1
|
Hamar R, Varga M. The zebrafish ( Danio rerio) snoRNAome. NAR Genom Bioinform 2025; 7:lqaf013. [PMID: 40046902 PMCID: PMC11880993 DOI: 10.1093/nargab/lqaf013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 02/08/2025] [Accepted: 02/14/2025] [Indexed: 04/16/2025] Open
Abstract
Small nucleolar RNAs (snoRNAs) are one of the most abundant and evolutionary ancient group of functional non-coding RNAs. They were originally described as guides of post-transcriptional rRNA modifications, but emerging evidence suggests that snoRNAs fulfil an impressive variety of cellular functions. To reveal the true complexity of snoRNA-dependent functions, we need to catalogue first the complete repertoire of snoRNAs in a given cellular context. While the systematic mapping and characterization of "snoRNAomes" for some species have been described recently, this has not been done hitherto for the zebrafish (Danio rerio). Using size-fractionated RNA sequencing data from adult zebrafish tissues, we created an interactive "snoRNAome" database for this species. Our custom-designed analysis pipeline allowed us to identify with high-confidence 67 previously unannotated snoRNAs in the zebrafish genome, resulting in the most complete set of snoRNAs to date in this species. Reanalyzing multiple previously published datasets, we also provide evidence for the dynamic expression of some snoRNAs during the early stages of zebrafish development and tissue-specific expression patterns for others in adults. To facilitate further investigations into the functions of snoRNAs in zebrafish, we created a novel interactive database, snoDanio, which can be used to explore small RNA expression from transcriptomic data.
Collapse
Affiliation(s)
- Renáta Hamar
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, 1117, Hungary
| | - Máté Varga
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, 1117, Hungary
| |
Collapse
|
2
|
Niu X, Melendez DL, Raj S, Cai J, Senadeera D, Mandelbaum J, Shestopalov IA, Martin SD, Zon LI, Schlaeger TM, Lai LP, McMahon AP, Craft AM, Galloway JL. A conserved transcription factor regulatory program promotes tendon fate. Dev Cell 2024; 59:3106-3123.e12. [PMID: 39216481 PMCID: PMC11781300 DOI: 10.1016/j.devcel.2024.08.006] [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: 04/03/2023] [Revised: 01/24/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Tendons, which transmit force from muscles to bones, are highly prone to injury. Understanding the mechanisms driving tendon fate would impact efforts to improve tendon healing, yet this knowledge is limited. To find direct regulators of tendon progenitor emergence, we performed a zebrafish high-throughput chemical screen. We established forskolin as a tenogenic inducer across vertebrates, functioning through Creb1a, which is required and sufficient for tendon fate. Putative enhancers containing cyclic AMP (cAMP) response elements (CREs) in humans, mice, and fish drove specific expression in zebrafish cranial and fin tendons. Analysis of these genomic regions identified motifs for early B cell factor (Ebf/EBF) transcription factors. Mutation of CRE or Ebf/EBF motifs significantly disrupted enhancer activity and specificity in tendons. Zebrafish ebf1a/ebf3a mutants displayed defects in tendon formation. Notably, Creb1a/CREB1 and Ebf1a/Ebf3a/EBF1 overexpression facilitated tenogenic induction in zebrafish and human pluripotent stem cells. Together, our work identifies the functional conservation of two transcription factors in promoting tendon fate.
Collapse
Affiliation(s)
- Xubo Niu
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Delmy L Melendez
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Suyash Raj
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Junming Cai
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dulanjalee Senadeera
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph Mandelbaum
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ilya A Shestopalov
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Scott D Martin
- Department of Sports Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Leonard I Zon
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Thorsten M Schlaeger
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lick Pui Lai
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - April M Craft
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| | - Jenna L Galloway
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
3
|
Xu J, Liu S, Ai Y, Zhang Y, Li S, Li Y. Establishment and transcriptome analysis of single blastomere-derived cell lines from zebrafish. J Genet Genomics 2024; 51:957-969. [PMID: 39097227 DOI: 10.1016/j.jgg.2024.07.018] [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: 04/09/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Maintaining chromosome euploidy in zebrafish embryonic cells is challenging because of the degradation of genomic integrity during cell passaging. In this study, we report the derivation of zebrafish cell lines from single blastomeres. These cell lines have a stable chromosome status attributed to BMP4 and exhibit continuous proliferation in vitro. Twenty zebrafish cell lines are successfully established from single blastomeres. Single-cell transcriptome sequencing analysis confirms the fidelity of gene expression profiles throughout long-term culturing of at least 45 passages. The long-term cultured cells are specialized into epithelial cells, exhibiting similar expression patterns validated by integrative transcriptomic analysis. Overall, this work provides a protocol for establishing zebrafish cell lines from single blastomeres, which can serve as valuable tools for in vitro investigations of epithelial cell dynamics in terms of life-death balance and cell fate determination during normal homeostasis.
Collapse
Affiliation(s)
- Jia Xu
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Siqi Liu
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yirui Ai
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yunbin Zhang
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shifeng Li
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yiping Li
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
4
|
Liu W, Ding Y, Shen Z, Xu C, Yi W, Wang D, Zhou Y, Zon LI, Liu JX. BF170 hydrochloride enhances the emergence of hematopoietic stem and progenitor cells. Development 2024; 151:dev202476. [PMID: 38940293 DOI: 10.1242/dev.202476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 05/14/2024] [Indexed: 06/29/2024]
Abstract
Generation of hematopoietic stem and progenitor cells (HSPCs) ex vivo and in vivo, especially the generation of safe therapeutic HSPCs, still remains inefficient. In this study, we have identified compound BF170 hydrochloride as a previously unreported pro-hematopoiesis molecule, using the differentiation assays of primary zebrafish blastomere cell culture and mouse embryoid bodies (EBs), and we demonstrate that BF170 hydrochloride promoted definitive hematopoiesis in vivo. During zebrafish definitive hematopoiesis, BF170 hydrochloride increases blood flow, expands hemogenic endothelium (HE) cells and promotes HSPC emergence. Mechanistically, the primary cilia-Ca2+-Notch/NO signaling pathway, which is downstream of the blood flow, mediated the effects of BF170 hydrochloride on HSPC induction in vivo. Our findings, for the first time, reveal that BF170 hydrochloride is a compound that enhances HSPC induction and may be applied to the ex vivo expansion of HSPCs.
Collapse
Affiliation(s)
- WenYe Liu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - YuYan Ding
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zheng Shen
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Cong Xu
- Stem Cell Program and Hematology/Oncology, Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - William Yi
- Stem Cell Program and Hematology/Oncology, Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Ding Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yi Zhou
- Stem Cell Program and Hematology/Oncology, Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Leonard I Zon
- Stem Cell Program and Hematology/Oncology, Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute/Children's Hospital, 300 Longwood Avenue, Karp 8, Boston, MA 02115, USA
| | - Jing-Xia Liu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| |
Collapse
|
5
|
Bomkamp C, Musgrove L, Marques DMC, Fernando GF, Ferreira FC, Specht EA. Differentiation and Maturation of Muscle and Fat Cells in Cultivated Seafood: Lessons from Developmental Biology. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2023; 25:1-29. [PMID: 36374393 PMCID: PMC9931865 DOI: 10.1007/s10126-022-10174-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 10/10/2022] [Indexed: 06/16/2023]
Abstract
Cultivated meat, also known as cultured or cell-based meat, is meat produced directly from cultured animal cells rather than from a whole animal. Cultivated meat and seafood have been proposed as a means of mitigating the substantial harms associated with current production methods, including damage to the environment, antibiotic resistance, food security challenges, poor animal welfare, and-in the case of seafood-overfishing and ecological damage associated with fishing and aquaculture. Because biomedical tissue engineering research, from which cultivated meat draws a great deal of inspiration, has thus far been conducted almost exclusively in mammals, cultivated seafood suffers from a lack of established protocols for producing complex tissues in vitro. At the same time, fish such as the zebrafish Danio rerio have been widely used as model organisms in developmental biology. Therefore, many of the mechanisms and signaling pathways involved in the formation of muscle, fat, and other relevant tissue are relatively well understood for this species. The same processes are understood to a lesser degree in aquatic invertebrates. This review discusses the differentiation and maturation of meat-relevant cell types in aquatic species and makes recommendations for future research aimed at recapitulating these processes to produce cultivated fish and shellfish.
Collapse
Affiliation(s)
- Claire Bomkamp
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| | - Lisa Musgrove
- University of the Sunshine Coast, Sippy Downs, Queensland Australia
| | - Diana M. C. Marques
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Gonçalo F. Fernando
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| | - Frederico C. Ferreira
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Elizabeth A. Specht
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| |
Collapse
|
6
|
Isosteviol improves cardiac function and promotes angiogenesis after myocardial infarction in rats. Cell Tissue Res 2021; 387:275-285. [PMID: 34820705 DOI: 10.1007/s00441-021-03559-9] [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: 05/21/2021] [Accepted: 11/12/2021] [Indexed: 10/19/2022]
Abstract
Isosteviol has been indicated as a cardiomyocyte protector. However, the underlying mechanism remains unclear. Thus, we sought to confirm the protective effect of isosteviol after myocardial infarction in a model of permanent coronary artery occlusion and investigate the potential proangiogenic activity in vitro and in vivo. A 4-week permanent coronary artery occlusion rat model was generated, and the protective effect of isosteviol was evaluated by echocardiographic imaging and hemodynamics assays. The coronary capillary density was tested by immunochemistry and micro-computed tomography (μCT) imaging. The effect of isosteviol on endothelial cells was determined in human umbilical vein endothelial cells (HUVECs) in vitro and Tg (kdrl: EGFP) zebrafish in vivo. We also examined the expression of related transcription factors by real-time polymerase chain reaction (RT-qPCR). Isosteviol increased ejection fraction (EF), fractional shortening (FS), cardiac systolic index (CI), maximum rate of increase of left ventricular pressure (Max dp/dt), and left ventricular systolic pressure (LVSP) by 32%, 40%, 25%, 26%, and 10%, respectively, in permanent coronary artery occlusion rats. Interestingly, it also promoted coronary capillary density by 2.5-fold. In addition, isosteviol promoted the proliferation and branching of HUVECs in vitro. It also rescued intersegmental vessel (ISV) development and improved endothelial cell proliferation by approximately fivefold (4-6) in zebrafish embryos in vivo. Isosteviol also upregulated the expression of hypoxia inducible factor-1α (HIF-1α) and vascular endothelial growth factor A (VEGFA) in zebrafish by fourfold and 3.5-fold, respectively. Our findings suggest that isosteviol is a proangiogenic agent and that this activity is related to its protective effects against myocardial ischemia. After using the permanent coronary artery occlusion model, we demonstrated that isosteviol promotes angiogenesis directly and increases capillary density in myocardial ischemia rats. Isosteviol promotes angiogenesis in zebrafish in vivo and increases vascular endothelial cell proliferation in HUVECs and zebrafish. The angiogenesis activity of isosteviol may be correlated with VEGFA and HIF-1α signaling.
Collapse
|
7
|
Tanaka K, Adachi H, Akasaka H, Tamaoki J, Fuse Y, Kobayashi M, Kitazawa T, Teraoka H. Oxidative stress inducers potentiate 2,3,7,8-tetrachlorodibenzo-p-dioxin-mediated pre-cardiac edema in larval zebrafish. J Vet Med Sci 2021; 83:1050-1058. [PMID: 34024870 PMCID: PMC8349820 DOI: 10.1292/jvms.21-0081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We reported the involvement of oxidative stress and prostaglandins including thromboxane and prostacyclin in pre-cardiac edema (early edema) caused by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). While the involvement of oxidative stress in TCDD-induced toxicity has been frequently reported, the mechanism of its action is still unclear. In the present study, oxidative stress inducers including paraquat, hydrogen peroxide (H2O2) and rotenone augmented early edema (edema) induced by a low concentration of TCDD (0.1 ppb) at 55 hr post fertilization (hpf), while each of them alone did not cause edema. Edema caused by TCDD plus oxidative stress inducers was almost abolished by antioxidants, an antagonist for thromboxane receptor (ICI-192,605) and an agonist for prostacyclin receptor (beraprost), suggesting that the site of action of these inducers was in the regular signaling pathway after activation of aryl hydrocarbon receptor type 2 (AHR2) by TCDD. Oxidative stress inducers also enhanced edema caused by an agonist for the thromboxane receptor (U46619), and the enhancement was also inhibited by antioxidants. Sulforaphane and auranofin, activators of Nrf2 that is a master regulator of anti-oxidative response, did not affect U46619-evoked edema but almost abolished TCDD-induced edema and potentiation by paraquat in both TCDD- and U46619-induced edema. Taken together, the results suggest that oxidative stress augments pre-cardiac edema caused by TCDD via activation of thromboxane receptor-mediated signaling in developing zebrafish. As paraquat and other oxidative stress inducers used also are environmental pollutants, interaction between dioxin-like compounds and exogenous source of oxidative stress should also be considered.
Collapse
Affiliation(s)
- Katsuki Tanaka
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Hikaru Adachi
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Hironobu Akasaka
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Junya Tamaoki
- Department of Molecular and Developmental Biology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuji Fuse
- Department of Molecular and Developmental Biology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Makoto Kobayashi
- Department of Molecular and Developmental Biology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Takio Kitazawa
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Hiroki Teraoka
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| |
Collapse
|
8
|
Ecklonia cava Extract and Its Derivative Dieckol Promote Vasodilation by Modulating Calcium Signaling and PI3K/AKT/eNOS Pathway in In Vitro and In Vivo Models. Biomedicines 2021; 9:biomedicines9040438. [PMID: 33921856 PMCID: PMC8073412 DOI: 10.3390/biomedicines9040438] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 12/22/2022] Open
Abstract
Nitric oxide (NO), an endothelial-derived relaxing factor synthesized by endothelial nitric oxide synthase (eNOS) in endothelial cells, enhances vasodilation by modulating vascular tone. The calcium concentration critically influences eNOS activation in endothelial cells. Thus, modulation of calcium-dependent signaling pathways may be a potential therapeutic strategy to enhance vasodilation. Marine algae reportedly possess protective effects against cardiovascular disorders, including hypertension and vascular dysfunction; however, the underlying molecular signaling pathways remain elusive. In the present study, we extracted and isolated dieckol from Ecklonia cava and investigated calcium transit-enhanced vasodilation. Calcium modulation via the well-known M3 muscarinic acetylcholine receptor (AchM3R), which is linked to NO formation, was investigated and the vasodilatory effect of dieckol was verified. Our results indicated that dieckol effectively promoted NO generation via the PI3K/Akt/eNOS axis and calcium transients influenced by AchM3R. We also treated Tg(flk: EGFP) transgenic zebrafish with dieckol to assess its vasodilatory effect. Dieckol promoted vasodilation by enlarging the dorsal aorta diameter, thus regulating blood flow velocity. In conclusion, our findings suggest that dieckol modulates calcium transit through AchM3R, increases endothelial-dependent NO production, and efficiently enhances vasodilation. Thus, E. cava and its derivative, dieckol, can be considered as potential natural vasodilators.
Collapse
|
9
|
Xin N, Jiang Y, Liu S, Zhou Y, Cheng Y. Effects of prednisolone on behavior and hypothalamic-pituitary-interrenal axis activity in zebrafish. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 75:103325. [PMID: 31924570 DOI: 10.1016/j.etap.2020.103325] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 03/17/2019] [Accepted: 01/02/2020] [Indexed: 06/10/2023]
Abstract
Prednisolone is a synthetic glucocorticoid used clinically for treating allergies, inflammation, and autoimmune diseases. Long-term prednisolone use has been shown to have negative effects on physiology and mood. We aimed to study the pharmacology and toxicology of glucocorticoid-like drugs by investigating behavioral and hypothalamic-pituitary-interrenal (HPI) axis effects in a zebrafish model. Zebrafish embryos 24 h post fertilization were exposed to 25 μM prednisolone. Their behavior was investigated 5 days post fertilization (dpf), and their HPI axis-related activity and related neurotransmitter levels were investigated 3, 4, 5, and 6 dpf. The behavior results showed that exposure to prednisolone resulted in decreased autonomic activity and low sensitivity to light. qRT-PCR and ELISA results showed decreased activity of the HPI axis and increased secretion of dopamine and serotonin after exposure to prednisolone. This study provides us with new insights into understanding the effects of glucocorticoids on the HPI axis.
Collapse
Affiliation(s)
- Ning Xin
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, China
| | - Yu Jiang
- Department of Orthopedics, The Affiliated Wuxi No. 2, People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Sha Liu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, China
| | - Yanlong Zhou
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, China
| | - Yanbo Cheng
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, China.
| |
Collapse
|
10
|
Patel BB, Clark KL, Kozik EM, Dash L, Kuhlman JA, Sakaguchi DS. Isolation and culture of primary embryonic zebrafish neural tissue. J Neurosci Methods 2019; 328:108419. [PMID: 31472190 DOI: 10.1016/j.jneumeth.2019.108419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/23/2019] [Accepted: 08/27/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Primary cell culture is a valuable tool to utilize in parallel with in vivo studies in order to maximize our understanding of the mechanisms surrounding neurogenesis and central nervous system (CNS) regeneration and plasticity. The zebrafish is an important model for biomedical research and primary neural cells are readily obtainable from their embryonic stages viatissue dissociation. Further, transgenic reporter lines with cell type-specific expression allows for observation of distinct cell populations within the dissociated tissue. NEW METHOD Here, we define an efficient method for ex vivo quantification and characterization of neuronal and glial tissue dissociated from embryonic zebrafish. RESULTS Zebrafish brain dissociated cells have been documented to survive in culture for at least 9 days in vitro (div). Anti-HuC/D and anti-Acetylated Tubulin antibodies were used to identify neurons in culture; at 3 div approximately 48% of cells were HuC/D positive and 85% expressed serotonin, suggesting our protocol can efficiently isolate neurons from whole embryonic zebrafish brains. Live time-lapse imaging was also carried out to analyze cell migration in vitro. COMPARISON WITH EXISTING METHODS Primary cultures of zebrafish neural cells typically have low rates of survivability in vitro. We have developed a culture system that has long term cell viability, enabling direct analysis of cell-cell and cell-extracellular matrix interactions. CONCLUSIONS These results demonstrate a practical method for isolating, dissociating and culturing of embryonic zebrafish neural tissue. This approach could further be utilized to better understand zebrafish regeneration in vitro.
Collapse
Affiliation(s)
- Bhavika B Patel
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, United States; Neuroscience Program, United States
| | - Kendra L Clark
- Department of Animal Science, Iowa State University, Ames, IA 50011, United States; Genetics and Genomics Program, United States
| | - Emily M Kozik
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, United States
| | - Linkan Dash
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, United States; Genetics and Genomics Program, United States
| | - Julie A Kuhlman
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, United States.
| | - Donald S Sakaguchi
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, United States; Neuroscience Program, United States.
| |
Collapse
|
11
|
Zhong T, Piao L, Kim HJ, Liu X, Jiang S, Liu G. Chlorogenic Acid-Enriched Extract of Ilex kudingcha C.J. Tseng Inhibits Angiogenesis in Zebrafish. J Med Food 2018; 20:1160-1167. [PMID: 29243968 DOI: 10.1089/jmf.2017.3987] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Kudingcha is a particularly bitter tasting tea that has been widely used in China to eliminate fever and itching eyes, and to clear blood toxins. Kudingcha is considered of value for its potential anticancer effects that are attributed to the presence of characteristic bioactive ingredients. The chlorogenic acid (CGA) derivatives 3-0-caffeoylquinic acid, 5-0-caffeoylquinic acid, 3,5-0-dicaffeoylquinic acid, and 4,5-0-dicaffeoylquinic acid were separated from Ilex kudingcha C.J. Tseng extract by high-performance liquid chromatography (HPLC)-photodiode array detector (PDA) and HPLC-nuclear magnetic resonance (NMR). In Tg(flk1:EGFP) zebrafish embryos at 52 hours postfertilization (hpf), angiogenesis was significantly inhibited by kudingcha extract (KDCE) at concentrations of 400 and 500 μg/mL and CGA also showed significant inhibition in embryos treated with 80, 100, and 130 μg/mL. Endothelial cell apoptosis showed a dose-dependent increase in response to KDCE and CGA. CGA derivatives from KDCE could have potential as anticancer agents against tumor angiogenesis.
Collapse
Affiliation(s)
- Tao Zhong
- 1 Department of Veterinary Medicine, Hainan University , 58 Renmin Road, Haikou, China
| | - Linghua Piao
- 2 Department of Physiology, Hainan Medical University , Haikou, China
| | - Hyun Jung Kim
- 3 College of Pharmacy and Natural Medicine Research Institute, Mokpo National University , Muan-gun, Korea
| | - Xiande Liu
- 1 Department of Veterinary Medicine, Hainan University , 58 Renmin Road, Haikou, China
| | - Shengnan Jiang
- 4 Department of Nuclear Medicine, Affiliated HaiKou Hospital, Xiangya School of Medicine, Central South University , Haikou, China
| | - Guomin Liu
- 5 Kudingcha Institute, Hainan University , Haikou, China
| |
Collapse
|
12
|
Hodgson P, Ireland J, Grunow B. Fish, the better model in human heart research? Zebrafish Heart aggregates as a 3D spontaneously cardiomyogenic in vitro model system. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 138:132-141. [PMID: 29729327 DOI: 10.1016/j.pbiomolbio.2018.04.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/04/2018] [Accepted: 04/27/2018] [Indexed: 12/25/2022]
Abstract
The zebrafish (ZF) has become an essential model for biomedical, pharmacological and eco-toxicological heart research. Despite the anatomical differences between fish and human hearts, similarities in cellular structure and conservation of genes as well as pathways across vertebrates have led to an increase in the popularity of ZF as a model for human cardiac research. ZF research benefits from an entirely sequenced genome, which allows us to establish and study cardiovascular mutants to better understand cardiovascular diseases. In this review, we will discuss the importance of in vitro model systems for cardiac research and summarise results of in vitro 3D heart-like cell aggregates, consisting of myocardial tissue formed spontaneously from enzymatically digested whole embryonic ZF larvae (Zebrafish Heart Aggregate - ZFHA). We will give an overview of the similarities and differences of ZF versus human hearts and highlight why ZF complement established mammalian models (i.e. murine and large animal models) for cardiac research. At this stage, the ZFHA model system is being refined into a high-throughput (more ZFHA generated than larvae prepared) and stable in vitro test system to accomplish the same longevity of previously successful salmonid models. ZFHA have potential for the use of high-throughput-screenings of different factors like small molecules, nucleic acids, proteins and lipids which is difficult to achieve in the zebrafish in vivo screening models with lethal mutations as well as to explore ion channel disorders and to find appropriate drugs for safety screening.
Collapse
Affiliation(s)
- Patricia Hodgson
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, UK; Salford Royal NHS Foundation Trust, Stott Lane, Salford M6 8HD, UK
| | - Jake Ireland
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, UK; School of Chemistry, Materials Science, and Engineering, Hilmer Building, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Bianka Grunow
- University Medicine Greifswald, Institute of Physiology, Greifswalder Str. 11C, 17495 Karlsburg, Germany; Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, UK.
| |
Collapse
|
13
|
Sassen WA, Lehne F, Russo G, Wargenau S, Dübel S, Köster RW. Embryonic zebrafish primary cell culture for transfection and live cellular and subcellular imaging. Dev Biol 2017; 430:18-31. [DOI: 10.1016/j.ydbio.2017.07.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 07/24/2017] [Accepted: 07/24/2017] [Indexed: 10/19/2022]
|
14
|
Chua JS, Tran VM, Kalita M, Quintero MV, Antelope O, Muruganandam G, Saijoh Y, Kuberan B. A glycan-based approach to therapeutic angiogenesis. PLoS One 2017; 12:e0182301. [PMID: 28763512 PMCID: PMC5538652 DOI: 10.1371/journal.pone.0182301] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 07/11/2017] [Indexed: 01/23/2023] Open
Abstract
Angiogenesis, the sprouting of new blood vessels from existing vasculature, involves multiple complex biological processes, and it is an essential step for hemostasis, tissue healing and regeneration. Angiogenesis stimulants can ameliorate human disease conditions including limb ischemia, chronic wounds, heart disease, and stroke. The current strategies to improve the bioavailability of pro-angiogenic growth factors, including VEGF and FGF2, have remained largely unsuccessful. This study demonstrates that small molecules, termed click-xylosides, can promote angiogenesis in the in vitro matrigel tube formation assay and the ex ovo chick chorioallantoic membrane assay, depending on their aglycone moieties. Xyloside treatment enhances network connectivity and cell survivability, thereby, maintaining the network structures on matrigel culture for an extended period of time. These effects were achieved via the secreted xyloside-primed glycosaminoglycans (GAG) chains that in part, act through an ERK1/2 mediated signaling pathway. Through the remodeling of GAGs in the extracellular matrix of endothelial cells, the glycan approach, involving xylosides, offers great potential to effectively promote therapeutic angiogenesis.
Collapse
Affiliation(s)
- Jie Shi Chua
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, United States of America
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Vy M. Tran
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, United States of America
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Mausam Kalita
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Maritza V. Quintero
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Orlando Antelope
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Geethu Muruganandam
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, United States of America
| | - Yukio Saijoh
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, United States of America
| | - Balagurunathan Kuberan
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, United States of America
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah, United States of America
- Department of Biology, University of Utah, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
15
|
Ibrahim M, Richardson MK. Beyond organoids: In vitro vasculogenesis and angiogenesis using cells from mammals and zebrafish. Reprod Toxicol 2017; 73:292-311. [PMID: 28697965 DOI: 10.1016/j.reprotox.2017.07.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/12/2017] [Accepted: 07/05/2017] [Indexed: 12/24/2022]
Abstract
The ability to culture complex organs is currently an important goal in biomedical research. It is possible to grow organoids (3D organ-like structures) in vitro; however, a major limitation of organoids, and other 3D culture systems, is the lack of a vascular network. Protocols developed for establishing in vitro vascular networks typically use human or rodent cells. A major technical challenge is the culture of functional (perfused) networks. In this rapidly advancing field, some microfluidic devices are now getting close to the goal of an artificially perfused vascular network. Another development is the emergence of the zebrafish as a complementary model to mammals. In this review, we discuss the culture of endothelial cells and vascular networks from mammalian cells, and examine the prospects for using zebrafish cells for this objective. We also look into the future and consider how vascular networks in vitro might be successfully perfused using microfluidic technology.
Collapse
Affiliation(s)
- Muhammad Ibrahim
- Animal Science and Health Cluster, Institute of Biology Leiden, Leiden University, The Netherlands; Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, Pakistan
| | - Michael K Richardson
- Animal Science and Health Cluster, Institute of Biology Leiden, Leiden University, The Netherlands.
| |
Collapse
|
16
|
van Rooijen E, Fazio M, Zon LI. From fish bowl to bedside: The power of zebrafish to unravel melanoma pathogenesis and discover new therapeutics. Pigment Cell Melanoma Res 2017; 30:402-412. [PMID: 28379616 PMCID: PMC6038924 DOI: 10.1111/pcmr.12592] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/22/2017] [Indexed: 12/28/2022]
Abstract
Melanoma is the most aggressive and deadliest form of skin cancer. A detailed knowledge of the cellular, molecular, and genetic events underlying melanoma progression is highly relevant to diagnosis, prognosis and risk stratification, and the development of new therapies. In the last decade, zebrafish have emerged as a valuable model system for the study of melanoma. Pathway conservation, coupled with the availability of robust genetic, transgenic, and chemical tools, has made the zebrafish a powerful model for identifying novel disease genes, visualizing cancer initiation, interrogating tumor-microenvironment interactions, and discovering new therapeutics that regulate melanocyte and melanoma development. In this review, we will give an overview of these studies, and highlight recent advancements that will help unravel melanoma pathogenesis and impact human disease.
Collapse
Affiliation(s)
- Ellen van Rooijen
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Maurizio Fazio
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
- PhD program in Biological and Biomedical Sciences, Harvard University, Boston, MA, USA
| | - Leonard I. Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
In vitro development of zebrafish vascular networks. Reprod Toxicol 2017; 70:102-115. [DOI: 10.1016/j.reprotox.2017.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/27/2017] [Accepted: 02/08/2017] [Indexed: 12/28/2022]
|
18
|
Directed Differentiation of Zebrafish Pluripotent Embryonic Cells to Functional Cardiomyocytes. Stem Cell Reports 2016; 7:370-382. [PMID: 27569061 PMCID: PMC5032289 DOI: 10.1016/j.stemcr.2016.07.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 07/26/2016] [Accepted: 07/27/2016] [Indexed: 12/24/2022] Open
Abstract
A cardiomyocyte differentiation in vitro system from zebrafish embryos remains to be established. Here, we have determined pluripotency window of zebrafish embryos by analyzing their gene-expression patterns of pluripotency factors together with markers of three germ layers, and have found that zebrafish undergoes a very narrow period of pluripotency maintenance from zygotic genome activation to a brief moment after oblong stage. Based on the pluripotency and a combination of appropriate conditions, we established a rapid and efficient method for cardiomyocyte generation in vitro from primary embryonic cells. The induced cardiomyocytes differentiated into functional and specific cardiomyocyte subtypes. Notably, these in vitro generated cardiomyocytes exhibited typical contractile kinetics and electrophysiological features. The system provides a new paradigm of cardiomyocyte differentiation from primary embryonic cells in zebrafish. The technology provides a new platform for the study of heart development and regeneration, in addition to drug discovery, disease modeling, and assessment of cardiotoxic agents. Zebrafish embryos may start to exit from pluripotency shortly after the oblong stage Beating cell clusters are efficiently generated from zebrafish blastomeres Beating cell clusters contain specific cardiomyocyte subtypes Induced cardiomyocytes possess normal electrophysiological features
Collapse
|
19
|
Zhao H, Osborne OJ, Lin S, Ji Z, Damoiseux R, Wang Y, Nel AE, Lin S. Lanthanide Hydroxide Nanoparticles Induce Angiogenesis via ROS-Sensitive Signaling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:4404-11. [PMID: 27383397 PMCID: PMC5240819 DOI: 10.1002/smll.201600291] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/11/2016] [Indexed: 05/26/2023]
Abstract
Recent studies suggest that the nanorods consisting of europium hydroxide could promote angiogenesis. In this study, it is sought to determine if additional types of nanoparticles are capable of enhancing angiogenesis and in addition, understand the underlying mechanisms. For this reason, a method is employed that combines a high throughput in vitro cell based screen coupled with an in vivo validation using vascular specific green fluorescent protein reporter transgenic zebrafish for examining proangiogenesis activity. After screening multiple types of nanoparticles, it is discovered that four of them, Eu(III) (OH)3 rods (Eu rods), Eu(III) (OH)3 spheres (Eu spheres), Tb(III) (OH)3 rods (Tb rods), and Tb(III) (OH)3 spheres (Tb spheres), are the most effective in promoting angiogenesis. It is also showed that ionic forms of europium nitrate [Eu(NO3 )3 ] (Eu) and terbium nitrate [Tb(NO3 )3 ] (Tb), the two lanthanide elements for these four nanoparticles, are also capable of enhancing angiogenesis. However, this effect is further enhanced by nanoparticle synthesis. Finally, it is demonstrated that reactive oxygen species H2 O2 is a key factor in the process of proangiogenesis by lanthanide elemental nanoparticles.
Collapse
Affiliation(s)
- Haishan Zhao
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510640, China
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Olivia J Osborne
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Sijie Lin
- College of Environmental Science and Engineering, Tongji University, Shanghai, China
| | - Zhaoxia Ji
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Robert Damoiseux
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Yuqiang Wang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510640, China
| | - André E Nel
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Shuo Lin
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
- College of Environmental Science and Engineering, Tongji University, Shanghai, China
| |
Collapse
|
20
|
Mattiazzi Usaj M, Styles EB, Verster AJ, Friesen H, Boone C, Andrews BJ. High-Content Screening for Quantitative Cell Biology. Trends Cell Biol 2016; 26:598-611. [PMID: 27118708 DOI: 10.1016/j.tcb.2016.03.008] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 12/25/2022]
Abstract
High-content screening (HCS), which combines automated fluorescence microscopy with quantitative image analysis, allows the acquisition of unbiased multiparametric data at the single cell level. This approach has been used to address diverse biological questions and identify a plethora of quantitative phenotypes of varying complexity in numerous different model systems. Here, we describe some recent applications of HCS, ranging from the identification of genes required for specific biological processes to the characterization of genetic interactions. We review the steps involved in the design of useful biological assays and automated image analysis, and describe major challenges associated with each. Additionally, we highlight emerging technologies and future challenges, and discuss how the field of HCS might be enhanced in the future.
Collapse
Affiliation(s)
| | - Erin B Styles
- The Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada
| | - Adrian J Verster
- The Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada
| | - Helena Friesen
- The Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada
| | - Charles Boone
- The Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada
| | - Brenda J Andrews
- The Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada.
| |
Collapse
|
21
|
Zhao H, Huang H, Lin S. Chemical approaches to angiogenesis in development and regeneration. Methods Cell Biol 2016; 134:369-76. [PMID: 27312498 DOI: 10.1016/bs.mcb.2016.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Vascular endothelial cells are essential building blocks of angiogenesis, which is required for normal embryonic development and tissue regeneration. In this chapter, we describe how to use transgenic zebrafish embryos expressing vascular-specific green fluorescent protein to evaluate differentiation, growth, and morphogenesis of endothelial cells. When combined with instrument automation and computational analysis, this method allows high-throughput screening for biologically active small chemical molecules that are effective in promoting angiogenesis. These molecules can be validated in mammalian endothelial cell differentiation and proliferation assays. These studies provide new reagents and therapeutic candidates for regenerative medicine studies.
Collapse
Affiliation(s)
- H Zhao
- University of California Los Angeles, Los Angeles, CA, United States; Jinan University, Guangzhou, China
| | - H Huang
- University of California Los Angeles, Los Angeles, CA, United States
| | - S Lin
- University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
22
|
Abstract
Zebrafish embryonic cell cultures have many useful properties that make them complementary to intact embryos for a wide range of studies. Embryonic cell cultures allow for maintenance of transient cell populations, control of chemical and mechanical cues received by cells, and facile chemical screening. Zebrafish cells can be cultured in either heterogeneous or homogeneous cultures from a wide range of developmental time points. Here we describe two methods with particular applicability to chemical screening: a method for the culture of blastomeres for directed differentiation toward the myogenic lineage and a method for the culture of neural crest cells in heterogeneous cultures from early somitogenesis embryos.
Collapse
Affiliation(s)
- C A Ciarlo
- Harvard Medical School and Children's Hospital, Boston, MA, United States
| | - L I Zon
- Children's Hospital and Dana Farber Cancer Institute, Boston, MA, United States; Harvard University, Cambridge, MA, United States
| |
Collapse
|
23
|
Webb AB, Lengyel IM, Jörg DJ, Valentin G, Jülicher F, Morelli LG, Oates AC. Persistence, period and precision of autonomous cellular oscillators from the zebrafish segmentation clock. eLife 2016; 5. [PMID: 26880542 PMCID: PMC4803185 DOI: 10.7554/elife.08438] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 02/11/2016] [Indexed: 12/11/2022] Open
Abstract
In vertebrate development, the sequential and rhythmic segmentation of the body axis
is regulated by a “segmentation clock”. This clock is comprised of a population of
coordinated oscillating cells that together produce rhythmic gene expression patterns
in the embryo. Whether individual cells autonomously maintain oscillations, or
whether oscillations depend on signals from neighboring cells is unknown. Using a
transgenic zebrafish reporter line for the cyclic transcription factor Her1, we
recorded single tailbud cells in vitro. We demonstrate that individual cells can
behave as autonomous cellular oscillators. We described the observed variability in
cell behavior using a theory of generic oscillators with correlated noise. Single
cells have longer periods and lower precision than the tissue, highlighting the role
of collective processes in the segmentation clock. Our work reveals a population of
cells from the zebrafish segmentation clock that behave as self-sustained, autonomous
oscillators with distinctive noisy dynamics. DOI:http://dx.doi.org/10.7554/eLife.08438.001 The timing and pattern of gene activity in cells can be very important. For example,
precise gene activity patterns in 24-hour circadian clocks help to set daily cycles
of rest and activity in organisms. In such scenarios, cells often communicate with
each other to coordinate the activity of their genes. To fully understand how the
behavior of the population emerges, scientists must first understand the gene
activity patterns in individual cells. Rhythmic gene activity is essential for the spinal column to form in fish and other
vertebrate embryos. A group of cells that switch genes on/off in a coordinated
pattern act like a clock to regulate the timing of the various steps in the process
of backbone formation. However, it is not clear if each cell is able to maintain a
rhythm of gene expression on their own, or whether they rely on messages from
neighboring cells to achieve it. Now, Webb et al. use time-lapse videos of individual cells isolated from the tail of
zebrafish embryos to show that each cell can maintain a pattern of rhythmic activity
in a gene called Her1. In the experiments, individual cells were
removed from zebrafish and placed under a microscope to record and track the activity
of Her1 over time using fluorescent proteins. These experiments show
that each cell is able to maintain a rhythmic pattern of Her1
expression on its own. Webb et al. then compared the Her1 activity patterns in individual
cells with the Her1 patterns present in a larger piece of zebrafish
tissue. The experiments showed that the rhythms in the individual cells are slower
and less precise in their timing than in the tissue. This suggests that groups of
cells must work together to create the synchronized rhythms of gene expression with
the right precision and timing needed for the spinal column to be patterned
correctly. In the future, further experiment with these cells will allow researchers to
investigate the genetic basis of the rhythms in single cells, and find out how
individual cells work together with their neighbors to allow tissues to work
properly. DOI:http://dx.doi.org/10.7554/eLife.08438.002
Collapse
Affiliation(s)
- Alexis B Webb
- MRC-National Institute for Medical Research, London, United Kingdom.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Iván M Lengyel
- Departamento de Física, FCEyN UBA and IFIBA, CONICET, Buenos Aires, Argentina
| | - David J Jörg
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
| | - Guillaume Valentin
- MRC-National Institute for Medical Research, London, United Kingdom.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Frank Jülicher
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
| | - Luis G Morelli
- Departamento de Física, FCEyN UBA and IFIBA, CONICET, Buenos Aires, Argentina
| | - Andrew C Oates
- MRC-National Institute for Medical Research, London, United Kingdom.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Department of Cell and Developmental Biology, University College London, London, United Kingdom
| |
Collapse
|
24
|
Hu Z, Brooks SA, Dormoy V, Hsu CW, Hsu HY, Lin LT, Massfelder T, Rathmell WK, Xia M, Al-Mulla F, Al-Temaimi R, Amedei A, Brown DG, Prudhomme KR, Colacci A, Hamid RA, Mondello C, Raju J, Ryan EP, Woodrick J, Scovassi AI, Singh N, Vaccari M, Roy R, Forte S, Memeo L, Salem HK, Lowe L, Jensen L, Bisson WH, Kleinstreuer N. Assessing the carcinogenic potential of low-dose exposures to chemical mixtures in the environment: focus on the cancer hallmark of tumor angiogenesis. Carcinogenesis 2015; 36 Suppl 1:S184-202. [PMID: 26106137 PMCID: PMC4492067 DOI: 10.1093/carcin/bgv036] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 12/12/2014] [Accepted: 12/15/2014] [Indexed: 01/09/2023] Open
Abstract
One of the important 'hallmarks' of cancer is angiogenesis, which is the process of formation of new blood vessels that are necessary for tumor expansion, invasion and metastasis. Under normal physiological conditions, angiogenesis is well balanced and controlled by endogenous proangiogenic factors and antiangiogenic factors. However, factors produced by cancer cells, cancer stem cells and other cell types in the tumor stroma can disrupt the balance so that the tumor microenvironment favors tumor angiogenesis. These factors include vascular endothelial growth factor, endothelial tissue factor and other membrane bound receptors that mediate multiple intracellular signaling pathways that contribute to tumor angiogenesis. Though environmental exposures to certain chemicals have been found to initiate and promote tumor development, the role of these exposures (particularly to low doses of multiple substances), is largely unknown in relation to tumor angiogenesis. This review summarizes the evidence of the role of environmental chemical bioactivity and exposure in tumor angiogenesis and carcinogenesis. We identify a number of ubiquitous (prototypical) chemicals with disruptive potential that may warrant further investigation given their selectivity for high-throughput screening assay targets associated with proangiogenic pathways. We also consider the cross-hallmark relationships of a number of important angiogenic pathway targets with other cancer hallmarks and we make recommendations for future research. Understanding of the role of low-dose exposure of chemicals with disruptive potential could help us refine our approach to cancer risk assessment, and may ultimately aid in preventing cancer by reducing or eliminating exposures to synergistic mixtures of chemicals with carcinogenic potential.
Collapse
Affiliation(s)
- Zhiwei Hu
- To whom correspondence should be addressed. Tel: +1 614 685 4606; Fax: +1-614-247-7205;
| | - Samira A. Brooks
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Valérian Dormoy
- INSERM U1113, team 3 “Cell Signalling and Communication in Kidney and Prostate Cancer”, University of Strasbourg, Facultée de Médecine, 67085 Strasbourg, France
- Department of Cell and Developmental Biology, University of California, Irvine, CA 92697, USA
| | - Chia-Wen Hsu
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Hsue-Yin Hsu
- Department of Life Sciences, Tzu-Chi University, Taiwan, Republic of China
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, Taipei Medical University, Taiwan, Republic of China
| | - Thierry Massfelder
- INSERM U1113, team 3 “Cell Signalling and Communication in Kidney and Prostate Cancer”, University of Strasbourg, Facultée de Médecine, 67085 Strasbourg, France
| | - W. Kimryn Rathmell
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Fahd Al-Mulla
- Department of Life Sciences, Tzu-Chi University, Taiwan, Republic of China
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Florence 50134, Italy
| | - Dustin G. Brown
- Department of Environmental and Radiological Health Sciences
, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523, USA
| | - Kalan R. Prudhomme
- Environmental and Molecular Toxicology, Environmental Health Science Center, Oregon State University, Corvallis, OR 97331, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna, Italy
| | - Roslida A. Hamid
- Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor, Malaysia
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Jayadev Raju
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Food Directorate
, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Elizabeth P. Ryan
- Department of Environmental and Radiological Health Sciences
, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523, USA
| | - Jordan Woodrick
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, WashingtonDC 20057, USA
| | - A. Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advance Research), King George’s Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna, Italy
| | - Rabindra Roy
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, WashingtonDC 20057, USA
| | - Stefano Forte
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Hosni K. Salem
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia B2N 1X5, Canada
| | - Lasse Jensen
- Division of Cardiovascular Medicine, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden and
| | - William H. Bisson
- Environmental and Molecular Toxicology, Environmental Health Science Center, Oregon State University, Corvallis, OR 97331, USA
| | - Nicole Kleinstreuer
- Integrated Laboratory Systems, Inc., in support of the National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, NIEHS, MD K2-16, RTP, NC 27709, USA
| |
Collapse
|
25
|
Kong P, Zhang BS, Lei P, Kong XD, Zhang SS, Li D, Zhang Y. Neurotoxicity of cerebro-spinal fluid from patients with Parkinson's disease on mesencephalic primary cultures as an in vitro model of dopaminergic neurons. Mol Med Rep 2015; 12:2217-24. [PMID: 25845313 DOI: 10.3892/mmr.2015.3575] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 01/29/2015] [Indexed: 11/06/2022] Open
Abstract
Parkinson's disease is a degenerative disorder of the central nervous system. In spite of extensive research, neither the cause nor the mechanisms have been firmly established thus far. One assumption is that certain toxic substances may exist in the cerebro-spinal fluid (CSF) of Parkinson's disease patients. To confirm the neurotoxicity of CSF and study the potential correlation between neurotoxicity and the severity of Parkinson's disease, CSF was added to cultured cells. By observation of cell morphology, changes in the levels of lactate dehydrogenase, the ratio of tyrosine hydroxylase-positive cells, and the expression of tyrosine hydroxylase mRNA and protein, the differences between the two groups were shown. The created in vitro model of dopaminergic neurons using primary culture of mouse embryonic mesencephalic tissue is suitable for the study of neurotoxicity. The observations of the present study indicated that CSF from Parkinson's disease patients contains factors that can cause specific injury to cultured dopaminergic neurons. However, no obvious correlation was found between the neurotoxicity of CSF and the severity of Parkinson's disease.
Collapse
Affiliation(s)
- Ping Kong
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300192, P.R. China
| | - Ben-Shu Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin 300192, P.R. China
| | - Ping Lei
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300192, P.R. China
| | - Xiao-Dong Kong
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300192, P.R. China
| | - Shi-Shuang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300192, P.R. China
| | - Dai Li
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300192, P.R. China
| | - Yun Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300192, P.R. China
| |
Collapse
|
26
|
Santoro MM. Zebrafish as a model to explore cell metabolism. Trends Endocrinol Metab 2014; 25:546-54. [PMID: 24997878 DOI: 10.1016/j.tem.2014.06.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 06/04/2014] [Accepted: 06/10/2014] [Indexed: 12/20/2022]
Abstract
Cell metabolism plays a key role in many essential biological processes. The recent availability of novel technologies and organisms to model cell metabolism in vivo is expanding current knowledge of cell metabolism. In this context, the zebrafish (Danio rerio) is emerging as a valuable model system to learn about the metabolic routes critical for cellular homeostasis. Here, the most recent methods and studies on cell metabolism are summarized, which support the overall value for the zebrafish model system not only to study metabolism but also metabolic disease states. It is envisioned that this small vertebrate system will help in the understanding of pathogenesis for numerous metabolic-related disorders in humans and in the identification of their therapeutic treatments.
Collapse
Affiliation(s)
- Massimo M Santoro
- Laboratory of Endothelial Molecular Biology, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven, B-3000, Belgium; Laboratory of Endothelial Molecular Biology, Vesalius Research Center, VIB, Leuven, B-3000, Belgium.
| |
Collapse
|
27
|
Yasgar A, Simeonov A. Current approaches for the discovery of drugs that deter substance and drug abuse. Expert Opin Drug Discov 2014; 9:1319-31. [PMID: 25251069 DOI: 10.1517/17460441.2014.956721] [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: 11/05/2022]
Abstract
INTRODUCTION Much has been presented and debated on the topic of drug abuse and its multidimensional nature, including the role of society and its customs and laws, economical factors, and the magnitude and nature of the burden. Given the complex nature of the receptors and pathways implicated in regulation of the cognitive and behavioral processes associated with addiction, a large number of molecular targets have been interrogated during recent years to discover starting points for development of small-molecule interventions. AREAS COVERED This review describes recent developments in the field of early drug discovery for drug abuse interventions with an emphasis on the advances published during the 2012 - 2014 period. EXPERT OPINION Technologically, the processes/platforms utilized in drug abuse drug discovery are nearly identical to those used in the other disease areas. A key complicating factor in drug abuse research is the enormous biological complexity surrounding the brain processes involved and the associated difficulty in finding 'good' targets and achieving exquisite selectivity of treatment agents. While tremendous progress has been made during recent years to use the power of high-throughput technologies to discover proof-of-principle molecules for many new targets, next-generation models will be especially important in this field. Examples include: seeking advantageous drug-drug combinations, the use of automated whole-animal behavioral screening systems, advancing our understanding of the role of epigenetics in drug addiction and the employment of organoid-level 3D test platforms (also referred to as tissue-chip or organs-on-chip).
Collapse
Affiliation(s)
- Adam Yasgar
- National Institutes of Health, NIH Chemical Genomics Center, National Center for Advancing Translational Sciences , Bethesda, MD , USA +1 301 217 5721 ; +1 301 217 5736 ;
| | | |
Collapse
|
28
|
Affiliation(s)
- Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
29
|
Xu C, Tabebordbar M, Iovino S, Ciarlo C, Liu J, Castiglioni A, Price E, Liu M, Barton ER, Kahn CR, Wagers AJ, Zon LI. A zebrafish embryo culture system defines factors that promote vertebrate myogenesis across species. Cell 2013; 155:909-921. [PMID: 24209627 PMCID: PMC3902670 DOI: 10.1016/j.cell.2013.10.023] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 08/29/2013] [Accepted: 10/11/2013] [Indexed: 01/05/2023]
Abstract
Ex vivo expansion of satellite cells and directed differentiation of pluripotent cells to mature skeletal muscle have proved difficult challenges for regenerative biology. Using a zebrafish embryo culture system with reporters of early and late skeletal muscle differentiation, we examined the influence of 2,400 chemicals on myogenesis and identified six that expanded muscle progenitors, including three GSK3β inhibitors, two calpain inhibitors, and one adenylyl cyclase activator, forskolin. Forskolin also enhanced proliferation of mouse satellite cells in culture and maintained their ability to engraft muscle in vivo. A combination of bFGF, forskolin, and the GSK3β inhibitor BIO induced skeletal muscle differentiation in human induced pluripotent stem cells (iPSCs) and produced engraftable myogenic progenitors that contributed to muscle repair in vivo. In summary, these studies reveal functionally conserved pathways regulating myogenesis across species and identify chemical compounds that expand mouse satellite cells and differentiate human iPSCs into engraftable muscle.
Collapse
Affiliation(s)
- Cong Xu
- Division of Hematology/Oncology, Children’s Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Mohammadsharif Tabebordbar
- Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Salvatore Iovino
- Harvard Medical School, Boston, MA 02115, USA
- Joslin Diabetes Center, Boston, MA 02115, USA
| | - Christie Ciarlo
- Division of Hematology/Oncology, Children’s Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Jingxia Liu
- Division of Hematology/Oncology, Children’s Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Alessandra Castiglioni
- Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA 02115, USA
- Joslin Diabetes Center, Boston, MA 02115, USA
- Vita-Salute San Raffaele University, Milan, 20132, Italy
| | - Emily Price
- Division of Hematology/Oncology, Children’s Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Min Liu
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elisabeth R. Barton
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - C. Ronald Kahn
- Harvard Medical School, Boston, MA 02115, USA
- Joslin Diabetes Center, Boston, MA 02115, USA
| | - Amy J. Wagers
- Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA 02115, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Joslin Diabetes Center, Boston, MA 02115, USA
| | - Leonard I. Zon
- Division of Hematology/Oncology, Children’s Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|