1
|
Mastrogiovanni M, Martínez-Navarro FJ, Bowman TV, Cayuela ML. Inflammation in Development and Aging: Insights from the Zebrafish Model. Int J Mol Sci 2024; 25:2145. [PMID: 38396822 PMCID: PMC10889087 DOI: 10.3390/ijms25042145] [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: 01/02/2024] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Zebrafish are an emergent animal model to study human diseases due to their significant genetic similarity to humans, swift development, and genetic manipulability. Their utility extends to the exploration of the involvement of inflammation in host defense, immune responses, and tissue regeneration. Additionally, the zebrafish model system facilitates prompt screening of chemical compounds that affect inflammation. This study explored the diverse roles of inflammatory pathways in zebrafish development and aging. Serving as a crucial model, zebrafish provides insights into the intricate interplay of inflammation in both developmental and aging contexts. The evidence presented suggests that the same inflammatory signaling pathways often play instructive or beneficial roles during embryogenesis and are associated with malignancies in adults.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Francisco Juan Martínez-Navarro
- Grupo de Telomerasa, Cáncer y Envejecimiento, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca, 30120 Murcia, Spain
| | - Teresa V. Bowman
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - María L. Cayuela
- Grupo de Telomerasa, Cáncer y Envejecimiento, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 30100 Murcia, Spain
| |
Collapse
|
2
|
Xia J, Liu M, Zhu C, Liu S, Ai L, Ma D, Zhu P, Wang L, Liu F. Activation of lineage competence in hemogenic endothelium precedes the formation of hematopoietic stem cell heterogeneity. Cell Res 2023; 33:448-463. [PMID: 37016019 PMCID: PMC10235423 DOI: 10.1038/s41422-023-00797-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/01/2023] [Indexed: 04/06/2023] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) are considered as a heterogeneous population, but precisely when, where and how HSPC heterogeneity arises remain largely unclear. Here, using a combination of single-cell multi-omics, lineage tracing and functional assays, we show that embryonic HSPCs originate from heterogeneous hemogenic endothelial cells (HECs) during zebrafish embryogenesis. Integrated single-cell transcriptome and chromatin accessibility analysis demonstrates transcriptional heterogeneity and regulatory programs that prime lymphoid/myeloid fates at the HEC level. Importantly, spi2+ HECs give rise to lymphoid/myeloid-primed HSPCs (L/M-HSPCs) and display a stress-responsive function under acute inflammation. Moreover, we uncover that Spi2 is required for the formation of L/M-HSPCs through tightly controlling the endothelial-to-hematopoietic transition program. Finally, single-cell transcriptional comparison of zebrafish and human HECs and human induced pluripotent stem cell-based hematopoietic differentiation results support the evolutionary conservation of L/M-HECs and a conserved role of SPI1 (spi2 homolog in mammals) in humans. These results unveil the lineage origin, biological function and molecular determinant of HSPC heterogeneity and lay the foundation for new strategies for induction of transplantable lineage-primed HSPCs in vitro.
Collapse
Affiliation(s)
- Jun Xia
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mengyao Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Caiying Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Shicheng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lanlan Ai
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Dongyuan Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ping Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Lu Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.
| | - Feng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Life Sciences, Shandong University, Qingdao, Shandong, China.
| |
Collapse
|
3
|
Binder V, Li W, Faisal M, Oyman K, Calkins DL, Shaffer J, Teets EM, Sher S, Magnotte A, Belardo A, Deruelle W, Gregory TC, Orwick S, Hagedorn EJ, Perlin JR, Avagyan S, Lichtig A, Barrett F, Ammerman M, Yang S, Zhou Y, Carson WE, Shive HR, Blachly JS, Lapalombella R, Zon LI, Blaser BW. Microenvironmental control of hematopoietic stem cell fate via CXCL8 and protein kinase C. Cell Rep 2023; 42:112528. [PMID: 37209097 PMCID: PMC10824047 DOI: 10.1016/j.celrep.2023.112528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 03/19/2023] [Accepted: 05/02/2023] [Indexed: 05/22/2023] Open
Abstract
Altered hematopoietic stem cell (HSC) fate underlies primary blood disorders but microenvironmental factors controlling this are poorly understood. Genetically barcoded genome editing of synthetic target arrays for lineage tracing (GESTALT) zebrafish were used to screen for factors expressed by the sinusoidal vascular niche that alter the phylogenetic distribution of the HSC pool under native conditions. Dysregulated expression of protein kinase C delta (PKC-δ, encoded by prkcda) increases the number of HSC clones by up to 80% and expands polyclonal populations of immature neutrophil and erythroid precursors. PKC agonists such as cxcl8 augment HSC competition for residency within the niche and expand defined niche populations. CXCL8 induces association of PKC-δ with the focal adhesion complex, activating extracellular signal-regulated kinase (ERK) signaling and expression of niche factors in human endothelial cells. Our findings demonstrate the existence of reserve capacity within the niche that is controlled by CXCL8 and PKC and has significant impact on HSC phylogenetic and phenotypic fate.
Collapse
Affiliation(s)
- Vera Binder
- Dr. von Hauner Childrens' Hospital, University Hospital Ludwig Maximillian's University, Department of Pediatric Hematology/Oncology, 80337 Munich, Germany
| | - Wantong Li
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | - Muhammad Faisal
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | - Konur Oyman
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | - Donn L Calkins
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | - Jami Shaffer
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | - Emily M Teets
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | - Steven Sher
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | - Andrew Magnotte
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | - Alex Belardo
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | - William Deruelle
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | - T Charles Gregory
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA; The Ohio State University College of Medicine, Department of Biomedical Informatics, Columbus, OH 43210, USA
| | - Shelley Orwick
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | - Elliott J Hagedorn
- Boston University School of Medicine, Department of Medicine, Boston, MA 02118, USA
| | - Julie R Perlin
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
| | - Serine Avagyan
- Dana-Farber/Boston Children's Hospital Cancer and Blood Disorders Center, Boston, MA 02115, USA
| | - Asher Lichtig
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
| | - Francesca Barrett
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
| | - Michelle Ammerman
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
| | - Song Yang
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
| | - Yi Zhou
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
| | - William E Carson
- The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | - Heather R Shive
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - James S Blachly
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA; The Ohio State University College of Medicine, Department of Biomedical Informatics, Columbus, OH 43210, USA
| | - Rosa Lapalombella
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | - Leonard I Zon
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA; Dana-Farber/Boston Children's Hospital Cancer and Blood Disorders Center, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA
| | - Bradley W Blaser
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA.
| |
Collapse
|
4
|
Li H, Luo Q, Cai S, Tie R, Meng Y, Shan W, Xu Y, Zeng X, Qian P, Huang H. Glia maturation factor-γ is required for initiation and maintenance of hematopoietic stem and progenitor cells. Stem Cell Res Ther 2023; 14:117. [PMID: 37122014 PMCID: PMC10150485 DOI: 10.1186/s13287-023-03328-1] [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: 04/24/2022] [Accepted: 04/05/2023] [Indexed: 05/02/2023] Open
Abstract
BACKGROUND In vertebrates, hematopoietic stem and progenitor cells (HSPCs) emerge from hemogenic endothelium in the floor of the dorsal aorta and subsequently migrate to secondary niches where they expand and differentiate into committed lineages. Glia maturation factor γ (gmfg) is a key regulator of actin dynamics that was shown to be highly expressed in hematopoietic tissue. Our goal is to investigate the role and mechanism of gmfg in embryonic HSPC development. METHODS In-depth bioinformatics analysis of our published RNA-seq data identified gmfg as a cogent candidate gene implicated in HSPC development. Loss and gain-of-function strategies were applied to study the biological function of gmfg. Whole-mount in situ hybridization, confocal microscopy, flow cytometry, and western blotting were used to evaluate changes in the number of various hematopoietic cells and expression levels of cell proliferation, cell apoptosis and hematopoietic-related markers. RNA-seq was performed to screen signaling pathways responsible for gmfg deficiency-induced defects in HSPC initiation. The effect of gmfg on YAP sublocalization was assessed in vitro by utilizing HUVEC cell line. RESULTS We took advantage of zebrafish embryos to illustrate that loss of gmfg impaired HSPC initiation and maintenance. In gmfg-deficient embryos, the number of hemogenic endothelium and HSPCs was significantly reduced, with the accompanying decreased number of erythrocytes, myelocytes and lymphocytes. We found that blood flow modulates gmfg expression and gmfg overexpression could partially rescue the reduction of HSPCs in the absence of blood flow. Assays in zebrafish and HUVEC showed that gmfg deficiency suppressed the activity of YAP, a well-established blood flow mediator, by preventing its shuttling from cytoplasm to nucleus. During HSPC initiation, loss of gmfg resulted in Notch inactivation and the induction of Notch intracellular domain could partially restore the HSPC loss in gmfg-deficient embryos. CONCLUSIONS We conclude that gmfg mediates blood flow-induced HSPC maintenance via regulation of YAP, and contributes to HSPC initiation through the modulation of Notch signaling. Our findings reveal a brand-new aspect of gmfg function and highlight a novel mechanism for embryonic HSPC development.
Collapse
Affiliation(s)
- Honghu Li
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Qian Luo
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Shuyang Cai
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Ruxiu Tie
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Ye Meng
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Wei Shan
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Yulin Xu
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Xiangjun Zeng
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Pengxu Qian
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China.
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China.
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, 310012, People's Republic of China.
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China.
- School of Medicine, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, People's Republic of China.
| | - He Huang
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China.
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China.
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, 310012, People's Republic of China.
| |
Collapse
|
5
|
Nakajima H, Ishikawa H, Yamamoto T, Chiba A, Fukui H, Sako K, Fukumoto M, Mattonet K, Kwon HB, Hui SP, Dobreva GD, Kikuchi K, Helker CSM, Stainier DYR, Mochizuki N. Endoderm-derived islet1-expressing cells differentiate into endothelial cells to function as the vascular HSPC niche in zebrafish. Dev Cell 2023; 58:224-238.e7. [PMID: 36693371 DOI: 10.1016/j.devcel.2022.12.013] [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: 11/20/2021] [Revised: 10/26/2022] [Accepted: 12/29/2022] [Indexed: 01/25/2023]
Abstract
Endothelial cells (ECs) line blood vessels and serve as a niche for hematopoietic stem and progenitor cells (HSPCs). Recent data point to tissue-specific EC specialization as well as heterogeneity; however, it remains unclear how ECs acquire these properties. Here, by combining live-imaging-based lineage-tracing and single-cell transcriptomics in zebrafish embryos, we identify an unexpected origin for part of the vascular HSPC niche. We find that islet1 (isl1)-expressing cells are the progenitors of the venous ECs that constitute the majority of the HSPC niche. These isl1-expressing cells surprisingly originate from the endoderm and differentiate into ECs in a process dependent on Bmp-Smad signaling and subsequently requiring npas4l (cloche) function. Single-cell RNA sequencing analyses show that isl1-derived ECs express a set of genes that reflect their distinct origin. This study demonstrates that endothelial specialization in the HSPC niche is determined at least in part by the origin of the ECs.
Collapse
Affiliation(s)
- Hiroyuki Nakajima
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan.
| | - Hiroyuki Ishikawa
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; AMED-CREST, AMED, Tokyo 100-0004, Japan; Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto 606-8507, Japan
| | - Ayano Chiba
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Hajime Fukui
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Keisuke Sako
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Moe Fukumoto
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Kenny Mattonet
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Hyouk-Bum Kwon
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Subhra P Hui
- S. N. Pradhan Centre for Neurosciences, University of Calcutta, Kolkata 700019, India
| | - Gergana D Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Kazu Kikuchi
- Department of Cardiac Regeneration Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Christian S M Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany; Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, Marburg 35043, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany.
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan.
| |
Collapse
|
6
|
Dudziak K, Nowak M, Sozoniuk M. One Host-Multiple Applications: Zebrafish ( Danio rerio) as Promising Model for Studying Human Cancers and Pathogenic Diseases. Int J Mol Sci 2022; 23:10255. [PMID: 36142160 PMCID: PMC9499349 DOI: 10.3390/ijms231810255] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/03/2022] [Accepted: 09/03/2022] [Indexed: 11/17/2022] Open
Abstract
In recent years, zebrafish (ZF) has been increasingly applied as a model in human disease studies, with a particular focus on cancer. A number of advantages make it an attractive alternative for mice widely used so far. Due to the many advantages of zebrafish, modifications can be based on different mechanisms and the induction of human disease can take different forms depending on the research goal. Genetic manipulation, tumor transplantation, or injection of the pathogen are only a few examples of using ZF as a model. Most of the studies are conducted in order to understand the disease mechanism, monitor disease progression, test new or alternative therapies, and select the best treatment. The transplantation of cancer cells derived from patients enables the development of personalized medicine. To better mimic a patient's body environment, immune-deficient models (SCID) have been developed. A lower immune response is mostly generated by genetic manipulation but also by irradiation or dexamethasone treatment. For many studies, using SCID provides a better chance to avoid cancer cell rejection. In this review, we describe the main directions of using ZF in research, explain why and how zebrafish can be used as a model, what kind of limitations will be met and how to overcome them. We collected recent achievements in this field, indicating promising perspectives for the future.
Collapse
Affiliation(s)
- Karolina Dudziak
- Chair and Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Michał Nowak
- Institute of Plant Genetics, Breeding and Biotechnology, University of Life Sciences in Lublin, 20-950 Lublin, Poland
| | - Magdalena Sozoniuk
- Institute of Plant Genetics, Breeding and Biotechnology, University of Life Sciences in Lublin, 20-950 Lublin, Poland
| |
Collapse
|
7
|
Dai Y, Wu S, Cao C, Xue R, Luo X, Wen Z, Xu J. Csf1rb regulates definitive hematopoiesis in zebrafish. Development 2022; 149:276084. [DOI: 10.1242/dev.200534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 07/07/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
In vertebrates, hematopoietic stem and progenitor cells (HSPCs) are capable of self-renewal and continuously replenishing all mature blood lineages throughout life. However, the molecular signaling regulating the maintenance and expansion of HSPCs remains incompletely understood. Colony-stimulating factor 1 receptor (CSF1R) is believed to be the primary regulator for the myeloid lineage but not HSPC development. Here, we show a surprising role of Csf1rb, a zebrafish homolog of mammalian CSF1R, in preserving the HSPC pool by maintaining the proliferation of HSPCs. Deficiency of csf1rb leads to a reduction in both HSPCs and their differentiated progenies, including myeloid, lymphoid and erythroid cells at early developmental stages. Likewise, the absence of csf1rb conferred similar defects upon HSPCs and leukocytes in adulthood. Furthermore, adult hematopoietic cells from csf1rb mutants failed to repopulate immunodeficient zebrafish. Interestingly, loss-of-function and gain-of-function assays suggested that the canonical ligands for Csf1r in zebrafish, including Csf1a, Csf1b and Il34, were unlikely to be ligands of Csf1rb. Thus, our data indicate a previously unappreciated role of Csf1r in maintaining HSPCs, independently of known ligands.
Collapse
Affiliation(s)
- Yimei Dai
- School of Medicine, South China University of Technology 1 Laboratory of Immunology & Regeneration , , Guangzhou 510006, China
| | - Shuting Wu
- State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, the Hong Kong University of Science and Technology 2 Division of Life Science , , Clear Water Bay, Kowloon, Hong Kong , People's Republic of China
| | - Canran Cao
- School of Medicine, South China University of Technology 1 Laboratory of Immunology & Regeneration , , Guangzhou 510006, China
| | - Rongtao Xue
- Nanfang Hospital, Southern Medical University 3 Department of Hematology , , Guangzhou, Guangdong 510515 , China
| | - Xuefen Luo
- State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, the Hong Kong University of Science and Technology 2 Division of Life Science , , Clear Water Bay, Kowloon, Hong Kong , People's Republic of China
| | - Zilong Wen
- State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, the Hong Kong University of Science and Technology 2 Division of Life Science , , Clear Water Bay, Kowloon, Hong Kong , People's Republic of China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen Peking University−Hong Kong University of Science and Technology Medical Center 4 , Shenzhen 518055 , China
| | - Jin Xu
- School of Medicine, South China University of Technology 1 Laboratory of Immunology & Regeneration , , Guangzhou 510006, China
| |
Collapse
|
8
|
Ando K, Ishii T, Fukuhara S. Zebrafish Vascular Mural Cell Biology: Recent Advances, Development, and Functions. Life (Basel) 2021; 11:1041. [PMID: 34685412 PMCID: PMC8537713 DOI: 10.3390/life11101041] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/29/2021] [Accepted: 09/29/2021] [Indexed: 12/14/2022] Open
Abstract
Recruitment of mural cells to the vascular wall is essential for forming the vasculature as well as maintaining proper vascular functions. In recent years, zebrafish genetic tools for mural cell biology have improved substantially. Fluorescently labeled zebrafish mural cell reporter lines enable us to study, with higher spatiotemporal resolution than ever, the processes of mural cell development from their progenitors. Furthermore, recent phenotypic analysis of platelet-derived growth factor beta mutant zebrafish revealed well-conserved organotypic mural cell development and functions in vertebrates with the unique features of zebrafish. However, comprehensive reviews of zebrafish mural cells are lacking. Therefore, herein, we highlight recent advances in zebrafish mural cell tools. We also summarize the fundamental features of zebrafish mural cell development, especially at early stages, and functions.
Collapse
Affiliation(s)
- Koji Ando
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School, Tokyo 113 8602, Japan; (T.I.); (S.F.)
| | | | | |
Collapse
|
9
|
Fañanas-Baquero S, Orman I, Becerra Aparicio F, Bermudez de Miguel S, Garcia Merino J, Yañez R, Fernandez Sainz Y, Sánchez R, Dessy-Rodríguez M, Alberquilla O, Alfaro D, Zapata A, Bueren JA, Segovia JC, Quintana-Bustamante O. Natural estrogens enhance the engraftment of human hematopoietic stem and progenitor cells in immunodeficient mice. Haematologica 2021; 106:1659-1670. [PMID: 32354868 PMCID: PMC8168497 DOI: 10.3324/haematol.2019.233924] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Indexed: 12/31/2022] Open
Abstract
Hematopoietic Stem and Progenitor Cells are crucial in the maintenance of lifelong production of all blood cells. These Stem Cells are highly regulated to maintain homeostasis through a delicate balance between quiescence, self-renewal and differentiation. However, this balance is altered during the hematopoietic recovery after Hematopoietic Stem and Progenitor Cell Transplantation. Transplantation efficacy can be limited by inadequate Hematopoietic Stem Cells number, poor homing, low level of engraftment, or limited self-renewal. As recent evidences indicate that estrogens are involved in regulating the hematopoiesis, we sought to examine whether natural estrogens (estrone or E1, estradiol or E2, estriol or E3 and estetrol or E4) modulate human Hematopoietic Stem and Progenitor Cells. Our results show that human Hematopoietic Stem and Progenitor Cell subsets express estrogen receptors, and whose signaling is activated by E2 and E4 on these cells. Additionally, these natural estrogens cause different effects on human Progenitors in vitro. We found that both E2 and E4 expand human Hematopoietic Stem and Progenitor Cells. However, E4 was the best tolerated estrogen and promoted cell cycle of human Hematopoietic Progenitors. Furthermore, we identified that E2 and, more significantly, E4 doubled human hematopoietic engraftment in immunodeficient mice without altering other Hematopoietic Stem and Progenitor Cells properties. Finally, the impact of E4 on promoting human hematopoietic engraftment in immunodeficient mice might be mediated through the regulation of mesenchymal stromal cells in the bone marrow niche. Together, our data demonstrate that E4 is well tolerated and enhances human reconstitution in immunodeficient mice, directly by modulating human Hematopoietic Progenitor properties and indirectly by interacting with the bone marrow niche. This application might have particular relevance to ameliorate the hematopoietic recovery after myeloablative conditioning, especially when limiting numbers of Hematopoietic Stem and Progenitor Cells are available.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - David Alfaro
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | - Agustin Zapata
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | | | | | | |
Collapse
|
10
|
Krishnan M, Kumar S, Kangale LJ, Ghigo E, Abnave P. The Act of Controlling Adult Stem Cell Dynamics: Insights from Animal Models. Biomolecules 2021; 11:biom11050667. [PMID: 33946143 PMCID: PMC8144950 DOI: 10.3390/biom11050667] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/02/2021] [Accepted: 04/09/2021] [Indexed: 12/12/2022] Open
Abstract
Adult stem cells (ASCs) are the undifferentiated cells that possess self-renewal and differentiation abilities. They are present in all major organ systems of the body and are uniquely reserved there during development for tissue maintenance during homeostasis, injury, and infection. They do so by promptly modulating the dynamics of proliferation, differentiation, survival, and migration. Any imbalance in these processes may result in regeneration failure or developing cancer. Hence, the dynamics of these various behaviors of ASCs need to always be precisely controlled. Several genetic and epigenetic factors have been demonstrated to be involved in tightly regulating the proliferation, differentiation, and self-renewal of ASCs. Understanding these mechanisms is of great importance, given the role of stem cells in regenerative medicine. Investigations on various animal models have played a significant part in enriching our knowledge and giving In Vivo in-sight into such ASCs regulatory mechanisms. In this review, we have discussed the recent In Vivo studies demonstrating the role of various genetic factors in regulating dynamics of different ASCs viz. intestinal stem cells (ISCs), neural stem cells (NSCs), hematopoietic stem cells (HSCs), and epidermal stem cells (Ep-SCs).
Collapse
Affiliation(s)
- Meera Krishnan
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Gurgaon-Faridabad Ex-pressway, Faridabad 121001, India; (M.K.); (S.K.)
| | - Sahil Kumar
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Gurgaon-Faridabad Ex-pressway, Faridabad 121001, India; (M.K.); (S.K.)
| | - Luis Johnson Kangale
- IRD, AP-HM, SSA, VITROME, Aix-Marseille University, 13385 Marseille, France;
- Institut Hospitalo Universitaire Méditerranée Infection, 13385 Marseille, France;
| | - Eric Ghigo
- Institut Hospitalo Universitaire Méditerranée Infection, 13385 Marseille, France;
- TechnoJouvence, 13385 Marseille, France
| | - Prasad Abnave
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Gurgaon-Faridabad Ex-pressway, Faridabad 121001, India; (M.K.); (S.K.)
- Correspondence:
| |
Collapse
|
11
|
A single-cell resolution developmental atlas of hematopoietic stem and progenitor cell expansion in zebrafish. Proc Natl Acad Sci U S A 2021; 118:2015748118. [PMID: 33785593 PMCID: PMC8040670 DOI: 10.1073/pnas.2015748118] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The caudal hematopoietic tissue (CHT) is characterized as a hematopoietic organ for fetal hematopoietic stem and progenitor cell (HSPC) expansion in zebrafish. In this study, we used scRNA-seq combined with functional assays to decode the developing CHT. First, we resolved fetal HSPC heterogeneity, manifested as lineage priming and metabolic gene signatures. We further analyzed the cellular interactions among nonhematopoietic niche components and HSPCs and identified an endothelial cell-specific factor, Gpr182, followed by experimental validation of its role in promoting HSPC expansion. Finally, we uncovered the conservation and divergence of developmental hematopoiesis between human fetal liver and zebrafish CHT. Our study provides a valuable resource for fetal HSPC development and clues to establish a supportive niche for HSPC expansion in vitro. During vertebrate embryogenesis, fetal hematopoietic stem and progenitor cells (HSPCs) exhibit expansion and differentiation properties in a supportive hematopoietic niche. To profile the developmental landscape of fetal HSPCs and their local niche, here, using single-cell RNA-sequencing, we deciphered a dynamic atlas covering 28,777 cells and 9 major cell types (23 clusters) of zebrafish caudal hematopoietic tissue (CHT). We characterized four heterogeneous HSPCs with distinct lineage priming and metabolic gene signatures. Furthermore, we investigated the regulatory mechanism of CHT niche components for HSPC development, with a focus on the transcription factors and ligand–receptor networks involved in HSPC expansion. Importantly, we identified an endothelial cell-specific G protein–coupled receptor 182, followed by in vivo and in vitro functional validation of its evolutionally conserved role in supporting HSPC expansion in zebrafish and mice. Finally, comparison between zebrafish CHT and human fetal liver highlighted the conservation and divergence across evolution. These findings enhance our understanding of the regulatory mechanism underlying hematopoietic niche for HSPC expansion in vivo and provide insights into improving protocols for HSPC expansion in vitro.
Collapse
|
12
|
Kasper DM, Hintzen J, Wu Y, Ghersi JJ, Mandl HK, Salinas KE, Armero W, He Z, Sheng Y, Xie Y, Heindel DW, Joo Park E, Sessa WC, Mahal LK, Lebrilla C, Hirschi KK, Nicoli S. The N-glycome regulates the endothelial-to-hematopoietic transition. Science 2020; 370:1186-1191. [PMID: 33273096 PMCID: PMC8312266 DOI: 10.1126/science.aaz2121] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 05/20/2020] [Accepted: 10/27/2020] [Indexed: 12/31/2022]
Abstract
Definitive hematopoietic stem and progenitor cells (HSPCs) arise from the transdifferentiation of hemogenic endothelial cells (hemECs). The mechanisms of this endothelial-to-hematopoietic transition (EHT) are poorly understood. We show that microRNA-223 (miR-223)-mediated regulation of N-glycan biosynthesis in endothelial cells (ECs) regulates EHT. miR-223 is enriched in hemECs and in oligopotent nascent HSPCs. miR-223 restricts the EHT of lymphoid-myeloid lineages by suppressing the mannosyltransferase alg2 and sialyltransferase st3gal2, two enzymes involved in protein N-glycosylation. ECs that lack miR-223 showed a decrease of high mannose versus sialylated sugars on N-glycoproteins such as the metalloprotease Adam10. EC-specific expression of an N-glycan Adam10 mutant or of the N-glycoenzymes phenocopied miR-223 mutant defects. Thus, the N-glycome is an intrinsic regulator of EHT, serving as a key determinant of the hematopoietic fate.
Collapse
Affiliation(s)
- Dionna M. Kasper
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA.,Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jared Hintzen
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA.,Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yinyu Wu
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA.,Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Joey J. Ghersi
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA.,Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Hanna K. Mandl
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA.,Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Kevin E. Salinas
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA.,Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
| | - William Armero
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA.,Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Zhiheng He
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA.,Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ying Sheng
- Department of Chemistry, University of California, Davis, CA 95616, USA
| | - Yixuan Xie
- Department of Chemistry, University of California, Davis, CA 95616, USA
| | - Daniel W. Heindel
- Biomedical Chemistry Institute, Department of Chemistry, New York University, New York, NY 10003, USA
| | - Eon Joo Park
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - William C. Sessa
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Lara K. Mahal
- Biomedical Chemistry Institute, Department of Chemistry, New York University, New York, NY 10003, USA.,Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Carlito Lebrilla
- Department of Chemistry, University of California, Davis, CA 95616, USA
| | - Karen K. Hirschi
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA.,Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.,Developmental Genomics Center, Cell Biology Department, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Stefania Nicoli
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA.,Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
13
|
Heck AM, Ishida T, Hadland B. Location, Location, Location: How Vascular Specialization Influences Hematopoietic Fates During Development. Front Cell Dev Biol 2020; 8:602617. [PMID: 33282876 PMCID: PMC7691428 DOI: 10.3389/fcell.2020.602617] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/30/2020] [Indexed: 01/22/2023] Open
Abstract
During embryonic development, sequential waves of hematopoiesis give rise to blood-forming cells with diverse lineage potentials and self-renewal properties. This process must accomplish two important yet divergent goals: the rapid generation of differentiated blood cells to meet the needs of the developing embryo and the production of a reservoir of hematopoietic stem cells to provide for life-long hematopoiesis in the adult. Vascular beds in distinct anatomical sites of extraembryonic tissues and the embryo proper provide the necessary conditions to support these divergent objectives, suggesting a critical role for specialized vascular niche cells in regulating disparate blood cell fates during development. In this review, we will examine the current understanding of how organ- and stage-specific vascular niche specialization contributes to the development of the hematopoietic system.
Collapse
Affiliation(s)
- Adam M. Heck
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Takashi Ishida
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Brandon Hadland
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
14
|
Breus O, Dickmeis T. Genetically encoded thiol redox-sensors in the zebrafish model: lessons for embryonic development and regeneration. Biol Chem 2020; 402:363-378. [PMID: 33021959 DOI: 10.1515/hsz-2020-0269] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022]
Abstract
Important roles for reactive oxygen species (ROS) and redox signaling in embryonic development and regenerative processes are increasingly recognized. However, it is difficult to obtain information on spatiotemporal dynamics of ROS production and signaling in vivo. The zebrafish is an excellent model for in vivo bioimaging and possesses a remarkable regenerative capacity upon tissue injury. Here, we review data obtained in this model system with genetically encoded redox-sensors targeting H2O2 and glutathione redox potential. We describe how such observations have prompted insight into regulation and downstream effects of redox alterations during tissue differentiation, morphogenesis and regeneration. We also discuss the properties of the different sensors and their consequences for the interpretation of in vivo imaging results. Finally, we highlight open questions and additional research fields that may benefit from further application of such sensor systems in zebrafish models of development, regeneration and disease.
Collapse
Affiliation(s)
- Oksana Breus
- Institute of Biological and Chemical Systems - Biological Information Processing, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, D-76344Eggenstein-Leopoldshafen, Germany
| | - Thomas Dickmeis
- Institute of Biological and Chemical Systems - Biological Information Processing, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, D-76344Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
15
|
Ong SLM, de Vos IJHM, Meroshini M, Poobalan Y, Dunn NR. Microfibril-associated glycoprotein 4 (Mfap4) regulates haematopoiesis in zebrafish. Sci Rep 2020; 10:11801. [PMID: 32678226 PMCID: PMC7366704 DOI: 10.1038/s41598-020-68792-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 06/19/2020] [Indexed: 12/13/2022] Open
Abstract
Microfibril-associated glycoprotein 4 (MFAP4) is an extracellular matrix protein belonging to the fibrinogen-related protein superfamily. MFAP4 is produced by vascular smooth muscle cells and is highly enriched in the blood vessels of the heart and lung, where it is thought to contribute to the structure and function of elastic fibers. Genetic studies in humans have implicated MFAP4 in the pathogenesis of Smith-Magenis syndrome, in which patients present with multiple congenital abnormalities and mental retardation, as well as in the severe cardiac malformation left-sided congenital heart disease. Comprehensive genetic analysis of the role of MFAP4 orthologues in model organisms during development and tissue homeostasis is however lacking. Here, we demonstrate that zebrafish mfap4 transcripts are detected embryonically, resolving to the macrophage lineage by 24 h post fertilization. mfap4 null mutant zebrafish are unexpectedly viable and fertile, without ostensible phenotypes. However, tail fin amputation assays reveal that mfap4 mutants have reduced numbers of macrophages, with a concomitant increase in neutrophilic granulocytes, although recruitment of both cell types to the site of injury was unaffected. Molecular analyses suggest that loss of Mfap4 alters the balance between myeloid and lymphoid lineages during both primitive and definitive haematopoiesis, which could significantly impact the downstream function of the immune system.
Collapse
Affiliation(s)
- Sheena L M Ong
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore, 138648, Singapore.,Department of Pathology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Ivo J H M de Vos
- Skin Research Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 11 Mandalay Road, Clinical Sciences Building, #17-01, Singapore, 308232, Singapore.,Department of Genetics, University Medical Center Groningen, 9700 RB, Groningen, The Netherlands
| | - M Meroshini
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, 11 Mandalay Road, Clinical Sciences Building, Singapore, 308232, Singapore
| | - Yogavalli Poobalan
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore, 138648, Singapore.,Engine Biosciences, 160 Robinson Road, 23-20 SBF Center, Singapore, 068914, Singapore
| | - N Ray Dunn
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore, 138648, Singapore. .,Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, 11 Mandalay Road, Clinical Sciences Building, Singapore, 308232, Singapore.
| |
Collapse
|
16
|
Oh CK, Kang JW, Lee Y, Myung K, Ha M, Kang J, Kwon EJ, Kim Y, Oh SO, Heo HJ, Kim S, Kim YH. Role of kif2c, A Gene Related to ALL Relapse, in Embryonic Hematopoiesis in Zebrafish. Int J Mol Sci 2020; 21:ijms21093127. [PMID: 32354205 PMCID: PMC7246619 DOI: 10.3390/ijms21093127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/17/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
Relapse of acute lymphoblastic leukemia (ALL) is dangerous and it worsens the prognosis of patients; however, prognostic markers or therapeutic targets for ALL remain unknown. In the present study, using databases such as TARGET, GSE60926 and GSE28460, we determined that KIF2C and its binding partner, KIF18B are overexpressed in patients with relapsed ALL compared to that in patients diagnosed with ALL for the first time. As 50% of the residues are exactly the same and the signature domain of KIF2C is highly conserved between human and zebrafish, we used zebrafish embryos as a model to investigate the function of kif2c in vivo. We determined that kif2c is necessary for lymphopoiesis in zebrafish embryos. Additionally, we observed that kif2c is not related to differentiation of HSCs; however, it is important for the maintenance of HSCs as it provides survival signals to HSCs. These results imply that the ALL relapse-related gene KIF2C is linked to the survival of HSCs. In conclusion, we suggest that KIF2C can serve as a novel therapeutic target for relapsed ALL.
Collapse
Affiliation(s)
- Chang-Kyu Oh
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan 44919, Korea; (C.-K.O.); (Y.L.); (K.M.)
| | - Ji Wan Kang
- Interdisciplinary Program of Genomic Science, Pusan National University, Yangsan 50612, Korea; (J.W.K.); (M.H.); (J.K.); (E.J.K.); (Y.K.)
| | - Yoonsung Lee
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan 44919, Korea; (C.-K.O.); (Y.L.); (K.M.)
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan 44919, Korea; (C.-K.O.); (Y.L.); (K.M.)
| | - Mihyang Ha
- Interdisciplinary Program of Genomic Science, Pusan National University, Yangsan 50612, Korea; (J.W.K.); (M.H.); (J.K.); (E.J.K.); (Y.K.)
| | - Junho Kang
- Interdisciplinary Program of Genomic Science, Pusan National University, Yangsan 50612, Korea; (J.W.K.); (M.H.); (J.K.); (E.J.K.); (Y.K.)
| | - Eun Jung Kwon
- Interdisciplinary Program of Genomic Science, Pusan National University, Yangsan 50612, Korea; (J.W.K.); (M.H.); (J.K.); (E.J.K.); (Y.K.)
| | - Youngjoo Kim
- Interdisciplinary Program of Genomic Science, Pusan National University, Yangsan 50612, Korea; (J.W.K.); (M.H.); (J.K.); (E.J.K.); (Y.K.)
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Korea; (S.-O.O.); (H.J.H.)
| | - Hye Jin Heo
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Korea; (S.-O.O.); (H.J.H.)
| | - Shin Kim
- Department of Immunology, School of Medicine, Keimyung University, Dalseo-gu, Daegu 42601, Korea
- Institute of Medical Science, Keimyung University, Dalseo-gu, Daegu 42601, Korea
- Correspondence: (S.K.); (Y.H.K.); Tel.: +82-53-258-7359 (S.K.); +82-51-510-8091 (Y.H.K.); Fax: +82-53-258-7355 (S.K.); +82-51-510-8049 (Y.H.K.)
| | - Yun Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Korea; (S.-O.O.); (H.J.H.)
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan 50612, Korea
- Correspondence: (S.K.); (Y.H.K.); Tel.: +82-53-258-7359 (S.K.); +82-51-510-8091 (Y.H.K.); Fax: +82-53-258-7355 (S.K.); +82-51-510-8049 (Y.H.K.)
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW The availability of organs for transplant fails to meet the demand and this shortage is growing worse every year. As the cost of not getting a suitable donor organ can mean death for patients, new tools and approaches that allows us to make advances in transplantation faster and provide a different vantage point are required. To address this need, we introduce the concept of using the zebrafish (Danio rerio) as a new model system in organ transplantation. The zebrafish community offers decades of research experience in disease modeling and a rich toolbox of approaches for interrogating complex pathological states. We provide examples of how already existing zebrafish assays/tools from cancer, regenerative medicine, immunology, and others, could be leveraged to fuel new discoveries in pursuit of solving the organ shortage. RECENT FINDINGS Important innovations have enabled several types of transplants to be successfully performed in zebrafish, including stem cells, tumors, parenchymal cells, and even a partial heart transplant. These innovations have been performed against a backdrop of an expansive and impressive list of tools designed to uncover the biology of complex systems that include a wide array of fluorescent transgenic fish that label specific cell types and mutant lines that are transparent, immune-deficient. Allogeneic transplants can also be accomplished using immune suppressed and syngeneic fish. Each of these innovations within the zebrafish community would provide several helpful tools that could be applied to transplant research. SUMMARY We highlight some examples of existing tools and assays developed in the zebrafish community that could be leveraged to overcome barriers in organ transplantation, including ischemia-reperfusion, short preservation durations, regeneration of marginal grafts, and acute and chronic rejection.
Collapse
|
18
|
Ferrero G, Gomez E, Lyer S, Rovira M, Miserocchi M, Langenau DM, Bertrand JY, Wittamer V. The macrophage-expressed gene (mpeg) 1 identifies a subpopulation of B cells in the adult zebrafish. J Leukoc Biol 2020; 107:431-443. [PMID: 31909502 PMCID: PMC7064944 DOI: 10.1002/jlb.1a1119-223r] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 11/07/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022] Open
Abstract
The mononuclear phagocytic system consists of many cells, in particular macrophages, scattered throughout the body. However, there is increasing evidence for the heterogeneity of tissue-resident macrophages, leading to a pressing need for new tools to discriminate mononuclear phagocytic system subsets from other hematopoietic lineages. Macrophage-expressed gene (Mpeg)1.1 is an evolutionary conserved gene encoding perforin-2, a pore-forming protein associated with host defense against pathogens. Zebrafish mpeg1.1:GFP and mpeg1.1:mCherry reporters were originally established to specifically label macrophages. Since then more than 100 peer-reviewed publications have made use of mpeg1.1-driven transgenics for in vivo studies, providing new insights into key aspects of macrophage ontogeny, activation, and function. Whereas the macrophage-specific expression pattern of the mpeg1.1 promoter has been firmly established in the zebrafish embryo, it is currently not known whether this specificity is maintained through adulthood. Here we report direct evidence that beside macrophages, a subpopulation of B-lymphocytes is marked by mpeg1.1 reporters in most adult zebrafish organs. These mpeg1.1+ lymphoid cells endogenously express mpeg1.1 and can be separated from mpeg1.1+ macrophages by virtue of their light-scatter characteristics using FACS. Remarkably, our analyses also revealed that B-lymphocytes, rather than mononuclear phagocytes, constitute the main mpeg1.1-positive population in irf8null myeloid-defective mutants, which were previously reported to recover tissue-resident macrophages in adulthood. One notable exception is skin macrophages, whose development and maintenance appear to be independent from irf8, similar to mammals. Collectively, our findings demonstrate that irf8 functions in myelopoiesis are evolutionary conserved and highlight the need for alternative macrophage-specific markers to study the mononuclear phagocytic system in adult zebrafish.
Collapse
Affiliation(s)
- Giuliano Ferrero
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium.,ULB Institute of Neuroscience (UNI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Etienne Gomez
- Department of Pathology and Immunology, University of Geneva, School of Medicine, Geneva, Switzerland
| | - Sowmya Lyer
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital Research Institute, Boston, Massachusetts, USA
| | - Mireia Rovira
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium.,ULB Institute of Neuroscience (UNI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Magali Miserocchi
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium.,ULB Institute of Neuroscience (UNI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - David M Langenau
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital Research Institute, Boston, Massachusetts, USA
| | - Julien Y Bertrand
- Department of Pathology and Immunology, University of Geneva, School of Medicine, Geneva, Switzerland
| | - Valérie Wittamer
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium.,ULB Institute of Neuroscience (UNI), Université Libre de Bruxelles (ULB), Brussels, Belgium.,WELBIO, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
19
|
Daniel MG, Rapp K, Schaniel C, Moore KA. Induction of developmental hematopoiesis mediated by transcription factors and the hematopoietic microenvironment. Ann N Y Acad Sci 2019; 1466:59-72. [PMID: 31621095 DOI: 10.1111/nyas.14246] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/30/2019] [Accepted: 09/13/2019] [Indexed: 12/11/2022]
Abstract
The induction of hematopoiesis in various cell types via transcription factor (TF) reprogramming has been demonstrated by several strategies. The eventual goal of these approaches is to generate a product for unmet needs in hematopoietic cell transplantation therapies. The most successful strategies hew closely to clues provided from developmental hematopoiesis in terms of factor expression and environmental cues. In this review, we aim to summarize the TFs that play important roles in developmental hematopoiesis primarily and to also touch on adult hematopoiesis. Several aspects of cellular and molecular biology coalesce in this process, with TFs and surrounding cellular signals playing a major role in the overall development of the hematopoietic lineage. We attempt to put these elements into the context of reprogramming and highlight their roles.
Collapse
Affiliation(s)
- Michael G Daniel
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York City, New York.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York City, New York.,The Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Katrina Rapp
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York City, New York.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Christoph Schaniel
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York City, New York.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York City, New York.,Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York.,Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Kateri A Moore
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York City, New York.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York City, New York
| |
Collapse
|
20
|
Vega-López A, Pagadala NS, López-Tapia BP, Madera-Sandoval RL, Rosales-Cruz E, Nájera-Martínez M, Reyes-Maldonado E. Is related the hematopoietic stem cells differentiation in the Nile tilapia with GABA exposure? FISH & SHELLFISH IMMUNOLOGY 2019; 93:801-814. [PMID: 31419534 DOI: 10.1016/j.fsi.2019.08.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/07/2019] [Accepted: 08/12/2019] [Indexed: 06/10/2023]
Abstract
The signaling mediated by small non-proteinogenic molecules, which probably have the capacity to serve as a bridge amongst complex systems is one of the most exiting challenges for the study. In the current report, stem cells differentiation of the immune system in Nile tilapia treated with sub-basal doses of GABA evaluated as c-kit+ and Sca-1+ cells disappearance on pronephros, thymus, spleen and peripheral blood mononuclear cells by flow cytometry was assessed. Explanation of biological response was performed by molecular docking approach and multiparametric analysis. Stem cell differentiation depends on a delicate balance of negative and positive interactions of this neurotransmitter with receptors and transcription factors involved in this process. This in turn depends on the type of interaction with hematopoietic niche to differentiate into primordial, early or late hematopoiesis as well as from the dose delivery. In fish treated with the low doses of GABA (0.1% over basal value) primordial hematopoiesis is regulated by interaction of glutamate (Glu) with the Ly-6 antigen. Early hematopoiesis was influenced by the bond of GABA near or adjacent to turns of FLTR3-Ig-IV domain. During late hematopoiesis, negative regulation by structural modifications on PU.1/IRF-4 complex, IL-7Rα and GM-CSFR mainly prevails. Results of molecular docking were in agreement with the percentages of the main blood cells lineages estimated in pronephros by flow cytometry. Current study provides the first evidences about the role of inhibitory and excitatory neurotransmitters such as GABA and Glu, respectively with the most transcriptional factors and receptors involved on hematopoiesis in adult Nile tilapia.
Collapse
Affiliation(s)
- Armando Vega-López
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Laboratorio de Toxicología Ambiental, Av. Wilfrido Massieu S/n, Unidad Profesional Zacatenco, México, CP 07738, Mexico.
| | | | - Brenda P López-Tapia
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Laboratorio de Toxicología Ambiental, Av. Wilfrido Massieu S/n, Unidad Profesional Zacatenco, México, CP 07738, Mexico
| | - Ruth L Madera-Sandoval
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Laboratorio de Toxicología Ambiental, Av. Wilfrido Massieu S/n, Unidad Profesional Zacatenco, México, CP 07738, Mexico
| | - Erika Rosales-Cruz
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Laboratorio de Citología, Carpio y Plan de Ayala S/n, Casco de Santo Tomás, México, CP 11340, Mexico
| | - Minerva Nájera-Martínez
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Laboratorio de Toxicología Ambiental, Av. Wilfrido Massieu S/n, Unidad Profesional Zacatenco, México, CP 07738, Mexico
| | - Elba Reyes-Maldonado
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Laboratorio de Citología, Carpio y Plan de Ayala S/n, Casco de Santo Tomás, México, CP 11340, Mexico
| |
Collapse
|
21
|
Rho SS, Kobayashi I, Oguri-Nakamura E, Ando K, Fujiwara M, Kamimura N, Hirata H, Iida A, Iwai Y, Mochizuki N, Fukuhara S. Rap1b Promotes Notch-Signal-Mediated Hematopoietic Stem Cell Development by Enhancing Integrin-Mediated Cell Adhesion. Dev Cell 2019; 49:681-696.e6. [DOI: 10.1016/j.devcel.2019.03.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 02/16/2019] [Accepted: 03/22/2019] [Indexed: 01/09/2023]
|
22
|
Grainger S, Nguyen N, Richter J, Setayesh J, Lonquich B, Oon CH, Wozniak JM, Barahona R, Kamei CN, Houston J, Carrillo-Terrazas M, Drummond IA, Gonzalez D, Willert K, Traver D. EGFR is required for Wnt9a-Fzd9b signalling specificity in haematopoietic stem cells. Nat Cell Biol 2019; 21:721-730. [PMID: 31110287 PMCID: PMC6559346 DOI: 10.1038/s41556-019-0330-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 04/11/2019] [Indexed: 12/31/2022]
Abstract
Wnt signalling drives a plethora of processes in development, homeostasis, and disease; however, the role and mechanism of individual ligand/receptor (Wnt/Frizzled, Fzd) interactions in specific biological processes remain poorly understood. Wnt9a is specifically required for the amplification of blood progenitor cells during development. Using genetic studies in zebrafish and human embryonic stem cells, paired with in vitro cell biology and biochemistry, we have determined that Wnt9a signals specifically through Fzd9b to elicit β-catenin-dependent Wnt signalling that regulates haematopoietic stem and progenitor cell emergence. We demonstrate that the epidermal growth factor receptor (EGFR) is required as a co-factor for Wnt9a/Fzd9b signalling. EGFR-mediated phosphorylation of one tyrosine residue on the Fzd9b intracellular tail in response to Wnt9a promotes internalization of the Wnt9a/Fzd9b/LRP signalosome and subsequent signal transduction. These findings provide mechanistic insights for specific Wnt/Fzd signals, which will be crucial for specific therapeutic targeting and regenerative medicine.
Collapse
Affiliation(s)
- Stephanie Grainger
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Nicole Nguyen
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jenna Richter
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA.,Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Jordan Setayesh
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Brianna Lonquich
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Chet Huan Oon
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jacob M Wozniak
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA.,Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, La Jolla, CA, USA.,Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Rocio Barahona
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Caramai N Kamei
- Massachusetts General Hospital Nephrology Division, Charlestown, MA, USA
| | - Jack Houston
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA.,Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Marvic Carrillo-Terrazas
- Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, La Jolla, CA, USA.,Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Iain A Drummond
- Massachusetts General Hospital Nephrology Division, Charlestown, MA, USA.,Harvard Medical School, Department of Genetics, Boston, MA, USA
| | - David Gonzalez
- Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, La Jolla, CA, USA.,Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Karl Willert
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA.
| | - David Traver
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA. .,Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
23
|
Bone marrow sinusoidal endothelium as a facilitator/regulator of cell egress from the bone marrow. Crit Rev Oncol Hematol 2019; 137:43-56. [DOI: 10.1016/j.critrevonc.2019.01.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 01/12/2019] [Accepted: 01/29/2019] [Indexed: 02/06/2023] Open
|
24
|
Gacha-Garay MJ, Niño-Joya AF, Bolaños NI, Abenoza L, Quintero G, Ibarra H, Gonzalez JM, Akle V, Garavito-Aguilar ZV. Pilot Study of an Integrative New Tool for Studying Clinical Outcome Discrimination in Acute Leukemia. Front Oncol 2019; 9:245. [PMID: 31024847 PMCID: PMC6465644 DOI: 10.3389/fonc.2019.00245] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 03/18/2019] [Indexed: 01/04/2023] Open
Abstract
Acute leukemia is a heterogeneous set of diseases affecting children and adults. Current prognostic factors are not accurate predictors of the clinical outcome of adult patients and the stratification of risk groups remains insufficient. For that reason, this study proposes a multifactorial analysis which integrates clinical parameters, ex vivo tumor characterization and behavioral in vivo analysis in zebrafish. This model represents a new approach to understand leukemic primary cells behavior and features associated with aggressiveness and metastatic potential. Xenotransplantation of primary samples from patients newly diagnosed with acute leukemia in zebrafish embryos at 48 hpf was used to asses survival rate, dissemination pattern, and metastatic potential. Seven samples from young adults classified in adverse, favorable or intermediate risk group were characterized. Tumor heterogeneity defined by Leukemic stem cell (LSC) proportion, was performed by metabolic and cell membrane biomarkers characterization. Thus, our work combines all these parameters with a robust quantification strategy that provides important information about leukemia biology, their relationship with specific niches and the existent inter and intra-tumor heterogeneity in acute leukemia. In regard to prognostic factors, leukemic stem cell proportion and Patient-derived xenografts (PDX) migration into zebrafish were the variables with highest weights for the prediction analysis. Higher ALDH activity, less differentiated cells and a broader and random migration pattern are related with worse clinical outcome after induction chemotherapy. This model also recapitulates multiple aspects of human acute leukemia and therefore is a promising tool to be employed not only for preclinical studies but also supposes a new tool with a higher resolution compared to traditional methods for an accurate stratification of patients into worse or favorable clinical outcome.
Collapse
Affiliation(s)
- María José Gacha-Garay
- Laboratory of Developmental Biology, Department of Biological Sciences, Universidad de los Andes, Bogotá, Colombia
| | - Andrés Felipe Niño-Joya
- Laboratory of Developmental Biology, Department of Biological Sciences, Universidad de los Andes, Bogotá, Colombia
| | - Natalia I Bolaños
- Biomedical Sciences Group, School of Medicine, Universidad de los Andes, Bogotá, Colombia
| | - Lina Abenoza
- Department of Oncology, Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | | | - Humberto Ibarra
- Microscopy Core, Vice-Presidency of Research, Universidad de los Andes, Bogotá, Colombia
| | - John M Gonzalez
- Biomedical Sciences Group, School of Medicine, Universidad de los Andes, Bogotá, Colombia
| | - Verónica Akle
- Laboratory of Neuroscience and Circadian Rhythms, School of Medicine, Universidad de los Andes, Bogotá, Colombia
| | - Zayra V Garavito-Aguilar
- Laboratory of Developmental Biology, Department of Biological Sciences, Universidad de los Andes, Bogotá, Colombia
| |
Collapse
|
25
|
STIL: a multi-function protein required for dopaminergic neural proliferation, protection, and regeneration. Cell Death Discov 2019; 5:90. [PMID: 31044090 PMCID: PMC6484007 DOI: 10.1038/s41420-019-0172-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 03/18/2019] [Indexed: 01/10/2023] Open
Abstract
Degeneration of dopaminergic (DA) neurons in the brain is the major cause for Parkinson’s disease (PD). While genetic loci and cellular pathways involved in DA neuron proliferation have been well documented, the genetic and molecular and cellular basis of DA cell survival remains to be elucidated. Recently, studies aimed to uncover the mechanisms of DA neural protection and regeneration have been reported. One of the most recent discoveries, i.e., multi-function of human oncogene SCL/TAL interrupting locus (Stil) in DA cell proliferation, neural protection, and regeneration, created a new field for studying DA cells and possible treatment of PD. In DA neurons, Stil functions through the Sonic hedgehog (Shh) pathway by releasing the inhibition of SUFU to GLI1, and thereby enhances Shh-target gene transcription required for neural proliferation, protection, and regeneration. In this review article, we will highlight some of the new findings from researches relate to Stil in DA cells using zebrafish models and cultured mammalian PC12 cells. The findings may provide the proof-of-concept for the development of Stil as a tool for diagnosis and/or treatment of human diseases, particularly those caused by DA neural degeneration.
Collapse
|
26
|
Making HSCs in vitro: don't forget the hemogenic endothelium. Blood 2018; 132:1372-1378. [PMID: 30089629 DOI: 10.1182/blood-2018-04-784140] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/04/2018] [Indexed: 12/29/2022] Open
Abstract
Generating a hematopoietic stem cell (HSC) in vitro from nonhematopoietic tissue has been a goal of experimental hematologists for decades. Until recently, no in vitro-derived cell has closely demonstrated the full lineage potential and self-renewal capacity of a true HSC. Studies revealing stem cell ontogeny from embryonic mesoderm to hemogenic endothelium to HSC provided the key to inducing HSC-like cells in vitro from a variety of cell types. Here we review the path to this discovery and discuss the future of autologous transplantation with in vitro-derived HSCs as a therapeutic modality.
Collapse
|