- Chiara Bongiovanni
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Francesca Sacchi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Silvia Da Pra
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Elvira Pantano
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Carmen Miano
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Marco Bruno Morelli
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Gabriele D'Uva
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| |
Collapse
|
43
|
Knockdown of vitamin D receptor genes impairs touch-evoked escape behavior in zebrafish. EXPERIMENTAL RESULTS 2021. [DOI: 10.1017/exp.2021.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Abstract
Vitamin D is a steroid hormone well-known for its role in calcium homeostasis and bone health. Biological actions of vitamin D are mediated through the vitamin D receptor (VDR) present in various cells and tissues. Vitamin D has been implicated in multiple aspects of neuromuscular functions. This study aimed to investigate the role of VDR signaling during early stage of locomotor development utilizing a gene knockdown approach. Zebrafish larvae deficient in VDR showed severe motor impairment and no obvious response to touch. These results indicate that VDR signaling is indispensable for the correct neuromuscular development and touch-evoked escape swimming behavior in zebrafish.
Collapse
|
44
|
Honkoop H, Nguyen PD, van der Velden VEM, Sonnen KF, Bakkers J. Live imaging of adult zebrafish cardiomyocyte proliferation ex vivo. Development 2021; 148:271839. [PMID: 34397091 PMCID: PMC8489017 DOI: 10.1242/dev.199740] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/04/2021] [Indexed: 12/24/2022]
Abstract
Zebrafish are excellent at regenerating their heart by reinitiating proliferation in pre-existing cardiomyocytes. Studying how zebrafish achieve this holds great potential in developing new strategies to boost mammalian heart regeneration. Nevertheless, the lack of appropriate live-imaging tools for the adult zebrafish heart has limited detailed studies into the dynamics underlying cardiomyocyte proliferation. Here, we address this by developing a system in which cardiac slices of the injured zebrafish heart are cultured ex vivo for several days while retaining key regenerative characteristics, including cardiomyocyte proliferation. In addition, we show that the cardiac slice culture system is compatible with live timelapse imaging and allows manipulation of regenerating cardiomyocytes with drugs that normally would have toxic effects that prevent their use. Finally, we use the cardiac slices to demonstrate that adult cardiomyocytes with fully assembled sarcomeres can partially disassemble their sarcomeres in a calpain- and proteasome-dependent manner to progress through nuclear division and cytokinesis. In conclusion, we have developed a cardiac slice culture system, which allows imaging of native cardiomyocyte dynamics in real time to discover cellular mechanisms during heart regeneration.
Collapse
Affiliation(s)
- Hessel Honkoop
- Hubrecht Institute-KNAW and Utrecht University Medical Center, Utrecht 3584CT, The Netherlands
| | - Phong D Nguyen
- Hubrecht Institute-KNAW and Utrecht University Medical Center, Utrecht 3584CT, The Netherlands
| | | | - Katharina F Sonnen
- Hubrecht Institute-KNAW and Utrecht University Medical Center, Utrecht 3584CT, The Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and Utrecht University Medical Center, Utrecht 3584CT, The Netherlands.,Department of Pediatric Cardiology, Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584EA, The Netherlands
| |
Collapse
|
45
|
Abstract
Mammalian cardiomyocytes mostly utilize oxidation of fatty acids to generate ATP. The fetal heart, in stark contrast, mostly uses anaerobic glycolysis. During perinatal development, thyroid hormone drives extensive metabolic remodeling in the heart for adaptation to extrauterine life. These changes coincide with critical functional maturation and exit of the cell cycle, making the heart a post-mitotic organ. Here, we review the current understanding on the perinatal shift in metabolism, hormonal status, and proliferative potential in cardiomyocytes. Thyroid hormone and glucocorticoids have roles in adult cardiac metabolism, and both pathways have been implicated as regulators of myocardial regeneration. We discuss the evidence that suggests these processes could be interrelated and how this can help explain variation in cardiac regeneration across ontogeny and phylogeny, and we note what breakthroughs are still to be made.
Collapse
Affiliation(s)
- Niall Graham
- 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 94143, 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 94143, USA
- Correspondence: Guo N Huang, Ph.D., University of California San Francisco, 555 Mission Bay Blvd South, Room 352V, San Francisco, CA 94158, USA.
| |
Collapse
|
46
|
Chen A, Han Y, Poss KD. Regulation of zebrafish fin regeneration by vitamin D signaling. Dev Dyn 2021; 250:1330-1339. [PMID: 33064344 PMCID: PMC8050121 DOI: 10.1002/dvdy.261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Vitamin D is an essential nutrient that has long been known to regulate skeletal growth and integrity. In models of major appendage regeneration, treatment with vitamin D analogs has been reported to improve aspects of zebrafish fin regeneration in specific disease or gene misexpression contexts, but also to disrupt pattern in regenerating salamander limbs. Recently, we reported strong mitogenic roles for vitamin D signaling in several zebrafish tissues throughout life stages, including epidermal cells and osteoblasts of adult fins. To our knowledge, molecular genetic approaches to dissect vitamin D function in appendage regeneration have not been described. RESULTS Using a knock-in GFP reporter for the expression of the vitamin D target gene and negative regulator cyp24a1, we identified active vitamin D signaling in adult zebrafish fins during tissue homeostasis and regeneration. Transgenic expression of cyp24a1 or a dominant-negative vitamin D receptor (VDR) inhibited regeneration of amputated fins, whereas global vitamin D treatment accelerated regeneration. Using tissue regeneration enhancer elements, we found that local enhancement of VDR expression could improve regeneration with low doses of a vitamin D analog. CONCLUSIONS Vitamin D signaling enhances the efficacy of fin regeneration in zebrafish.
Collapse
Affiliation(s)
- Anzhi Chen
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, 27710, USA
- Regeneration Next, Duke University, Durham, North Carolina, 27710, USA
| | - Yanchao Han
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, 27710, USA
- Regeneration Next, Duke University, Durham, North Carolina, 27710, USA
- Institute for Cardiovascular Science, Medical College, Soochow University, Suzhou, China
| | - Kenneth D. Poss
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, 27710, USA
- Regeneration Next, Duke University, Durham, North Carolina, 27710, USA
| |
Collapse
|
47
|
Paredes A, Santos-Clemente R, Ricote M. Untangling the Cooperative Role of Nuclear Receptors in Cardiovascular Physiology and Disease. Int J Mol Sci 2021; 22:ijms22157775. [PMID: 34360540 PMCID: PMC8346021 DOI: 10.3390/ijms22157775] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
The heart is the first organ to acquire its physiological function during development, enabling it to supply the organism with oxygen and nutrients. Given this early commitment, cardiomyocytes were traditionally considered transcriptionally stable cells fully committed to contractile function. However, growing evidence suggests that the maintenance of cardiac function in health and disease depends on transcriptional and epigenetic regulation. Several studies have revealed that the complex transcriptional alterations underlying cardiovascular disease (CVD) manifestations such as myocardial infarction and hypertrophy is mediated by cardiac retinoid X receptors (RXR) and their partners. RXRs are members of the nuclear receptor (NR) superfamily of ligand-activated transcription factors and drive essential biological processes such as ion handling, mitochondrial biogenesis, and glucose and lipid metabolism. RXRs are thus attractive molecular targets for the development of effective pharmacological strategies for CVD treatment and prevention. In this review, we summarize current knowledge of RXR partnership biology in cardiac homeostasis and disease, providing an up-to-date view of the molecular mechanisms and cellular pathways that sustain cardiomyocyte physiology.
Collapse
|
48
|
Mateus R, Fuhrmann JF, Dye NA. Growth across scales: Dynamic signaling impacts tissue size and shape. Curr Opin Cell Biol 2021; 73:50-57. [PMID: 34182209 DOI: 10.1016/j.ceb.2021.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 05/06/2021] [Indexed: 12/20/2022]
Abstract
Organ and tissue growth result from an integration of biophysical communication across biological scales, both in time and space. In this review, we highlight new insight into the dynamic connections between control mechanisms operating at different length scales. First, we consider how the dynamics of chemical and electrical signaling in the shape of gradients or waves affect spatiotemporal signal interpretation. Then, we discuss the mechanics underlying dynamic cell behavior during oriented tissue growth, followed by the connections between signaling at the tissue and organismal levels.
Collapse
Affiliation(s)
- Rita Mateus
- Max Planck Institute for Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany.
| | - Jana F Fuhrmann
- Max Planck Institute for Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Natalie A Dye
- Max Planck Institute for Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany; Mildred Scheel Nachwuchszentrum (MSNZ) P2, Medical Faculty, Technische Universität Dresden, Germany.
| |
Collapse
|
49
|
Tellez-Garcia AA, Álvarez-Martínez R, López-Martínez JM, Arellano-Carbajal F. Transcriptome analysis during early regeneration of Lumbriculus variegatus. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101050] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
50
|
Tp53 Suppression Promotes Cardiomyocyte Proliferation during Zebrafish Heart Regeneration. Cell Rep 2021; 32:108089. [PMID: 32877671 PMCID: PMC7494019 DOI: 10.1016/j.celrep.2020.108089] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/06/2020] [Accepted: 08/07/2020] [Indexed: 01/04/2023] Open
Abstract
Zebrafish regenerate heart muscle through division of pre-existing cardiomyocytes. To discover underlying regulation, we assess transcriptome datasets for dynamic gene networks during heart regeneration and identify suppression of genes associated with the transcription factor Tp53. Cardiac damage leads to fluctuation of Tp53 protein levels, concomitant with induced expression of its central negative regulator, mdm2, in regenerating cardiomyocytes. Zebrafish lacking functional Tp53 display increased indicators of cardiomyocyte proliferation during regeneration, whereas transgenic Mdm2 blockade inhibits injury-induced cardiomyocyte proliferation. Induced myocardial overexpression of the mitogenic factors Nrg1 or Vegfaa in the absence of injury also upregulates mdm2 and suppresses Tp53 levels, and tp53 mutations augment the mitogenic effects of Nrg1. mdm2 induction is spatiotemporally associated with markers of de-differentiation in injury and growth contexts, suggesting a broad role in cardiogenesis. Our findings reveal myocardial Tp53 suppression by mitogen-induced Mdm2 as a regulatory component of innate cardiac regeneration.
Collapse
|